Next Article in Journal
Erratum: Pérez-Belmonte, S., et al. Subtypes of Depression: Latent Class Analysis in Spanish Old People with Depressive Symptoms. Life 2020, 10, 70
Next Article in Special Issue
Retrotransposons as Drivers of Mammalian Brain Evolution
Previous Article in Journal
Nutrient and Dietary Patterns in Relation to the Pathogenesis of Postmenopausal Osteoporosis—A Literature Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mobile Elements in Ray-Finned Fish Genomes

Dipartimento di Scienze della Vita e dell’Ambiente, Università Politecnica delle Marche, 60131 Ancona, Italy
*
Author to whom correspondence should be addressed.
Life 2020, 10(10), 221; https://doi.org/10.3390/life10100221
Submission received: 29 July 2020 / Revised: 18 September 2020 / Accepted: 22 September 2020 / Published: 25 September 2020
(This article belongs to the Special Issue The Mobilome in Vertebrate Genomes)

Abstract

:
Ray-finned fishes (Actinopterygii) are a very diverse group of vertebrates, encompassing species adapted to live in freshwater and marine environments, from the deep sea to high mountain streams. Genome sequencing offers a genetic resource for investigating the molecular bases of this phenotypic diversity and these adaptations to various habitats. The wide range of genome sizes observed in fishes is due to the role of transposable elements (TEs), which are powerful drivers of species diversity. Analyses performed to date provide evidence that class II DNA transposons are the most abundant component in most fish genomes and that compared to other vertebrate genomes, many TE superfamilies are present in actinopterygians. Moreover, specific TEs have been reported in ray-finned fishes as a possible result of an intricate relationship between TE evolution and the environment. The data summarized here underline the biological interest in Actinopterygii as a model group to investigate the mechanisms responsible for the high biodiversity observed in this taxon.

1. Introduction

Actinopterygii is one of the most diverse groups of vertebrates, with over 30,000 species [1]. This taxonomic group includes Polypteriformes, Acipenseriformes, Holostei, and Teleostei. The order Polypteriformes comprises a unique family, Polypteridae, with two extant genera, Erpetoichthys (including only one species) and Polypterus (including eleven species). These fish appeared during the Cretaceous and inhabit equatorial and subequatorial freshwater areas in Africa. Several molecular phylogenetic analyses have shown that bichirs are a basal lineage of the ray-finned fishes [2,3,4,5]. Data about genome size are available for only four species, with values ranging from 3.69 to 7.25 pg/N [6] (Figure 1), while entire genomic DNA sequencing has been performed for only Erpetoichthys calabaricus (Ensembl release 99—January 2020). The order Acipenseriformes includes two families: Acipenseridae (sturgeons), with four extant genera (Acipenser, Huso, Scaphirhynchus, and Pseudoscaphirhynchus), contains 24 species, and Polyodontidae (paddlefish), with two extant genera (Polyodon and Psephurus), contains three species. These fish are widely distributed in the rivers, lakes, and seas of Northern Hemisphere countries. Data from 17 species reveal that these taxa have a very wide range of genome sizes, spanning from 1.22 to 9.32 pg/N [6]. Moreover, the high value (9.32 pg/N) recorded in Acipenser brevirostrum is the highest value among ray-finned fishes (Figure 1).
Holostei and Teleostei are part of the Neopterygii, a group of fishes that appeared in the late Permian and were characterized by better swimming capabilities and feeding mechanisms that allowed them to colonize a wider range of habitats. The infraclass Holostei comprises two orders: Amiiformes (bowfins) has a single family, Amiidae, which includes only one living species, Amia calva; Lepisosteiformes also has a single family, Lepisosteidae (gars), including seven species subdivided into two genera, Atractosteus (three species) and Lepisosteus (four species). Amia calva is found in freshwater rivers and lakes of North America, while gars are distributed in fresh, brackish, and, sometimes, marine waters of North and Central America. Genome sizes are available for only three species of Holostei, with an average of 1.29 pg/N [6], and only the genome of Lepisosteus oculatus has been fully sequenced (Ensembl release 99—January 2020). Teleostei is the most successful group of ray-finned fishes, with more than 24,000 species subdivided into 72 orders [7]. Teleosts appeared in the Late Triassic, and their evolutionary radiation occurred during the Mesozoic and Cenozoic. They have adapted to aquatic environments worldwide, from salt to fresh waters, from cold to warm seas, and from high-elevation mountain lakes to extreme sea depths [8]. In the genome size database, more than 1800 teleost species are listed, with values ranging from 0.34 pg/N (the most compact genome among vertebrates, recorded in a species of the Tetraodontidae family) to 4.90 pg/N (in a species belonging to the Salmonidae) [6], and the genomes of more than 300 species have been sequenced (NCBI genome database).
Knowledge of genome composition and architecture is fundamental in the comprehension of the evolutionary processes responsible for fish radiation. The advent of high-throughput sequencing technologies and bioinformatics has provided a great amount of genomic data, which has been extremely useful for obtaining insights into the evolution of ray-finned fish genomes. Indeed, the identification of gene loss and duplication events, genomic arrangements, variation in base composition, and different selection pressures on specific genomic regions have been highlighted through comparative genomics [8]. Moreover, it is known that, as in other vertebrates, the genome of actinopterygians underwent two rounds of whole-genome duplication (WGD) [9,10,11]. A third event occurred in teleosts approximately 226–350 Mya, leading to duplicated genes that were probably responsible for the radiation of these clades [11,12,13]. A fourth round of whole-genome duplication occurred independently in Neotropical Corydoradinae catfishes between 54 and 66 Mya [14], in salmonids approximately 88–103 Mya [15,16] and in cyprinids, approximately 5.6–11.3 Mya [17]. Another interesting aspect of the fish genome is the GC content. On the basis of GC percentage, genomes can be divided into five isochores, i.e., regions longer than 300 kb, with a high degree of uniformity in guanine and cytosine: two light isochores, L1 with 34–36% and L2 with 37–40%, and three heavy isochores, H1 with 41–45%, H2 with 46–52%, and H3 with more than 53%. While the genomes of mammals and birds contain heavy isochores, resulting in GC heterogeneity, fish and amphibian genomes have isochores with low CG content and typically have two light isochores [18]. Excluding salmonids, a negative correlation between genome size and genomic GC% in fish has been reported [19]. In contrast, nonteleost gars possess an AT/GC compartmentalized genome [20], and their closest living relatives, the bowfin Amia calva, have a typical teleost-like AT/GC homogenous genome, despite being nonteleosts [19].
Finally, the presence of mobile elements in fish genomes most likely contribute to shaping their genomes, providing advantageous features that have allowed them to successfully adapt to different environments [21,22,23]. Given the ability to move throughout the genome, the impact of mobile elements on genome evolution is higher than commonly supposed, and several papers have recognized the role of these elements as one of the most powerful evolutionary tools [23]. On the basis of these premises, this review is focused on the importance of mobile genetic elements for the genomes of actinopterygian, one of the most diverse vertebrate groups (Table 1).

2. Mobile Elements

Mobile elements are genetic elements capable of moving throughout the genome by a transposition mechanism. The effect of their movement can be deleterious for the host genome if they interrupt genes; in contrast, it can lead to advantageous innovations, creating new genes or regulatory sequences through a process called molecular domestication.
On the basis of their transposition intermediate, either RNA or DNA, mobile elements can be distinguished into two main classes, according to the classification proposed by Wicker and colleagues [153] (Figure 1).
Class I elements transpose via RNA intermediaries and are characterized by a copy and paste transposition mechanism. Their RNA intermediate is reverse-transcribed into its complementary DNA by a reverse transcriptase (RT) encoded by the mobile element. Reverse transcription is followed by reintegration into the host genome. Through the copy and paste mechanism of transposition, Class I elements are the main source of increased repetitive fractions, thereby having a major impact in large genomes [153,154,155]. Class I mobile elements are composed of long terminal repeat (LTR) and non-LTR subclasses. LTR retrotransposons are characterized by long terminal repeats that confer the ability to transpose. For exogenous retroviruses, LTR retrotransposons are structurally composed of gag and pol genes; gag genes encode viral structural particles and pol genes encode the whole retrotranscription machinery (reverse transcriptase, ribonuclease H, and integrase; Figure 2). In contrast to LTR retrotransposons, exogenous retroviruses possess the env gene, which encodes the viral envelope. However, traces of the env gene have been found in LTR retrotransposons [156]. DIRS, considered more complex LTR retroelements [37], are structurally characterized by a tyrosine recombinase (YR) instead of an integrase and by inverted terminal repeats. Long and short interspersed nuclear elements (LINEs and SINEs) are non-LTR retrotransposons. Of these, LINEs are autonomous retroelements constituted by two open reading frames (ORFs) and a poly A tail at the 3′ end. Generally, ORF2 encodes a reverse transcriptase and an endonuclease protein [153]. In contrast, SINEs are RT-lacking retroelements, and they need RT encoded by autonomous elements to transpose [157]. Finally, another group of Class I elements, Penelope retroelements, must be considered separately due to their very large diversity in terms of structural features. The common components are pseudo-LTRs (pLTRs), a reverse transcriptase, and an endonuclease [158,159].
Class II mobile elements use a DNA intermediate to transpose their genomic DNA copies into a novel chromosomal position [160,161] and can be divided into subclasses I and II. Subclass I consists of two main elements: TIR and Crypton. TIRs are autonomous elements characterized by terminal inverted repeats (TIRs) and a transposase through which transposition occurs via a cut and paste mechanism, in which both DNA strands are cleaved. The DNA transposons hAT, Merlin, Mutator, PiggyBac, PIF-Harbinger, Tc1-Mariner, and Transib can be found in this subclass. Crypton elements use a tyrosine recombinase (YR) in a transposition mechanism, probably involving recombination between a circular intermediate and the DNA target [37]. Helitrons and Maverick are the two major representative elements of subclass II. These DNA elements transpose via a copy and paste mechanism [153]. Helitron DNA transposons replicate using a rolling-circle mechanism and encode for replication initiation (Rep) and a DNA helicase (Hel) [162], while Maverick transposons encode for an integrase, an ORF, and polymerase B. For polymerase B, transposition involves a single-strand excision phase, extrachromosomal replication, and consequent reintegration into a new location [163]. Miniature Inverted Transposable Elements (MITEs), also grouped in Class II, do not encode a transposase; therefore, they exploit transposases encoded by autonomous elements to move throughout the genome [164].

3. Transposable Elements in Actinopterygians

The evolutionary dynamics of TEs are different in several lineages, which strongly support their pivotal role in genome evolution. The evaluation of mobile element impact on the actinopterygian genome is a fundamental step toward understanding the biodiversity of this taxon. With increasing genomic resources, a clear positive correlation between genome size and the percentage of TEs has been found in ray-finned fish [21,37,68,164,165]. Moreover, a wide range of TE amounts has been recorded in this taxon, with only 6% in the compact pufferfish genome and 55% in the zebrafish genome [165] (Figure 1).
Data published to date suggest that compared to other vertebrate genomes, class II DNA transposons are the most abundant component in most fish genomes [32,165]. Most TE superfamilies (i.e., Gypsy, BEL/Pao, ERV, DIRS, Penelope, Rex6/Dong, R2, L1, RTE, L2, Rex1/Babar, Jockey, Helitron, Maverick, Zisupton, Tcl-Mariner, hAT, PIF-Harbinger, PiggyBac, and EnSpm) are present in the actinopterygian genome, evidencing a higher diversity than that in other vertebrates [165]. Among them, Tc/mariner, hAT, L1, L2, and Gypsy are the most widespread and predominant TE superfamilies in fish genomes [31,68]. Comparing the distribution of the transposon superfamilies among the actinopterygians, the Cyprinidae family presents the highest level of TE diversity [165]. However, some organisms present a predominance of specific TE superfamilies, such as Gypsy in Boleophthalmus pectinirostris, L2 and RTE in Nothobranchius furzeri, Tc/mariner in Astyanax mexicanus, and hAT in Danio rerio [165]. These elements have been preserved in the genomes of these organisms, and, thus, they could have had a pivotal role in their evolution. Shao and colleagues [165] proposed that the interaction between TEs and host genomes is comparable to that between organisms and their environments, explained by the Red Queen paradigm: harmful TEs are eliminated by host genomes, while beneficial TEs are instead preserved. Moreover, a critical role of CR1 in vertebrate evolution has been reported by the same authors. The low copy number of CR1 elements found in teleosts, contrary to primitive fishes and sarcopterygians, suggests the preservation and proliferation of these elements during the transition from water to land in tetrapods [165].
In the deeply branched nonteleost ray-finned fishes, the mobilome has been inferred from the genomes of the sturgeon Acipenser ruthenus and the spotted gar, L. oculatus. The former has a similar pattern to that observed in teleosts [166], while the latter shows a predominance of non-LTR retrotransposons [32,37]. The condition observed in spotted gar is also common to the elephant shark, Callorhinchus milii, and the lamprey, Petromyzon marinus [32]. The amount of non-LTR in bony fishes might be due to the presence of mechanisms restricting the invasion of retroelements in their genomes [31].
Another interesting feature of the ray-finned fish mobilome is the presence of more recent TE copies than those found in other vertebrate lineages. In particular, cod, stickleback, and fugu have very recent TE copies, and differences in TE activity can also be observed between species closely related to medaka and platyfish [37]. Kimura distance-based copy divergence analysis performed on 35 actinopterygians shows one or, at most, two TE amplification bursts [32,37,68,165]. These events were preceded by periods in which new elements arose through genetic mutations or where TEs invaded the host genome through horizontal transfer. Subsequently, natural selection and defense mechanisms of the host genome select beneficial mobile elements, and a period of coexistence between TEs and the host genome begins. These steps, which occurred during the history of TE activity, are associated with species radiation [148,167,168,169,170], suggesting that TEs are responsible for important evolutionary events.
L. oculatus is a nonteleost ray-finned fish that has not undergone further WGDs after those that have occurred at the base of vertebrates (1R and 2R WGDs). The quantitative analysis of TEs showed no differences among teleosts. This finding does not support any link between ancestral genome duplication and TE expansion in the teleost lineage [32]. The analysis of the Salmo salar genome revealed an expansion of DNA transposons, with a return to the diploid state after the 4R WGD [106]. The rediploidization is also achieved through the contraction of the genome associated with TE loss. This could explain the loss of Rex3, a teleost-specific non-LTR retroelement, absent in salmonids [61].
A positive correlation has been reported between the GC content of TEs and genomes [19]. Analyzing the GC% in the main TE groups, Class I retrotransposons, with 45.6%, are more GC-rich than Class II DNA transposons, with 40.1%; DIRS are the TEs with the highest GC content (53.8%), while the CMC transposons are the mobile elements with the lowest GC content (35.8%) in fish genomes. The GC-poor DNA transposons seem to be responsible for the overall GC homogenization of fish genomes.

4. Rex Retroelements

Rex retroelements are repeated elements that are widely distributed among teleost genomes and were deeply active during the evolution of this lineage [59,60,61]. Published in 1999 by Volff and his research team [59], the first report of three reverse transcriptase (RT)-carrying retrotransposons in the model fish Xiphophorus maculatus is attributable to the origin of the name Rex for this class of fish-specific retroelements.
A sequence derived from the Y chromosome of X. maculatus of the Rio Jamapa allowed Volff and colleagues firstly to isolate a truncated copy Rex1-Ximj, and then to evidence many other copies of this non-LTR retrotransposon in different teleost species, defining a second class of Rex retroelements named Rex1. Concerning their main structural features, Rex1 non-LTRs are characterized by an RT, an apurinic/apyrimidinic (A/P*) site that can be located upstream or downstream of the RT-encoding region, and a 3′-UTR region. On the other hand, Rex3 and Rex6 retroelements harbor a gene encoding an endonuclease (EN) in addition to RT.
A high copy number of a novel class of Rex retroelements, the so-called Rex6 elements, was further evidenced by Volff and colleagues in 2001 in the genomes of several teleosts [61]. Rex6 is a member of the R4 family [171] of non-LTR retrotransposons and it encodes a specific type of endonuclease, closely related to the type IIS restriction enzymes isolated from trypanosomes, nematodes, and arthropods [172]. Rex6 elements have been found in many teleost orders: Anguilliformes [55,59], Beloniformes [60,61], Carangiformes [129], Centrarchiformes [59], Characiformes [79,82,149,173,174,175], Cichliformes [60,132,133,136,176,177,178,179,180], Cypriniformes [59], Cyprinodontiformes [59], Esociformes [60], Perciformes [60,181], Salmoniformes [60], Siluriformes [174,182,183,184], and Tetraodontiformes [60,61,134,142,185].
Although no evolutionary relationship among Rex1, Rex3, and Rex6 has emerged to date, they are usually considered together in fluorescence in situ hybridization (FISH) studies, demonstrating their key role in karyotype evolution in fish (for review, see Carducci et al. [186]). Overall, their localization has been observed in heterochromatin at telomeric [82,129,173,183], pericentromeric, and centromeric regions [149,177,180,181,183] and in supernumerary chromosomes [79,176]. Of extreme interest is the non-negligible number of papers underlying the localization of Rex retroelements at the euchromatic level [129,134,179,182,184], strongly supporting the relatively high rate of gene-linkage disruption and chromosomal rearrangements in teleost genomes [149]. In general, the distribution of Rex retroelements in chromosomes varies considerably between teleost orders and families [186].
All the papers reviewed herein highlight the significant role of the Rex retroelements in the rapid evolution of teleosts, in particular, acting on karyotype and genome structure.

5. Endogenous Retroviruses

Retroviruses are viruses constituted by a single-stranded positive-sense RNA. After infection, a retrovirus reaches the cytoplasm of the host cell, where a reverse transcriptase (RT) converts its ssRNA into cDNA, ready to be integrated into the nuclear genome of the infected host cell. Once integrated, the provirus will exploit the nuclear machinery of the host cell to transcribe and translate its components. There are four main components of the basic toolkit of retrovirus genomes: long terminal repeats (LTRs), which carry a promoter sequence that mediates the interaction with integrase for retrovirus integration into the host cell genome; gag (group-specific antigen) genes, which encode structural protein components; pol (polymerase) genes, which enclose the RT, protease, and integrase domains; env (envelope) genes, which encode coat proteins [187]. Structurally, retroviruses differ from retrotransposons by the presence of genes encoding envelope proteins. Moreover, a characteristic hallmark that allows the identification of a past retrovirus infection is provided by the solo-LTR derived from ectopic homologous recombination between two LTRs [187]. Infection by a retrovirus may occur within a germline, leading to the generation of endogenous viral elements, the so-called endogenous retroviruses (ERVs).
ERVs are inherited through vertical transmission and consequently maintained within the host genome over millions of years [188]. Identified in all vertebrate lineages [189] and all belonging to the Retroviridae family, ERVs can be approximately grouped into three main classes based on the phylogenetic relationships between the seven exogenous retrovirus genera identified: Class I (closely related to Gammaretroviruses and Epsilonretroviruses), Class II (closely related to Betaretroviruses), and Class III (Spumavirus-like elements) [190]. Hayward and colleagues [189] have identified two further clades: human endogenous retroviruses S/L (HERVS/L)-like and snakehead fish retrovirus (SnRV)-like elements.
Naville and Volff [191] have shown that the overall ERV content in fish genomes ranges from 0.01 to 1%. In particular, epsilon-related retroviruses are the most frequent ERVs in ray-finned fishes [192]. The lowest value reported is 0.033% for Takifugu rubripes (with approximately 1800 insertions), and the maximum is 0.76% in Danio rerio (with more than 30,000 insertions) [191]. The best-studied ERV element in teleosts is Zebrafish Endogenous Retrovirus (ZFERV), isolated from zebrafish [193].
In addition to epsilon-related retroviruses and Snakehead fish retrovirus (SnRV)-like elements, endogenous foamy virus (EFV) sequences have been detected in different teleost species, including cod, platyfish, and zebrafish [194,195]. No reports of gamma or Class II elements have been described to date [189].
Whereas the evolutionary importance of ERVs, as a source of new genes [196] and, in general, as a mediator of gene expression [197,198] in catalyzing genome evolution, has been evidenced in mammals, nothing is known about the roles of ERVs in teleost evolution [191].
The complex evolutionary history of retroviruses has been recently investigated by Xu and colleagues [190]. Through an extensive genomic and phylogenetic analysis performed on species representing the main evolutionary lineages, of which 66 were ray-finned fishes, the authors unveiled the role of teleosts and turtles, as vehicles for retrovirus transmission, in overcoming the water–land barrier.

6. TEs and Sex Chromosomes

The wide chromosomal diversity in teleosts (e.g., interspecific diploid number variation; the presence or absence of sex and supernumerary chromosomes) has been suggested to be correlated with the ability to incorporate transposable elements [199]. The evolutionary success of TEs in a given population is strictly linked to their persistence, which is obtainable through TE vertical transmission in the germline, from one to the next generation [199]. Moreover, the accumulation of repetitive sequences is a common phenomenon in sex chromosomes, characterized by the absence of recombination [200].
Several papers have reported the involvement of Rex retrotransposons in the differentiation of sex chromosomes [149,173,175,181], with a key role played by Rex6 [173]. These elements have been mapped on the sex chromosomes of four species belonging to the Characiformes [173,175] and one species of Perciformes [181] and on the largest pair of chromosomes recognized as sexual chromosomes in one species belonging to the Cichliformes [180]. Other convincing examples of the role of TEs in the control of sexual development and function have been recently reviewed by Dechaud and colleagues [199]. A clear example of TE control in a germline through cis-regulation was reported in the medaka, Oryzias latipes: a LINE/Rex1 retroelement was found within the nonautonomous P element Izanagi, corresponding to the upstream region of the master sex-determining gene (dmrt1bY) in medaka. In particular, the LINE/Rex1-derived sequence located within the Izanagi element carries the binding site for Sox5, a transcriptional factor involved in the regulation of dmrt1bY [201,202]. A role of TEs in the determination of sex chromosome structure and evolution has also been observed in X. maculatus, in which the accumulation and spreading of Texim genes in only the Y chromosome is due to the activity of Helitron transposons, deeply influencing the evolution of this chromosome in platyfish [203]. Finally, in salmonids, analyses performed on the boundary regions of the master sex-determining gene (sdY) have shown a certain accumulation of TEs, which is probably responsible for the different sdY gene chromosomal locations despite their conservation [105,204].

7. Fish Transposons and the Environment

Both abiotic and biotic factors are continuously changing, resulting in new selective pressures that challenge population survival. To cope with these changes, organisms colonize new habitats and exploit their phenotypic plasticity and/or adaptive evolutionary traits. Natural selection allows organisms with features appropriate for a specific environment to survive and, thus, to reproduce, increasing their fitness. Genetic variants will be transmitted to the next generation, increasing in frequency in the population. Genetic variation can be caused by not only point mutations and whole-genome duplications but also TE activity. Moreover, transposons can be co-opted and exapted, creating regulatory sequences, coding exons, or entirely new genes useful for the host genome [22,23,37,205]. Indeed, a great number of reports have suggested the responsiveness and susceptibility of TEs to environmental changes or stressful conditions [206,207,208,209,210,211,212,213,214]. Yuan and colleagues [31] analyzed 52 fishes and reported an increase in the DNA transposons in bony fish living in freshwaters and an abundance of tandem repeats in marine species that was not explained by phylogenetic relationships. In particular, among DNA transposons, Tc1 is the most well-represented in freshwater bony fishes. This association clearly suggests a potential role of TEs in the adaptation of fish to their living environments. Freshwater environments might encourage the proliferation and spread of DNA transposons, probably because transposition can cause new genetic variants useful for host adaptation to the environment. According to these authors, the large number of repetitive elements can contribute to the generation of novel genes useful for adaptability to the environment. Moreover, the presence of such a high content of repetitive elements can cause unstable genomes due to recombination and splicing events. Due to natural selection, uncontrollable increases in genome size do not occur. Auvinet and colleagues [148] reported a preferential accumulation of four families of DIRS1 in specific chromosomal locations of the Antarctic teleost species belonging to the Trematomus genus. According to these authors, the concentration of these TEs in pericentromeric and centromeric areas could have been mediated by multiple glacial-interglacial cycles that took place in the Antarctic region. The variation in temperature probably led to changes in epigenetic regulation that have allowed TE bursts. An interesting correlation between TEs and the environmental temperature has also been evidenced by our group in a recent publication [207], in which a phylogenetic analysis was performed on the partial reverse transcriptase of the Rex3 retroelement in 39 teleost species. Surprisingly, in this investigation, there was a lack of correspondence with the canonical taxonomy relationships. Indeed, the Rex3 sequences analyzed clustered into two groups, strictly related to the environmental temperature in which these species live, suggesting a selective role of temperature on specific TE sequence variants.

8. Conclusions

Actinopterygii is a taxon characterized by a high diversity of species adapted to a wide range of environments. There is generally a positive correlation between genome size and TE coverage, and the major contributors to the genome size variation are DNA transposons. The data summarized here show that the ray-finned fish genomes are unique among vertebrates in their overall TE composition. The high level of TE diversity suggests that these genetic elements represent an important evolutionary tool that has had a pivotal role in fish evolution. However, it is not clear whether repetitive elements lead to environmental adaptation or vice versa [31].
Moreover, significant differences are also evident in TE activity, which might be linked to body temperature and host defense mechanisms. Indeed, body temperature is influenced by environmental conditions, which could affect the activity of the proteins involved in transposition mechanisms; the capacity to replicate and compete with other TEs is influenced by host defense mechanisms, such as piRNAs and methylation. However, information about genome size and data on genome sequencing in ray-finned fishes is still limited. Such is the case for the deep-branching nonteleost ray-finned fishes belonging to the Polypteriformes; the investigation of the genomes of these taxa could be extremely useful for providing information on the common ancestor of TEs among actinopterygian species.

Author Contributions

Conceptualization, M.A.B. and F.C.; writing—original draft preparation, F.C., M.B., A.C., E.C. and M.A.B.; supervision, M.A.B. and A.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by a grant from Ministero della Ricerca e dell’Istruzione (project number 59, 2020).

Conflicts of Interest

The authors declare no conflict of interests. The funder did not participate in any phase of the study, including the publication of data.

References

  1. Froese, R.; Pauly, D. FishBase. World Wide Web Electronic Publication. Version (12/2019). Available online: www.fishbase.org (accessed on 5 June 2020).
  2. Noack, K.; Zardoya, R.; Meyer, A. The complete mitochondrial DNA sequence of the bichir (Polypterus ornatipinnis), a basal ray-finned fish: Ancient establishment of the consensus vertebrate gene order. Genetics 1996, 144, 1165–1180. [Google Scholar]
  3. Venkatesh, B.; Erdmann, M.V.; Brenner, S. Molecular synapomorphies resolve evolutionary relationships of extant jawed vertebrates. Proc. Natl. Acad. Sci. USA 2001, 98, 11382–11387. [Google Scholar]
  4. Inoue, J.G.; Miya, M.; Tsukamoto, K.; Nishida, M. Basal actinopterygian relationships: A mitogenomic perspective on the phylogeny of the “ancient fish”. Mol. Phylogenet. Evol. 2003, 26, 110–120. [Google Scholar]
  5. Kikugawa, K.; Katoh, K.; Kuraku, S.; Sakurai, H.; Ishida, O.; Iwabe, N.; Miyata, T. Basal jawed vertebrate phylogeny inferred from multiple nuclear DNA-coded genes. BMC Evol. Biol. 2004, 2, 3. [Google Scholar]
  6. Gregory, T.R. Animal Genome Size Database. Version (12/2019). Available online: http://www.genomesize.com (accessed on 5 June 2020).
  7. Betancur-R, R.; Wiley, E.O.; Arratia, G.; Acero, A.; Bailly, N.; Miya, M.; Lecointre, G.; Ortí, G. Phylogenetic classification of bony fishes. BMC Evol. Biol. 2017, 17, 162. [Google Scholar]
  8. Bian, C.; Huang, Y.; Li, J.; You, X.; Yi, Y.; Ge, W.; Shi, Q. Divergence, evolution and adaptation in ray-finned fish genomes. Sci. China Life Sci. 2019, 62, 1003–1018. [Google Scholar]
  9. Dehal, P.; Boore, J.L. Two rounds of whole genome duplication in the ancestral vertebrate. PLoS Biol. 2005, 3, e314. [Google Scholar]
  10. Postlethwait, J.H.; Woods, I.G.; Ngo-Hazelett, P.; Yan, Y.L.; Kelly, P.D.; Chu, F.; Huang, H.; Hill-Force, A.; Talbot, W.S. Zebrafish comparative genomics and the origins of vertebrate chromosomes. Genome Res. 2000, 10, 1890–1902. [Google Scholar]
  11. Vandepoele, K.; de Vos, W.; Taylor, J.S.; Meyer, A.; Van de Peer, Y. Major events in the genome evolution of vertebrates: Paranome age and size differ considerably between ray-finned fishes and land vertebrates. Proc. Natl. Acad. Sci. USA 2004, 101, 1638–1643. [Google Scholar]
  12. Christoffels, A.; Koh, E.G.L.; Chia, J.M.; Brenner, S.; Aparicio, S.; Venkatesh, B. Fugu genome analysis provides evidence for a whole-genome duplication early during the evolution of ray-finned fishes. Mol. Biol. Evol. 2004, 21, 1146–1151. [Google Scholar]
  13. Meyer, A.; Van de Peer, Y. From 2R to 3R: Evidence for a fish-specific genome duplication (FSGD). Bioessays 2005, 27, 937–945. [Google Scholar]
  14. Marburger, S.; Alexandrou, M.A.; Taggart, J.B.; Creer, S.; Carvalho, G.; Oliveira, C.; Taylor, M.I. Whole genome duplication and transposable element proliferation drive genome expansion in Corydoradinae catfishes. Proc. R. Soc. B Biol. Sci. 2018, 285, 20172732. [Google Scholar]
  15. Alexandrou, M.A.; Swartz, B.A.; Matzke, N.J.; Oakley, T.H. Genome duplication and multiple evolutionary origins of complex migratory behavior in Salmonidae. Mol. Phylogenet. Evol. 2013, 69, 514–523. [Google Scholar]
  16. Macqueen, D.J.; Johnston, I.A. A well-constrained estimate for the timing of the salmonid whole genome duplication reveals major decoupling from species diversification. Proc. R. Soc. B 2014, 281, 2013–2881. [Google Scholar]
  17. Wang, J.T.; Li, J.T.; Zhang, X.F.; Sun, X.W. Transcriptome analysis reveals the time of the fourth round of genome duplication in common carp (Cyprinus carpio). BMC Genom. 2012, 13, 96. [Google Scholar]
  18. Costantini, M.; Cammarano, R.; Bernardi, G. The evolution of isochore patterns in vertebrate genomes. BMC Genom. 2009, 10, 146. [Google Scholar]
  19. Symonová, R.; Suh, A. Nucleotide composition of transposable elements likely contributes to AT/GC compositional homogeneity of teleost fish genomes. Mob. DNA 2019, 10, 49. [Google Scholar]
  20. Symonová, R.; Majtánová, Z.; Arias-Rodriguez, L.; Morkovský, L.; Korìnková, T.; Cavin, L.; Johnson Pokorná, M.; Dolezálková, M.; Flajshans, M.; Normandeau, E.; et al. Genome compositional organization in gars shows more similarities to mammals than to other ray-finned fish. J. Exp. Zool. Mol. Dev. Evol. 2016, 328, 607–619. [Google Scholar]
  21. Canapa, A.; Barucca, M.; Biscotti, M.A.; Forconi, M.; Olmo, E. Transposons, genome size, and evolutionary insights in animals. Cytogenet. Genome Res. 2015, 147, 217–239. [Google Scholar]
  22. Carducci, F.; Biscotti, M.A.; Barucca, M.; Canapa, A. Transposable elements in vertebrates: Species evolution and environmental adaptation. Eur. Zool. J. 2019, 86, 497–503. [Google Scholar]
  23. Warren, I.A.; Naville, M.; Chalopin, D.; Levin, P.; Berger, C.S.; Galiana, D.; Volff, J.N. Evolutionary impact of transposable elements on genomic diversity and lineage-specific innovation in vertebrates. Chrom. Res. 2015, 23, 505–531. [Google Scholar]
  24. Pujolar, J.M.; Astolfi, L.; Boscari, E.; Vidotto, E.; Barbisan, F.; Bruson, A.; Congiu, L. Tana1, a new putatively active Tc1-like transposable element in the genome of sturgeons. Mol. Phylogenet. Evol. 2013, 66, 223–232. [Google Scholar]
  25. Cheng, P.; Huang, Y.; Du, H.; Li, C.; Lv, Y.; Ruan, R.; Ye, H.; Bian, C.; You, X.; Xu, J.; et al. Draft genome and complete Hox-cluster characterization of the sterlet (Acipenser ruthenus). Front. Genet. 2019, 10, 776. [Google Scholar]
  26. Braasch, I.; Gehrke, A.R.; Smith, J.J.; Kawasaki, K.; Manousaki, T.; Pasquier, J.; Amores, A.; Desvignes, T.; Batze, P.; Catchen, J.; et al. The spotted gar genome illuminates vertebrate evolution and facilitates human-teleost comparisons. Nat. Genet. 2016, 48, 4. [Google Scholar]
  27. Hao, S.; Han, K.; Meng, L.; Huang, X.; Shi, C.; Zhang, M.; Wang, Y.; Liu, Q.; Zhang, Y.; Seim, I.; et al. Three genomes of Osteoglossidae shed light on ancient teleost evolution. BioRxiv 2020. [Google Scholar] [CrossRef]
  28. Jiang, J.; Yuan, H.; Zheng, X.; Wang, Q.; Kuang, T.; Li, J.; Liu, J.; Song, S.; Wang, W.; Cheng, F.; et al. Gene markers for exon capture and phylogenomics in ray-finned fishes. Ecol. Evol. 2019, 9, 3973–3983. [Google Scholar]
  29. Rodriguez, F.; Arkhipova, I.R. Transposable elements and polyploid evolution in animals. Curr. Opin. Genet. Dev. 2018, 49, 115–123. [Google Scholar]
  30. Symonová, R.; Howell, W.M. Vertebrate genome evolution in the light of fish cytogenomics and rDNAomics. Genes 2018, 9, 96. [Google Scholar]
  31. Yuan, Z.; Liu, S.; Zhou, T.; Tian, C.; Bao, L.; Dunham, R.; Liu, Z. Comparative genome analysis of 52 fish species suggests differential associations of repetitive elements with their living aquatic environments. BMC Genom. 2018, 19, 141. [Google Scholar]
  32. Chalopin, D.; Volff, J.N. Analysis of the spotted gar genome suggests absence of causative link between ancestral genome duplication and transposable element diversification in teleost fish. J. Exp. Zool. B Mol. Dev. Evol. 2017, 328, 629–637. [Google Scholar]
  33. Pasquier, J.; Braasch, I.; Batzel, P.; Cabau, C.; Montfort, J.; Nguyen, T.; Jouanno, E.; Berthelot, C.; Klopp, C.; Journot, L.; et al. Evolution of gene expression afterwhole-genome duplication: New insights from the spotted gar genome. J. Exp. Zool. Mol. Dev. Evol. 2017, 328, 709–721. [Google Scholar]
  34. Gornung, E. Twenty years of physical mapping of major ribosomal RNA genes across the teleosts: A review of research. Cytogenet. Genome Res. 2013, 141, 90–102. [Google Scholar]
  35. Spaink, H.P.; Jansen, H.J.; Dirks, R.P. Advances in genomics of bony fish. Brief. Funct. Genomics 2013, 13, 144–156. [Google Scholar]
  36. Kajikawa, M.; Okada, N. LINEs mobilize SINEs in the eel through a shared 3′ sequence. Cell 2002, 111, 433–444. [Google Scholar]
  37. Chalopin, D.; Naville, M.; Plard, F.; Galiana, D.; Volff, J.N. Comparative analysis of transposable elements highlights mobilome diversity and evolution in vertebrates. Genome Biol. Evol. 2015, 7, 567–580. [Google Scholar]
  38. Coluccia, E.; Pichiri, G.; Nieddu, M.; Coni, P.; Manconi, S.; Deiana, A.M.; Salvadori, S.; Mezzanotte, R. Identification of two new repetitive elements and chromosomal mapping of repetitive DNA sequences in the fish Gymnothorax unicolor (Anguilliformes: Muraenidae). Eur. J. Histochem. 2011, 55, e12. [Google Scholar]
  39. Durif, C.M.F.; Dufour, S.; Elie, P. Impact of silvering stage, age, body size and condition on reproductive potential of the European eel. Mar. Ecol. Prog. Ser. 2006, 327, 171–181. [Google Scholar]
  40. Frankowski, J.; Bastrop, R. Identification of Anguilla anguilla (L.) and Anguilla rostrata (LeSueur) and their hybrids based on a diagnostic single nucleotide polymorphism in nuclear 18SrDNA. Mol. Ecol. Res. 2010, 10, 173–176. [Google Scholar]
  41. Geeraerts, C.; Belpaire, C. The effects of contaminants in European eel: A review. Ecotoxicology 2010, 19, 239–266. [Google Scholar]
  42. Gong, X.L.; Li, S.F.; Cai, W.C.; Wang, C.H. Isolation and characterization of polymorphic microsatellite markers from Australia short-finned eel (Anguilla australis Richardson). Mol. Ecol. Res. 2008, 8, 777–779. [Google Scholar]
  43. Ishikawa, S.; Tsukamoto, K.; Nishida, M. Characterization of microsatellite loci from the Japanese eel Anguilla japonica. Mol. Ecol. Notes 2001, 1, 140–142. [Google Scholar]
  44. Itoi, S.; Nakaya, M.; Kaneko, G.; Kondo, H.; Sezaki, K.; Watabe, S. Rapid identification of eels Anguilla japonica and Anguilla anguilla by polymerase chain reaction with single nucleotide polymorphism-based specific probes. Fish Sci. 2005, 71, 1356–1364. [Google Scholar]
  45. Jansen, H.J.; Liem, M.; Jong-Raadsen, S.A.; Dufour, S.; Weltzien, F.A.; Swinkels, W.; Koelewijn, A.; Palstra, A.P.; Pelster, B.; Spaink, H.P.; et al. Rapid de novo assembly of the European eel genome from nanopore sequencing reads. Sci. Rep. 2017, 7, 7213. [Google Scholar]
  46. Kitaoka, M.; Okamura, N.; Ichinose, H.; Goto, M. Detection of SNPs in fish DNA: Application of the Fluorogenic Ribonuclease Protection (FRIP) assay for the authentication of food contents. J. Agric. Food Chem. 2008, 56, 6246–6251. [Google Scholar]
  47. Li, Z.; Chen, F.; Huang, C.; Zheng, W.; Yu, C.; Cheng, H.; Zhou, R. Genome-wide mapping and characterization of microsatellites in the swamp eel genome. Sci. Rep. 2017, 7, 3157. [Google Scholar]
  48. Maes, G.E.; Pujolar, J.M.; Raeymaekers, J.A.M.; Dannewitz, J.; Volckaert, F.A.M. Microsatellite conservation and bayesian individual assignment in four Anguilla species. Mar. Ecol. Prog. Ser. 2006, 319, 251–261. [Google Scholar]
  49. Pierron, F.; Daffe, G.; Lambert, P.; Couture, P.; Baudrimont, M. Retrotransposon methylation and activity in wild fish (A. anguilla): A matter of size. Environ. Poll. 2019, 245, 494–503. [Google Scholar]
  50. Pujolar, J.M.; Maes, G.E.; Volckaert, F.A.M. Genetic patchiness among recruits in the European eel Anguilla anguilla. Mar. Ecol. Prog. Ser. 2006, 307, 209–217. [Google Scholar]
  51. Suntronpong, A.; Singchat, W.; Kruasuwan, W.; Prakhongcheep, O.; Sillapaprayoon, S.; Muangmai, N.; Somyong, S.; Indananda, C.; Kraichak, E.; Peyachoknagul, S.; et al. Characterization of centromeric satellite DNAs (MALREP) in the Asian swamp eel (Monopterus albus) suggests the possible origin of repeats from transposable elements. Genomics 2020, 112, 3097–3107. [Google Scholar]
  52. Pujolar, J.M.; Maes, G.E.; van Houdt, J.K.J.; Zane, L. Isolation and characterization of expressed sequence tag-linked microsatellite loci for the European eel (Anguilla anguilla). Mol. Ecol. Res. 2009, 9, 233–235. [Google Scholar]
  53. Pujolar, J.M.; Jacobsen, M.W.; Als, T.D.; Frydenberg, J.; Magnussen, E.; Jonsson, B.; Jiang, X.; Cheng, L.; Bekkevold, D.; Maes, G.E.; et al. Assessing patterns of hybridization between North Atlantic eels using diagnostic single-nucleotide polymorphisms. Heredity 2014, 112, 627–637. [Google Scholar]
  54. Rozenfeld, C.; Blanca, J.; Gallego, V.; Garcìa-Carpintero, V.; Herranz-Jusdado, J.G.; Pérez, L.; Asturiano, J.F.; Cañizares, J.; Peñaranda, D.S. De novo European eel transcriptome provides insights into the evolutionary history of duplicated genes in teleost lineages. PLoS ONE 2019, 14, e0218085. [Google Scholar]
  55. Suntronpong, A.; Thapana, W.; Twilprawat, P.; Prakhongcheep, O.; Somyong, S.; Muangmai, N.; Peyachoknagul, S.; Srikulnath, K. Karyological characterization and identification of four repetitive element groups (the 18S–28S rRNA gene, telomeric sequences, microsatellite repeat motifs, Rex retroelements) of the Asian swamp eel (Monopterus albus). Comp. Cytogenet. 2017, 11, 435–462. [Google Scholar]
  56. Tseng, M.C.; Chen, C.A.; Kao, H.W.; Tzeng, W.N.; Lee, S.C. Polymorphisms of GA/GT microsatellite loci from Anguilla japonica. Mar. Biotechnol. 2001, 3, 275–280. [Google Scholar]
  57. Tu, Z.; Li, S.; Mao, C. The changing tails of a novel short interspersed element in Aedes aegypti: Genomic evidence for slippage retrotransposition and the relationship between 3′ tandem repeats and the poly(dA) tail. Genetics 2004, 168, 2037–2047. [Google Scholar]
  58. Utsunomia, R.; Pansonato-Alves, J.C.; Cardim Scacchetti, P.; Oliveira, C.; Foresti, F. Scattered organization of the histone multigene family and transposable elements in Synbranchus. Genet. Mol. Biol. 2014, 37, 30–36. [Google Scholar]
  59. Volff, J.N.; Körting, C.; Sweeney, K.; Schartl, M. The non-LTR retrotransposon Rex3 from the Fish Xiphophorus is widespread among teleosts. Mol. Biol. Evol. 1999, 16, 1427–1438. [Google Scholar]
  60. Volff, J.N.; Körting, C.; Schartl, M. Multiple lineages of the non-LTR retrotransposon Rex1 with varying success in invading fish genomes. Mol. Biol. Evol. 2000, 17, 1673–1684. [Google Scholar]
  61. Volff, J.N.; Körting, C.; Froschauer, A.; Sweeney, K.; Schartl, M. Non-LTR retrotransposons encoding a restriction enzyme-like endonuclease in vertebrates. J. Mol. Evol. 2001, 52, 351–360. [Google Scholar]
  62. Yi, M.; Chen, F.; Luo, M.; Cheng, Y.; Zhao, H.; Cheng, H.; Zhou, R. Rapid Evolution of piRNA Pathway in the Teleost Fish: Implication for an Adaptation to Transposon Diversity. Genome Biol. Evol. 2014, 6, 1393–1407. [Google Scholar]
  63. Ráb, P.; Yano, C.F.; Lavoué, S.; Jegede, O.I.; Bertollo, L.A.C.; Ezaz, T.; Majtánová, Z.; de Oliveira, E.A.; Cioffi, M.B. Karyotype and mapping of repetitive DNAs in the African butterfly fish Pantodon buchholzi, the sole species of the family Pantodontidae. Cytogenet. Genome Res. 2016, 149, 312–320. [Google Scholar]
  64. de Souza, F.H.S.; Perez, M.F.; Bertollo, L.A.C.; de Oliveira, E.A.; Lavoué, S.; Gestich, C.C.; Ráb, P.; Ezaz, T.; Liehr, T.; Viana, P.F.; et al. Interspecific Genetic Differences and Historical Demography in South American Arowanas (Osteoglossiformes, Osteoglossidae, Osteoglossum). Genes 2019, 10, 693. [Google Scholar]
  65. Mohindra, V.; Dangi, T.; Tripathi, R.K.; Kumar, R.; Singh, R.K.; Jena, J.K.; Mohapatra, T. Draft genome assembly of Tenualosa ilisha, Hilsa shad, provides resource for osmoregulation studies. Sci. Rep. 2019, 9, 16511. [Google Scholar]
  66. Machado, A.M.; Tørresen, O.K.; Kabeya, N.; Couto, A.; Petersen, B.; Felício, M.; Campos, P.F.; Fonseca, E.; Bandarra, N.; Lopes-Marques, M.; et al. “Out of the can”: A draft genome assembly, liver transcriptome, and nutrigenomics of the european sardine, Sardina pilchardus. Genes 2018, 9, 485. [Google Scholar]
  67. Swathi, A.; Shekhar, M.S.; Katneni, V.K.; Vijayan, K.K. Genome size estimation of brackishwater fishes and penaeid shrimps by flow cytometry. Mol. Biol. Rep. 2018, 45, 951–960. [Google Scholar]
  68. Gao, B.; Shen, D.; Xue, S.; Chen, C.; Cui, H.; Song, C. The contribution of transposable elements to size variations between four teleost genomes. Mob. DNA 2016, 7, 4. [Google Scholar]
  69. Guo, X.M.; Zhang, Q.Q.; Sun, Y.W.; Jiang, X.Y.; Zou, S.M. Tc1-like Transposase Thm3 of silver carp (Hypophthalmichthys molitrix) can mediate gene transposition in the genome of blunt snout bream (Megalobrama amblycephala). G3 2015, 5, 2601–2610. [Google Scholar]
  70. Micale, L.; Loviglio, M.N.; Manzoni, M.; Fusco, C.; Augello, B.; Migliavacca, E.; Cotugno, G.; Monti, E.; Borsani, G.; Reymond, A.; et al. A fish-specific transposable element shapes the repertoire of p53 target genes in zebrafish. PLoS ONE 2012, 7, e46642. [Google Scholar]
  71. Pereira, C.S.A.; Pazian, M.F.; Ráb, P.; Collares-Pereira, M.J. Dynamics of Rex3 in the genomes of endangered Iberian Leuciscinae (Teleostei, Cyprinidae) and their natural hybrids. Mol. Cytogenet. 2015, 8, 81. [Google Scholar]
  72. Ahn, S.J.; Kim, M.S.; Jang, J.H.; Lim, S.U.; Lee, H.H. MMTS, a new subfamily of Tc1-like transposons. Mol. Cells. 2008, 26, 387–395. [Google Scholar]
  73. Ivics, Z.; Izsvak, Z.; Minter, A.; Hackett, P.B. Identification of functional domains and evolution of Tc1-like transposable elements. Proc. Natl. Acad. Sci. USA 1996, 93, 5008–5013. [Google Scholar]
  74. Liu, D.; You, C.; Liu, S.; Liu, L.; Duan, W.; Chen, S.; Yan, J.; Liu, Y. Characterization of a novel Tc1-like transposon from bream (Cyprinidae, Megalobrama) and its genetic variation in the polyploidy progeny of bream-red crucian carp crosses. J. Mol. Evol. 2009, 69, 395–403. [Google Scholar]
  75. Ishiyama, S.; Yamazaki, K.; Kurihara, F.; Yamashita, D.; Sao, K.; Hattori, A.; Koga, A. DNA-based transposable elements with nucleotide sequence similar to Tol2 from medaka fish are prevalent in cyprinid fishes. Gene Rep. 2017, 9, 37–45. [Google Scholar]
  76. Xu, P.; Xu, J.; Liu, G.; Chen, L.; Zhou, Z.; Peng, W.; Jiang, Y.; Zhao, Z.; Jia, Z.; Sun, Y.; et al. The allotetraploid origin and asymmetrical genome evolution of the common carp Cyprinus carpio. Nat. Commun. 2019, 10, 4625. [Google Scholar]
  77. Dulz, T.A.; Lorscheider, C.A.; Nascimento, V.D.; Noleto, R.B.; Moreira-Filho, O.; Nogaroto, V.; Vicari, M.R. Comparative cytogenetics among Leporinus friderici and Leporellus vittatus populations (Characiformes, Anostomidae): Focus on repetitive DNA elements. Comp. Cytogenet. 2019, 13, 105–120. [Google Scholar]
  78. Barbosa, P.; de Oliveira, L.A.; Pucci, M.B.; Santos, M.E.; Moreira-Filho, O.; Vicari, M.R.; Nogaroto, V.; de Almeida, M.C.; Ferreira Artoni, R. Identification and chromosome mapping of repetitive elements in the Astyanax scabripinnis (Teleostei: Characidae) species complex. Genetica 2015, 143, 55–62. [Google Scholar]
  79. Daniel, S.N.; Penitente, M.; Silva, D.M.Z.A.; Hashimoto, D.T.; Ferreira, D.C.; Foresti, F.; Porto-Foresti, F. Organization and chromosomal distribution of histone genes and transposable Rex elements in the genome of Astyanax bockmanni (Teleostei, Characiformes). Cytogenet. Genome Res. 2015, 146, 311–318. [Google Scholar]
  80. Gao, B.; Chen, W.; Shen, D.; Wang, S.; Chen, C.; Zhang, L.; Wang, W.; Wang, X.; Song, C. Characterization of autonomous families of Tc1/Mariner transposon in neoteleost genomes. Mar. Genom. 2017, 34, 67–77. [Google Scholar]
  81. Martins, C.; Ferreira, I.A.; Oliveira, C.; Foresti, F.; Galetti, P.M., Jr. A tandemly repetitive centromeric DNA sequence of the fish Hoplias malabaricus (Characiformes: Erythrinidae) is derived from 5S rDNA. Genetica 2006, 127, 133–141. [Google Scholar]
  82. Silva, D.M.Z.A.; Pansonato-Alves, J.C.; Utsunomia, R.; Daniel, S.N.; Hashimoto, D.T.; Oliveira, C.; Porto-Foresti, F.; Foresti, F. Chromosomal organization of repetitive DNA sequences in Astyanax bockmanni (Teleostei, Characiformes): Dispersive location, association and co-localization in the genome. Genetica 2013, 141, 329–336. [Google Scholar]
  83. Pessenda, G.; Piscor, D.; da Silva, E.L.; Centofante, L.; Parise-Maltempi, P.P. Chromosomal organization of retrotransposon Rex1 in Astyanax species (Characiformes, Characidae). Caryologia 2018, 71, 80–84. [Google Scholar]
  84. Ferreira, V.A.M.; Marajo, L.; Matoso, D.A.; Ribeiro, L.B.; Feldberg, E. Chromosomal mapping of Rex retrotransposons in Tambaqui (Colossoma macropomum Cuvier, 1818) exposed to three climate change scenarios. Cytogenet. Genome Res. 2019, 159, 39–47. [Google Scholar]
  85. Pucci, M.B.; Nogaroto, V.; Moreira-Filho, O.; Vicari, M.R. Dispersion of transposable elements and multigene families: Microstructural variation in Characidium (Characiformes: Crenuchidae) genomes. Genet. Mol. Biol. 2018, 41, 585–592. [Google Scholar]
  86. Schemberger, M.O.; Nascimento, V.D.; Coan, R.; Ramos, E.; Nogaroto, V.; Ziemniczak, K.; Valente, G.T.; Moreira-Filho, O.; Martins, C.; Vicari, M.R. DNA transposon invasion and microsatellite accumulation guide W chromosome differentiation in a Neotropical fish genome. Chromosoma 2019, 128, 547–560. [Google Scholar]
  87. Schemberger, M.O.; Oliveira, J.I.; Nogaroto, V.; Almeida, M.C.; Artoni, R.F.; Cestari, M.M.; Moreira-Filho, O.; Vicari, M.R. Construction and characterization of a repetitive DNA library in Parodontidae (Atinopterygii:Characiformes): A genomic and evolutionary approach to the degeneration of the W sex chromosome. Zebrafish 2014, 11, 518–527. [Google Scholar]
  88. Schemberger, M.O.; Nogaroto, V.; Almeida, M.C.; Ferreira Artoni, R.; Valente, G.T.; Martins, C.; Moreira-Filho, O.; Cestari, M.M.; Vicari, M.R. Sequence analyses and chromosomal distribution of the Tc1/Mariner element in Paraodontidae fish (Teleostei: Characiforme). Gene 2016, 593, 308–314. [Google Scholar]
  89. Terencio, M.L.; Schneider, C.H.; Gross, M.C.; Vicari, M.R.; Farias, I.P.; Passos, K.B.; Feldberg, E. Evolutionary dynamics of repetitive DNA in Semaprochilodus (Characiformes, Prochilodontidae): A fish model for sex chromosome differentiation. Sex Dev. 2013, 7, 325–333. [Google Scholar]
  90. Yano, C.; Bertollo, L.A.C.; Ezaz, T.; Trifonov, V.; Sember, A.; Liehr, T.; Cioffi, M.B. Highly conserved Z and molecularly diverged W chromosomes in the fish genus Triportheus (Characiformes, Triportheidae). Heredity 2017, 118, 276–283. [Google Scholar]
  91. Yano, C.F.; Merlo, M.A.; Portela-Bens, S.; Cioffi, M.B.; Bertollo, L.A.C.; Santos-Júnior, C.D.; Rebordinos, L. Evolutionary dynamics of multigene families in Triportheus (Characiformes, Triportheidae): A transposon mediated mechanism? Front. Mar. Sci. 2020, 7, 6. [Google Scholar]
  92. de Sene, V.F.; Pansonato-Alves, J.C.; Ferreira, D.C.; Utsumonia, R.; Oliveira, C.; Foresti, F. Mapping of the retrotransposable elements Rex1 and Rex3 in chromosomes of eigenmannia (Teleostei, Gymnotigformes, Sternopygidae). Cytogenet. Genome Res. 2015, 146, 319–324. [Google Scholar]
  93. da Silva, M.; Matoso, D.A.; Vicari, M.R.; de Almeida, M.C.; Margarido, V.P.; Artoni, R.F. Physical Mapping of 5S rDNA in two species of knifefishes: Gymnotus pantanal and Gymnotus paraguensis (Gymnotiformes). Cytogenet. Genome Res. 2011, 134, 303–307. [Google Scholar]
  94. da Silva, M.; Barbosa, P.; Artoni, R.F.; Feldberg, E. Evolutionary dynamics of 5S rDNA and recurrent association of transposable elements in electric fish of the family Gymnotidae (Gymnotiformes): The case of Gymnotus mamiraua. Cytogenet. Genome Res. 2016, 149, 297–303. [Google Scholar]
  95. Ferreira, D.C.; Oliveira, C.; Foresti, F. Chromosome mapping of retrotransposable elements Rex1 and Rex3 in three fish species in the subfamily Hypoptopomatinae (Teleostei, Siluriformes, Loricariidae). Cytogenet. Genome Res. 2011, 132, 64–70. [Google Scholar]
  96. Gonzalez-Gouveia, J.; Wolf, I.R.; Vilas-Boas, L.A.; Heslop-Harrison, J.S.; Schwarzacher, T.; Dias, A.L. Repetitive DNA in the catfish genome: rDNA, microsatellites, and Tc1-Mariner transposon sequences in Imparfinis species (Siluriformes, Heptapteridae). J. Hered. 2017, 108, 650–657. [Google Scholar]
  97. Liu, Z.; Li, P.; Kucuktas, H.; Dunham, R. Characterization of nonautonomous Tc1-like transposable elements of channel catfish (Ictalurus punctatus). Fish Physiol. Biochem. 1999, 21, 65–72. [Google Scholar]
  98. Primo, C.C.; Glugoski, L.; Vicari, M.R.; Nogaroto, V. Chromosome mapping and molecular characterization of the Tc1/Mariner element in Rineloricaria (Siluriformes: Loricariidae). Braz. Arch. Biol. Technol. 2018, 61, e18170623. [Google Scholar]
  99. Traldi, J.B.; Lui, R.L.; de Martinez, J.F.; Vicari, M.R.; Nogaroto, V.; Filho, O.M.; Blanco, D. R Chromosomal distribution of the retroelements Rex1, Rex3 and Rex6 in species of the genus Harttia and Hypostomus (Siluriformes: Loricariidae). Neotrop. Ichthyol. 2019, 17, e190010. [Google Scholar]
  100. da Silva, K.R.; Mariotto, S.; Centofante, L.; Parise-Maltempi, P.P. Chromosome mapping of a Tc1-like transposon in species of the catfish Ancistrus. Comp. Cytogenet. 2017, 11, 65–79. [Google Scholar]
  101. Hughes, L.C.; Ortí, G.; Huang, Y.; Sun, Y.; Baldwin, C.C.; Thompson, A.W.; Arcila, D.; Betancur-R, R.; Li, C.; Becker, L.; et al. Comprehensive phylogeny of ray-finned fishes (Actinopterygii) based on transcriptomic and genomic data. Proc. Natl. Acad. Sci. USA 2018, 115, 6249–6254. [Google Scholar]
  102. Ybazeta, G. Molecular systematics and biogeography of the Galaxiidae. Ph.D. Thesis, Univeristy of Toronto, Toronto, ON, Canada, 2012. [Google Scholar]
  103. Pan, Q.; Feron, R.; Yano, A.; Guyomard, R.; Jouanno, E.; Vigouroux, E.; Wen, M.; Busnel, J.M.; Bobe, J.; Concordet, J.P.; et al. Identification of the master sex determining gene in Northern pike (Esox lucius) reveals restricted sex chromosome differentiation. PLoS Genet. 2019, 15, e1008013. [Google Scholar]
  104. de Boer, J.G.; Yazawa, R.; Davidson, W.S.; Koop, B.F. Bursts and horizontal evolution of DNA transposon in the speciation of pseudotetraploid salmonids. BMC Genom. 2007, 8, 422. [Google Scholar]
  105. Faber-Hammond, J.J.; Phillips, R.B.; Brown, K.H. Comparative analysis of the shared sex-determination region (SDR) among salmonid fishes. Genome Biol. Evol. 2015, 7, 1972–1987. [Google Scholar]
  106. Lien, S.; Koop, B.K.; Sandve, S.R.; Miller, J.R.; Kent, M.P.; Nome, T.; Hvidsten, T.R.; Leong, J.S.; Minkley, D.R.; Zimin, A.; et al. The Atlantic salmon genome provides insights into rediploidization. Nature 2016, 533, 200–205. [Google Scholar]
  107. Sävilammi, T.; Primmer, C.R.; Varadharajan, S.; Guyomard, R.; Guiguen, Y.; Sandve, S.R.; Vøllestad, L.A.; Papakostas, S.; Lien, S. The chromosome-level genome assembly of european grayling reveals aspects of a unique genome evolution process within salmonids. G3 2019, 9, 1283–1294. [Google Scholar]
  108. 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 icefish, Protosalanx hyalocranius. GigaScience 2017, 6, giw012. [Google Scholar]
  109. Malmstrøm, M.; Matschiner, M.; Tørresen, O.K.; Jakobsen, K.S.; Jentoft, S. Whole genome sequencing data and de novo draft assemblies for 66 teleost species. Sci. Data 2017, 4, 160132. [Google Scholar]
  110. Ota, K.; Tateno, Y.; Gobojori, T. Highly differentiated and conserved sex chromosome in fish species (Aulopus japonicus: Teleostei, Aulopidae). Gene 2003, 317, 187–193. [Google Scholar]
  111. Brynildsen, W. Transposable Elements in Teleost Fish: In Silico Exploration of TE Activity, Diversity and Abundance across 74 Teleost Fish Genomes. Master’s Thesis, University Oslo, Oslo, Norway, 2016. Available online: http://urn.nb.no/URN:NBN:no-55565 (accessed on 4 May 2020).
  112. Tørresen, O.K.; Star, B.; Jentoft, S.; Reinar, W.B.; Grove, H.; Miller, J.R.; Walenz, B.P.; Knight, J.; Ekholm, J.M.; Peluso, P.; et al. An improved genome assembly uncovers prolific tandem repeats in Atlantic cod. BMC Genom. 2017, 18, 95. [Google Scholar]
  113. Lima, L.C.B.; de Martinez, P.A.; Molina, W.F. Cytogenetic characterization of three Balistoidea fish species from the Atlantic with inferences on chromosomal evolution in the families Monacanthidae and Balistidae. Comp. Cytogenet. 2011, 5, 61–69. [Google Scholar]
  114. Nirchio, M.; Rossi, A.R.; Foresti, F.; Oliveira, C. Chromosome evolution in fishes: A new challenging proposal from Neotropical species. Neotrop. Ichthyol. 2014, 12, 761–770. [Google Scholar]
  115. Pérez Bielsa, N. Is the Small Genome Size of Pleuronectiformes Related to Small Intron Size? Master’s Thesis, Universitat de Girona, Girona, Spain, 2013. [Google Scholar]
  116. Tarallo, A. Genome Composition Plasticity in Marine Organisms. Ph.D. Thesis, University of Naples “Federico II”, Naples, Italy, 2016. [Google Scholar]
  117. Dettleff, P.; Hormazabal, E.; Aedo, J.; Fuentes, M.; Meneses, C.; Molina, A.; Valdes, J.A. Identification and evaluation of long noncoding RNAs in response to handling stress in red cusk-eel (Genypterus chilensis) via RNA-seq. Mar. Biotechnol. 2020, 22, 94–108. [Google Scholar]
  118. Small, C.M.; Bassham, S.; Catchen, J.; Amores, A.; Fuiten, A.M.; Brown, R.S.; Jones, A.G.; Cresko, W.A. The genome of the Gulf pipefish enables understanding of evolutionary innovations. Genome Biol. 2016, 17, 258. [Google Scholar]
  119. Cioffi, M.; Bertollo, L.A.; Villa, M.A.; de Oliveira, E.A.; Tanomtong, A.; Yano, C.F.; Supiwong, W.; Chaveerach, A. Genomic organization of repetitive DNA elements and its implications for the chromosomal evolution of channid fishes (Actinopterygii, Perciformes). PLoS ONE 2015, 10, e0130199. [Google Scholar]
  120. García, E.; Cross, I.; Portela-Bens, S.; Rodríguez, M.E.; García-Angulo, A.; Molina, B.; Cuadrado, A.; Liehr, T.; Rebordinos, L. Integrative genetic map of repetitive DNA in the sole Solea senegalensis genome shows a Rex transposon located in a proto-sex chromosome. Sci. Rep. 2019, 9, 17146. [Google Scholar]
  121. Roberts, B.H.; Morrongiello, J.R.; King, A.J.; Morgan, D.L.; Saunders, T.M.; Woodhead, J.; Crook, D.A. Migration to freshwater increases growth rates in a facultatively catadromous tropical fish. Oecologia 2019, 191, 253–260. [Google Scholar]
  122. Rodríguez, M.E.; Molina, B.; Merlo, M.A.; Arias-Pérez, A.; Portela-Bens, S.; García-Angulo, A.; Cross, I.; Liehr, T.; Rebordinos, L. Evolution of the proto sex-chromosome in Solea senegalensis. Int. J. Mol. Sci. 2019, 20, 5111. [Google Scholar]
  123. Bohne, A.; Zhou, Q.; Darras, A.; Schmidt, C.; Schartl, M.; Galiana-Arnoux, D.; Volff, J.N. Zisupton—A novel superfamily of DNA transposable elements recently active in fish. Mol. Biol. Evol. 2011, 29, 631–645. [Google Scholar]
  124. Koga, A.; Shimada, A.; Shima, A.; Sakaizumi, M.; Tachida, H.; Hori, H. Evidence for recent invasion of the medaka fish genome by the Tol2 transposable element. Genetics 2000, 155, 273–281. [Google Scholar]
  125. Koga, A.; Hori, H. The Tol2 transposable element of the medaka fish: An active DNA-based element naturally occurring in a vertebrate genome. Genes Genet. Syst. 2001, 76, 1–8. [Google Scholar]
  126. Koga, A.; Sakaizumi, M.; Hori, H. Transposable elements in medaka fish. Zool. Sci. 2002, 19, 1–6. [Google Scholar]
  127. Dennenmoser, S.; Sedlazeck, F.J.; Iwaszkiewicz, E.; Li, X.-Y.; Altmuller, J.; Nolte, A.W. Copy number increases of transposable elements and protein-coding genes in an invasive fish of hybrid origin. Mol. Ecol. 2017, 26, 4712–4724. [Google Scholar]
  128. Xiao, Y.; Xiao, Z.; Ma, D.; Liu, J.; Li, J. Genome sequence of the barred knifejaw Oplegnathus fasciatus (Temminck & Schlegel, 1844): The first chromosome-level draft genome in the family Oplegnathidae. GigaScience 2019, 8, giz013. [Google Scholar]
  129. Costa, G.W.W.F.; Cioffi, M.B.; Bertollo, L.A.C.; Molina, W.F. Transposable elements in fish chromosomes: A study in the marine cobia species. Cytogenet. Genome Res. 2013, 141, 126–132. [Google Scholar]
  130. Almeida, L.A.H.; Nunes, L.A.; Bitencourt, J.A.; Molina, W.F.; Affonso, P.R.A.M. Chromosomal evolution and cytotaxonomy in wrasses (Perciformes; Labridae). J. Hered. 2017, 108, 239–253. [Google Scholar]
  131. Jacobina, U.P.; Cioffi, M.B.; Souza, L.G.R.; Calado, L.L.; Tavares, M.; Manzella, J., Jr.; Bertollo, L.A.C.; Molina, W.F. Chromosome mapping of repetitive sequences in Rachycentron canadum (Perciformes: Rachycentridae): Implications for karyotypic evolution and perspectives for biotechnological uses. J. Biomed. Biotechnol. 2011, 2011, 218231. [Google Scholar]
  132. Gross, M.C.; Schneider, C.H.; Valente, G.T.; Porto, J.I.R.; Martins, C.; Feldeberg, E. Comparative cytogenetic analysis of the genus Symphysodon (Discus Fishes, Cichlidae): Chromosomal characteristics of retrotransposons and minor ribosomal DNA. Cytogenet. Genome Res. 2010, 127, 43–53. [Google Scholar]
  133. de Freitas Mourão, A.A.; Daniel, S.N.; Hashimoto, D.T.; Ferreira, C.D.; Porto-Foresti, F. Organization and distribution of repetitive DNA classes in the Cichla kelberi and Cichla piquiti Genome. Cytologia 2017, 82, 193–197. [Google Scholar]
  134. Fischer, C.; Bouneau, L.; Coutanceau, J.P.; Weissenbach, J.; Volff, J.N.; Ozouf-Costaz, C. Global heterochromatic colocalization of transposable elements with minisatellites in the compact genome of the pufferfish Tetraodon nigroviridis. Gene 2004, 336, 175–183. [Google Scholar]
  135. Santos, F.A.; Marques, D.F.; Terencio, M.L.; Feldberg, E.; Rodrigues, L.R. Cytogenetic variation of repetitive DNA elements in Hoplias malabaricus (Characiformes-Erythrinidae) from white, black and clear water rivers of the Amazon basin. Genet. Mol. Biol. 2016, 39, 40–48. [Google Scholar]
  136. Schneider, C.H.; Gross, M.C.; Terencio, M.L.; do Carmo, E.J.; Martins, C.; Feldberg, E. Evolutionary dynamics of retrotransposable elements Rex1, Rex3 and Rex6 in neotropical cichlid genomes. BMC Evol. Biol. 2013, 13, 152. [Google Scholar]
  137. Capriglione, T.; De Paolo, S.; Cocca, E. Helinoto, a Helitron2 transposon from the icefish Chionodraco hamatus, contains a region with three deubiquitinase-like domains that exhibit transcriptional activity. Comp. Biochem. Physiol. Part D Genom. Proteom. 2014, 11, 49–58. [Google Scholar]
  138. Capriglione, T.; Odierna, G.; Caputo, V.; Canapa, A.; Olmo, E. Characterization of a Tc1- like transposon in the Antarctic ice-fish, Chionodraco hamatus. Gene 2002, 295, 193–198. [Google Scholar]
  139. Cocca, E.; De Iorio, S.; Capriglione, T. Identification of a novel helitron transposon in the genome of Antarctic fish. Mol. Phylogenet. Evol. 2011, 58, 439–446. [Google Scholar]
  140. Hendry, T.A.; Freed, L.L.; Fader, D.; Fenolio, D.; Sutton, T.T.; Lopex, J.V. Ongoing transposon-mediated genome reduction in the luminous bacterial symbionts of deep-sea ceratioid anglerfishes. Am. Soc. Microbiol. mBio 2018, 9. [Google Scholar] [CrossRef] [Green Version]
  141. Xu, D.; Molina, W.F.; Yano, C.F.; Zhang, Y.; de Oliveira, E.A.; Lou, B.; Cioffi, M.B. Comparative cytogenetics in three Sciaenid species (Teleostei, Perciformes): Evidence of interspecific chromosomal diversification. Mol. Cytogenet. 2017, 10, 37. [Google Scholar]
  142. Dasilva, C.; Hadji, H.; Ozouf-Costaz, C.; Nicaud, S.; Jaillon, O.; Weissenbach, J.; Crollius, H.R. Remarkable compartmentalization of transposable elements and pseudogenes in the heterochromatin of the Tetraodon nigroviridis genome. Proc. Natl. Acad. Sci. USA 2002, 99, 13636–13641. [Google Scholar]
  143. Guo, M.; Wang, S.; Su, Y.; Zhou, Y.; Liu, M.; Wang, J. Molecular cytogenetic analyses of Epinephelus bruneus and Epinephelus moara (Perciformes, Epinephelidae). PeerJ 2014, 2, e412. [Google Scholar]
  144. Zong, W.; Gao, B.; Diaby, M.; Shen, D.; Wang, S.; Wang, Y.; Sang, Y.; Chen, C.; Wang, X.; Song, C. Traveler, a new DD35E family of Tc1/Mariner transposons, invaded vertebrates very recently. Genome Biol. Evol. 2020, 12, 66–76. [Google Scholar]
  145. Costa, G.W.; Cioffi, M.B.; Bertollo, L.A.; Molina, W.F. The evolutionary dynamics of ribosomal genes, histone H3, and transposable Rex elements in the genome of Atlantic snappers. J. Hered. 2016, 107, 173–180. [Google Scholar]
  146. Dalziel, A.C.; Tirbhowan, S.; Drapeau, H.F.; Power, C.; Jonah, L.S.; Gbotsyo, Y.A.; Dion-Côté, A.M. Using asexual vertebrates to study genome evolution and animal physiology: Banded (Fundulus diaphanus) x common killifish (F. heteroclitus) hybrid lineages as a model system. Evol. Appl. 2020, 13, 1214–1239. [Google Scholar]
  147. Collares-Pereira, M.J.; Matos, I.; Morgado-Santos, M.; Coelho, M.M. Natural pathways towards polyploidy in animals: The Squalius alburnoides fish complex as a model system to study genome size and genome reorganization in polyploids. Cytogenet. Genome Res. 2013, 140, 97–116. [Google Scholar]
  148. Auvinet, J.; Graça, P.; Belkadi, L.; Petit, L.; Bonnivard, E.; Dettai, A.; Detrich, W.H.; Ozouf-Costaz, C.; Higuet, D. Mobilization of retrotransposons as a cause of chromosomal diversification and rapid speciation: The case for the Antarctic teleost genus Trematomus. BMC Genom. 2018, 19, 339. [Google Scholar]
  149. Cioffi, M.B.; Martins, C.; Bertollo, L.A.C. Chromosome spreading of associated transposable elements and ribosomal DNA in the fish Erythrinus erythrinus. Implications for genome change and karyoevolution in fish. BMC Evol. Biol. 2010, 10, 271. [Google Scholar]
  150. Motta-Neto, C.C.; Cioffi, M.D.; Costa, G.W.; Amorim, K.D.; Bertollo, L.A.; Artoni, R.F.; Molina, W.F. Overview on karyotype stasis in atlantic grunts (Eupercaria, Haemulidae) and the evolutionary extensions for other marine fish groups. Front. Mar. Sci. 2019, 6, 628. [Google Scholar]
  151. Auvinet, J.; Graça, P.; Ghigliotti, L.; Pisano, E.; Dettaï, A.; Ozouf-Costaz, C.; Higuet, D. Insertion hot spots of DIRS1 retrotransposon and chromosomal diversifications among the Antarctic teleosts Nototheniidae. Int. J. Mol. Sci. 2019, 20, 701. [Google Scholar]
  152. Volff, J.N.; Bouneau, L.; Ozouf-Costaz, C.; Fischer, C. Diversity of retrotransposable elements in compact pufferfish genomes. Trends Genet. 2003, 19, 12. [Google Scholar]
  153. Wicker, T.; Sabot, F.; Hua-Van, A.; Bennetzen, J.L.; Capy, P.; Chalhoub, B.; Flavell, A.; Leroy, P.; Morgante, M.; Panaud, O.; et al. A unified classification system for eukaryotic transposable elements. Nat. Rev. Genet. 2007, 8, 973–982. [Google Scholar]
  154. Sun, C.; Shepard, D.B.; Chong, R.A.; López Arriaza, J.; Hall, K.; Castoe, T.A.; Feschotte, C.; Pollock, D.D.; Mueller, R.L. LTR retrotransposons contribute to genomic gigantism in plethodontid salamanders. Genome Biol. Evol. 2012, 40, 168–183. [Google Scholar]
  155. Makałowski, W.; Gotea, V.; Pande, A.; Makałowska, I. Transposable elements: Classification, identification, and their use as a tool for comparative genomics. In Evolutionary Genomics: STATISTICAL and Computational Methods, Methods in Molecular Biology; Anisimova, M., Ed.; Springer: New York, NY, USA, 2019; Volume 1910. [Google Scholar] [CrossRef] [Green Version]
  156. Kazazian, H.H., Jr. Mobile elements: Drivers of genome evolution. Science 2004, 303, 1626–1632. [Google Scholar]
  157. Kramerov, D.A.; Vassetzky, N.S. Origin and evolution of SINEs in eukaryotic genomes. Heredity 2011, 107, 487–495. [Google Scholar]
  158. Evgen’ev, M.B.; Arkhipova, I.R. Penelope-like elements–A new class of retroelements: Distribution, function and possible evolutionary significance. Cytogenet. Genome Res. 2005, 110, 510–521. [Google Scholar]
  159. Arkhipova, I.R.; Yushenova, I.A.; Rodriguez, F. Endonuclease-containing Penelope retrotransposons in the bdelloid rotifer Adineta vaga exhibit unusual structural features and play a role in expansion of host gene families. Mob. DNA 2013, 4, 19. [Google Scholar]
  160. Bourque, G.; Burns, K.H.; Gehring, M.; Goburnovam, V.; Scluanov, A.; Hammell, M.; Imbeault, M.; Izsvák, Z.; Levin, H.L.; Macfarlan, T.S.; et al. Ten things you should know about transposable elements. Genome Biol. 2018, 19, 199. [Google Scholar]
  161. Goerner-Potvin, P.; Bourque, G. Computational tools to unmask transposable elements. Nat. Rev. Genet. 2018, 19, 688–704. [Google Scholar]
  162. Grabundzija, I.; Messing, S.A.; Thomas, J.; Cosby, R.L.; Bilic, I.; Miskey, C.; Gogol-Döring, A.; Kapitonov, V.; Diem, T.; Dalda, A.; et al. Helitron transposon reconstructed from bats reveals a novel mechanism of genome shuffling in eukaryotes. Nat. Commun. 2016, 7, 10716. [Google Scholar]
  163. Kapitonov, V.V.; Jurka, J. Self-synthesizing DNA transposons in eukaryotes. Proc. Natl. Acad. Sci. USA 2006, 103, 4540–4545. [Google Scholar]
  164. Feschotte, C.; Swamy, L.; Wessler, S.R. Genome-wide analysis of Mariner-like transposable elements in rice reveals complex relationships with stowaway miniature inverted repeat transposable elements (MITEs). Genetics 2003, 163, 747–758. [Google Scholar]
  165. Shao, F.; Han, M.; Peng, Z. Evolution and diversity of transposable elements in fish genomes. Sci. Rep. 2019, 9, 15399. [Google Scholar]
  166. Du, K.; Stöck, M.; Kneitz, S.; Klopp, C.; Woltering, J.M.; Adolfi, M.C.; Feron, R.; Prokopov, D.; Makunin, A.; Kichigin, I.; et al. The sterlet sturgeon genome sequence and the mechanisms of segmental rediploidization. Nat. Ecol. Evol. 2020, 4, 841–852. [Google Scholar]
  167. Dion-Cote, A.M.; Renaut, S.; Normandeau, E.; Bernatchez, L. RNA-seq reveals transcriptomic shock involving transposable elements reactivation in hybrids of young lake whitefish species. Mol. Biol. Evol. 2014, 31, 1188–1199. [Google Scholar]
  168. Oliver, K.R.; Greene, W.K. Mobile DNA and the TE-Thrust hypothesis: Supporting evidence from the primates. Mob. DNA 2011, 2, 8. [Google Scholar]
  169. Grahn, R.A.; Rinehart, T.A.; Cantrell, M.A.; Wichman, H.A. Extinction of LINE-1 activity coincident with a major mammalian radiation in rodents. Cytogenet. Genome Res. 2005, 110, 407–415. [Google Scholar]
  170. Platt, R.N.; Vandewege, M.W.; Kern, C.; Schmidt, C.J.; Hoffmann, F.G.; Ray, D.A. Large numbers of novel miRNAs originate from DNA transposons and are coincident with a large species radiation in bats. Mol. Biol. Evol. 2014, 31, 1536–1545. [Google Scholar]
  171. Burke, W.D.; Muller, F.; Eickbush, T.H. R4, a non-LTR retrotransposon specific to the large subunit rRNA genes of nematodes. Nucleic Acid Res. 1995, 23, 4628–4634. [Google Scholar]
  172. Yang, J.; Malik, H.S.; Eickbush, T.H. Identification of the endonuclease domain encoded by R2 and other site-specific, non-long terminal repeat retrotransposable elements. Proc. Natl. Acad. Sci. USA 1999, 96, 7847–7852. [Google Scholar]
  173. Yano, C.F.; Poltronieri, J.; Bertollo, L.A.C.; Ferreira Artoni, R.; Liehr, T.; Cioffi, M.B. Chromosomal mapping of repetitive DNAs in Triportheus trifurcatus (Characidae, Characiformes): Insights into the differentiation of the Z and W chromosomes. PLoS ONE 2014, 9, e90946. [Google Scholar]
  174. Pansonato-Alves, J.C.; Serrano, E.A.; Utsumomia, R.; Scacchetti, P.C.; Oliveira, C.; Foresti, F. Mapping five repetitive DNA classes in sympatric species of Hypostomus (Teleostei: Siluriformes: Loricariidae): Analysis of chromosomal variability. Rev. Fish. Biol. Fisher. 2013, 4, 447–489. [Google Scholar]
  175. Splendore de Borba, R.; Lourenço da Silva, E.; Parise-Maltempi, P.P. Chromosome mapping of retrotransposable elements Rex1 and Rex3 in Leporinus Spix, 1829 species (Characiformes: Anostomidae) and its relationships among heterochromatic segments and W sex chromosome. Mob. Genet. Elem. 2013, 3, e27460. [Google Scholar]
  176. Fantinatti, B.E.A.; Mazzuchelli, J.; Valente, G.T.; Cabral-de-Mello, D.C.; Martins, C. Genomic content and new insights on the origin of the B chromosome of the cichlid fish Astatotilapia latifasciata. Genetica 2011, 139, 1273–1282. [Google Scholar]
  177. Valente, G.T.; Mazzuchelli, J.; Ferreira, I.A.; Poletto, A.B. Cytogenetic mapping of the retroelements Rex1, Rex3 and Rex6 among cichlid fish: New insights on the chromosomal distribution of transposable elements. Cytogenet. Genome Res. 2011, 133, 34–42. [Google Scholar]
  178. Mazzuchelli, J.; Martins, C. Genomic organization of repetitive DNAs in the cichlid fish Astronotus ocellatus. Genetica 2009, 136, 461–469. [Google Scholar]
  179. Teixeira, W.G.; Ferreira, I.A.; Cabral-de-Mello, D.C.; Mazzuchelli, J.; Valente, G.T.; Pinhal, D.; Poletto, A.B.; Venere, P.C.; Martins, C. Organization of repeated DNA elements in the genome of the cichlid fish Cichla kelberi and its contributions to the knowledge of fish genomes. Cytogenet. Genome Res. 2009, 125, 224–234. [Google Scholar]
  180. Griffin, D.K.; Harvey, S.C.; Campos-Ramos, R.; Ayling, L.J.; Bromage, N.R.; Masabanda, J.S.; Penman, D.J. Early origins of the X and Y chromosomes: Lessons from tilapia. Cytogenet. Genome Res. 2002, 99, 157–163. [Google Scholar]
  181. Ozouf-Costaz, C.; Brandt, J.; Korting, C.; Pisano, E.; Bonillo, C.; Coutanceau, J.P.; Volff, J.N. Genome dynamics and chromosomal localization of the non-LTR retrotransposons Rex1 and Rex3 in Antarctic fish. Antarct. Sci. 2004, 16, 51–57. [Google Scholar]
  182. Supiwong, W.; Liehr, T.; Cioffi, M.B.; Chaveerach, A.; Kosyakova, N.; Pinthong, K.; Tanee, T.; Tanomtong, A. Chromosomal evolution in naked catfishes (Bagridae, Siluriformes): A comparative chromosome mapping study. Zool. Anz. 2014, 253, 316–320. [Google Scholar]
  183. Favarato, R.M.; Ribeiro, L.B.; Feldberg, E.; Matoso, D.A. Chromosomal mapping of transposable elements of the Rex family in the bristlenose catfish, ancistrus (Siluriformes, Loricariidae), from the amazonian region. J. Hered. 2017, 108, 254–261. [Google Scholar]
  184. Ferreira, D.C.; Porto-Foresti, F.; Oliveira, C.; Foresti, F. Transposable elements as a potential source for understanding the fish genome. Mob. Genet. Elements 2011, 1, 112–117. [Google Scholar]
  185. Bouneau, L.; Fischer, C.; Ozouf-Costaz, C.; Froschauer, A.; Jaillon, O.; Coutanceau, J.P.; Korting, C.; Weissenbach, J.; Bernot, A.; Volff, J.N. An active non-LTR retrotransposon with tandem structure in the compact genome of pufferfish Tetraodon nigroviridis. Genome Res. 2003, 13, 1686–1695. [Google Scholar]
  186. Carducci, F.; Barucca, M.; Canapa, A.; Biscotti, M.A. Rex retroelements and teleost genomes: An overview. Int. J. Mol. Sci. 2018, 19, 3653. [Google Scholar]
  187. Malik, H.S.; Henikoff, S.; Eickbush, T.H. Poised for contagion: Evolutionary origins of the infectious abilities of invertebrate retroviruses. Genome Res. 2000, 10, 1307–1318. [Google Scholar]
  188. Feschotte, C.; Gilbert, C. Endogenous viruses: Insights into viral evolution and impact on host biology. Nat. Rev. Genet. 2012, 13, 283–296. [Google Scholar]
  189. Hayward, A.; Cornwallis, C.K.; Jern, P. Pan-vertebrate comparative genomics unmasks retrovirus macroevolution. Proc. Natl. Acad. Sci. USA 2015, 112, 464–469. [Google Scholar]
  190. Xu, X.; Zhao, H.; Gong, Z.; Han, G.-Z. Endogenous retroviruses of non-avian/mammalian vertebrates illuminate diversity and deep history of retroviruses. Plos Pathog. 2018, 14, e1007072. [Google Scholar]
  191. Naville, M.; Volff, J.N. Endogenous retroviruses in fish genomes: From relics of past infections to evolutionary innovations? Front. Microbiol. 2016, 7, 1197. [Google Scholar]
  192. Basta, H.A.; Cleveland, S.B.; Clinton, R.A.; Dimitrov, A.G.; McClure, M.A. Evolution of teleost fish retroviruses: Characterization of new retroviruses with cellular genes. J. Virol. 2009, 83, 10152–10162. [Google Scholar]
  193. Shen, C.H.; Steiner, L.A. Genome structure and thymic expression of an endogenous retrovirus in zebrafish. J. Virol. 2004, 78, 899–911. [Google Scholar]
  194. Llorens, C.; Muñoz-Pomer, A.; Bernad, L.; Botella, H.; Moya, A. Network dynamics of eukaryotic LTR retroelements beyond phylogenetic trees. Biol. Direct. 2009, 4, 41. [Google Scholar]
  195. Schartl, M.; Walter, R.B.; Shen, Y.; Garcia, T.; Catchen, J.; Amores, A.; Braasch, I.; Chalopin, D.; Volff, J.N.; Lesch, K.P.; et al. The genome of the platyfish, Xiphophorus maculatus, provides insights into evolutionary adaptation and several complex traits. Nat. Genet. 2013, 45, 567–572. [Google Scholar]
  196. Naville, M.; Warren, I.A.; Haftek-Terreau, Z.; Chalopin, D.; Brunet, F.; Levin, P.; Galiana, D.; Volff, J.N. Not so bad after all: Retroviruses and long terminal repeat retrotransposons as a source of new genes in vertebrates. Clin. Microbiol. Infect. 2016, 22, 312–323. [Google Scholar]
  197. Feschotte, C. Transposable elements and the evolution of regulatory networks. Nat. Rev. Genet. 2008, 9, 397–405. [Google Scholar]
  198. Chuong, E.B.; Elde, N.C.; Feschotte, C. Regulatory evolution of innate immunity through co-option of endogenous retroviruses. Science 2016, 351, 1083–1087. [Google Scholar]
  199. Dechaud, C.; Volff, J.; Schartl, M.; Naville, M. Sex and the TEs: Transposable elements in sexual development and function in animals. Mob. DNA 2019, 10, 42. [Google Scholar]
  200. Ezaz, T.; Deakin, J.E. Repetitive sequence and sex chromosome evolution in vertebrates. Adv. Evol. Biol. 2014, 2014, 104683. [Google Scholar]
  201. Herpin, A.; Braasch, I.; Kraeussling, M.; Schmidt, C.; Thoma, E.C.; Nakamura, S.; Tanaka, M.; Schartl., M. Transcriptional rewiring of the sex determining dmrt1 gene duplicate by transposable elements. PLoS Genet. 2010, 6, e1000844. [Google Scholar]
  202. Schartl, M.; Schories, S.; Wakamatsu, Y.; Nagao, Y.; Hashimoto, H.; Bertin, C.; Mourot, B.; Schmidt, C.; Wilhelm, D.; Centanin, L.; et al. Sox5 is involved in germ-cell regulation and sex determination in medaka following co-option of nested transposable elements. BMC Biol. 2018, 16, 16. [Google Scholar]
  203. Tomaszkiewicz, M.; Chalopin, D.; Schartl, M.; Galiana, D.; Volff, J.N. A multicopy Y chromosomal SGNH hydrolase gene expressed in the testis of the platyfish has been captured and mobilized by a Helitron transposon. BMC Genet. 2014, 15, 44. [Google Scholar]
  204. Lubieniecki, K.P.; Lin, S.; Cabana, E.I.; Li, J.; Lai, Y.Y.Y.; Davidson, W.S. Genomic instability of the sex-determining locus in Atlantic salmon (Salmo salar). G3 2015, 5, 2513–2522. [Google Scholar]
  205. Sotero-Caio, C.G.; Platt, R.N., II; Suh, A.; Ray, D.A. Evolution and diversity of transposable elements in vertebrate genomes. Genome Biol. Evol. 2017, 9, 161–177. [Google Scholar]
  206. Casacuberta, E.; González, J. The impact of transposable elements in environmental adaptation. Mol. Ecol. 2013, 6, 1503–1517. [Google Scholar]
  207. Carducci, F.; Biscotti, M.A.; Forconi, M.; Barucca, M.; Canapa, A. An intriguing relationship between teleost Rex3 retroelement and environmental temperature. Biol. Lett. 2019, 15, 20190279. [Google Scholar]
  208. Schmidt, A.L.; Anderson, L.M. Repetitive DNA elements as mediators of genomic change in response to environmental cues. Biol. Rev. Camb. Philos. Soc. 2006, 81, 531–543. [Google Scholar]
  209. Fablet, M.; Vieira, C. Evolvability, epigenetics and transposable elements. Biomol. Concepts 2011, 2, 331–341. [Google Scholar]
  210. Canapa, A.; Biscotti, M.A.; Barucca, M.; Carducci, F.; Carotti, E.; Olmo, E. Shedding light upon the complex net of genome size, genome composition and environment in chordates. Eur. Zool. J. 2020, 87, 192–202. [Google Scholar]
  211. Biscotti, M.A.; Barucca, M.; Canapa, A. New insights into the genome repetitive fraction of the Antarctic bivalve Adamussium colbecki. PLoS ONE 2018, 13, e0194502. [Google Scholar]
  212. Chu, C.G.; Tan, C.T.; Yu, G.T.; Zhong, S.; Xu, S.S.; Yan, L. A novel retrotransposon inserted in the dominant Vrn-B1 allele confers spring growth habit in tetraploid wheat (Triticum turgidum L.). G3 2011, 1, 637–645. [Google Scholar]
  213. Fujino, K.; Hashida, S.N.; Ogawa, T.; Natsume, T.; Uchiyama, T.; Mikami, T.; Kishima, Y. Temperature controls nuclear import of Tam3 transposase in Antirrhinum. Plant J. 2011, 65, 146–155. [Google Scholar]
  214. Makarevitch, I.; Waters, A.J.; West, P.T.; Stitzer, M.; Hirsch, C.N.; Ross-Ibarra, J.; Springer, N.M. Transposable elements contribute to activation of maize genes in response to abiotic stress. PLoS Genet. 2015, 11, e1004915. [Google Scholar]
Figure 1. On the left: cladogram showing the relationships of bony fishes (modified from Betancur-R et al. [7]), with the relative DNA content from the genome size database [6]. The teleost orders lacking genome size information are shown in red. Whole-genome duplications (WGDs) are shown in blue: WGD 1R and 2R occurred in the common ancestor of vertebrates; WGD 3R occurred in the common ancestor of teleosts. The blue stars indicate taxa that underwent further independent WGD events. On the right: a separated cladogram of the Eupercaria clade with its relative nuclear DNA content. Order-level incertae sedis (O level in this figure) includes families awaiting evidence to clarify their phylogenetic status [7].
Figure 1. On the left: cladogram showing the relationships of bony fishes (modified from Betancur-R et al. [7]), with the relative DNA content from the genome size database [6]. The teleost orders lacking genome size information are shown in red. Whole-genome duplications (WGDs) are shown in blue: WGD 1R and 2R occurred in the common ancestor of vertebrates; WGD 3R occurred in the common ancestor of teleosts. The blue stars indicate taxa that underwent further independent WGD events. On the right: a separated cladogram of the Eupercaria clade with its relative nuclear DNA content. Order-level incertae sedis (O level in this figure) includes families awaiting evidence to clarify their phylogenetic status [7].
Life 10 00221 g001
Figure 2. Mobile genetic element classification based on transposition mechanisms (according to Wicker et al. [153]). The main structural components of the elements are reported on the right (modified from Makalowsky et al. [155]).
Figure 2. Mobile genetic element classification based on transposition mechanisms (according to Wicker et al. [153]). The main structural components of the elements are reported on the right (modified from Makalowsky et al. [155]).
Life 10 00221 g002
Table 1. Table summarizing works published to date about transposable elements in teleost orders.
Table 1. Table summarizing works published to date about transposable elements in teleost orders.
Teleost OrdersReferences
Polypteriformes
Acipenseriformes[24,25]
Amiiformes
Lepisosteiformes[19,20,26,27,28,29,30,31,32,33,34,35]
Elopiformes[36]
Albuliformes
Notacanthiformes
Anguilliformes[24,32,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62]
Osteoglossomorpha[63,64]
Clupeiformes[65,66]
Alepocephaliformes
Gonorynchiformes[67]
Cypriniformes[14,68,69,70,71,72,73,74,75,76]
Characiformes[77,78,79,80,81,82,83,84,85,86,87,88,89,90,91]
Gymnotiformes[92,93,94]
Siluriformes[14,31,95,96,97,98,99,100]
Lepidogalaxiiformes
Argentiniformes[101]
Galaxiiformes[102]
Esociformes[103]
Salmoniformes[104,105,106,107]
Osmeriformes[108,109]
Stomiatiformes[109]
Ateleopodiformes[109]
Aulopiformes[109,110]
Myctiophiformes[109]
Lampriformes[109]
Percopsiformes[109]
Zeiformes[109]
Stylephoriformes[109]
Gadiformes[111,112,109]
Polymixiiformes[109]
Trachicthtyiformes
Beryciformes[34,109,113,114,115,116]
Holocentriformes[109]
Ophidiaria[109,111,117]
Batrachoidiaria[109]
Pelagiaria[109]
Synghatharia[118]
Gobiaria[109]
Anabantaria[109,119]
Carangaria[109,120,121,122]
Ovalentaria[109,123,124,125,126]
Eupercaria[30,31,109,119,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152]

Share and Cite

MDPI and ACS Style

Carducci, F.; Barucca, M.; Canapa, A.; Carotti, E.; Biscotti, M.A. Mobile Elements in Ray-Finned Fish Genomes. Life 2020, 10, 221. https://doi.org/10.3390/life10100221

AMA Style

Carducci F, Barucca M, Canapa A, Carotti E, Biscotti MA. Mobile Elements in Ray-Finned Fish Genomes. Life. 2020; 10(10):221. https://doi.org/10.3390/life10100221

Chicago/Turabian Style

Carducci, Federica, Marco Barucca, Adriana Canapa, Elisa Carotti, and Maria Assunta Biscotti. 2020. "Mobile Elements in Ray-Finned Fish Genomes" Life 10, no. 10: 221. https://doi.org/10.3390/life10100221

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