Next Article in Journal
Abalone Collagen Extracts Potentiate Stem Cell Properties of Human Epidermal Keratinocytes
Next Article in Special Issue
Whole Genome Sequencing of the Giant Grouper (Epinephelus lanceolatus) and High-Throughput Screening of Putative Antimicrobial Peptide Genes
Previous Article in Journal
Pinnatoxins’ Deleterious Effects on Cholinergic Networks: From Experimental Models to Human Health
Previous Article in Special Issue
Whole Genome Sequencing of the Blue Tilapia (Oreochromis aureus) Provides a Valuable Genetic Resource for Biomedical Research on Tilapias
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Genome Sequencing of the Japanese Eel (Anguilla japonica) for Comparative Genomic Studies on tbx4 and a tbx4 Gene Cluster in Teleost Fishes

1
BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China
2
Shenzhen Key Lab of Marine Genomics, Guangdong Provincial Key Lab of Molecular Breeding in Marine Economic Animals, BGI Academy of Marine Sciences, BGI Marine, BGI, Shenzhen 518083, China
3
BGI Zhenjiang Institute of Hydrobiology, Zhenjiang 212000, China
4
Guangdong Provincial Key Laboratory for Aquatic Economic Animals, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this project.
Mar. Drugs 2019, 17(7), 426; https://doi.org/10.3390/md17070426
Submission received: 17 June 2019 / Revised: 17 July 2019 / Accepted: 18 July 2019 / Published: 20 July 2019
(This article belongs to the Special Issue Genetics of Marine Organisms Associated with Human Health)

Abstract

:
Limbs originated from paired fish fins are an important innovation in Gnathostomata. Many studies have focused on limb development-related genes, of which the T-box transcription factor 4 gene (tbx4) has been considered as one of the most essential factors in the regulation of the hindlimb development. We previously confirmed pelvic fin loss in tbx4-knockout zebrafish. Here, we report a high-quality genome assembly of the Japanese eel (Anguilla japonica), which is an economically important fish without pelvic fins. The assembled genome is 1.13 Gb in size, with a scaffold N50 of 1.03 Mb. In addition, we collected 24 tbx4 sequences from 22 teleost fishes to explore the correlation between tbx4 and pelvic fin evolution. However, we observed complete exon structures of tbx4 in several pelvic-fin-loss species such as Ocean sunfish (Mola mola) and ricefield eel (Monopterus albus). More interestingly, an inversion of a special tbx4 gene cluster (brip1-tbx4-tbx2b- bcas3) occurred twice independently, which coincides with the presence of fin spines. A nonsynonymous mutation (M82L) was identified in the nuclear localization sequence (NLS) of the Japanese eel tbx4. We also examined variation and loss of hindlimb enhancer B (HLEB), which may account for pelvic fin loss in Tetraodontidae and Diodontidae. In summary, we generated a genome assembly of the Japanese eel, which provides a valuable genomic resource to study the evolution of fish tbx4 and helps elucidate the mechanism of pelvic fin loss in teleost fishes. Our comparative genomic studies, revealed for the first time a potential correlation between the tbx4 gene cluster and the evolutionary development of toxic fin spines. Because fin spines in teleosts are usually venoms, this tbx4 gene cluster may facilitate the genetic engineering of toxin-related marine drugs.

1. Introduction

The Japanese eel, Anguilla japonica, is a world-famous teleost fish due to its unique migration pattern and economic importance. Without any effective breeding technology, aquaculture in Asian countries has to depend on catching wild glass eels each year. To provide genetic resources for biological and practical studies on this teleost, we initiated a Japanese eel genome project in China.
The emergence of paired appendages has improved the movability and defensive capability of ancient vertebrates. In fishes, pectoral fins first appeared in extinct jawless fishes, whereas pelvic fins initially developed in the most primitive extinct jawed fishes—placoderms—which existed ~525 million years ago (Mya) in the middle Cambrian [1,2,3,4,5]. Tetrapods evolved from a fish-like ancestor; subsequently, paired fins evolved into limbs to adapt to terrestrial environments. Hence, it was a common thought that tetrapod forelimbs and hindlimbs are homologous to fish pectoral and pelvic fins, respectively. In recent years, the evolution of paired appendages has drawn considerable attention. Similar to the Japanese eel, the tiger tail seahorse (Hippocampus comes) exhibits a pelvic- fin-loss phenotype, which may be due to the loss of the T-box transcription factor 4 (tbx4), as we have confirmed pelvic fin loss in tbx4-knockout zebrafish [6].
T-box genes encode a family of transcription factors that are related to the metazoan development [7]. Their protein sequences possess a highly conserved DNA-binding domain (i.e., T-box domain). Within the T-box family, tbx2/3/4/5 subfamilies have been extensively studied because of their important roles in development of vertebrate appendages, heart and eyes [8]. At least 600 Mya, tbx2/3 and tbx4/5 diverged via tandem duplication, which maintained a tight linkage in most species; subsequently, either of these duplicated into two paralogous genes due to a whole genome duplication event [8]. Tbx4 has been a principal effective gene for the development of pelvic fins or hindlimbs in vertebrates, as knocking out tbx4 in zebrafish disrupts pelvic fin formation [6]. To date, numerous studies on tbx4 have mainly focused on mammals, especially on humans, whereas those involving fishes are limited. With over 38,000 extant species, teleost fishes comprise the largest group of living vertebrates [9] for in-depth investigations on the evolution of the tbx4 gene.
The development of high-throughput sequencing technologies has facilitated sequencing of the genomes of over 60 teleost species [10]. These data provide a good opportunity for us to perform a comparative genomic study on tbx4 isotypes and to identify variations in tbx4 sequences and gene structures across teleost fishes. In this study, we generated a high-quality genome assembly of the Japanese eel and then performed phylogenetic and synteny analyses, as well as variation determination of 24 tbx4 sequences in 22 representative teleost species. Interestingly, for the first time, we determined an inversion of a special tbx4 gene cluster that is potentially correlated with the evolutionary development of teleost fin spines, which may facilitate the development of marine drugs.

2. Results

2.1. Summary of Genome Survey and De Novo Assembly

A total of 268.61 Gb of raw reads were generated by an Illumina HiSeq X-Ten platform (see more details in Section 4.2 and Table S1). After removal of low-quality reads, index and adapter sequences, and PCR duplicates using SOAPfilter v2.2 (BGI, Shenzhen, China) [11], we obtained 184.05 Gb of clean reads (Table S1) for subsequent assembly and annotation. Based on a 17-mer distribution (Figure 1), we estimated that the genome size of the Japanese eel is approximately 1.03 Gb (Table S2) [12].
We employed SOAPdenovo [11] to obtain a primary draft, which consisted of 1,227,464 contigs and 462,272 scaffolds, with a contig N50 of 2.00 kb and a scaffold N50 of 383.80 kb. Subsequently, GapCloser and SSPACE [13] were employed to fill the gaps and elongate the scaffolds. As a result, we assembled a 1.23-Gb genome with 608,352 contigs and 351,879 scaffolds (Table 1). To assess the completeness of this assembly, we employed actinopterygii_bod9 as reference for a BUSCO analysis [14,15], which demonstrated that the benchmarking value was fair up to 83.9% (Table 2).
After performing an additional series of filtering manually for those heterozygous redundant scaffolds (sequencing depth <40×; see more information in Section 4.2), we generated a final 1.13-GB genome assembly (Table 1) with 256,649 contigs and 41,687 scaffolds, and the contig N50 and scaffold N50 values reached 11.47 kb and 1.03 Mb, respectively.
In our assembly, repetitive sequences accounted for 22.94% of the entire genome. The detailed categories are summarized in Table S3. Finally, a total of 17,147 genes with an average of 7.6 exons were predicted (see more details in Table S4).

2.2. Conservation of the Vertebrate tbx4 Genes in Gene Structure

The examined various vertebrate species, including amphioxus (Branchiostoma floridae), a shark (Chiloscyllium punctatum), the zebrafish (Danio rerio), a frog (Xenopus tropicalis), a turtle (Chelonia mydas), a chicken (Gallus gallus) and humans (Homo sapiens), were chosen as representative of Cephalochordata, cartilaginous fishes, bony fishes, amphibians, reptiles, and mammals, respectively (see more details in Section 4.3 and Figure 2). From amphioxus to human, we observed that the tbx4 genes in all vertebrate species contain eight exons, and each of them have a similar length in various species (Figure 2). Although divergent at ~699 Mya [16], this gene seems to be highly conserved in terms of gene structure in vertebrates.

2.3. Conservation of the T-box Region

All T-box genes share a common T-box domain, which is composed of 180 to 190 amino acid (aa) residues [17]. Multiple sequence alignment of 24 tbx4 sequences from 22 representative species revealed that the T-box domains of the tbx4 genes are highly conserved in vertebrates (Figure 3). Usually, the central T-box domain is composed of exons 3 to 5, partial exon 2 and exon 6. Interestingly, a cave-restricted barbel fish (Sinocyclocheilus anshuiensis) [18] and anadromous Chinook salmon (Oncorhynchus tshawytscha) [19] both possess two copies of the tbx4 gene, most likely due to an extra whole genome duplication event.

2.4. The Tbx4 Gene of the Japanese Eel

The nuclear localization sequence (NLS) of tbx4 genes usually consists of 13 aa and lies in the conserved DNA-binding motif (T-box domain; Figure 3). Comparative analysis revealed a nonsynonymous variant in the Japanese eel tbx4 (M82L; Figure 4), which may be related to pelvic fin loss [20]. To confirm this variant, PCR was performed (data not shown).
A previous research [21] showed that all substitutions except for K11 of tbx5 NLS could cause cytoplasmic localization of fusion proteins. Another study [20] identified two nonsynonymous mutations in the NLS of zebrafish tbx4, which impaired nuclear localization of the protein and thereby disrupted pelvic fin development.
The entire protein sequence of the Japanese eel tbx4 gene is presented in Figure 5. Its alignment with zebrafish TBX4 revealed a high conservation between the two fish species. Localization of the same NLS sequence in both fishes is marked in a red box for a detailed comparison (upper panel in Figure 5).

2.5. Phylogenetic Analysis and Synteny Comparison

We constructed a phylogenetic tree of 24 tbx4 protein sequences using spotted gar (Lepisosteus oculatus) as the outgroup (Figure 6). Eels (Elopomorpha) showed the earliest branching in teleosts, whereas the other lineages diverged later. Because the length of each branch represents the evolution rate of the examined gene, we speculate that the Japanese eel has a high evolution rate in tbx4 (see more details in the left tree of Figure 6).

2.6. The Brip1-tbx4-tbx2b-bcas3 Cluster

We observed that a cluster composed of four genes (brip1, tbx4, tbx2b and bcas3) maintains the same arrangement in teleosts (Figure 6 and Figure 7), despite chromosomal rearrangements occurring since the divergence of bony vertebrates approximately 465 Mya [22,23,24,25]. Previous studies involving mammals demonstrate a putative limb enhancer at the interior of bcas3 [26]. The hindlimb enhancer A (HLEA) and HLEB are located at the interval of tbx2-tbx4 and tbx4-brip1, respectively [27]. Tbx2 encodes a transcriptional repressor that is related to digit development [28]. Bcas3 has shown to be overexpressed in breast cancer cells [29], and brip1 encodes a protein that interacts with bcas3 [30]. However, current understanding of brip1 and bcas3 in fishes is limited.
Interestingly, an inversion of the brip1-tbx4-tbx2b-bcas3 cluster occurred twice independently in teleosts. One inversion happened in Acanthopterygii, and the another appeared in a subclade of Otophysa—which includes Characiformes, Gymnotiformes and Siluriformes—but not Cypriniformes (see more details in the two highlighted black boxes of Figure 6). Lineages with one of the inversions are armed with fin spines. In fact, Acanthopterygii is named for the representative sharp and bony rays in their dorsal fins, anal fins or pelvic fins. Members of Siluriformes are armed with spines in the anal, dorsal, caudal, adipose, and paired fins [31,32,33]. Additionally, fin spines are characterized in some members of the Characiformes [34]. Gymnotiformes, although divergent from Siluriformes [35], is an outlier due to its absence of pelvic fins and dorsal fins.
Chinese yellow catfish (Pelteobagrus fulvidraco) secretes venom through its fin spines, which has been proposed by us to be essential for the development of marine drugs [36]. Eeltail catfish (Siluriformes), scorpionfish and stonefish (Scorpaeniformes) also have venomous fin spines that can severely injure other animals [37]. As we determined in the present study, an inversion of the brip1-tbx4-tbx2b-bcas3 cluster occurred in these Acanthopterygii fishes (Figure 7), which are in line with the existence of fin spines.

2.7. HLEB

HLEB is a highly conserved enhancer of the tbx4 genes from mammals to cartilaginous fishes, which play an important role in hindlimb development [27]. Here, we compared 11 HLEB sequences across Acanthopterygii fishes against three-spined stickleback (Gasterosteus aculeatu). Previous studies suggested that Tetraodontiformes had undergone reductions or increases in pelvic complexes [38]. We found that the HLEB of Ocean sunfish (Mola mola) was very similar to three-spined stickleback than other four related puffer fishes (Takifugu bimaculatus, T. obscurus, T. rubripes, and Tetraodon nigroviridi; see a VISTA plot [39,40] in Figure 8), which might be responsible for the loss of pelvic fins in Tetraodontidae. However, another Tetraodontiformes species, spot-fin porcupinefish (Diodon hystrix) as well as tiger tail seahorse may have lost the HLEB sequence (unpublished data).

3. Discussion

3.1. Various Genetic Mechanisms for Pelvic Fin Development

Since the emergence of two paired appendages, one or both of these were secondarily lost in many animal lineages and showed a corresponding high level of disparity. For example, eels (Anguilliformes), ricefield eel (Synbranchiformes), and electric eel (Electrophorus electricus; Gymnotiformes) have completely lost their pelvic fins completely; for puffer fishes and filefishes (Tetraodontiformes), however, there exists a great diversity in their pelvic fins ranging from acquired to various degrees of reduction [38]. It has been proposed that an altered hoxd9a expression may account for the loss of pelvic fins in Japanese puffer (Takifugu rubripes; Tetraodontiformes) [41]. Basal snakes (boa and python) retained a vestigial pelvic girdle and rudimentary hindlimbs, whereas advanced snakes (viper, rattlesnake, king cobra, and corn snake), representing the majority (>85%) of all extant snake species, completely lost all skeletal limb structures due to a 17-bp deletion in the zone of the polarizing activity (ZPA) regulatory sequence [42]. The ZPA has proven to be a limb-specific enhancer of the Sonic hedgehog (Shh) gene, which is indispensable for limb development [43,44,45,46,47,48,49,50]. In addition, another research demonstrated that the HLEB, a highly conserved putative pitx1 binding site, had lost the function for limb development in snakes [27].
Two nonsynonymous mutations within the tbx4 NLS (A78V, G79A) are enough to disrupt pelvic fin development in zebrafish [20]. Pitx1, a homeobox-containing transcription factor with importance in hindlimb identity and outgrowth [51,52,53], has been associated with pelvic fin variations in natural populations of three-spined stickleback (Gasterosteiformes) [54]. Pitx1–mediated pelvic reduction was also observed in ninespine stickleback (Pungitius pungitius; Gasterosteiformes), and even in distantly related species such as manatees [54]. Furthermore, some lizards and mammals have more or less lost their paired appendages, although the detailed mechanisms remain unclear.
Embryonic development of limbs or pelvic fins mainly undergoes three main steps, including positioning, initiation and outgrowth. It is a comprehensive process and involves several genes, such as tbx4, pitx1, hoxa13, hoxb9, hoxc9, hoxd9, hoxd10, hoxd13, wnt2b, wnt8c, wnt3a/3, shh, fgf10, and fgf8 [41,42,43,44,45,46,47,48,49,50,51,52,53,54,55]. At the first step of embryonic development of vertebrate paired appendages, homeobox (hox) genes expressed in the lateral plate mesoderm and somitic mesoderm specify the position of the limbs and interlimb region. Subsequently, tbx4 and tbx5 in the lateral plate mesoderm activate the expression of downstream wnt8c/fgf10 and wnt2b/fgf10 in hindlimbs and forelimbs, respectively. Then, Wnt/Fgf signaling feedbacks on tbx4 and tbx5 to maintain their expression. After that, fgf10 activates wnt3a/3/fgf8 signals in the limb ectoderm and induces the formation of apical ectodermal ridge shh expression in the posterior limb bud, which has been maintained by fibroblast growth factor (FGF) signaling in the apical ectodermal ridge and the FGF signaling feedback on shh to maintain its expression. In addition, tbx4 expression is regulated by pitx1 and to a lesser extent by pitx2 [53].
Here, we selected one of the most important factors for the involvement in hindlimb development—tbx4—to perform comparative genomic studies in teleost fishes. We provided new information about tbx4 genes, including gene structure, variation, synteny, enhancement sequence and phylogenetic status. We also revealed that the genetic backgrounds for pelvic fin loss in various species might be diverse. Monopterus albus has lost pelvic fins despite a complete gene structure for tbx4 and normal HLEB. The genetic mechanisms of pelvic fin loss in a given group of species, such as pufferfishes, may significantly vary. Previous studies have suggested that altered hoxd9a expression may result in pelvic loss in Japanese puffer [41]. According to our present results, however, variations in HLEB may be also involved in the regulation of pelvic phenotypes.

3.2. Potential Importance of the Tbx4 Gene Cluster for the Evolutionary Development of Toxic Fin Spines

Structural conservation often indicates stable function(s). We observed that the tbx4 gene cluster brip1-tbx4-tbx2b-bcas3 widely exists in teleost fishes. Previous studies have demonstrated that this cluster linkage may result from the shared regulatory domains required for coordinated expression [8]. The NLS of tbx4 plays a key role in protein nuclear transport, and its structure must be intact to play its essential role in the induction of the pelvic fin outgrowth [21]. For the Japanese eel, a nonsynonymous mutation was detected in the NLS of tbx4 (Figure 4), which is considered to be correlated with pelvic fin development.
Many fishes have developed fin spines for defense or hunting purposes. In our previous reports [36,56,57], we predicted several toxin genes from the venom glands of fin spines in Chinese yellow catfish using a combination of genomic, transcriptomic, and proteomic sequencing. The contribution of more toxins to future drugs seems to be more promising [57], and we will therefore sequence and analyze more fish species with fin spines. In Figure 6, we observed an inversion of the tbx4 gene cluster, which may be correlated with the development of toxic fin spines. Hence, we propose a deep investigation of the synthetic biological application of this cluster, which may benefit the development of novel marine drugs.

4. Materials and Methods

4.1. Sample Collection

A female Japanese eel was collected from a local aquaculture base of BGI Marine in Huizhou, Guangdong Province, China. Species identification with cloning of the COI sequence was performed immediately after collection of muscle samples. All experiments were conducted in accordance with the guidelines of the Animal Ethics Committee and were approved by the Institutional Review Board on Bioethics and Biosafety of BGI (No. FT1510).

4.2. Genome Sequencing, Assembling and Annotation

Genomic DNA (for the genome sequencing) and total RNA (for the transcriptome sequencing) were extracted from the muscle samples as previously described [36]. For whole-genome sequencing, we constructed seven paired-end sequencing libraries, including three short-insert (270, 500, and 800 bp) and four long-insert (2, 5, 10, and 20 kb). Finally, paired-end sequencing was performed on an Illumina HiSeq X-ten platform (San Diego, CA, USA).
After genome sequencing, we employed the SOAPdenovo (version 2.04) to assemble the draft genome with the parameter “-k 27 –M 1”. Subsequently, krskgf, Gapcloser1.12, and Gapcloser1.10 were used to fill gaps in the primary assembly successively. After that, SSPACE was used to elongate the scaffolds produced by Gapcloser1.10. These steps were described in detail in our previous studies [12,18,36]. We also manually filtered those redundant scaffolds caused by the high heterozygosity. Because a heterozygous scaffold contains remarkably lower sequencing depth than a normal scaffold, we could remove these scaffolds with the sequencing depth <40× (~1/4 of the average sequencing depth). Our genome assembly of the Japanese eel has been deposited in the NCBI under the project ID PRJNA533944 with an accession code of VDMF00000000.
We used RepeatModeller v1.08 (Institute for System Biology, Seattle, CA, USA) along with LTR-FINDER v1.06 [58] for de novo repeat sequence prediction, and Tandem Repeat Finder (Trf), RepeatMasker v4.06 [59] along with RepeatProteinMask v4.06 for homology prediction by aligning to the RepBase v21.01 [60]. Finally, we integrated the results produced by the above-mentioned two prediction methods.
For whole gene set annotation, we masked the repetitive elements of the assembling genome and then adopted three different strategies, namely, ab initio annotation, homologous annotation, and transcriptome-based annotation, as previously reported [36]. We used AUGUSTUS v2.5 [61] and GENSCAN v1.0 [62] for ab initio prediction. For homologous annotation, we downloaded the protein sequences (release version 89) of eight vertebrate species from the ensemble, including zebrafish, Atlantic cod (Gadus morhua), spotted gar (Lepisosteus oculatus), Nile tilapia (Oreochromis niloticus), medaka (Oryzias latipes), Japanese puffer, spotted green pufferfish (Tetraodon nigroviridis), and sea lamprey (Petromyzon marinus) to search for the best-hit alignments in the Japanese eel genome by TblastN program [63]. Subsequently, GeneWise v2.2.0 [64] was used to identify the gene structure of alignment produced by TblastN. For transcriptome-based prediction, we used Tophat v2.1.1 [65] and Cufflinks v2.2.1 (University of Maryland, College Park, MD, USA) to predict the gene set with the transcriptomic data of liver and gill sequenced by an Illumina Hiseq2500 platform. Finally, EVidenceModeler [66] was employed to integrate the consensus results of the three prediction methods. The predicted gene set was used to identify the functional motifs and domains by mapping to five public functional databases, including KEGG [67], NCBI-Nr, Swiss-Prot, TrEMBL [68], and Interpro [69] using BLAST.

4.3. Collection of the Genome Sequences

We downloaded 26 fish genomes and 27 protein sequences of T-box family as well as seven adjacent proteins of tbx4 from NCBI (see more details in Table 3). The Chinese clearhead icefish (Protosalanx hyalocranius) and Northern snakehead (Channa argus) genomes were downloaded from GigaDB [70,71]. The genomes of spot-fin porcupinefish (Diodon hystrix) and river fugu (Takifugu obscurus) were obtained from our laboratory (unpublished data).

4.4. Collection of the Tbx4 Sequences

We picked out seven genes with adjacent locations to the tbx4 gene(s) in most of the downloaded fish genomes. To avoid mapping onto other paralogous genes in the T-box family and to ensure the accuracy to find tbx4 homolog, we merged the 27 T-box protein sequences and seven adjacent genes (see Table 3) as a whole reference to build an alignment index. Subsequently, we aligned the reference to all the examined genomes using TBLASTN to acquire tbx4 homolog sequence(s). We then selected those with alignments to at least three adjacent genes on the same chromosome or scaffold (with tbx4 gene). Subsequently, we used Exonerate [70] and GeneWise v2.2.0 [62] to calculate the amino acid sequence of each tbx4 gene, and we corrected errors manually according to the zebrafish TBX4 protein sequence. Finally, we obtained 24 TBX4 protein sequences from 22 representative teleost fishes.
Due to the limitations of sequencing and assembly, the Japanese eel tbx4 sequence of our assembly was truncated. We filled the gap and completed the synteny information using a chromosome-level assembly version of the Japanese eel genome, GCA_003597225.1, from NCBI [72].

4.5. Sequence Alignment, Phylogenetic Analysis and Identification of Conserved Synteny

We extracted the T-box domain of these tbx4 proteins and performed a multiple alignment by Muscle [73,74]. After that, we colorized the alignment results using TEXshade [75]. These collected TBX4 protein sequences were then employed to predict their best nucleotide substitution model under the Akaike Information Criterion (AIC) [76], which was implemented in prottest-3.4.2 [77]. We also performed multiple alignments of these collected tbx4 protein sequences by MEGA-7.0 [78] and constructed phylogenetic topologies with 1,000 replicates to evaluate branch supports with the maximum likelihood (ML) method by phyML-3.1 [79,80]. To assess the collinearity conservation and assure confidence of the collected tbx4 sequences, we detected arrangement orders of the seven adjacent genes of tbx4 in each species.

4.6. HLEB Analysis

We obtained the 873-bp HLEB sequence of the three-spined stickleback by examining the reported primers on the genome (GCA_000180675.1). Then, we mapped this HLEB sequence onto the examined genomes for acquisition of corresponding homologous sequences by using LAGAN [81]. The alignment results were visualized by VISTA plot [39,40].

5. Conclusions

We sequenced and assembled a 1.13-Gb genome of the Japanese eel for a comparative genomic study on the tbx4 gene cluster. The tbx4 gene apparently harbors a nonsynonymous mutation in an important site of the NLS, which was considered to be correlated with the pelvic fin development. Interestingly, its adjacent brip1 gene was also lost. We investigated 24 tbx4 sequences from 22 teleost lineages and detected an inversion that occurred twice independently in teleost fishes, which coincides with the presence of fin spines. Additionally, the change or loss of HLEB may be responsible for the disappearance of pelvic fins in some Tetraodontiformes species. This is the first report describing the potential correlation of the inversed tbx4 gene cluster with the development of fin spines, which may benefit the development of novel marine drugs.

Supplementary Materials

The following materials are available online at https://www.mdpi.com/1660-3397/17/7/426/s1. Table S1: Summary of the genome sequencing data for the Japanese eel. Table S2: Genome size estimation based on the 17-mer frequencies. Table S3: Statistics of the repeat sequences in the genome assembly of the Japanese eel. Table S4: Statistics of the gene annotation for the assembled genome of the Japanese eel.

Author Contributions

Q.S., X.L. and C.B. conceived and designed the experiments. S.Y., R.G., and J.X. collected samples. L.Y., Z.W. and X.Z. performed the experiments. W.C. analyzed data and prepared the manuscript. Q.S. revised the manuscript. X.Y., C.B., J.L., and Y.L. participated in discussions and data analysis. S.Y., X.L., and Q.S. provided financial support.

Acknowledgments

The work was supported by Shenzhen Special Program for Development of Emerging Strategic Industries (No. JSGG20170412153411369), and Shenzhen Dapeng Special Program for Industrial Development (Nos. KY20190108, KY20180205, and KY20160307).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

appbp2amyloid protein-binding protein 2
brip1BRCA1 interacting protein C-terminal helicase 1
bcas3breast carcinoma amplified sequence 3
eomesaeomesodermin homolog A
eomesbeomesodermin homolog B
fgf familyfibroblast growth factor family
HLEAhindlimb enhancer A
HLEBhindlimb enhancer B
hox geneshomeobox genes
lhx1aLIM homeobox transcription factor 1, alpha
mgamax gene-associated protein
mgalmax gene-associated protein-like
NLSnuclear localization sequence
pitx1paired-like homeodomain 1
ppm1dprotein phosphatase 1D
shhSonic hedgehog
tabrachyury homolog A
tbbrachyury homolog B
tbr1aT-box brain protein 1A
tbr1bT-box brain protein 1B
tbx genesT-box transcription factors
tbx2T-Box transcription factor 2
tbx2bT-Box transcription factor 2B
tbx3T-Box transcription factor 3
tbx4T-Box transcription factor 4
tbx5T-Box transcription factor 5
usp32ubiquitin specific peptidase 32
vegtvegetal T-box transcription factor
wnt familywingless-type MMTV integration site family

References

  1. Sansom, R.S.; Gabbott, S.E.; Purnell, M.A. Unusual anal fin in a Devonian jawless vertebrate reveals complex origins of paired appendages. Biol. Lett. 2013, 9, 5. [Google Scholar] [CrossRef] [PubMed]
  2. Forey, P.L. Agnathans recent and fossil, and the origin of jawed vertebrates. Rev. Fish Biol. Fisher. 1995, 5, 267–303. [Google Scholar] [CrossRef]
  3. Zhu, M.; Yu, X.B.; Choo, B.; Wang, J.Q.; Jia, L.T. An antiarch placoderm shows that pelvic girdles arose at the root of jawed vertebrates. Biol. Lett. 2012, 8, 453–456. [Google Scholar] [CrossRef] [PubMed]
  4. Don, E.K.; Currie, P.D.; Cole, N.J. The evolutionary history of the development of the pelvic fin/hindlimb. J. Anat. 2013, 222, 114–133. [Google Scholar] [CrossRef]
  5. Blair, J.E.; Hedges, S.B. Molecular phylogeny and divergence times of deuterostome animals. Mol. Biol. Evol. 2005, 22, 2275–2284. [Google Scholar] [CrossRef]
  6. Lin, Q.; Fan, S.; Zhang, Y.; Xu, M.; Zhang, H.; Yang, Y.; Lee, A.P.; Woltering, J.M.; Ravi, V.; Gunter, H.M.; et al. The seahorse genome and the evolution of its specialized morphology. Nature 2016, 540, 395–399. [Google Scholar] [CrossRef] [Green Version]
  7. Papaioannou, V.E. T-box genes in development: From hydra to humans. Int. Rev. Cytol. 2001, 207, 1–70. [Google Scholar]
  8. Horton, A.C.; Mahadevan, N.R.; Minguillon, C.; Osoegawa, K.; Rokhsar, D.S.; Ruvinsky, I.; de Jong, P.J.; Logan, M.P.; Gibson-Brown, J.J. Conservation of linkage and evolution of developmental function within the Tbx2/3/4/5 subfamily of T-box genes: Implications for the origin of vertebrate limbs. Dev. Genes Evol. 2008, 218, 613–628. [Google Scholar] [CrossRef]
  9. Jackson, L.M.; Fernando, P.C.; Hanscom, J.S.; Balhoff, J.P.; Mabee, P.M. Automated integration of trees and traits: A case study using paired fin loss across teleost fishes. Syst. Biol. 2018, 67, 559–575. [Google Scholar] [CrossRef]
  10. Bian, C.; Huang, Y.; Li, J.; You, X.; Yi, Y.; Ge, W.; Shi, Q. Divergence, evolution and adaptation in ray-fined fish genomes. Sci. China Life Sci. 2019, 62. [Google Scholar] [CrossRef]
  11. Luo, R.; Liu, B.; Xie, Y.; Li, Z.; Huang, W.; Yuan, J.; He, G.; Chen, Y.; Pan, Q.; Liu, Y.; et al. SOAPdenovo2: An empirically improved memory-efficient short-read de novo assembler. GigaScience 2012, 1, 18. [Google Scholar] [CrossRef] [PubMed]
  12. Song, L.; Bian, C.; Luo, Y.; Wang, L.; You, X.; Li, J.; Qiu, Y.; Ma, X.; Zhu, Z.; Ma, L.; et al. Draft genome of the Chinese mitten crab, Eriocheir sinensis. GigaScience 2016, 5, 5. [Google Scholar] [CrossRef] [PubMed]
  13. Boetzer, M.; Henkel, C.V.; Jansen, H.J.; Butler, D.; Pirovano, W. Scaffolding pre-assembled contigs using SSPACE. Bioinformatics 2011, 15, 578–579. [Google Scholar] [CrossRef]
  14. Simao, F.A.; Waterhouse, R.M.; Ioannidis, P.; Kriventseva, E.V.; Zdobnov, E.M. BUSCO: Assessing genome assembly and annotation completeness with single-copy orthologs. Bioinformatics 2015, 31, 3210–3212. [Google Scholar] [CrossRef] [PubMed]
  15. Zdobnov, E.M.; Tegenfeldt, F.; Kuznetsov, D.; Waterhouse, R.M.; Simão, F.A.; Ioannidis, P.; Seppey, M.; Loetscher, A.; Kriventseva, E.V. OrthoDB v9.1: Cataloging evolutionary and functional annotations for animal, fungal, plant, archaeal, bacterial and viral orthologs. Nucleic Acids Res. 2017, 45, 744–749. [Google Scholar] [CrossRef] [PubMed]
  16. Parfrey, L.W.; Lahr, D.J.; Knoll, A.H.; Katz, L.A. Estimating the timing of early eukaryotic diversification with multigene molecular clocks. Proc. Natl. Acad. Sci. USA 2011, 108, 13624–13629. [Google Scholar] [CrossRef] [Green Version]
  17. Tada, M.; Smith, J.C. T-targets: Clues to understanding the functions of T-box proteins. Dev. Growth Differ. 2001, 43, 1–11. [Google Scholar] [CrossRef] [PubMed]
  18. Yang, J.; Chen, X.; Bai, J.; Fang, D.; Qiu, Y.; Jiang, W.; Yuan, H.; Bian, C.; Lu, J.; He, S.; et al. The Sinocyclocheilus cavefish genome provides insights into cave adaptation. BMC Biol. 2016, 14, 1. [Google Scholar] [CrossRef]
  19. Christensen, K.A.; Leong, J.S.; Sakhrani, D.; Biagi, C.A.; Minkley, D.R.; Withler, R.E.; Rondeau, E.B.; Koop, B.F.; Devlin, R.H. Chinook salmon (Oncorhynchus tshawytscha) genome and transcriptome. PLoS ONE 2018, 13, e0195461. [Google Scholar] [CrossRef]
  20. Don, E.K.; de Jong-Curtain, T.A.; Doggett, K.; Hall, T.E.; Heng, B.; Badrock, A.P.; Winnick, C.; Nicholson, G.A.; Guillemin, G.J.; Currie, P.D.; et al. Genetic basis of hindlimb loss in a naturally occurring vertebrate model. Biol. Open 2016, 5, 359–366. [Google Scholar] [CrossRef] [Green Version]
  21. Collavoli, A.; Hatcher, C.J.; He, J.; Okin, D.; Deo, R.; Basson, C.T. TBX5 nuclear localization is mediated by dual cooperative intramolecular signals. J. Mol. Cell. Cardiol. 2003, 35, 1191–1195. [Google Scholar] [CrossRef]
  22. Betancur-R, R.; Broughton, R.E.; Wiley, E.O.; Carpenter, K.; López, J.A.; Li, C.; Holcroft, N.I.; Arcila, D.; Sanciangco, M.; Cureton, J.C., II; et al. The tree of life and a new classification of bony fishes. PLoS Curr. 2013, 5. [Google Scholar] [CrossRef] [PubMed]
  23. Dos Reis, M.; Thawornwattana, Y.; Angelis, K.; Telford, M.J.; Donoghue, P.C.; Yang, Z. Uncertainty in the timing of origin of animals and the limits of precision in molecular timescales. Curr. Biol. 2015, 25, 2939–2950. [Google Scholar] [CrossRef] [PubMed]
  24. Inoue, J.G.; Miya, M.; Lam, K.; Tay, B.H.; Danks, J.A.; Bell, J.; Walker, T.I.; Venkatesh, B. Evolutionary origin and phylogeny of the modern holocephalans (Chondrichthyes: Chimaeriformes): A mitogenomic perspective. Mol. Biol. Evol. 2010, 27, 2576–2586. [Google Scholar] [CrossRef] [PubMed]
  25. Betancur-R, R.; Orti, G.; Pyron, A.R. Fossil-based comparative analyses reveal ancient marine ancestry erased by extinction in ray-finned fishes. Ecol. Lett. 2015, 18, 441–450. [Google Scholar] [CrossRef] [PubMed]
  26. Infante, C.R.; Park, S.; Mihala, A.G.; Kingsley, D.M.; Menke, D.B. Pitx1 broadly associates with limb enhancers and is enriched on hindlimb cis-regulatory elements. Dev. Biol. 2013, 374, 234–244. [Google Scholar] [CrossRef] [PubMed]
  27. Menke, D.B.; Guenther, C.; Kingsley, D.M. Dual hindlimb control elements in the Tbx4 gene and region-specific control of bone size in vertebrate limbs. Development 2008, 135, 2543–2553. [Google Scholar] [CrossRef] [PubMed]
  28. Farin, H.F.; Lüdtke, T.H.; Schmidt, M.K.; Placzko, S.; Schuster-Gossler, K.; Petry, M.; Christoffels, V.M.; Kispert, A. Tbx2 terminates Shh/Fgf signaling in the developing mouse limb bud by direct repression of Gremlin1. PLoS Genet. 2013, 9, e1003467. [Google Scholar] [CrossRef] [PubMed]
  29. Bärlund, M.; Monni, O.; Weaver, J.D.; Kauraniemi, P.; Sauter, G.; Heiskanen, M.; Kallioniemi, O.P.; Kallioniemi, A. Cloning of BCAS3 (17q23) and BCAS4 (20q13) genes that undergo amplification, overexpression, and fusion in breast cancer. Genes Chromosomes Cancer 2003, 35, 311–317. [Google Scholar] [CrossRef] [PubMed]
  30. Yu, X.; Chini, C.C.; He, M.; Mer, G.; Chen, J. The BRCT domain is a phospho-protein binding domain. Science 2003, 302, 639–642. [Google Scholar] [CrossRef] [PubMed]
  31. Kubicek, K.M.; Britz, R.; Conway, K.W. Ontogeny of the catfish pectoral-fin spine (Teleostei: Siluriformes). J. Morphol. 2019, 280, 339–359. [Google Scholar] [CrossRef] [PubMed]
  32. Souza, C.S.; Costa-Silva, G.J.; Roxo, F.F.; Foresti, F.; Oliveira, C. Genetic and morphological analyses demonstrate that Schizolecis guntheri (Siluriformes: Loricariidae) is likely to be a species complex. Front. Genet. 2018, 9, 69. [Google Scholar] [CrossRef] [PubMed]
  33. Stewart, T.A.; Bonilla, M.M.; Ho, R.K.; Hale, M.E. Adipose fin development and its relation to the evolutionary origins of median fins. Sci. Rep. 2019, 9, 512. [Google Scholar] [CrossRef] [PubMed]
  34. Marinho, M.M.F.; Bastos, D.A.; Menezes, N.A. New species of miniature fish from Marajó Island, Pará, Brazil, with comments on its relationships (Characiformes: Characidae). Neotrop. Ichthyol. 2013, 11, 739–746. [Google Scholar] [CrossRef]
  35. Albert, J.; Crampton, W. Diversity and phylogeny of neotropical electric fishes (Gymnotiformes). In Electroreception; Bullock, T.H., Hopkins, C.D., Popper, A.N., Fay, R.R., Eds.; Springer: New York, NY, USA, 2005; pp. 360–409. [Google Scholar]
  36. Zhang, S.; Li, J.; Qin, Q.; Liu, W.; Bian, C.; Yi, Y.; Wang, M.; Zhong, L.; You, X.; Tang, S.; et al. Whole-genome sequencing of Chinese yellow catfish provides a valuable genetic resource for high-throughput identification of toxin genes. Toxins 2018, 10, 448. [Google Scholar] [CrossRef] [PubMed]
  37. Galloway, K.A.; Porter, M.E. Mechanical properties of the venomous spines of Pterois volitans and morphology among lionfish species. J. Exp. Biol. 2019, 222, jeb197905. [Google Scholar] [CrossRef] [PubMed]
  38. Yamanoue, Y.; Setiamarga, D.H.; Matsuura, K.J. Pelvic fins in teleosts: Structure, function and evolution. Fish Biol. 2010, 77, 1173–1208. [Google Scholar] [CrossRef]
  39. Mayor, C.; Brudno, M.; Schwartz, J.R.; Poliakov, A.; Rubin, E.M.; Frazer, K.A.; Pachter, L.S.; Dubchak, I. VISTA: Visualizing Global DNA Sequence Alignments of Arbitrary Length. Bioinformatics 2000, 16, 1046. [Google Scholar] [CrossRef]
  40. Frazer, K.A.; Pachter, L.; Poliakov, A.; Rubin, E.M.; Dubchak, I. VISTA: Computational tools for comparative genomics. Nucleic Acids Res. 2004, 32, 273–279. [Google Scholar] [CrossRef]
  41. Tanaka, M.; Hale, L.A.; Amores, A.; Yan, Y.L.; Cresko, W.A.; Suzuki, T.; Postlethwait, J.H. Developmental genetic basis for the evolution of pelvic fin loss in the pufferfish Takifugu rubripes. Dev. Biol. 2005, 281, 227–239. [Google Scholar] [CrossRef]
  42. Kvon, E.Z.; Kamneva, O.K.; Melo, U.S.; Barozzi, I.; Osterwalder, M.; Mannion, B.J.; Tissières, V.; Pickle, C.S.; Plajzer-Frick, I.; Lee, E.A.; et al. Progressive loss of function in a limb enhancer during snake evolution. Cell 2016, 167, 633–642. [Google Scholar] [CrossRef]
  43. Infante, C.R.; Mihala, A.G.; Park, S.; Wang, J.S.; Johnson, K.K.; Lauderdale, J.D.; Menke, D.B. Shared enhancer activity in the limbs and phallus and functional divergence of a limb-genital cis-regulatory element in snakes. Dev. Cell 2015, 35, 107–119. [Google Scholar] [CrossRef]
  44. Lettice, L.A.; Heaney, S.J.; Purdie, L.A.; Li, L.; de Beer, P.; Oostra, B.A.; Goode, D.; Elgar, G.; Hill, R.E.; de Graaff, E. A long-range Shh enhancer regulates expression in the developing limb and fin and is associated with preaxial polydactyly. Hum. Mol. Genet. 2003, 12, 1725–1735. [Google Scholar] [CrossRef]
  45. Lettice, L.A.; Williamson, I.; Wiltshire, J.H.; Peluso, S.; Devenney, P.S.; Hill, A.E.; Essafi, A.; Hagman, J.; Mort, R.; Grimes, G.; et al. Opposing functions of the ETS factor family define Shh spatial expression in limb buds and underlie polydactyly. Dev. Cell 2012, 22, 459–467. [Google Scholar] [CrossRef]
  46. Sagai, T.; Hosoya, M.; Mizushina, Y.; Tamura, M.; Shiroishi, T. Elimination of a long-range cis-regulatory module causes complete loss of limb-specific Shh expression and truncation of the mouse limb. Development 2005, 132, 797–803. [Google Scholar] [CrossRef]
  47. Lettice, L.A.; Hill, A.E.; Devenney, P.S.; Hill, R.E. Point mutations in a distant sonic hedgehog cis-regulator generate a variable regulatory output responsible for preaxial polydactyly. Hum. Mol. Genet. 2008, 17, 978–985. [Google Scholar] [CrossRef]
  48. Lettice, L.A.; Williamson, I.; Devenney, P.S.; Kilanowski, F.; Dorin, J.; Hill, R.E. Development of five digits is controlled by a bipartite long-range cis-regulator. Development 2014, 141, 1715–1725. [Google Scholar] [CrossRef]
  49. Sagai, T.; Masuya, H.; Tamura, M.; Shimizu, K.; Yada, Y.; Wakana, S.; Gondo, Y.; Noda, T.; Shiroishi, T. Phylogenetic conservation of a limb-specific, cis-acting regulator of Sonic hedgehog (Shh). Mamm. Genom. 2004, 15, 23–34. [Google Scholar] [CrossRef]
  50. Zeller, R.; Zuniga, A. Shh and Gremlin1 chromosomal landscapes in development and disease. Curr. Opin. Genet. Dev. 2007, 17, 428–434. [Google Scholar] [CrossRef]
  51. Thompson, A.C.; Capellini, T.D. A novel enhancer near the Pitx1 gene influences development and evolution of pelvic appendages in vertebrates. eLife 2018, 7, 38555. [Google Scholar] [CrossRef]
  52. Logan, M.; Tabin, C.J. Role of Pitx1 upstream of Tbx4 in specification of hindlimb identity. Science 1999, 283, 1736–1739. [Google Scholar] [CrossRef]
  53. Marcil, A.; Dumontier, E.; Chamberl, M.; Camper, S.A.; Drouin, J. Pitx1 and Pitx2 are required for development of hindlimb buds. Development 2003, 130, 45–55. [Google Scholar] [CrossRef]
  54. Shapiro, M.D.; Marks, M.E.; Peichel, C.L.; Blackman, B.K.; Nereng, K.S.; Jónsson, B.; Schluter, D.; Kingsley, D.M. Genetic and developmental basis of evolutionary pelvic reduction in threespine sticklebacks. Nature 2004, 428, 717–723. [Google Scholar] [CrossRef]
  55. Shapiro, M.D.; Bell, M.A.; Kingsley, D.M. Parallel genetic origins of pelvic reduction in vertebrates. Proc. Natl. Acad. Sci. USA 2006, 103, 13753–13758. [Google Scholar] [CrossRef] [Green Version]
  56. Xie, B.; Li, X.; Lin, Z.; Ruan, Z.; Wang, M.; Liu, J.; Tong, T.; Li, J.; Huang, Y.; Wen, B.; et al. Prediction of toxin genes from Chinese yellow catfish based on transcriptomic and proteomic sequencing. Int. J. Mol. Sci. 2016, 17, 556. [Google Scholar] [CrossRef]
  57. Xie, B.; Huang, Y.; Baumann, K.; Fry, B.G.; Shi, Q. From marine venoms to drugs: Efficiently supported by a combination of transcriptomics and proteomics. Mar. Drugs 2017, 15, 103. [Google Scholar] [CrossRef]
  58. Xu, Z.; Wang, H. LTR_FINDER: An efficient tool for the prediction of full-length LTR retrotransposons. Nucleic Acids Res. 2007, 35, 265–268. [Google Scholar] [CrossRef]
  59. Tarailo-Graovac, M.; Chen, N. Using RepeatMasker to identify repetitive elements in genomic sequences. Curr. Protoc. Bioinform. 2009, 25, 4–10. [Google Scholar]
  60. Jurka, J.; Kapitonov, V.V.; Pavlicek, A.; Klonowski, P.; Kohany, O.; Walichiewicz, J. Repbase Update, a database of eukaryotic repetitive elements. Cytogenet. Genome Res. 2005, 110, 462–467. [Google Scholar] [CrossRef]
  61. Stanke, M.; Keller, O.; Gunduz, I.; Hayes, A.; Waack, S.; Morgenstern, B. AUGUSTUS: Ab initio prediction of alternative transcripts. Nucleic Acids Res. 2006, 34, 435–439. [Google Scholar] [CrossRef]
  62. Burge, C.; Karlin, S. Prediction of complete gene structures in human genomic DNA. J. Mol. Biol. 1997, 268, 78–94. [Google Scholar] [CrossRef] [Green Version]
  63. Mount, D.W. Using the basic local alignment search tool (blast). Cold Spring Harb. Protoc. 2007, 2007. [Google Scholar] [CrossRef]
  64. Birney, E.; Durbin, R. Using genewise in the drosophila annotation experiment. Genome Res. 2000, 10, 547–548. [Google Scholar] [CrossRef]
  65. Trapnell, C.; Pachter, L.; Salzberg, S.L. TopHat: Discovering splice junctions with RNA-Seq. Bioinformatics 2009, 25, 1105–1111. [Google Scholar] [CrossRef]
  66. Haas, B.J.; Salzberg, S.L.; Zhu, W.; Pertea, M.; Allen, J.E.; Orvis, J.; White, O.; Buell, C.R.; Wortman, J.R. Automated eukaryotic gene structure annotation using EVidenceModeler and the Program to Assemble Spliced Alignments. Genome Biol. 2008, 9, R7. [Google Scholar] [CrossRef]
  67. Kanehisa, M.; Goto, S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 2000, 27, 29–34. [Google Scholar] [CrossRef]
  68. Bairoch, A.; Apweiler, R. The SWISS-PROT protein sequence database and its supplement TrEMBL in 2000. Nucleic Acids Res. 2000, 28, 45–48. [Google Scholar] [CrossRef]
  69. Hunter, S.; Apweiler, R.; Attwood, T.K.; Bairoch, A.; Bateman, A.; Binns, D.; Bork, P.; Das, U.; Daugherty, L.; Duquenne, L.; et al. InterPro: The integrative protein signature database. Nucleic Acids Res. 2009, 37, 211–215. [Google Scholar] [CrossRef]
  70. Liu, K.; Xu, D.; Li, J.; Bian, C.; Duan, J.; Zhou, Y.; Zhang, M.; You, X.; You, Y.; Chen, J.; et al. Whole genome sequencing of Chinese clearhead icefishe, Protosalanx hyalocranius. GigaScience 2017, 6, 1–6. [Google Scholar] [CrossRef]
  71. Xu, J.; Bian, C.; Chen, K.; Liu, G.; Jiang, Y.; Luo, Q.; You, X.; Peng, W.; Li, J.; Huang, Y.; et al. Draft genome of the northern snakehead, Channa argus. Gigascience 2017, 6, 1–5. [Google Scholar] [CrossRef]
  72. Nomura, K.; Fujiwara, A.; Iwasaki, Y.; Nishiki, I.; Matsuura, A.; Ozaki, A.; Sudo, R.; Tanaka, H. Genetic parameters and quantitative trait loci analysis associated with body size and timing at metamorphosis into glass eels in captive-bred Japanese eels (Anguilla japonica). PLoS ONE 2018, 13, e0201784. [Google Scholar] [CrossRef]
  73. Slater, G.S.; Birney, E. Automated generation of heuristics for biological sequence comparison. BMC Bioinform. 2005, 6, 31. [Google Scholar] [CrossRef]
  74. Edgar, R.C. Muscle: A multiple sequence alignment method with reduced time and space complexity. BMC Bioinform. 2004, 5, 113. [Google Scholar] [CrossRef]
  75. Beitz, E. Texshade: Shading and labeling of multiple sequence alignments using latex2 epsilon. Bioinformatics 2000, 16, 135–139. [Google Scholar] [CrossRef]
  76. Posada, D.; Buckley, T.R. Model selection and model averaging in phylogenetics: Advantages of akaike information criterion and bayesian approaches over likelihood ratio tests. Syst. Biol. 2004, 53, 793–808. [Google Scholar] [CrossRef]
  77. Darriba, D.; Taboada, G.L.; Doallo, R.; Posada, D. ProtTest 3: Fast selection of best-fit models of protein evolution. Bioinformatics 2011, 27, 1164–1165. [Google Scholar] [CrossRef]
  78. Kumar, S.; Stecher, G.; Tamura, K. MEGA7: Molecular evolutionary genetics analysis version 7.0 for bigger datasets. Mol. Biol. Evol. 2016, 33, 1870–1874. [Google Scholar] [CrossRef]
  79. Guindon, S.; Dufayard, J.F.; Lefort, V.; Anisimova, M.; Hordijk, W.; Gascuel, O. New algorithms and methods to estimate maximum-likelihood phylogenies: Assessing the performance of PhyML 3.0. Syst. Biol. 2010, 59, 307–321. [Google Scholar] [CrossRef]
  80. Guindon, S.; Delsuc, F.; Dufayard, J.F.; Gascuel, O. Estimating Maximum Likelihood Phylogenies with PhyML. Methods Mol. Biol. 2009, 537, 113–137. [Google Scholar] [Green Version]
  81. Brudno, M.; Do, C.B.; Cooper, G.M.; Kim, M.F.; Davydov, E.; Green, E.D.; Sidow, A.; Batzoglou, S. NISC Comparative Sequencing Program. LAGAN and Multi-LAGAN: Efficient Tools for Large-Scale Multiple Alignment of Genomic DNA. Genome Res. 2003, 13, 721–731. [Google Scholar] [CrossRef]
Figure 1. A 17-mer distribution of the Japanese eel genome sequencing. Only the sequencing data from short-insert libraries (500 and 800 bp) were used for the k-mer analysis. The x-axis is the sequencing depth of each unique 17-mer, and the y-axis is the percentage of these unique 17-mers. The peak depth (K_depth) is 37, and the corresponding k-mer number (N) is 37,982,773,125. We therefore calculated the genome size (G) to be ~1.03 Gb based on the following formula [12]: G=N/K_depth.
Figure 1. A 17-mer distribution of the Japanese eel genome sequencing. Only the sequencing data from short-insert libraries (500 and 800 bp) were used for the k-mer analysis. The x-axis is the sequencing depth of each unique 17-mer, and the y-axis is the percentage of these unique 17-mers. The peak depth (K_depth) is 37, and the corresponding k-mer number (N) is 37,982,773,125. We therefore calculated the genome size (G) to be ~1.03 Gb based on the following formula [12]: G=N/K_depth.
Marinedrugs 17 00426 g001
Figure 2. Structures of the tbx4 genes in various vertebrate species. Green boxes and lines represent exons and introns, respectively. Numbers inside the boxes are the exact amino acid numbers, indicating their similarity among various species.
Figure 2. Structures of the tbx4 genes in various vertebrate species. Green boxes and lines represent exons and introns, respectively. Numbers inside the boxes are the exact amino acid numbers, indicating their similarity among various species.
Marinedrugs 17 00426 g002
Figure 3. Similarity of the T-box domain in the tbx4 genes of various vertebrate species. The blue color represents conserved sites. The yellow color represents the similarity >80% and white shows less conserved sites. The colored dots beneath the sequence alignment indicate conservation track, ranging from blue (non-conserved) to red (the most conserved). The red box highlights the NLS region of the tbx4 genes. A red arrow indicates a nonsynonymous mutation in the Japanese eel tbx4 gene.
Figure 3. Similarity of the T-box domain in the tbx4 genes of various vertebrate species. The blue color represents conserved sites. The yellow color represents the similarity >80% and white shows less conserved sites. The colored dots beneath the sequence alignment indicate conservation track, ranging from blue (non-conserved) to red (the most conserved). The red box highlights the NLS region of the tbx4 genes. A red arrow indicates a nonsynonymous mutation in the Japanese eel tbx4 gene.
Marinedrugs 17 00426 g003
Figure 4. Visualization of the tbx4 NLS regions in different vertebrate species using Bioedit (Tom Hall Ibis Therapeutics, Carlsbad, CA, USA). Red characters represent the codons and amino acids of nonsynonymous mutations.
Figure 4. Visualization of the tbx4 NLS regions in different vertebrate species using Bioedit (Tom Hall Ibis Therapeutics, Carlsbad, CA, USA). Red characters represent the codons and amino acids of nonsynonymous mutations.
Marinedrugs 17 00426 g004
Figure 5. Alignment of the TBX4 protein sequences of Japanese eel against zebrafish. The red box highlights the NLS regions (same as Figure 4).
Figure 5. Alignment of the TBX4 protein sequences of Japanese eel against zebrafish. The red box highlights the NLS regions (same as Figure 4).
Marinedrugs 17 00426 g005
Figure 6. Phylogenetic and synteny comparisons of the tbx4 genes in vertebrates. The figure in the left is a Bayesian tree. Numbers on the branches are bootstrap supports (black) obtained from the phyML-3.1 reconstruction. Spotted gar was used as the outgroup. The figure in the right represents the synteny of tbx4. Distances between genes and the gene length are not drawn to scale. The red branches and the two black boxes highlight two remarkable inversions of the brip1-tbx4-tbx2b-bcas3 cluster in teleost species. The five-point star represents the determined pelvic fin loss in the examined species.
Figure 6. Phylogenetic and synteny comparisons of the tbx4 genes in vertebrates. The figure in the left is a Bayesian tree. Numbers on the branches are bootstrap supports (black) obtained from the phyML-3.1 reconstruction. Spotted gar was used as the outgroup. The figure in the right represents the synteny of tbx4. Distances between genes and the gene length are not drawn to scale. The red branches and the two black boxes highlight two remarkable inversions of the brip1-tbx4-tbx2b-bcas3 cluster in teleost species. The five-point star represents the determined pelvic fin loss in the examined species.
Marinedrugs 17 00426 g006
Figure 7. The brip1-tbx4-tbx2b-bcas3 cluster. Colored boxes and lines represent genes and intergenic regions, respectively. The distance between two adjacent genes is indicated underneath the lines, while the length of exons is drawn to scale. Genes in the same orientation as tbx4 are marked above the horizontal lines; however, genes in the opposite orientation are placed below the lines.
Figure 7. The brip1-tbx4-tbx2b-bcas3 cluster. Colored boxes and lines represent genes and intergenic regions, respectively. The distance between two adjacent genes is indicated underneath the lines, while the length of exons is drawn to scale. Genes in the same orientation as tbx4 are marked above the horizontal lines; however, genes in the opposite orientation are placed below the lines.
Marinedrugs 17 00426 g007
Figure 8. A VISTA plot to compare the HLEB sequences of Acanthopterygii fishes against the reported 873-bp HLEB from three-spined stickleback (Gasterosteus aculeatus). Sequence identity along the y-axis, ranging from 50% to 100%, is shown in 100-bp sliding windows across the examined region (x-axis, bp). The pink shadows stand for regions with <100-bp continuous bases at ≥70% identity.
Figure 8. A VISTA plot to compare the HLEB sequences of Acanthopterygii fishes against the reported 873-bp HLEB from three-spined stickleback (Gasterosteus aculeatus). Sequence identity along the y-axis, ranging from 50% to 100%, is shown in 100-bp sliding windows across the examined region (x-axis, bp). The pink shadows stand for regions with <100-bp continuous bases at ≥70% identity.
Marinedrugs 17 00426 g008
Table 1. Summary of the assembling results.
Table 1. Summary of the assembling results.
StepSoftwareContig N50 (bp)Scaffold N50 (bp)Contig numberScaffold numberTotal length
(bp)
Primary assemblingSOAPdenovo1,999383,7981,227,464462,2721,167,219,893
Gap fillingkrskgf3,868375,823850,121462,2721,150,479,312
Gapclose1.125,372376,296761,523462,2721,154,146,689
Gapclose1.1010,215376,491624,151462,2721,154,798,407
Scaffold extending
Filtering
SSPACE
---
10,236
11,468
858,288
1,033,285
608,352
256,649
351,879
41,687
1,228,736,536
1,132,698,062
Table 2. A BUSCO assessment of our Japanese eel assembly.
Table 2. A BUSCO assessment of our Japanese eel assembly.
ParameterNumberPercentage (%)
Complete BUSCOs (C)384783.9
Complete and single-copy BUSCOs (S)334673.0
Complete and duplicated BUSCOs (D)50110.9
Fragmented BUSCOs (F)3808.3
Missing BUSCOs (M)3577.8
Total BUSCO groups searched (n)4584---
Table 3. Accession numbers for the 27 T-box family members and seven adjacent genes of tbx4.
Table 3. Accession numbers for the 27 T-box family members and seven adjacent genes of tbx4.
GeneSpecies NameAccession Number
eomesaDanio rerioAAH67719.1
eomesbD. rerioNP_001077044.1
mgalD. rerioXP_021324416.1
mgaD. rerioADA61227.1
taD. rerioQ07998.1
tbD. rerioXP_001343633.3
tbr1aD. rerioXP_693121.1
tbr1bD. rerioAAG48249.1
tbx15D. rerioAAM54074.1
tbx16D. rerioAAI65213.1
tbx18D. rerioAAI63460.1
tbx19D. rerioXP_003198807.1
tbx11D. rerioXP_017206601.2
tbx1D. rerioQ8AXX2.1
tbx20D. rerioAAF64322.1
tbx21D. rerioNP_001164070.1
tbx22D. rerioACU00296.1
tbx2aD. rerioAAH68364.1
tbx2bD. rerioQ7ZTU9.4
tbx3aD. rerioNP_001095140.2
tbx3bD. rerioXP_002662050.2
tbx4D. rerioAAI62554.1
tbx5aD. rerioQ9IAK8.2
tbx5bD. rerioADX53331.1
tbx6lD. rerioP79742.1
tbx6D. rerioQ8JIS6.2
vegtFundulus heteroclitusJAQ45978.1
lhx1aD. rerioQ90476.1
brip1Aphyosemion striatumSBP21433.1
bcas3Nothobranchius furzeriSBP60348.1
ppm1daN. furzeriSBP60348.1
appbp2N. furzeriSBP60348.1
usp32N. kuhntaeSBP60348.1

Share and Cite

MDPI and ACS Style

Chen, W.; Bian, C.; You, X.; Li, J.; Ye, L.; Wen, Z.; Lv, Y.; Zhang, X.; Xu, J.; Yang, S.; et al. Genome Sequencing of the Japanese Eel (Anguilla japonica) for Comparative Genomic Studies on tbx4 and a tbx4 Gene Cluster in Teleost Fishes. Mar. Drugs 2019, 17, 426. https://doi.org/10.3390/md17070426

AMA Style

Chen W, Bian C, You X, Li J, Ye L, Wen Z, Lv Y, Zhang X, Xu J, Yang S, et al. Genome Sequencing of the Japanese Eel (Anguilla japonica) for Comparative Genomic Studies on tbx4 and a tbx4 Gene Cluster in Teleost Fishes. Marine Drugs. 2019; 17(7):426. https://doi.org/10.3390/md17070426

Chicago/Turabian Style

Chen, Weiwei, Chao Bian, Xinxin You, Jia Li, Lizhen Ye, Zhengyong Wen, Yunyun Lv, Xinhui Zhang, Junmin Xu, Shaosen Yang, and et al. 2019. "Genome Sequencing of the Japanese Eel (Anguilla japonica) for Comparative Genomic Studies on tbx4 and a tbx4 Gene Cluster in Teleost Fishes" Marine Drugs 17, no. 7: 426. https://doi.org/10.3390/md17070426

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop