- Review
- Open access
- Published:
Advancing genetic improvement in the omics era: status and priorities for United States aquaculture
BMC Genomics volume 26, Article number: 155 (2025)
Abstract
Background
The innovations of the “Omics Era” have ushered in significant advancements in genetic improvement of agriculturally important animal species through transforming genetics, genomics and breeding strategies. These advancements were often coordinated, in part, by support provided over 30 years through the 1993–2023 National Research Support Project 8 (NRSP8, National Animal Genome Research Program, NAGRP) and affiliate projects focused on enabling genomic discoveries in livestock, poultry, and aquaculture species. These significant and parallel advances demand strategic planning of future research priorities. This paper, as an output from the May 2023 Aquaculture Genomics, Genetics, and Breeding Workshop, provides an updated status of genomic resources for United States aquaculture species, highlighting major achievements and emerging priorities.
Main text
Finfish and shellfish genome and omics resources enhance our understanding of genetic architecture and heritability of performance and production traits. The 2023 Workshop identified present aims for aquaculture genomics/omics research to build on this progress: (1) advancing reference genome assembly quality; (2) integrating multi-omics data to enhance analysis of production and performance traits; (3) developing resources for the collection and integration of phenomics data; (4) creating pathways for applying and integrating genomics information across animal industries; and (5) providing training, extension, and outreach to support the application of genome to phenome. Research focuses should emphasize phenomics data collection, artificial intelligence, identifying causative relationships between genotypes and phenotypes, establishing pathways to apply genomic information and tools across aquaculture industries, and an expansion of training programs for the next-generation workforce to facilitate integration of genomic sciences into aquaculture operations to enhance productivity, competitiveness, and sustainability.
Conclusion
This collective vision of applying genomics to aquaculture breeding with focus on the highlighted priorities is intended to facilitate the continued advancement of the United States aquaculture genomics, genetics and breeding research community and industries. Critical challenges ahead include the practical application of genomic tools and analytical frameworks beyond academic and research communities that require collaborative partnerships between academia, government, and industry. The scope of this review encompasses the use of omics tools and applications in the study of aquatic animals cultivated for human consumption in aquaculture settings throughout their life-cycle.
Background
The “Omics Era” has facilitated the development of genome-enabled breeding strategies by allowing scientists to routinely generate individual sequence data and assess the relationships between genetic, genomic, and phenotypic variations across production systems of agriculturally important species. Aquaculture is agriculture and one food production sector that has and continues to benefit greatly from the omics revolution in multiple ways, ranging from direct impacts on the propagated organisms, and indirectly through understanding the pests, pathogens, and aquatic environment that influence production efficiencies [1]. Recent advancements in aquaculture genomics have been driven by technological and methodological innovations. In the United States (U.S.) these research contributions have largely been facilitated via the National Research Support Project 8 (NRSP8) National Animal Genome Research Program (NAGRP)Footnote 1 for U.S. agricultural animal species (1993–2023) and complementary research projects supported by United States Department of Agriculture (USDA) Agricultural Research Service (ARS) and USDA National Institute of Food and Agriculture (NIFA), among other organizations. The aquaculture component of the NRSP8 program began in 2003 and focused on species that had established industries (i.e., existing producers and markets) during the funding period. The NRSP8 provided a forum for aquaculture scientists to network, form collaborations, facilitate and implement large-scale projects, offer feedback to funding agencies on national research priorities, provide students with an opportunity to present research, engage with international crop and animal scientists, and seek future employment opportunities in aquaculture genomics and related areas of study (i.e., omics).
Since the last comprehensive assessment of the U.S. aquaculture genomics, genetics and breeding sector [2], there has been considerable advancement within a ‘Science to Practice’ framework [3]. Specifically, a focus was placed on developing “infrastructure,” including genomics and bioinformatics tools, databases, and genetic resources, to enable genomics-oriented discovery science that informs on-the-ground breeding and production of aquaculture animals. Figure 1 presents the current status of omics resources that have been generated for U.S. aquaculture species, with a focus on species with established industries (“Established”) in addition to species with emergent industries (“Newly Cultured”) and those cultured as a means of protecting wild populations (“Protected”). The aquaculture genomics community achieved this infrastructure goal across a broad range of vertebrate and invertebrate species propagated in freshwater and marine environments through the generation of new or vastly improved reference genome assemblies, genotyping arrays, and, for some species in particular, an expansive suite of high-throughput omics tools (Fig. 1; citations associated with each species and category are provided in File S1).
Current status of omics resources for species cultivated in United States aquaculture. The current status is indicated with various colors: Dark green: good status; light green, outstanding progress has been made, but additional work still needed; dark yellow: significant progress has been made, but significant amount of additional work still needed; light yellow, some progress has been made
Interrogation and use of whole genome data as an anchor for a multitude of applications (e.g., genomic selection, GS; marker-assisted selection, MAS; genetic engineering, population structure analysis, demographic monitoring) has occurred in several finfish and shellfish species [4,5,6,7,8,9]. Finfish species with large economies of scale (e.g., salmonids) have experienced rapid advancements that have directly impacted commercial industry due to improved genetic infrastructure. These advancements include applying genome and omics data to improve growth [10,11,12], develop disease resistant lines [13,14,15,16], achieve monosex and sterilization [17, 18], evaluate environmental pollution burdens [19], and enhance seafood traceability [20, 21]. Impactful advancements have also been made within smaller scale and newly cultured species, such that improved germplasm created using novel, and continually developing, omics-based tools are available to commercial industries as they expand [22]. Beyond the aquaculture species, omics applications to studying pests and pathogens have enabled vaccine development [23], and studies of microbiota that inhabit the host and the aquatic environment have led to improvements in animal production and production system operation [24,25,26,27]. In all, the rapid genome infrastructure advancements made to date have enabled the exploration of the genetic architecture associated with production traits, allowing for the more precise evaluation of key quantitative genetic parameters and assessment of artificial selection potential.
While it is clear the aquaculture genetics, genomics, and breeding community has made significant strides since the last comprehensive review [2], new priorities and needs have emerged. Genetic resource development remains a continuous need within aquaculture due to the high diversity of cultured species [28]. There has been a growing focus on data integration strategies that facilitate trait dissection (e.g., genotype-to-phenotype, G2P, associations) within programs and among species, although this has been especially challenging as the number and dimensionality of data have grown exponentially with the rapid and continuous development of advanced technologies. The aquaculture genomics community has been highly successful in continuing to expand libraries of knowledge (data and analyses), but integration of these data and adoption of new tools (e.g., artificial intelligence, AI, and machine learning, ML) are hurdles that require technological cost reduction, improved accessibility of data and methodologies, and teaching and training of practitioners at multiple levels. Indeed, convergence of AI/ML and genomics will be a major component of the future of agriculture, including aquaculture [29]. Moreover, the practical application of these data and analytical frameworks to industry programs outside of the academic research community is contingent on teaching and training the next-generation workforce. Doing so is critical as we enter a “post-genomics era” in which these tools have been developed, are readily available, and require educated and trained personnel to apply these advancements towards industry priorities.
Here we update the current state of genomic resources for established and newly cultured aquaculture species in the U.S. and outline future directions and priorities for the aquaculture omics research community. The update is organized by the present aims of the aquaculture omics community, with examples of achievements that have been made within each aim and descriptions of components that have yet to be realized. These aims are: (1) advancing reference genome assembly quality; (2) integrating multi-omics data to enhance analysis of production and performance traits; (3) developing resources for the collection and integration of phenomics data; (4) creating pathways for applying and integrating genomics information across animal industries; and (5) providing training, extension, and outreach to support the application of genome to phenome. The scope of this review is primarily on aquaculture species produced commercially in the U.S. for human consumption. Although the focus is primarily on U.S.-based aquaculture research pertaining to food production, references to research conducted for fisheries management and conservation and the international community are included in cases where this research represents a tool, technique, or method that is utilized by U.S.-based aquaculture researchers, or the work represents the leading-edge of research globally.
Aim 1: Advancing reference genome assembly quality
Genome sequencing has progressed rapidly since the advent of next-generation sequencing, with current technology allowing for high-quality genome assemblies to be produced relatively quickly and inexpensively. A current challenge is understanding the structures and functions of genomes and how they impact phenotypes, which has guided research efforts since the publication of the 2018–2027 Genome to Phenome Blueprint [3]. The aquaculture genomics community must continue its efforts to generate high-quality genome assemblies as they facilitate research on functional variants, spatial and epigenomic regulation of performance traits, and genome-enabled breeding (i.e., GS), which have direct and indirect applications to U.S. commercial aquaculture. A high-quality genome assembly is defined by having (1) high completeness, (2) high accuracy, (3) high contiguity, (4) sequences anchored to chromosomes, (5) sex chromosome sequences (if present), and (6) haplotypes resolved. These criteria were presented as priority goals in the prior review [2] and largely encompass the ethos of telomere-to-telomere (T2T) initiatives to routinely produce phased, complete, and gapless diploid genomes. In advanced research domains, a T2T genome can be produced in hours with complete variant discovery for all chromosomes [30]. For example, there are now instances in which the initial genome assembly meets these “high-quality” criteria (e.g., Sablefish, Anoplopoma fimbria [31]) and we expect this to become more prevalent. However, attaining these criteria remains among the highest needs for most aquaculture species.
References for the most recent genome assemblies for established and newly cultured aquaculture species are provided in Table 1. This list includes genome assemblies accessible through National Center for Biotechnology Information (NCBI) GenBank® and indicates if presently included in AquaMine (v1.2; https://aquamine.elsiklab.missouri.edu/, Elsik Lab, University of Missouri), a data mining system that facilitates integration and comparison of genomic data across species. There are too many genome assemblies to comprehensively cover within the text, however, examples of recent advancements toward improving genome assemblies and meeting the “high-quality” criteria include: anchoring sequences to chromosomes in Pacific oyster (Magallana gigas) [32, 33], eastern oyster (Crassostrea virginica) [34], Atlantic salmon (Salmo salar) [35], Nile tilapia (Oreochromis niloticus) GIFT strain [36], and Pacific white shrimp (Litopenaeus vannamei) [37]; improving genome assembly contiguity and sequencing the sex determination gene on the Y chromosome of rainbow trout (Oncorhynchus mykiss) [38]; and reducing gaps in chromosomes and scaffold assemblies in blue (Ictalurus furcatus) and channel catfish (Ictalurus punctatus) [39,40,41].
Genome-enabled breeding going forward will likely utilize multiple types of genomic variation, including single nucleotide polymorphisms (SNPs) and structural variants (SVs), alongside gene annotation data to understand the heritable basis of production traits and to identify causative genome variants. Further, in the future this will be greatly facilitated by rapid and accurate AI/ML approaches. With the rapid and significant cost reduction of next-generation sequencing, low-coverage whole-genome sequencing followed by genotype imputation is becoming a cost-effective alternative to SNP array genotyping [79]. However, the production and use of SNP genotyping arrays will continue, and development of other cost-effective genotyping technologies is still necessary to comprehensively dissect genetic differences between lines, breeds, and populations of aquaculture species. An example of such advancements can be found in rainbow trout aquaculture, where new lines have been sequenced [38, 80], SVs have been identified in breeding populations [81], and the sex-determining gene was included in the Y chromosome sequence [38]. Maintaining SVs may also have important impacts withing breeding programs, for example, depletion of SVs has been identified among inbred lines of eastern oyster when compared to wild-type, suggesting that selective breeding can influence SVs [82]. This extensive library of data is foundational for breeding programs given the tremendous diversity of genetic architecture within and between populations of aquaculture species. Collectively these data enhance G2P assessments and increase precision of individual performance prediction within a breeding population.
One avenue for breeding is the identification of functional variants for specific selection of targets and traits. Genome-wide association studies (GWAS) are now commonplace [6, 83,84,85,86,87,88,89,90,91,92,93] and genome editing technologies, like CRISPR, can be used to validate gene function through genetic engineering [94,95,96,97,98,99,100,101,102,103]. These approaches have been used to produce distinct phenotypes [97, 104, 105], such as improved growth [99, 102, 106,107,108], disease resistance [99, 109], and sterility [97, 110, 111]. The emerging use of CRISPR genome editing technology in aquaculture will expand capabilities to identify functional variants helping to improve aquaculture production traits [112]. Evolving regulatory frameworks are facilitating the use of gene editing technologies in food animals in many countries, including the U.S. [113, 114]. The acceptance, implementation, and regulation of gene editing technologies in food production varies widely [115,116,117] and will likely continue to evolve as data accumulates on the health and safety of end products.
A different breeding approach is required for traits with polygenic structure, as no singular gene contributes a significant amount to variation of the trait. In such cases, pangenomic analyses that incorporate additional forms of genomic variation may identify networks that explain phenotypic variance or increase accuracy in estimating genetic relatedness, which drive increased effectiveness of GS. A pangenome is the set of core and dispensable genes within a species, and pangenomic variation is often structural, which has not yet become widely utilized in GWAS analyses [118]. High-quality genomes will benefit pangenomic analyses broadly through SV identification and give researchers the ability to interrogate SVs for phenotypic associations [119,120,121,122], species hybridization [40], and also give breeders the ability to discover, preserve, and utilize all genomic diversity within a species or line [82]. Approaches incorporating AI/ML techniques will also greatly enhance analysis and incorporation of pangenome data.
There is considerable variation in the spectrum of genome resources currently available for U.S. aquaculture species. While a number of established species have high-quality genome assemblies that have enabled advanced GS and genome-enabled breeding approaches to be utilized, other newly cultured species may be earlier in their development and at the initial stages of generating a reference genome (Table 1). High-quality genomes are better suited for functional annotation which further increases their applicability. Ongoing efforts are currently in place to create functional annotations for aquaculture species. In Europe, the AQUA-FAANG (Functional Annotation of ANimal Genomes)Footnote 2 project aims to provide functional annotation information (e.g., quantify chromatin accessibility via assay for transposase-accessible chromatin with sequencing, or ATAC-seq) for the six aquaculture species with major industries in the European Union [123] including rainbow trout, Atlantic salmon, and common carp (Cyprinus carpio). In the U.S., the FAANG project has included aquaculture species, and at present the primary focus is on rainbow trout [124]. It is important to note that genomic resources and tools are not confined to specific geographic regions and therefore advancements in this area, such as those made through international FAANG projects, can benefit aquaculture globally and should be shared and leveraged to best achieve sweeping progress in aquaculture.
The broad variation among species highlights the number of diverse species cultured in the U.S. aquaculture industry, and new species are added to the aquaculture portfolio every year [125,126,127,128,129]. Thus, while high-quality genome assembly is a current leading edge of aquaculture genomics, it must be acknowledged and embraced that the process of developing and implementing genomic resources in U.S. aquaculture will be a long road that will continue to include reference genome and genotyping tool production, with many species needing to start at the simplest of genome resource development. Information on the established broodstocks for U.S. aquaculture species maintained by public institutions (i.e., not for profit) can be found in Table 2 in addition to information for some broodstocks maintained by organizations supported by private or public–private partnerships.
Aim 2: Integrating multi-omics data to enhance analysis of production and performance traits
While genetic information is encoded in the genome sequences, the realization of performance and production traits are regulated at multiple levels between the genotype, observed phenotype, and the environment. Regarding these levels as separate scientific disciplines, the primary omics areas used to decipher molecular components and cellular processes in aquaculture organisms are genomics, epigenomics, transcriptomics, proteomics, microbiomics, and metagenomics. A single omics layer provides a massively parallel set of data for a given component. As examples: Genomics is the study of the complete set of DNA sequences within an organism, tissue, or cell that enables investigations of genetic variation and heredity. Epigenomics is the study of non-mutational modifications to DNA and RNA that provide information on gene regulation, chromatin structure, chemical modifications, and epigenetic inheritance. Transcriptomics is the study of RNA transcripts that allows for quantification of gene expression patterns, alternative splicing, and regulatory mechanisms, including that of non-coding RNA expression. Proteomics is the study of the entire set of proteins that explores protein structure, function, and interactions. Metabolomics is the study of the metabolites or chemical signatures that provide information on cellular processes, pathways, and metabolic regulation. Microbiomics is the study of the composition and function of microbial communities associated with the host and/or their environment; and metagenomics is the study of all genetic material collected from a sample that can include DNA from any organism present in the environment beyond the host organism (Fig. 2). The incorporation of two or more of these layers in an analysis is a “multi-omics” approach (also referred to as poly-omics, pan-omics, trans-omics, integrative omics, vertical omics, and systems omics) [29].
Many transcriptome analyses of aquaculture species have demonstrated both the complexity and power of transcriptomics for decoding phenotypes. For instance, transcriptome analyses led to the identification of genes related to low oxygen response in channel catfish [130, 131] and patterns of gene expression related to Flavobacterium columnare susceptibility and resistance in white bass (Morone chrysops) and hybrid striped bass (M. chrysops x M. saxatilis) [132]. Disease challenges can induce expression of a large numbers of genes, and further comparisons of resistant and susceptible lines (e.g., distinct populations) provide insights into important genes underlying resistance (e.g., catfish [133, 134] and eastern oyster [135,136,137]). Studies of post-transcriptional processing, such as RNA splicing and poly-adenylation have also led to powerful discoveries, especially in the context of the environmental impact on genetic regulation (i.e., gene-by-environment, GxE interaction), presumably through induction of environment-sensitive promoters and enhancers, and through epigenetic regulation. For instance, bacterial infection [138, 139] and heat stress [140] drastically increased the level of alternative splicing, and alternative polyadenylation [141] in channel catfish. Similarly, an Iso-Seq (isoform sequencing) study used to improve the rainbow trout genome annotation identified alternative splicing associated with economically important phenotypes, including resistance to bacterial cold-water disease and stress tolerance [14, 142]. Analysis of transcriptomic, proteomic, and/or metabolomic data that incorporate AI/ML algorithms have been applied to determine omic signatures of growth and body size (e.g., hybrid striped bass [143]), predict egg quality (e.g., striped bass, Morone saxatilis, [144, 145]), identify responses to chemical exposure or developmental state (e.g., striped bass [146] and white perch, Morone americana [147]), predict disease resistance (e.g., common carp [148]), and detect seafood traceability signatures (e.g., wild and aquacultured shrimp [149]). Further, advanced sequencing technologies now allow for transcriptomic resolution at the single-cell and spatial levels, the findings of which are anticipated to be of similarly high value and impact to aquaculture [150,151,152,153,154]. Continued investment in high-resolution functional annotations of reference genomes is crucial for maximizing the utility of transcriptomics and other omics-based tools.
In the last decade, huge progress has been made in understanding the impact of epigenomic regulation on phenotypes and related research strongly supports the contribution of epigenetic modifications to additional layers of variation that can be targeted to improve production-relevant traits related to reproduction, health, growth, and nutrition of agricultural animals [155,156,157,158]. Although such studies are still at the infancy stage with aquaculture species, several have demonstrated the epigenetic regulation of important production traits. For instance, an epigenetically marked locus was identified to be associated with sex determination in channel catfish [159] and allelic expression of hydin-1, the candidate master sex determination gene of channel catfish, was found to be regulated by DNA methylation [160]. Gong et al. (2023) [161] found that spatial regulation is crucial for the expression of the yellow catfish (Pelteobagrus fulvidraco) master sex determination gene, pfpdz1. Differentially methylated regions have been identified on the promoter regions of cell signaling and embryonic development genes in Atlantic salmon fed diets of differing micronutrient concentrations [162], and associated with high- and sub-fertility in male striped bass [163], polyploidy and muscle atrophy in rainbow trout [164], and the immune response of eastern oyster following infection with the protistan parasite Perkinsus marinus [165]. In addition to the identification of core biomarkers that are opportunities for epigenetic selection, leveraging of environmental manipulation and epigenetic memory to improve phenotype can be directly employed to enhance aquaculture [157]. In some cases, improved performance has been realized in the absence of characterizing the precise epigenetic mechanism (e.g., Olympia oyster, Ostrea lurida [166]; Pacific geoduck, Panopea generosa [167]), whereas in other studies the functional role of DNA methylation in contributing carryover effects have been described (e.g., Pacific geoduck [168]). There has also been valuable work regarding the degree and genomic mechanism by which genetic and epigenetic variation are associated, providing insight into ecotoxicological pollution burdens [169] and how selection and manipulation can be used in the future to improve phenotypes (e.g., Olympia oyster [170]). The overall roles and the diversity of epigenetic mechanisms across major taxa and insights into their potential applications in the culture of aquatic animals are vast and have been recently reviewed [155,156,157,158].
Microbiomics and metagenomics studies in aquaculture have largely focused on two areas: (1) gut/intestinal microbiota with the aim of examining fish health, welfare, digestion, and immune response, and (2) environmental samples, with the aim of improving fish health and production efficiency by targeting environment-associated problems in aquaculture systems. The gut microbiome is crucial in maintaining good health, homeostasis, and metabolism by acting as a barrier against harmful bacteria and the production of vital vitamins and metabolites. Composition of the gut microbiome is considered an "intermediate phenotype" that results from host genetics and environmental influences [171], and is considered part of the whole genome, or "hologenome.” As the host's genetics significantly shape the gut microbiome [172], the term "microbiability" has been introduced to describe a concept similar to heritability. Awany et al. (2019) [173] reviewed the interaction between the host and the microbiome and its impact on the host's physical characteristics broadly. Since then, studies have demonstrated the association between rainbow trout gut microbiome, growth, and muscle percentage [174,175,176], and shellfish microbiome differences that influence growth [177] and survival against viral disease [178].
Investigating the microbial organisms present in aquaculture environments provides information on the diversity and composition of both beneficial and harmful microorganisms that can serve as indicators of environmental health, inform disease prevention and management strategies, influence nutrient cycling and waste management, and help optimize feed composition and regimes. Shotgun metagenomics and, more commonly, amplicon sequencing, have been utilized for microbiome profiling in various aquaculture production systems (RAS, or recirculating aquaculture systems; biological floc, or biofloc; and ponds) and for conservation purposes in areas with coexisting native fish. These studies have focused on identifying optimal sample preparation/sequencing methods [26, 179], evaluating the effects of different environmental parameters/conditions [180], and characterizing communities associated with different aquaculture systems [181,182,183]. Environmental DNA (eDNA) coupled with amplicon sequencing have been used to monitor abundances of fish pathogens [184] and invasive fish species [185]. Additionally, metagenomics coupled with metaproteomics has been used to describe the relative functional role of the microbial community in a bivalve hatchery under different conditions [186].
The integrative multi-omics approach of simultaneously analyzing multiple omics layers of a system, ranging from single cell to whole organism, provides a more comprehensive, or holistic, understanding of intricate molecular networks that govern biological processes. In agriculture, multi-omics approaches can enhance breeding by providing greater insight into the genetic and molecular basis of desirable traits and subsequently improving selection accuracy [187, 188]. Multi-omics approaches can similarly improve resource optimization by identifying key molecular factors that influence traits such as growth, disease resistance, and stress tolerance. There are many considerations for the integration of multi-omics data and interpretation of results as data generation and quantification methods differ between omics fields. These considerations include, but are not limited to, data standardization, cross-platform compatibility, data and metadata handling and completeness (e.g., missing, imbalanced, etc.), analysis (i.e., reduction) methods, data reporting, and biological validation approaches. Cross-platform compatibility is a significant challenge with ML analyses of multi-omics data, principally due to differences in data reporting and standardization which, if unaccounted for, can lead to biased statistical outputs [189,190,191]. Workflows and applications to integrate multi-omics data for analysis and/or visualization are continuously being developed as the number of technologies that facilitate the collection of these data and the accessibility thereof increase. Examples include: MOMIC [192], OmicsSuite [193], Bioconductor MultiAssayExperiment [194], IDARE2 (Integrated DAtanodes of REgulation) and Cytoscape [195, 196], multi-omics [197], and those listed in Chakraborty et al. (2022) [187]. Additionally, analytical pipelines based in AI/ML are continuously being developed to integrate these data types and address cross-platform issues as the models employed are not restricted in the same manner as traditional statistical approaches [198]. The suitability of these and other multi-omics integration tools must be evaluated case-by-case, as many have been developed using data generated from plant or model animal species and therefore may not sufficiently account for nuances of some aquaculture species (e.g., whole genome duplication). Further, the aquatic environment and effects thereof present an opportunity to incorporate additional data that is different from plants and terrestrial organisms. As such, developing tools specifically suited to aquaculture data and expanding our knowledge of them is a much-needed critical step forward.
The integration of multi-omics approaches in agricultural sciences is a paradigm shift, offering unprecedented insights into the complexity of biological systems. The holistic perspective of distilling these complex interactions into quantifiable factors associated with phenotypic variation advances basic science and holds tremendous promise for practical applications, such as breeding, genome editing, and improved selection accuracy using genome-enabled selection [199]. Establishing standards for data integration and the development of comprehensive workflows will greatly contribute to the robustness and reliability of multi-omics studies, further positioning them as indispensable tools in the pursuit of sustainable and efficient food production.
Aim 3: Developing resources for the collection and integration of phenomics data
Phenomics is the study of observable traits that contribute to the expression of a phenotype [200]. Phenotypic variation is equally, if not more, important to genetic selection programs than genetic and genomic data [199]. Recent advances in sequencing technologies have allowed the application of multiple genome-enabled selection strategies to be at the fingertips and keyboards of aquaculture scientists. While cost and technical laboratory challenges of genetic data production remain hurdles, they no longer represent the major barrier for advancing animal breeding. The most significant barrier is currently high-throughput phenotyping and the creation of multi-trait phenomes that can be quantitatively assessed [201].
Zebrafish (Danio rerio) lead all finfish in phenomics data resources (reviewed in Fuentes et al., 2018 [202]), and this species represents the exception within aquatic species rather than the rule. Zebrafish are a model species for biomedical researchers due to the ease of husbandry, short generation time, and fewer genome duplication events than other fishes. Understanding individual gene action to phenome expression is laborious, expensive, and development rates are dependent on the generation time of the animal (i.e., age to sexual maturation) and ease of gene manipulation. Some aquaculture species lend themselves to this model (e.g., tilapia, Oreochromis spp.), while others have longer generation times (e.g., salmonids, catfish) or are challenging to introduce gene modifications (e.g., shellfish). Thus, the development of phenomics datasets via gene knockout or gene modification is not likely to be widely applied within aquaculture species at scale until the discipline matures and technologies are available that can reliably and efficiently modify organisms at various life cycle stages (e.g., eggs/zygote).
An area where phenomics has the potential to significantly influence aquaculture is via automated, high-throughput data capture. Advances in this area are occurring through the combination of imaging and/or sensor technologies with AI/ML models being able to identify diseased individuals in intensive aquaculture settings [203], count individuals within an enclosure [204], produce real-time body size data [205], estimate biomass changes [206], signal an alert system based on detected mortality events [207], classify shape of an organism (e.g., oysters, [208]), determine feed conversion efficiency [209], predict readiness to spawn [210], identify shellfish gonads (e.g., Pacific oyster, [211]), monitor interactions between marine aquaculture animals and wild populations [177], and estimate moisture, glycogen, and ploidy in shellfish (e.g., eastern oyster, [212]), among numerous other applications [213]. These advances set the stage for production-level questions to be answered for entire lots of animals with potentially lower labor costs and fewer data collection errors (e.g., reduced subjectivity). Additionally, post-harvest traits of quality, such as seafood product freshness, may be assessed via e-systems (e.g., e-eye, e-nose, e-tongue) that quantitatively assess quality and represent a leap forward in data automation for product processing from farm to market [214, 215].
Multiple challenges exist for integrating phenomics data into aquaculture programs. Aquaculture occurs in environments harsh on equipment, and development of sensors and cameras that are able to withstand aquatic environments and extreme weather events is needed. Efforts to install infrastructure and implement strategies to obtain phenomics data from these sensors and cameras (e.g., via internet connection, collection of memory cards) will also have to be emphasized, as this is similarly challenging in aquatic environments. Moreover, accessible software capable of processing raw data into tangible datasets that can be used for phenomics trait production must also be developed. The scales and methods of aquaculture operations vary widely, with large programs (e.g., salmon and trout) being more likely to adopt and afford these high-throughput systems. Accessibility and democratization across program scale and affordability for smaller scale programs (e.g., shellfish, newly cultured finfish species, etc.) will be necessary for aquaculture as a discipline to reap the benefits of phenomics. Initial efforts in generating genomic and omics resources for newly cultured species, and others at similar stages of industry development, should focus on developing high-quality reference assemblies that will anchor the layers of omics and phenomics data.
While production may benefit from population or group level measurements, breeding efforts require integration of individual phenotypic variation with genomic data. This is especially challenging in aquaculture environments where animal tagging is difficult and/or expensive to conduct and maintain. PIT (passive integrated transponder) tags can be implanted, yet to date there are limited records of combining tag readers with non-lethal automated data collection [216,217,218]. Developing technology that can simultaneously identify individuals and collect phenotypic data is a frontier for advancing aquaculture phenomics (e.g., Babu et al., 2022 [204]). The rapid advancements that are occurring in genomics and multi-omics cannot be fully exploited without similar advancement in and integration with phenomics.
Aim 4: Creating pathways for applying and integrating genomics information across animal industries
The integration and application of genomics data across animal agriculture sectors plays a pivotal role in driving impactful changes and improvements in these industries. Specifically, sharing omics resources and strategies used in diverse animal industries facilitates the increased leveraging of these data and tools to benefit myriad groups, including academic and industry researchers, Extension specialists, producers, and other stakeholders. For example, resources for genome data mining and visualization such as AquaMine (http://aquamine.org; Elsik Lab, University of Missouri) and AgAnimalGenomes [219] enable the exploration of genomic and omics data within and between animal production sectors without requiring advanced training. These and similar data exploration tools lend themselves to powerful comparisons, which, among many other potential findings, can help identify conserved pathways between species that can be exploited to enhance production-relevant traits. The field of aquaculture is inherently and uniquely positioned to collaborate with neighboring fields like fisheries management, molecular ecology, and conservation genomics given the overlap of study species and experimental approaches used to answer fundamental questions. This interdisciplinary nature of aquaculture research can and should be expanded to other agriculture sectors as we collectively work towards supporting global food security.
The application and integration of data-based tools and resources within and across animal industries relies on the generation, maintenance, and provision of high-quality omics data and detailed metadata that meets agreed-upon standards of interpretability and availability. Several initiatives have developed such standards for genomic data and analysis, including the MIxS (Minimum Information about any (x) Sequence) checklist for metadata produced by the Genomics Standards Consortium (GSC) [220]; recommendations for analysis standards from the Earth BioGenome Project [221]; and published guidelines for protocols and data standards for multiple omics project types published by the ENCODE Consortium (Encyclopedia of DNA Elements) [222, 223]. Broader guidelines for scientific data reporting, such as FAIR Principles (Findable, Accessible, Interoperable, Reusable) [224], and tools that improve FAIRness of resources, such the Bioschemas effort to embed distinct markup into published data (e.g., genes, chemicals, proteins), workflows, and training programs [225,226,227,228], have also been developed and should be considered and incorporated into the determined set of data and metadata standards. The current aims of the latest iteration of the NRSP8,Footnote 3 Building Applied Genomic Capacity for Animal Industries, echo these sentiments.
Further, consensus on joint investment and participation of researchers, Extension specialists, agency personnel, and other stakeholders is necessary to best facilitate the sharing of feedback, ideas, and innovations. Funding organizations, institutions, and individuals with an interest in animal and plant production should prioritize hosting and/or participating in forums for feedback, discussion panels, workshops, etc., such that persons and groups invested in U.S. agriculture can collaborate. As examples, USDA NIFA accepts investigator-initiated proposals for conferences and workshops year round, and the Aquaculture Information Exchange,Footnote 4 an online forum to facilitate discussion between members of the public and private sectors with interests in U.S. aquaculture, was launched in 2023 by Virginia Sea Grant, with support from the National Oceanic and Atmospheric Administration (NOAA) National Sea Grant Office, NOAA Fisheries Office of Aquaculture, USDA Agricultural Research Service (ARS), and USDA NIFA. The Plant and Animal Genome Conference (PAG) held annually in San Diego, California served as a meeting place for those involved in or affiliated with the initial NRSP8 NAGRP, where interaction between agriculture sectors was nurtured and facilitated. Consistency to this end (i.e., location) provided an ease of knowing at least one avenue for involvement of academic and government researchers, students, industry professionals, and other contributors (Fig. 3). However, a single annual meeting is not sufficient for establishing long-term, productive relationships within and among animal sectors, especially when the strengths come from the diversity of species groups, analytical approaches, and stakeholders from private sector (industry), government, and academia. These interactions are invaluable for integrating new insights, technologies, and analysis types into agricultural practices that have real-world impacts. As such, support and energy behind collaborative efforts must continue, if not increase, with the ever-advancing omics- and data-intensive technologies to optimally and most efficiently leverage these tools toward the improvement and strengthening of U.S. agriculture.
Aim 5: Providing training, extension, and outreach to support the application of genome to phenome
“Genome to phenome” broadly refers to the linkage between the genetic blueprint (genome) and the observable traits of an organism (phenotypes, or phenome [3, 229, 230]. Research with this focus is designed to further our understanding of how molecular components and environmental factors influence complex phenotypes and is highly relevant to agriculture. Applying G2P research across agriculture sectors is critical to addressing food security challenges that continue to escalate with the growing global population, unpredictable and more frequent environmental impacts (e.g., hurricanes, floods, droughts, etc.), and biological events (e.g., emergent animal disease outbreaks, COVID-19 pandemic). Consortiums such as the Agricultural Genome to Phenome Initiative (AG2PI; https://www.ag2pi.org/) have been developed to address challenges and advance G2P research across agriculture sectors, including data sharing and integration, and the development of tools to evaluate and measure phenotypes [229, 230].
The establishment and continued support of programs that train early-career professionals with the requisite skills to navigate the complexities of genomics in agriculture and facilitate their integration into the workforce is paramount to achieving the ultimate goals of G2P research and delivering nutritious aquatic animal products to a growing global population. Specifically, this workforce must be efficacious in conducting and reporting scientific research, developing and commercializing tools, and communicating outcomes to farmers, consumers, legislators, and future generations. Educational programs that provide a foundational understanding of genomics, phenomics, and their integration into animal breeding and management practices must be developed and made broadly available at general and advanced levels to support the range of workforce personnel needs. An example is the interdisciplinary Genomics, Phenomics, and Bioinformatics (GPB) graduate (PhD and MS) program at North Dakota State University, which is describedFootnote 5 as:
“The program is designed to provide students the necessary skills and intellectual background to work cooperatively with others in a research area that take a systems-wide approach to the study of the organization of life and expression and regulation of genes in an organism. Students in the program will perform advanced study, training and research in areas that focus on functional genomics, high-throughput phenotyping, and computation analysis of genomic and phenomics data. Students will learn and master the multi-omics approaches for research in many frontiers. Exposure to modern techniques, instrumentation, computational and statistical methods will prepare the student for success in both industrial and academic careers."
Another example of this effort is the recently-funded (2024) USDA NIFA project “Graduate Education in Livestock Phenomics and Quantitative Genomics” awarded to Texas A&M University (Research, Education, and Economics Information System, REEIS, accession no. 1031671) that aims to “(1) recruit, (2) develop, and (3) produce outstanding graduates in the animal production targeted expertise shortage area who will become the next generation of leaders in animal breeding equipped with expertise in phenomics and associated quantitative genomics methods.”
Internships, work-study, and similar programs should be designed by or in collaboration with industry to ensure that early-career professionals are gaining relevant hands-on experience and, if possible, direct training for future positions. This can include partnerships with industry, or other relevant groups, such that successful participants who wish to pursue a career in the field can be placed efficiently into the workforce. There are several examples of these programs for plant crops, such as the USDA NIFA Research and Extension Experience for Undergraduates (REEU) program for Phenomics Big Data Management at Washington State University.Footnote 6 Similar programs should be established for animal/aquaculture production and include the spectrum of involved parties. For example, Extension personnel affiliated with U.S. Land-grant Colleges and Universities (1862, 1890, and 1994) and NOAA Sea Grant Programs are often the first line of communication with aquaculture producers who would like to apply genomics in their production systems. Therefore, inclusion of Extension personnel in research partnerships and training programs greatly expands opportunities for technology transfer to industry stakeholders as well as workforce development and professional relationship building.
Further, supporting the attendance and participation of early-career professionals at conferences, stakeholder meetings, workshops, etc. should also be emphasized as these opportunities allow for advanced and continued learning, sharing of ideas, and the development of professional relationships within and outside of each specific area or field of work/research.
The establishment and support of comprehensive training, Extension, and outreach programs are pivotal to achieving the goals of G2P research and to efficiently and effectively transfer developed technologies to the commercial aquaculture industry. By preparing a well-equipped workforce, fostering industry collaboration, and promoting continued professional development, we can enhance the productivity, sustainability, and resilience of agriculture to benefit both producers and consumers.
Future research priorities & directions
One thrust of the 2023 Workshop for Aquaculture Genomics, Genetics, and Breeding was to identify priorities for the next decade and beyond based upon what is anticipated to become the cutting edge of aquaculture omics research in the U.S. and projections of what will be needed to utilize such research to help meet the needs of the global population. Here we outline these ideas as a suggested guide to where U.S. aquaculture genetics, genomics and breeding research resources should be directed in the years ahead. It must also be acknowledged that global research advancement and international collaboration will only benefit aquaculture omics research and is one of the most efficacious ways to produce formidable advancement.
Genome-enabled selection models and applications
The identification of functional genes has the potential to rapidly advance U.S. aquaculture and this will be greatly facilitated by the inclusion of AI/ML approaches in analysis pipelines [123, 148, 213]. Genes underlying reproductive sterility, growth, stress, and disease resistance provide commercial breeders with essential data to produce animal populations better adapted to production environments and systems [4, 231]. Creating sterile populations could significantly enhance the social license of aquaculture to operate by significantly reducing, if not functionally eliminating, the potential for introgression between captive and wild/natural populations. While identifying traits underlain by simple genetic architectures and individual genes (or small subsets of genes) will advance the aquaculture breeding field, relatively few traits will be classified as such. The phenotypes targeted through selection (i.e., breeding) programs are often quantitative, complex traits that are influenced by a large number of loci with small effect sizes [4].
For traits with polygenic architecture, the continued development of GS workflows (from genotyping to statistical analysis) is vital [11, 232,233,234,235]. These traits are best suited for classical quantitative genetic analysis, using genome-wide measures of relationship between individuals, rather than focusing on shared genetic variation from a subset of chromosomal segments. Genome-enabled selection programs will become more common throughout aquaculture as genotyping costs decrease, sample throughput increases, and the potential benefits of improved selection accuracy are realized into tangible production outcomes (i.e., more rapid genetic gain) [236]. How quickly U.S. aquaculture breeding programs adopt GS is predicated on the availability of trained staff, funding, and reliable, affordable tools to genotype and phenotype animals.
Cost reduction is especially important in aquaculture programs [237], as there are high numbers of breeding candidates in a given population and the market value of each individual is low relative to other livestock animal industries. Continued development of inexpensive, high-throughput tools for genomic/omics data generation is needed. Our understanding of how large (i.e., total number of loci assayed) a genotyping panel must be to maximize GS accuracy is changing as studies have found that only hundreds to thousands of loci are needed to achieve highest accuracy [83, 90, 238, 239]. Panel size reduction may be possible using imputation-based methods [11] and can be combined with pedigree information [232] and microhaplotype variation [233, 240] to further reduce panel sizes via increased statistical power. It is likely that geneticists will soon be able to routinely produce the genome-level data required to maximize the accuracy of GS models. However, the advancement of GS application will be constrained by the cost of genotyping and a lack of efficient, scalable, and cost-effective phenotyping methods for aquaculture species.
Pangenomics
The substantial diversity within and among cultured species and production methods sets aquaculture uniquely apart from other major animal production industries. This is a challenge in the limited portability of tools across aquaculture species, but an opportunity is also present as the genetic variation within aquaculture species is primarily untapped. In this vein, the continued advancement of pangenomics in aquatic species is important to discover, utilize, and preserve the genetic diversity available to breeding programs. Pangenomics also stands to introduce new types of genetic variation into GWAS analyses (e.g., SVs, gene deletions) that may influence trait expression. Pangenomic studies in cattle revealed that deletions were associated with breeds and subspecies delineations [241]; in plants, pangenomic studies have identified disease tolerance and yield associated genes [242]. Livestock and poultry have been at the forefront of the domestic animal pangenomics field, yet despite being in their early stages, multiple studies have already identified genetic variants associated with production traits that could not be quantified using singular genomic resources [243]. For pangenomics to impact aquaculture breeding, it is likely that bespoke pangenomes will be necessary for individual operations, given the decentralized model of hatchery production and limited direct link between programs compared to livestock and plants. The degree to which this is necessary will depend on many factors, but largely be driven by evolutionary relationships among broodstocks used by independent hatchery suppliers. Ultimately, quantifying pangenome variation allows for more comprehensive assessment of trait variation and increases the potential to identify functional regions and/or genes [244]. The opportunity of pangenomic research in aquaculture is enticing and necessary, as variation among ecotypes within species and across the multitude of closely related species will provide substantial fodder for trait and phenotypic dissection.
Phenomes (traits) of the future
Defining and identifying phenomes of interest for aquaculture is needed, as the ability to produce and integrate phenotype data on a production scale, rather than the production of omics data, is presently limiting aquaculture advancement [3]. Aquaculture has historically focused on growth, reproduction/sexual dimorphism, and feed conversion as selection traits. Rapidly changing environments, dietary (feed) components, and pests and pathogens in production environments are shifting selection targets toward resiliency phenotypes [245, 246]. In rainbow trout and Nile tilapia, resilience was estimated via a body weight phenotype across multiple temporal and spatial scales (production environments) with resiliency found to be heritable [247] with significant variance among lines [248]. These studies empirically demonstrate resilience can be applied in selective breeding to increase uniformity in the face of disturbance. In eastern oyster, production in lower salinity coastal environments is influenced by intensifying seasonal precipitation and freshwater diversions from major rivers for coastline stability, thus resilience defined by tolerance to low salinity stress is emerging as a breeding target with progress made on utilizing GS to advance this trait [84, 249, 250]. Disease resistance against viral [251,252,253,254,255,256], bacterial [257, 258], and parasitic pathogens [135, 252, 259] constitutes a core theme of resiliency phenotypes as well [5, 260].
To this end, defining resilience for each species and production environment is crucial. For example, shellfish reared in an intertidal environment may have a unique set of resilience traits that differ from resilience-conferring traits in a finfish program sited in the same waterbody. Within a single species, it is possible that suites of traits that define resilience phenomes vary based upon location (environmental characteristics) and/or production environment/system (e.g., open water net pen vs. recirculating aquaculture system). A common resilience trait among aquaculture species is the ability to survive and efficiently grow in dynamic conditions with the potential ability to respond to different stressors over time. Interestingly, resilient phenomes of multiple livestock and aquaculture species reared in varied agricultural systems have been found to share stress-associated pathways that contribute significantly to the phenotypes [261]. Therefore, resilience phenomes among species may be more similar than we currently hypothesize. Overall, identifying and developing the phenomes of the future will require significant effort. Effective methods are likely to be those which identify animals with high growth rate, high feed conversion efficiency, and enhanced resistance to disease and environmental stress in variable conditions.
While there is great promise of integrating multiple data streams into a composite phenome, advancements on specific traits can be made and should be prioritized. For example, producing value-added, healthier human food through targeting specific fatty acids in salmonids. Omega-3 long-chain polyunsaturated fatty acids (LC-PUFA) are essential for the health of both humans and fish, particularly for heart and brain function. However, aquaculture-produced salmon must consume approximately 2% of their diet from unsustainable sources of wild fish oil to maintain sufficient levels of LC-PUFA [171]. In this case, an individual trait such as LC-PUFA synthesis may benefit from phenomics approaches and reveal unexpected relationships that can be targeted to yield genetic gain.
Education, training, and program participation
Investment in education, training, and retention initiatives is imperative to the continued operation, future success, and fortification of U.S. agriculture, including aquaculture. Programs, such as those described above, will create a well-equipped workforce capable of advancing genomic/omics research and its applications, subsequently leading to improved agricultural productivity, sustainability, and food security. This includes establishing and maintaining programs to recruit students and interns for training as future educators, researchers, Extension personnel, and industry leaders. Strengthening collaborative ties between research, Extension, industry, and other stakeholders through the creation and utilization of centralized communication networks and information forums is vital to moving the aquaculture industry forward. Participation at any level is necessary and strongly encouraged, whether through securing or providing funding, establishing training-to-workforce pipelines, organizing or leading workshops and meetings, or contributing in other ways (e.g., Extension, outreach, public education). Collectively, these efforts ensure ongoing commitment and “buy-in” from all stakeholders, maximizing impact and promoting the continuous advancement of aquaculture and agriculture.
Conclusion
The aquaculture genetics, genomics, and breeding community has made significant advancements over a short period, yet new and exciting challenges lie ahead. The next frontier in U.S. aquaculture production is to harness high-quality genome assemblies and integrate them with multi-omics and pangenomic approaches to distill the genetic variance associated with phenomes. However, it should not be overlooked that the expansion of U.S. aquaculture production via continual introduction of new species will require generation of novel genetic tools and the creation of robust phenotyping technologies. Unpredictable global events, such as those highlighted by the COVID-19 pandemic, have brought the importance of food security and food chain stability across the world into focus. The opportunity afforded by the U.S. aquaculture sector to help address food security is vast, and with continued development of genome-enabled breeding and genome editing strategies, omics integration, phenomics curation, and robust training, the future looks bright for the continued expansion of finfish, crustacean, and mollusk aquaculture in the United States.
Based on the present status of aquaculture genetics, genomics, and breeding research, and industry needs, the following areas should be prioritized to continue developing and strengthening the U.S. aquaculture sector:
-
1.
Improvement of reference genome assemblies: Produce genome sequences of newly cultured aquaculture species and continue advancing the assembly and functional annotation of existing reference genomes including epigenome annotation, Genotype-Tissue Expression (GTEx), eQTL and epi-QTL (expression and epigenetic quantitative trait locus, respectively) analyses.
-
2.
Advancement of pangenomics: Quantify and utilize all forms of genetic variation within and among species to advance G2P understanding and enhance breeding programs.
-
3.
Expansion of genome-enabled workflows: Develop cost-effective, scalable methods of selecting and utilizing genomic/omics data to increase selection accuracy and improve genome editing technologies to facilitate a greater functional understanding of G2P in aquaculture species. AI/ML will play a vital role in such workflows.
-
4.
Defining resilient phenomes: Use genomic/omics data to identify and integrate both production-relevant traits (e.g., growth, disease resistance) and traits associated with environmental stress tolerance across varied culture environments and production systems.
-
5.
Data integration and utilization: Adopt data and metadata standards to improve the accessibility and integration of multiple data types within and between species and to enhance applicable use of these data (e.g., comparative omics).
-
6.
Education and training programs: Invest in initiatives to train a skilled workforce capable of advancing and applying genomic technologies in the aquaculture sector.
Data availability
No datasets were generated or analysed during the current study.
The authors declare no competing interests.
Change history
17 March 2025
A Correction to this paper has been published: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-025-11447-7
Notes
NRSP8 NAGRP Bioinformatics web page: https://www.animalgenome.org/bioinfo/community/mission
AQUA-FAANG website: https://www.aqua-faang.eu/about-us.html
2023-2028 NRSP8 project website: https://nimss.org/projects/view/18969
Aquaculture Information Exchange registration page: https://aquainfoexchange.org/register/
North Dakota State University GPB program: https://catalog.ndsu.edu/programs-study/graduate/genomics-bioinformatics/
USDA NIFA REEU for Phenomics Big Data Management: https://labs.wsu.edu/sankaran-phenomics/undergrad-research-exp/
References
Biga P, Rexroad C, Bart A, Sullivan T, Green C, Fuglie K, Kim G, Surface J, Jermolowicz A, Thomason T et al: Aquaculture is agriculture colloquium: USDA's role in supporting farmers of fish, shellfish, and aquatic plants. In.: United States Department of Agriculture (USDA); 2021.
Abdelrahman H, ElHady M, Alcivar-Warren A, Allen S, Al-Tobasei R, Bao L, Beck B, Blackburn H, Bosworth B, Buchanan J, et al. Aquaculture genomics, genetics and breeding in the United States: current status, challenges, and priorities for future research. BMC Genomics. 2017;18:1–23. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-017-3557-1.
Rexroad C, Vallet J, Matukumalli LK, Reecy J, Bickhart D, Blackburn H, Boggess M, Cheng H, Clutter A, Cockett N. Genome to phenome: improving animal health, production, and well-being–a new USDA blueprint for animal genome research 2018–2027. Front Genet. 2019;10:327. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2019.00327.
Houston RD, Bean TP, Macqueen DJ, Gundappa MK, Jin YH, Jenkins TL, Selly SLC, Martin SAM, Stevens JR, Santos EM, et al. Harnessing genomics to fast-track genetic improvement in aquaculture. Nat Rev Genet. 2020;21(7):389–409. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41576-020-0227-y.
Huang Y, Li Z, Li M, Zhang X, Shi Q, Xu Z. Fish genomics and its application in disease-resistance breeding. Rev Aquac. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/raq.12973.
Yáñez JM, Barría A, López ME, Moen T, Garcia BF, Yoshida GM, Xu P. Genome-wide association and genomic selection in aquaculture. Rev Aquac. 2022;15(2):645–75. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/raq.12750.
Wang J, Cheng Y, Su B, Dunham RA. Genome manipulation advances in selected aquaculture organisms. Rev Aquac. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/raq.12988.
Hollenbeck CM, Johnston IA. Genomic tools and selective breeding in molluscs. Front Genet. 2018;9:253. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2018.00253.
Zenger KR, Khatkar MS, Jones DB, Khalilisamani N, Jerry DR, Raadsma HW. Genomic selection in aquaculture: Application, limitations and opportunities with special reference to marine shrimp and pearl oysters. Front Genet. 2018;9:693. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2018.00693.
Barría A, Benzie JAH, Houston RD, De Koning D-J, de Verdal H. Genomic selection and genome-wide association study for feed-efficiency traits in a farmed Nile Tilapia (Oreochromis niloticus) population. Front Genet. 2021;12:737906. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2021.737906.
Verbyla KL, Kube PD, Evans BS. Commercial implementation of genomic selection in Tasmanian Atlantic salmon: Scheme evolution and validation. Evol Appl. 2022;15(4):631–44. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/eva.13304.
Tsai H-Y, Hamilton A, Tinch AE, Guy DR, Gharbi K, Stear MJ, Matika O, Bishop SC, Houston RD. Genome wide association and genomic prediction for growth traits in juvenile farmed Atlantic salmon using a high density SNP array. BMC Genomics. 2015;16(1):969. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-015-2117-9.
Robledo D, Matika O, Hamilton A, Houston RD. Genome-wide association and genomic selection for resistance to amoebic gill disease in Atlantic salmon. G3 (Bethesda). 2018;8(4):1195–203. https://doiorg.publicaciones.saludcastillayleon.es/10.1534/g3.118.200075.
Vallejo RL, Leeds TD, Gao G, Parsons JE, Martin KE, Evenhuis JP, Fragomeni BO, Wiens GD, Palti Y. Genomic selection models double the accuracy of predicted breeding values for bacterial cold water disease resistance compared to a traditional pedigree-based model in rainbow trout aquaculture. Genet Sel Evol. 2017;49(1):17. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12711-017-0293-6.
Fraslin C, Koskinen H, Nousianen A, Houston RD, Kause A. Genome-wide association and genomic prediction of resistance to Flavobacterium columnare in a farmed rainbow trout population. Aquaculture. 2022;557(738332):738332. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2022.738332.
Yoshida GM, Bangera R, Carvalheiro R, Correa K, Figueroa R, Lhorente JP, Yáñez JM. Genomic prediction accuracy for resistance against Piscirickettsia salmonis in farmed rainbow trout. G3 (Bethesda). 2018;8(2):719–26. https://doiorg.publicaciones.saludcastillayleon.es/10.1534/g3.117.300499.
Wang H-P, Shen Z-G, Yao H, O'Bryant P, Rapp D. Sex determination and monosex female production in yellow perch. In: Sex Control in Aquaculture. Chichester: John Wiley & Sons, Ltd; 2018. p. 429–443. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/9781119127291.ch20.
Xu L, Zhao M, Ryu JH, Hayman ES, Fairgrieve WT, Zohar Y, Luckenbach JA, Wong T-T. Reproductive sterility in aquaculture: A review of induction methods and an emerging approach with application to Pacific Northwest finfish species. Rev Aquac. 2023;15(1):220–41. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/raq.12712.
Nam S-E, Bae D-Y, Ki J-S, Ahn C-Y, Rhee J-S. The importance of multi-omics approaches for the health assessment of freshwater ecosystems. Mol Cell Toxicol. 2023;19(1):3–11. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13273-022-00286-2.
Cusa M. St John Glew K, Trueman C, Mariani S, Buckley L, Neat F, Longo C: A future for seafood point-of-origin testing using DNA and stable isotope signatures. Rev Fish Biol Fish. 2022;32(2):597–621. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11160-021-09680-w.
Wei S, Yun B, Liu S, Ding T. Multiomics technology approaches in blue foods. Curr Opin Food Sci. 2022;45(100833):100833. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cofs.2022.100833.
Valenza-Troubat N, Hilario E, Montanari S, Morrison-Whittle P, Ashton D, Ritchie P, Wellenreuther M. Evaluating new species for aquaculture: A genomic dissection of growth in the New Zealand silver trevally (Pseudocaranx georgianus). Evol Appl. 2022;15(4):591–602. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/eva.13281.
Shivam S, El-Matbouli M, Kumar G. Development of fish parasite vaccines in the OMICs era: Progress and opportunities. Vaccines (Basel). 2021;9(2):179. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/vaccines9020179.
Bledsoe JW, Small BC. Finfish Microbiota and direct-fed microbial applications in aquaculture. In: Direct-Fed Microbials and Prebiotics for Animals. Cham: Springer International Publishing; 2023. p. 249–300. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/978-3-031-40512-9_10.
Older CE, Griffin MJ, Richardson BM, Waldbieser GC, Reifers JG, Goodman PM, Ware C, Gatlin DM 3rd, Wise DJ, Yamamoto FY. Influence of probiotic and prebiotic supplementation on intestinal microbiota and resistance to Edwardsiella ictaluri infection in channel catfish (Ictalurus punctatus) following florfenicol administration. J Fish Dis. 2024;47(4):e13910. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jfd.13910.
Older CE, Richardson BM, Wood M, Waldbieser GC, Ware C, Griffin MJ, Ott BD. Evaluating nanopore sequencing for microbial community characterization in catfish pond water. J World Aquaculture Soc. 2024;55(1):289–301. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jwas.13002.
Older CE, Yamamoto FY, Griffin MJ, Ware C, Heckman TI, Soto E, Bosworth BG, Waldbieser GC. Comparison of high-throughput sequencing methods for bacterial microbiota profiling in catfish aquaculture. N Am J Aquac. 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/naaq.10309.
Cai J, Chan HL, Yan X, Leung P. A global assessment of species diversification in aquaculture. Aquaculture. 2023;576(739837):739837. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2023.739837.
Bashura J, Burke M, Lapitan R, Mikulec J, Owens T, Reading BJ, Richt JA, Spencer D, Valdivia-Granda W, Weekes J et al. Treats to food and agricultural resources. In. Edited by United States DoHS, Office of Intelligence and Analysis (I&A) Public-Private Analytic Exchange Program (AEP); 2021.
Miga KH, Eichler EE. Envisioning a new era: complete genetic information from routine, telomere-to-telomere genomes. Am J Human Genet. 2023;110(11):1832–40. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ajhg.2023.09.011.
Flores A-M, Christensen KA, Campbell B, Koop BF, Taylor JS. Sablefish (Anoplopoma fimbria) chromosome-level genome assembly. G3: Genes, Genomes, Genetics 2023;13(7). https://doiorg.publicaciones.saludcastillayleon.es/10.1093/g3journal/jkad089.
Penaloza C, Gutierrez AP, Eöry L, Wang S, Guo X, Archibald AL, Bean TP, Houston RD. A chromosome-level genome assembly for the Pacific oyster Crassostrea gigas. GigaScience. 2021;10(3):giab020. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/gigascience/giab020.
Mrowicki R, Uhl R. The genome sequence of the Pacific oyster, Magallana gigas (Thunberg, 1793). Wellcome Open Res. 2024;9:284. https://doiorg.publicaciones.saludcastillayleon.es/10.12688/wellcomeopenres.22255.1.
Puritz JB, Guo X, Hare M, He Y, Hillier LW, Jin S, Liu M, Lotterhos KE, Minx P, Modak T. A second unveiling: Haplotig masking of the eastern oyster genome improves population-level inference. Mol Ecol Resour. 2024;24(1):e13801. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/1755-0998.13801.
Gao G, Waldbieser GC, Youngblood RC, Zhao D, Pietrak MR, Allen MS, Stannard JA, Buchanan JT, Long RL, Milligan M. The generation of the first chromosome-level de novo genome assembly and the development and validation of a 50K SNP array for the St. John River aquaculture strain of North American Atlantic salmon. G3: Genes, Genomes, Genetics. 2023;13(9):jkad138. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/g3journal/jkad138.
Etherington GJ, Nash W, Ciezarek A, Mehta TK, Barria A, Peñaloza C, Khan MGQ, Durrant A, Forrester N, Fraser F et al. Chromosome-level genome sequence of the Genetically Improved Farmed Tilapia (GIFT, Oreochromis niloticus) highlights regions of introgression with O. mossambicus. BMC Genomics. 2022;23(1):832. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-022-09065-8.
Peng M, Chen X, Yang C, Liu Q, Li Q, Zhang B, Wang H, Zhu W, Feng P, Zeng D, et al. A high-quality genome assembly of the Pacific white shrimp (Litopenaeus vannamei) provides insights into its evolution and adaptation. Aquac Rep. 2023;33(101859):101859. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aqrep.2023.101859.
Gao G, Magadan S, Waldbieser GC, Youngblood RC, Wheeler PA, Scheffler BE, Thorgaard GH, Palti Y. A long reads-based de-novo assembly of the genome of the Arlee homozygous line reveals chromosomal rearrangements in rainbow trout. G3 Genes|Genomes|Genetics. 2021;11(4). https://doiorg.publicaciones.saludcastillayleon.es/10.1093/g3journal/jkab052.
Liu Z, Liu S, Yao J, Bao L, Zhang J, Li Y, Jiang C, Sun L, Wang R, Zhang Y, et al. The channel catfish genome sequence provides insights into the evolution of scale formation in teleosts. Nat Comm. 2016;7(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ncomms11757.
Waldbieser GC, Liu S, Yuan Z, Older CE, Gao D, Shi C, Bosworth BG, Li N, Bao L, Kirby MA, et al. Reference genomes of channel catfish and blue catfish reveal multiple pericentric chromosome inversions. BMC Biol. 2023;21(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12915-023-01556-8.
Wang H, Su B, Butts IAE, Dunham RA, Wang X. Chromosome-level assembly and annotation of the blue catfish Ictalurus furcatus, an aquaculture species for hybrid catfish reproduction, epigenetics, and heterosis studies. GigaScience. 2022;11. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/gigascience/giac070.
Pettersson ME, Rochus CM, Han F, Chen J, Hill J, Wallerman O, Fan G, Hong X, Xu Q, Zhang H, et al. A chromosome-level assembly of the Atlantic herring genome—detection of a supergene and other signals of selection. Genome Res. 2019;29(11):1919–28. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/gr.253435.119.
Mérot C, Stenløkk KSR, Venney C, Laporte M, Moser M, Normandeau E, Árnyasi M, Kent M, Rougeux C, Flynn JM, et al. Genome assembly, structural variants, and genetic differentiation between lake whitefish young species pairs (Coregonus sp.) with long and short reads. Mol Ecol. 2022;32(6):1458–77. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/mec.16468.
Gómez-Chiarri M, Warren WC, Guo X, Proestou D. Developing tools for the study of molluscan immunity: The sequencing of the genome of the eastern oyster. Crassostrea virginica. Fish & Shellfish Immunology. 2015;46(1):2–4. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.fsi.2015.05.004.
Rondeau EB, Minkley DR, Leong JS, Messmer AM, Jantzen JR, von Schalburg KR, Lemon C, Bird NH, Koop BF. The Genome and Linkage Map of the Northern Pike (Esox lucius): Conserved Synteny Revealed between the Salmonid Sister Group and the Neoteleostei. PLoS ONE. 2014;9(7):e102089. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0102089.
Rhie A, McCarthy SA, Fedrigo O, Damas J, Formenti G, Koren S, Uliano-Silva M, Chow W, Fungtammasan A, Kim J, et al. Towards complete and error-free genome assemblies of all vertebrate species. Nature. 2021;592(7856):737–46. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-021-03451-0.
Reid BN, Moran RL, Kopack CJ, Fitzpatrick SW. Rapture-ready darters: Choice of reference genome and genotyping method (whole-genome or sequence capture) influence population genomic inference in Etheostoma. Mol Ecol Resour. 2020;21(2):404–20. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/1755-0998.13275.
Tørresen OK, Star B, Jentoft S, Reinar WB, Grove H, Miller JR, Walenz BP, Knight J, Ekholm JM, Peluso P, et al. An improved genome assembly uncovers prolific tandem repeats in Atlantic cod. BMC Genomics. 2017;18(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-016-3448-x.
Griffiths JS, Sahasrabudhe RM, Marimuthu MPA, Chumchim N, Nguyen OH, Beraut E, Escalona M, Whitehead A. A draft reference genome of the red abalone, Haliotis rufescens, for conservation genomics. J Hered. 2022;113(6):673–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/jhered/esac047.
Einfeldt AL, Kess T, Messmer A, Duffy S, Wringe BF, Fisher J, den Heyer C, Bradbury IR, Ruzzante DE, Bentzen P. Chromosome level reference of Atlantic halibut Hippoglossus hippoglossus provides insight into the evolution of sexual determination systems. Mol Ecol Resour. 2021;21(5):1686–96. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/1755-0998.13369.
Jasonowicz AJ, Simeon A, Zahm M, Cabau C, Klopp C, Roques C, Iampietro C, Lluch J, Donnadieu C, Parrinello H, et al. Generation of a chromosome-level genome assembly for Pacific halibut (Hippoglossus stenolepis) and characterization of its sex-determining genomic region. Mol Ecol Resour. 2022;22(7):2685–700. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/1755-0998.13641.
Polinski JM, Zimin AV, Clark KF, Kohn AB, Sadowski N, Timp W, Ptitsyn A, Khanna P, Romanova DY, Williams P, et al. The American lobster genome reveals insights on longevity, neural, and immune adaptations. Sci Adv. 2021;7(26). https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.abe8290.
Braasch I, Gehrke AR, Smith JJ, Kawasaki K, Manousaki T, Pasquier J, Amores A, Desvignes T, Batzel P, Catchen J, et al. The spotted gar genome illuminates vertebrate evolution and facilitates human-teleost comparisons. Nat Genet. 2016;48(4):427–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ng.3526.
Song H, Guo X, Sun L, Wang Q, Han F, Wang H, Wray GA, Davidson P, Wang Q, Hu Z, et al. The hard clam genome reveals massive expansion and diversification of inhibitors of apoptosis in Bivalvia. BMC Biol. 2021;19(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12915-020-00943-9.
Farhat S, Bonnivard E, Pales Espinosa E, Tanguy A, Boutet I, Guiglielmoni N, Flot J-F, Allam B. Comparative analysis of the Mercenaria mercenaria genome provides insights into the diversity of transposable elements and immune molecules in bivalve mollusks. BMC Genomics. 2022;23(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-021-08262-1.
Sun C, Li J, Dong J, Niu Y, Hu J, Lian J, Li W, Li J, Tian Y, Shi Q, et al. Chromosome-level genome assembly for the largemouth bass Micropterus salmoides provides insights into adaptation to fresh and brackish water. Mol Ecol Resour. 2020;21(1):301–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/1755-0998.13256.
Sun B, Huang Y, Castro LFC, Yang S, Huang S, Jin W, Zhou H, Ijiri S, Luo Y, Gao J, et al. The chromosome-level genome and key genes associated with mud-dwelling behavior and adaptations of hypoxia and noxious environments in loach (Misgurnus anguillicaudatus). BMC Biol. 2023;21(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12915-023-01517-1.
Shekhar MS, Katneni VK, Jangam AK, Krishnan K, Prabhudas SK, Jani Angel JR, Sukumaran K, Kailasam M, Jena J. First Report of Chromosome-Level Genome Assembly for Flathead Grey Mullet, Mugil cephalus (Linnaeus, 1758). Front Genet. 2022;13. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2022.911446.
Christensen KA, Rondeau EB, Sakhrani D, Biagi CA, Johnson H, Joshi J, Flores A-M, Leelakumari S, Moore R, Pandoh PK, et al. The pink salmon genome: Uncovering the genomic consequences of a two-year life cycle. PLoS ONE. 2021;16(12):e0255752. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0255752.
Rondeau EB, Christensen KA, Sakhrani D, Biagi CA, Wetklo M, Johnson HA, Despins CA, Leggatt RA, Minkley DR, Withler RE, et al. Genome assembly, transcriptome and SNP database for chum salmon (Oncorhynchus keta). In.: Cold Spring Harbor Laboratory; 2021. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/2021.12.27.474290.
Rondeau EB, Christensen KA, Johnson HA, Sakhrani D, Biagi CA, Wetklo M, Despins CA, Leggatt RA, Minkley DR, Withler RE, et al. Insights from a chum salmon (Oncorhynchus keta) genome assembly regarding whole-genome duplication and nucleotide variation influencing gene function. G3 (Bethesda). 2023;13(8). https://doiorg.publicaciones.saludcastillayleon.es/10.1093/g3journal/jkad127.
Kim J-H, Leong JS, Koop BF, Devlin RH. Multi-tissue transcriptome profiles for coho salmon (Oncorhynchus kisutch), a species undergoing rediploidization following whole-genome duplication. Mar Genomics. 2016;25:33–7. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.margen.2015.11.008.
Christensen KA, Rondeau EB, Minkley DR, Sakhrani D, Biagi CA, Flores A-M, Withler RE, Pavey SA, Beacham TD, Godin T, et al. The sockeye salmon genome, transcriptome, and analyses identifying population defining regions of the genome. PLoS ONE. 2020;15(10):e0240935. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0240935.
Christensen KA, Leong JS, Sakhrani D, Biagi CA, Minkley DR, Withler RE, Rondeau EB, Koop BF, Devlin RH. Chinook salmon (Oncorhynchus tshawytscha) genome and transcriptome. PLoS ONE. 2018;13(4):e0195461. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0195461.
Conte MA, Joshi R, Moore EC, Nandamuri SP, Gammerdinger WJ, Roberts RB, Carleton KL, Lien S, Kocher TD. Chromosome-scale assemblies reveal the structural evolution of African cichlid genomes. GigaScience. 2019;8(4). https://doiorg.publicaciones.saludcastillayleon.es/10.1093/gigascience/giz030.
Gundappa MK, Peñaloza C, Regan T, Boutet I, Tanguy A, Houston RD, Bean TP, Macqueen DJ. Chromosome‐level reference genome for European flat oyster (Ostrea edulis L.). Evol Appl. 2022;15(11):1713–29. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/eva.13460.
Van Quyen D, Gan HM, Lee YP, Nguyen DD, Nguyen TH, Tran XT, Nguyen VS, Khang DD, Austin CM. Improved genomic resources for the black tiger prawn (Penaeus monodon). Mar Genomics. 2020;52:100751. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.margen.2020.100751.
Zhang X, Yuan J, Sun Y, Li S, Gao Y, Yu Y, Liu C, Wang Q, Lv X, Zhang X et al. Penaeid shrimp genome provides insights into benthic adaptation and frequent molting. Nat Comm. 2019;10(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-018-08197-4.
Feron R, Zahm M, Cabau C, Klopp C, Roques C, Bouchez O, Eché C, Valière S, Donnadieu C, Haffray P, et al. Characterization of a Y-specific duplication/insertion of the anti-Mullerian hormone type II receptor gene based on a chromosome-scale genome assembly of yellow perch Perca flavescens. Mol Ecol Resour. 2020;20(2):531–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/1755-0998.13133.
Xu Z, Gao T, Xu Y, Li X, Li J, Lin H, Yan W, Pan J, Tang J. A chromosome-level reference genome of red swamp crayfish Procambarus clarkii provides insights into the gene families regarding growth or development in crustaceans. Genomics. 2021;113(5):3274–84. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ygeno.2021.07.017.
Xu R, Martelossi J, Smits M, Iannello M, Peruzza L, Babbucci M, Milan M, Dunham JP, Breton S, Milani L, et al. Multi-tissue RNA-Seq Analysis and Long-read-based Genome Assembly Reveal Complex Sex-specific Gene Regulation and Molecular Evolution in the Manila Clam. Genome Biol Evol. 2022;14(12). https://doiorg.publicaciones.saludcastillayleon.es/10.1093/gbe/evac171.
Lien S, Koop BF, Sandve SR, Miller JR, Kent MP, Nome T, Hvidsten TR, Leong JS, Minkley DR, Zimin A, et al. The Atlantic salmon genome provides insights into rediploidization. Nature. 2016;533(7602):200–5. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nature17164.
Pasquier J, Cabau C, Nguyen T, Jouanno E, Severac D, Braasch I, Journot L, Pontarotti P, Klopp C, Postlethwait JH, et al. Gene evolution and gene expression after whole genome duplication in fish: the PhyloFish database. BMC Genomics. 2016;17(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-016-2709-z.
Araki K. Aokic J-y, Kawase J, Hamada K, Ozaki A, Fujimoto H, Yamamoto I, Usuki H: Whole Genome Sequencing of Greater Amberjack (Seriola dumerili) for SNP Identification on Aligned Scaffolds and Genome Structural Variation Analysis Using Parallel Resequencing. International Journal of Genomics. 2018;2018:1–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1155/2018/7984292.
Purcell CM, Seetharam AS, Snodgrass O, Ortega-García S, Hyde JR, Severin AJ. Insights into teleost sex determination from the Seriola dorsalis genome assembly. BMC Genomics. 2018;19(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-017-4403-1.
Ramesh B, Small CM, Healey H, Johnson B, Barker E, Currey M, Bassham S, Myers M, Cresko WA, Jones AG. Improvements to the Gulf pipefish Syngnathus scovelli genome. Gigabyte. 2023;2023:1–11. https://doiorg.publicaciones.saludcastillayleon.es/10.46471/gigabyte.76.
Ciezarek AG, Dunning LT, Jones CS, Noble LR, Humble E, Stefanni SS, Savolainen V. Substitutions in the Glycogenin-1 Gene Are Associated with the Evolution of Endothermy in Sharks and Tunas. Genome Biol Evol. 2016;8(9):3011–21. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/gbe/evw211.
Wu B, Feng C, Zhu C, Xu W, Yuan Y, Hu M, Yuan K, Li Y, Ren Y, Zhou Y, et al. The Genomes of Two Billfishes Provide Insights into the Evolution of Endothermy in Teleosts. Mol Biol Evol. 2021;38(6):2413–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/molbev/msab035.
Liu S, Martin KE, Snelling WM, Long R, Leeds TD, Vallejo RL, Wiens GD, Palti Y. Accurate genotype imputation from low-coverage whole-genome sequencing data of rainbow trout. G3: Genes, Genomes, Genetics 2024;14(9):jkae168. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/g3journal/jkae168.
Pearse DE, Barson NJ, Nome T, Gao G, Campbell MA, Abadía-Cardoso A, Anderson EC, Rundio DE, Williams TH, Naish KA, et al. Sex-dependent dominance maintains migration supergene in rainbow trout. Nat Ecol Evol. 2019;3(12):1731–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41559-019-1044-6.
Liu S, Gao G, Layer RM, Thorgaard GH, Wiens GD, Leeds TD, Martin KE, Palti Y. Identification of high-confidence structural variants in domesticated rainbow trout using whole-genome sequencing. Front Genet. 2021;12. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2021.639355.
Modak TH, Literman R, Puritz JB, Johnson KM, Roberts EM, Proestou D, Guo X, Gomez-Chiarri M, Schwartz RS. Extensive genome-wide duplications in the eastern oyster (Crassostrea virginica). Philos Trans R Soc Lond B Biol Sci. 1825;2021(376):20200164. https://doiorg.publicaciones.saludcastillayleon.es/10.1098/rstb.2020.0164.
Gutierrez AP, Bean TP, Hooper C, Stenton CA, Sanders MB, Paley RK, Rastas P, Bryrom M, Matika O, Houston RD. A genome-wide association study for host resistance to ostreid herpesvirus in Pacific oysters (Crassostrea gigas). G3: Genes, Genomes, Genetics. 2018;8(4):1273–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1534/g3.118.200113.
McCarty AJ, Allen Jr SK, Plough LV. Genome-wide analysis of acute low salinity tolerance in the eastern oyster Crassostrea virginica and potential of genomic selection for trait improvement. G3. 2022;12(1):jkab368. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/g3journal/jkab368.
Li N, Zhou T, Geng X, Jin Y, Wang X, Liu S, Xu X, Gao D, Li Q, Liu Z. Identification of novel genes significantly affecting growth in catfish through GWAS analysis. Mol Genet Genomics. 2018;293:587–99. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00438-017-1406-1.
Reis Neto RV, Yoshida GM, Lhorente JP, Yáñez JM. Genome-wide association analysis for body weight identifies candidate genes related to development and metabolism in rainbow trout (Oncorhynchus mykiss). Mol Genet Genomics. 2019;294:563–71. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00438-018-1518-2.
Sánchez-Roncancio C, García B, Gallardo-Hidalgo J, Yáñez JM. GWAS on imputed whole-genome sequence variants reveal genes associated with resistance to Piscirickettsia salmonis in rainbow trout (Oncorhynchus mykiss). Genes. 2022;14(1):114. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/genes14010114.
Tan S, Zhou T, Wang W, Jin Y, Wang X, Geng X, Luo J, Yuan Z, Yang Y, Shi H. GWAS analysis using interspecific backcross progenies reveals superior blue catfish alleles responsible for strong resistance against enteric septicemia of catfish. Mol Genet Genomics. 2018;293:1107–20. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00438-018-1443-4.
Ahmed RO, Ali A, Al-Tobasei R, Leeds T, Kenney B, Salem M. Weighted single-step GWAS identifies genes influencing fillet color in rainbow trout. Genes. 2022;13(8):1331. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/genes13081331.
Al-Tobasei R, Ali A, Garcia AL, Lourenco D, Leeds T, Salem M. Genomic predictions for fillet yield and firmness in rainbow trout using reduced-density SNP panels. BMC Genomics. 2021;22:1–11. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-021-07404-9.
Salem M, Al-Tobasei R, Ali A, Lourenco D, Gao G, Palti Y, Kenney B, Leeds TD. Genome-wide association analysis with a 50K transcribed gene SNP-chip identifies QTL affecting muscle yield in rainbow trout. Front Genet. 2018;9:387. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2018.00387.
Vallejo RL, Liu S, Gao G, Fragomeni BO, Hernandez AG, Leeds TD, Parsons JE, Martin KE, Evenhuis JP, Welch TJ, et al. Similar genetic architecture with shared and unique quantitative trait loci for bacterial cold water disease resistance in two rainbow trout breeding populations. Front Genet. 2017;8. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2017.00156.
Vela-Avitúa S, LaFrentz BR, Lozano CA, Shoemaker CA, Ospina-Arango JF, Beck BH, Rye M. Genome-wide association study for Streptococcus iniae in Nile tilapia (Oreochromis niloticus) identifies a significant QTL for disease resistance. Front Genet. 2023;14:1078381. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2023.1078381.
Su B, Shang M, Grewe PM, Patil JG, Peatman E, Perera DA, Cheng Q, Li C, Weng C-C, Li P. Suppression and restoration of primordial germ cell marker gene expression in channel catfish, Ictalurus punctatus, using knockdown constructs regulated by copper transport protein gene promoters: Potential for reversible transgenic sterilization. Theriogenology. 2015;84(9):1499–512. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.theriogenology.2015.07.037.
Elaswad A, Khalil K, Ye Z, Liu Z, Liu S, Peatman E, Odin R, Vo K, Drescher D, Gosh K. Effects of CRISPR/Cas9 dosage on TICAM1 and RBL gene mutation rate, embryonic development, hatchability and fry survival in channel catfish. Sci Rep. 2018;8(1):16499. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-018-34738-4.
Khalil K, Elaswad A, Abdelrahman H, Michel M, Chen W, Liu S, Odin R, Ye Z, Drescher D, Vo K. Editing the melanocortin-4 receptor gene in channel catfish using the CRISPR-cas9 system. Fishes. 2023;8(2):116. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/fishes8020116.
Wargelius A, Leininger S, Skaftnesmo KO, Kleppe L, Andersson E, Taranger GL, Schulz RW, Edvardsen RB. Dnd knockout ablates germ cells and demonstrates germ cell independent sex differentiation in Atlantic salmon. Sci Rep. 2016;6(1):21284. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/srep21284.
Cleveland BM, Yamaguchi G, Radler LM, Shimizu M. Editing the duplicated insulin-like growth factor binding protein-2b gene in rainbow trout (Oncorhynchus mykiss). Sci Rep. 2018;8(1):16054. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-018-34326-6.
Coogan M, Alston V, Su B, Khalil K, Elaswad A, Khan M, Simora RM, Johnson A, Xing D, Li S. CRISPR/Cas-9 induced knockout of myostatin gene improves growth and disease resistance in channel catfish (Ictalurus punctatus). Aquaculture. 2022;557:738290. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2022.738290.
Datsomor AK, Olsen RE, Zic N, Madaro A, Bones AM, Edvardsen RB, Wargelius A, Winge P. CRISPR/Cas9-mediated editing of Δ5 and Δ6 desaturases impairs Δ8-desaturation and docosahexaenoic acid synthesis in Atlantic salmon (Salmo salar L.). Sci Rep. 2019;9(1):16888. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-019-53316-w.
Jiang D, Chen J, Fan Z, Tan D, Zhao J, Shi H, Liu Z, Tao W, Li M, Wang D. CRISPR/Cas9-induced disruption of wt1a and wt1b reveals their different roles in kidney and gonad development in Nile tilapia. Dev Biol. 2017;428(1):63–73. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ydbio.2017.05.017.
Kishimoto K, Washio Y, Yoshiura Y, Toyoda A, Ueno T, Fukuyama H, Kato K, Kinoshita M. Production of a breed of red sea bream Pagrus major with an increase of skeletal muscle mass and reduced body length by genome editing with CRISPR/Cas9. Aquaculture. 2018;495:415–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2018.05.055.
Raudstein M, Straume AH, Kjærner-Semb E, Barvik M, Ellingsen S, Edvardsen RB. Highly efficient in vivo C-to-T base editing in Atlantic salmon (Salmo salar)–A step towards aquaculture precision breeding. Aquaculture. 2024;581:740487. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2023.740487.
Wang C, Xu J, Kocher TD, Li M, Wang D. CRISPR knockouts of pmela and pmelb engineered a golden tilapia by regulating relative pigment cell abundance. J Hered. 2022;113(4):398–413. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/jhered/esac018.
López-Porras A, Berg RS, Burgerhout E, Hansen ØJ, Györkei Á, Qiao S-W, Johansen F-E. CRISPR-Cas9/Cas12a-based genome editing in Atlantic cod (Gadus morhua). Aquaculture. 2024;581:740440. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2023.740440.
Kim J, Cho JY, Kim J-W, Kim H-C, Noh JK, Kim Y-O, Hwang H-K, Kim W-J, Yeo S-Y, An CM. CRISPR/Cas9-mediated myostatin disruption enhances muscle mass in the olive flounder Paralichthys olivaceus. Aquaculture. 2019;512:734336. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2019.734336.
Ohama M, Washio Y, Kishimoto K, Kinoshita M, Kato K. Growth performance of myostatin knockout red sea bream Pagrus major juveniles produced by genome editing with CRISPR/Cas9. Aquaculture. 2020;529: 735672. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2020.735672.
Khalil K, Elayat M, Khalifa E, Daghash S, Elaswad A, Miller M, Abdelrahman H, Ye Z, Odin R, Drescher D. Generation of myostatin gene-edited channel catfish (Ictalurus punctatus) via zygote injection of CRISPR/Cas9 system. Sci Rep. 2017;7(1):7301. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-017-07223-7.
Wang J, Su B, Xing D, Bruce TJ, Li S, Bern L, Shang M, Johnson A, Simora RMC, Coogan M. Generation of eco-friendly and disease-resistant channel catfish (Ictalurus punctatus) harboring the alligator cathelicidin gene via CRISPR/Cas9 engineering. Engineering. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.eng.2023.12.005.
Güralp H, Skaftnesmo KO, Kjærner-Semb E, Straume AH, Kleppe L, Schulz RW, Edvardsen RB, Wargelius A. Rescue of germ cells in dnd crispant embryos opens the possibility to produce inherited sterility in Atlantic salmon. Sci Rep. 2020;10(1):18042. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-020-74876-2.
Jin YH, Liao B, Migaud H, Davie A. Physiological impact and comparison of mutant screening methods in piwil2 KO founder Nile tilapia produced by CRISPR/Cas9 system. Sci Rep. 2020;10(1):12600. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-020-69421-0.
Mokrani A, Liu S. Harnessing CRISPR/Cas9 system to improve economic traits in aquaculture species. Aquaculture. 2024;579:740279. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2023.740279.
Wray-Cahen D, Bodnar A, Rexroad C, Siewerdt F, Kovich D. Advancing genome editing to improve the sustainability and resiliency of animal agriculture. CABI Agriculture and Bioscience. 2022;3(1):1–17. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s43170-022-00091-w.
Copper S. Following the Framework: Intentional Genomic Alterations in Animals. J Food L & Pol’y. 2022;18:117.
Rudenko L, Plunkett LM, Kornum A, Röcklinsberg H, Sørensen DB, Gjerris M. An overview of the regulation of genetically altered animals in research. Biotech Animals Res. 2024:23–72.
Van Eenennaam AL, De Figueiredo SF, Trott JF, Zilberman D. Genetic engineering of livestock: the opportunity cost of regulatory delay. Annual Review of Animal Biosciences. 2021;9(1):453–78. https://doiorg.publicaciones.saludcastillayleon.es/10.1146/annurev-animal-061220-023052.
Watson O, Hayta S. Precision breeding in agriculture and food systems in the United Kingdom. Transgenic Res. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s11248-024-00397-7.
Lemay M-A, Malle S. A practical guide to using structural variants for genome-wide association studies. In: Methods in Molecular Biology. Springer US; 2022. p. 161–172. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/978-1-0716-2237-7_10.
Ruigrok M, Xue B, Catanach A, Zhang M, Jesson L, Davy M, Wellenreuther M. The relative power of structural genomic variation versus SNPs in explaining the quantitative trait growth in the marine teleost Chrysophrys auratus. Genes. 2022;13(7):1129. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/genes13071129.
Bertolotti AC, Layer RM, Gundappa MK, Gallagher MD, Pehlivanoglu E, Nome T, Robledo D, Kent MP, Røsæg LL, Holen MM, et al. The structural variation landscape in 492 Atlantic salmon genomes. Nat Comm. 2020;11(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41467-020-18972-x.
Lecomte L, Árnyasi M, Ferchaud AL, Kent M, Lien S, Stenløkk K, Sylvestre F, Bernatchez L, Mérot C. Investigating structural variant, indel and single nucleotide polymorphism differentiation between locally adapted Atlantic salmon populations. Evol Appl. 2024;17(3). https://doiorg.publicaciones.saludcastillayleon.es/10.1111/eva.13653.
Gerdol M, Moreira R, Cruz F, Gómez-Garrido J, Vlasova A, Rosani U, Venier P, Naranjo-Ortiz MA, Murgarella M, Balseiro P, et al. Massive gene presence/absence variation in the mussel genome as an adaptive strategy: first evidence of a pan-genome in Metazoa. In.: Cold Spring Harbor Laboratory; 2019. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/781377.
Johnston IA, Kent MP, Boudinot P, Looseley M, Bargelloni L, Faggion S, Merino GA, Ilsley GR, Bobe J, Tsigenopoulos CS. Advancing fish breeding in aquaculture through genome functional annotation. Aquaculture. 2024:740589. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2024.740589.
Salem M, Al-Tobasei R, Ali A, An L, Wang Y, Bai X, Bi Y. H Z: Functional annotation of regulatory elements in rainbow trout uncovers roles of the epigenome in genetic selection and genome evolution. GigaScience. 2025;13:1–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/gigascience/giae092.
NOAA. NOAA Aquaculture Strategic Plan (2023–2028). In: National Oceanic and Atmospheric Administration (NOAA); 2022.
Aquaculture NSaTCSo: A National Strategic Plan for Aquaculture Research. In.: National Science and Technology Council Subcommittee on Aquaculture; 2022.
Rexroad C, Rust MB, Riche M, Wills P, Davis M. Opportunities for US marine finfish aquaculture. J World Aquaculture Soc. 2021;52(3):501–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jwas.12791.
Froehlich HE, Gentry RR, Lester SE, Rennick M, Lemoine HR, Tapia-Lewin S, Gardner L. Piecing together the data of the US marine aquaculture puzzle. J Environ Manage. 2022;308:114623. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jenvman.2022.114623.
Kumar G, Hegde S, van Senten J, Engle C, Boldt N, Parker M, Quagrainie K, Posadas B, Asche F, Dey M. Economic contribution of US aquaculture farms. J World Aquacul Soc. 2024:e13091. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jwas.13091.
Yang Y, Fu Q, Wang X, Liu Y, Zeng Q, Li Y, Gao S, Bao L, Liu S, Gao D. Comparative transcriptome analysis of the swimbladder reveals expression signatures in response to low oxygen stress in channel catfish Ictalurus punctatus. Physiol Genomics. 2018;50(8):636–47. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/physiolgenomics.00125.2017.
Ott BD, Hulse-Kemp AM, Duke MV, Griffin MJ, Peterson BC, Scheffler BE, Torrans EL, Allen PJ. Hypothalamic transcriptome response to simulated diel earthen pond hypoxia cycles in channel catfish (Ictalurus punctatus). Physiol Genomics. 2024;56(8):519–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1152/physiolgenomics.00007.2024.
Andersen LK, Abernathy JW, Farmer BD, Lange MD, McEntire ME, Rawles SD. Gene expression profiles of white bass (Morone chrysops) and hybrid striped bass (M. chrysops x M. saxatilis) gill tissue following Flavobacterium covae infection. Comparative Immunol Rep. 2024:200144. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cirep.2024.200144.
Wang R, Sun L, Bao L, Zhang J, Jiang Y, Yao J, Song L, Feng J, Liu S, Liu Z. Bulk segregant RNA-seq reveals expression and positional candidate genes and allele-specific expression for disease resistance against enteric septicemia of catfish. BMC Genomics. 2013;14:1–18. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/1471-2164-14-929.
Wang X, Liu S, Yang Y, Fu Q, Abebe A, Liu Z. Identification of NF-κB related genes in channel catfish and their expression profiles in mucosal tissues after columnaris bacterial infection. Dev Comp Immunol. 2017;70:27–38. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.dci.2017.01.003.
Proestou DA, Sullivan ME. Variation in global transcriptomic response to Perkinsus marinus infection among eastern oyster families highlights potential mechanisms of disease resistance. Fish Shellfish Immunol. 2020;96:141–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.fsi.2019.12.001.
Proestou DA, Sullivan ME, Lundgren KM, Ben-Horin T, Witkop EM, Hart KM. Understanding Crassostrea virginica tolerance of Perkinsus marinus through global gene expression analysis. Front Genet. 2023;14:1054558. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2023.1054558.
Sullivan ME, Proestou DA. Survival and transcriptomic responses to different Perkinsus marinus exposure methods in an Eastern oyster family. Aquaculture. 2021;542(736831):736831. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2021.736831.
Tan S, Wang W, Zhong X, Tian C, Niu D, Bao L, Zhou T, Jin Y, Yang Y, Yuan Z. Increased alternative splicing as a host response to Edwardsiella ictaluri infection in catfish. Mar Biotechnol. 2018;20:729–38. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10126-018-9844-2.
Tan S, Wang W, Tian C, Niu D, Zhou T, Yang Y, Gao D, Liu Z. Post-transcriptional regulation through alternative splicing after infection with Flavobacterium columnare in channel catfish (Ictalurus punctatus). Fish Shellfish Immunol. 2019;91:188–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.fsi.2019.05.008.
Tan S, Wang W, Tian C, Niu D, Zhou T, Jin Y, Yang Y, Gao D, Dunham R, Liu Z. Heat stress induced alternative splicing in catfish as determined by transcriptome analysis. Comp Biochem Physiol D: Genomics Proteomics. 2019;29:166–72. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cbd.2018.11.008.
Tan S, Wang W, Zhou T, Yang Y, Gao D, Dunham R, Liu Z. Polyadenylation sites and their characteristics in the genome of channel catfish (Ictalurus punctatus) as revealed by using RNA-Seq data. Comp Biochem Physiol D: Genomics Proteomics. 2019;30:248–55. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.cbd.2019.03.008.
Ali A, Thorgaard GH, Salem M. PacBio Iso-Seq improves the rainbow trout genome annotation and identifies alternative splicing associated with economically important phenotypes. Front Genet. 2021;12:683408. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2021.683408.
Rajab SA, Andersen LK, Kenter LW, Berlinsky DL, Borski RJ, McGinty AS, Ashwell CM, Ferket PR, Daniels HV, Reading BJ. Combinatorial metabolomic and transcriptomic analysis of muscle growth in hybrid striped bass (female white bass Morone chrysops x male striped bass M. saxatilis). BMC Genomics. 2024;25(1):580. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-024-10325-y.
Chapman RW, Reading BJ, Sullivan CV. Ovary transcriptome profiling via artificial intelligence reveals a transcriptomic fingerprint predicting egg quality in striped bass, Morone saxatilis. PLoS ONE. 2014;9(5):e96818. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0096818.
Sullivan CV, Chapman RW, Reading BJ, Anderson PE. Transcriptomics of mRNA and egg quality in farmed fish: Some recent developments and future directions. Gen Comp Endocrinol. 2015;221:23–30. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ygcen.2015.02.012.
Reading BJ, Williams VN, Chapman RW, Williams TI, Sullivan CV. Dynamics of the striped bass (Morone saxatilis) ovary proteome reveal a complex network of the translasome. J Proteome Res. 2013;12(4):1691–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/pr3010293.
Schilling J, Nepomuceno A, Schaff JE, Muddiman DC, Daniels HV, Reading BJ. Compartment proteomics analysis of white perch (Morone americana) ovary using support vector machines. J Proteome Res. 2014;13(3):1515–26. https://doiorg.publicaciones.saludcastillayleon.es/10.1021/pr401067g.
Palaiokostas C. Predicting for disease resistance in aquaculture species using machine learning models. Aquaculture Reports. 2021;20: 100660. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aqrep.2021.100660.
Ellisor DL, Bayless AL, Schock TB, Davis WC, Knott BT, Seghers J, Leys H, Emteborg H. Multi-omics characterization of NIST seafood reference materials and alternative matrix preparations. Anal Bioanal Chem. 2024;416(3):773–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00216-023-04928-9.
Daniels RR, Taylor RS, Robledo D, Macqueen DJ. Single cell genomics as a transformative approach for aquaculture research and innovation. Rev Aquac. 2023;15(4):1618–37. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/raq.12806.
Aldersey JE, Lange MD, Beck BH, Abernathy JW. Single-nuclei transcriptome analysis of channel catfish spleen provides insight into the immunome of an aquaculture-relevant species. PLoS ONE. 2024;19(9):e0309397. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0309397.
Cao M, Xue T, Huo H, Zhang X, Wang NN, Yan X, Li C. Spatial transcriptomes and microbiota reveal immune mechanism that respond to pathogen infection in the posterior intestine of Sebastes schlegelii. Open Biol. 2023;13(2):220302. https://doiorg.publicaciones.saludcastillayleon.es/10.1098/rsob.220302.
Sveen LR, Robinson N, Krasnov A, Daniels RR, Vaadal M, Karlsen C, Ytteborg E, Robledo D, Salisbury S, Dagnachew B et al. Transcriptomic landscape of Atlantic salmon (Salmo salar L.) skin. G3 (Bethesda). 2023;13(11). https://doiorg.publicaciones.saludcastillayleon.es/10.1093/g3journal/jkad215.
Taylor RS, Ruiz Daniels R, Dobie R, Naseer S, Clark TC, Henderson NC, Boudinot P, Martin SAM, Macqueen DJ. Single cell transcriptomics of Atlantic salmon (Salmo salar L.) liver reveals cellular heterogeneity and immunological responses to challenge by Aeromonas salmonicida. Front Immunol. 2022;13:984799. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fimmu.2022.984799.
Wang H-P, Shen Z-G. The potential role of epigenetics in aquaculture: insights from different taxa to diverse teleosts. 2023:1–43. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/9781119821946.ch1.
Piferrer F, Wang H-P. Epigenetics in aquaculture: knowledge gaps, challenges, and future prospects. 2023:451–63. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/9781119821946.ch20.
Gavery MR, Roberts SB. Epigenetic considerations in aquaculture. PeerJ. 2017;5(e4147):e4147. https://doiorg.publicaciones.saludcastillayleon.es/10.7717/peerj.4147.
Liu Z, Zhou T, Gao D. Genetic and epigenetic regulation of growth, reproduction, disease resistance and stress responses in aquaculture. Front Genet. 2022;13:994471. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2022.994471.
Yang Y, Zhou T, Liu Y, Tian C, Bao L, Wang W, Zhang Y, Liu S, Shi H, Tan S, et al. Identification of an epigenetically marked locus within the sex determination region of channel catfish. Int J Mol Sci. 2022;23(10):5471. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms23105471.
Wang W, Yang Y, Tan S, Zhou T, Liu Y, Tian C, Bao L, Xing D, Su B, Wang J. Genomic imprinting-like monoallelic paternal expression determines sex of channel catfish. Sci Adv. 2022;8(51):eadc8786. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/sciadv.adc8786.
Gong G, Xiong Y, Xiao S, Li X-Y, Huang P, Liao Q, Han Q, Lin Q, Dan C, Zhou L. Origin and chromatin remodeling of young X/Y sex chromosomes in catfish with sexual plasticity. Nat Sci Rev. 2023;10(2):nwac239. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/nsr/nwac239.
Saito T, Whatmore P, Taylor JF, Fernandes JMO, Adam A-C, Tocher DR, Espe M, Skjærven KH. Micronutrient supplementation affects DNA methylation in male gonads with potential intergenerational epigenetic inheritance involving the embryonic development through glutamate receptor-associated genes. BMC Genomics. 2022;23(1):115. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-022-08348-4.
Woods LC III, Li Y, Ding Y, Liu J, Reading BJ, Fuller SA, Song J. DNA methylation profiles correlated to striped bass sperm fertility. BMC Genomics. 2018;19:1–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-018-4548-6.
Salem M, Al-Tobasei R, Ali A, Kenney B. Integrated analyses of DNA methylation and gene expression of rainbow trout muscle under variable ploidy and muscle atrophy conditions. Genes. 2022;13(7):1151. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/genes13071151.
Johnson KM, Sirovy KA, Casas SM, La Peyre JF, Kelly MW. Characterizing the Epigenetic and Transcriptomic Responses to Perkinsus marinus Infection in the Eastern Oyster Crassostrea virginica. Front Mar Sci. 2020;7. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmars.2020.00598.
Spencer LH, Venkataraman YR, Crim R, Ryan S, Horwith MJ, Roberts SB. Carryover effects of temperature and pCO2 across multiple Olympia oyster populations. Ecol Appl. 2020;30(3):e02060. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/eap.2060.
Gurr SJ, Wanamaker SA, Vadopalas B, Roberts SB, Putnam HM. Repeat exposure to hypercapnic seawater modifies growth and oxidative status in a tolerant burrowing clam. J Exp Biol. 2021;224(13). https://doiorg.publicaciones.saludcastillayleon.es/10.1242/jeb.233932.
Putnam HM, Trigg SA, White SJ, Spencer LH, Vadopalas B, Natarajan A, Hetzel J, Jaeger E, Soohoo J, Gallardo-Escárate C, et al. Dynamic DNA methylation contributes to carryover effects and beneficial acclimatization in geoduck clams. 2022. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/2022.06.24.497506.
Šrut M. Ecotoxicological epigenetics in invertebrates: Emerging tool for the evaluation of present and past pollution burden. Chemosphere. 2021;282(131026):131026. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.chemosphere.2021.131026.
Silliman K, Spencer LH, White SJ, Roberts SB. Epigenetic and genetic population structure is coupled in a marine invertebrate. Genome Biol Evol. 2023;15(2). https://doiorg.publicaciones.saludcastillayleon.es/10.1093/gbe/evad013.
Jobling M. National Research Council (NRC): Nutrient requirements of fish and shrimp. Aquacult Int. 2011;20(3):601–2. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s10499-011-9480-6.
Bonder MJ, Kurilshikov A, Tigchelaar EF, Mujagic Z, Imhann F, Vila AV, Deelen P, Vatanen T, Schirmer M, Smeekens SP, et al. The effect of host genetics on the gut microbiome. Nat Genet. 2016;48(11):1407–12. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/ng.3663.
Awany D, Allali I, Dalvie S, Hemmings S, Mwaikono KS, Thomford NE, Gomez A, Mulder N, Chimusa ER. Host and microbiome genome-wide association studies: current state and challenges. Front Genet. 2019;9. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2018.00637.
Chapagain P, Arivett B, Cleveland BM, Walker DM, Salem M. Analysis of the fecal microbiota of fast- and slow-growing rainbow trout (Oncorhynchus mykiss). BMC Genomics. 2019;20(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-019-6175-2.
Chapagain P, Walker D, Leeds T, Cleveland BM, Salem M. Distinct microbial assemblages associated with genetic selection for high- and low- muscle yield in rainbow trout. BMC Genomics. 2020;21(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-020-07204-7.
Raymo G, Ali A, Ahmed RO, Salem M. Early-life fecal transplantation from high muscle yield rainbow trout to low muscle yield recipients accelerates somatic growth through respiratory and mitochondrial efficiency modulation. Microorganisms. 2024;12(2):261. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/microorganisms12020261.
English G, Lawrence MJ, McKindsey CW, Lacoursière-Roussel A, Bergeron H, Gauthier S, Wringe BF, Trudel M. A review of data collection methods used to monitor the associations of wild species with marine aquaculture sites. Rev Aquac. 2024;16(3):1160–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/raq.12890.
King WL, Siboni N, Williams NL, Kahlke T, Nguyen KV, Jenkins C, Dove M, O’Connor W, Seymour JR, Labbate M. Variability in the composition of Pacific oyster microbiomes across oyster families exhibiting different levels of susceptibility to OsHV-1 μvar disease. Front Microbiol. 2019;10:473. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fmicb.2019.00473.
Rieder J, Kapopoulou A, Bank C, Adrian-Kalchhauser I. Metagenomics and metabarcoding experimental choices and their impact on microbial community characterization in freshwater recirculating aquaculture systems. Environmental microbiome. 2023;18(1):8. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40793-023-00459-z.
Stevick RJ, Post AF, Gómez-Chiarri M. Functional plasticity in oyster gut microbiomes along a eutrophication gradient in an urbanized estuary. Animal microbiome. 2021;3:1–17. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s42523-020-00066-0.
Cardona E, Gueguen Y, Magré K, Lorgeoux B, Piquemal D, Pierrat F, Noguier F, Saulnier D. Bacterial community characterization of water and intestine of the shrimp Litopenaeus stylirostris in a biofloc system. BMC Microbiol. 2016;16:1–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12866-016-0770-z.
Drønen K, Roalkvam I, Dahle H, Olsen AB, Nilsen H, Wergeland H. Microbiome dataset from a marine recirculating aquaculture system (RAS) for salmon post-smolt production in Norway. Data Brief. 2022;40: 107767. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.dib.2021.107767.
Rajeev M, Jung I, Lim Y, Kim S, Kang I, Cho J-C. Metagenome sequencing and recovery of 444 metagenome-assembled genomes from the biofloc aquaculture system. Scientific data. 2023;10(1):707. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41597-023-02622-0.
Stilwell JM, Camus AC, Ware C, Walker CM, Stanton JB, Leary JH, Khoo LH, Wise DJ, Waldbieser GC, Griffin MJ. Influence of channel catfish and channel × blue catfish hybrids on myxozoan community composition in catfish aquaculture ponds. N Am J Aquac. 2023;85(3):241–51. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/naaq.10293.
Pukk L, Kanefsky J, Heathman AL, Weise EM, Nathan LR, Herbst SJ, Sard NM, Scribner KT, Robinson JD. eDNA metabarcoding in lakes to quantify influences of landscape features and human activity on aquatic invasive species prevalence and fish community diversity. Divers Distrib. 2021;27(10):2016–31. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/ddi.13370.
Timmins-Schiffman E, White SJ, Thompson RE, Vadopalas B, Eudeline B, Nunn BL, Roberts SB. Coupled microbiome analyses highlights relative functional roles of bacteria in a bivalve hatchery. Environ Microbiome. 2021;16(1):7. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40793-021-00376-z.
Chakraborty D, Sharma N, Kour S, Sodhi SS, Gupta MK, Lee SJ, Son YO. Applications of omics technology for livestock selection and improvement. Front Genet. 2022;13:774113. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2022.774113.
Yang Y, Saand MA, Huang L, Abdelaal WB, Zhang J, Wu Y, Li J, Sirohi MH, Wang F. Applications of multi-omics technologies for crop improvement. Front Plant Sci. 2021;12:563953. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fpls.2021.563953.
López de Maturana E, Alonso L, Alarcón P, Martín-Antoniano IA, Pineda S, Piorno L, Calle ML, Malats N. Challenges in the integration of omics and non-omics data. Genes. 2019;10(3):238. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/genes10030238.
Graw S, Chappell K, Washam CL, Gies A, Bird J, Robeson MS, Byrum SD. Multi-omics data integration considerations and study design for biological systems and disease. Molecular omics. 2021;17(2):170–85. https://doiorg.publicaciones.saludcastillayleon.es/10.1039/D0MO00041H.
Subramanian I, Verma S, Kumar S, Jere A, Anamika K. Multi-omics data integration, interpretation, and its application. Bioinform Biol Insights. 2020;14:1177932219899051.
Madrid-Márquez L, Rubio-Escudero C, Pontes B, González-Pérez A, Riquelme JC, Sáez ME. MOMIC: A multi-omics pipeline for data analysis, integration and interpretation. Appl Sci. 2022;12(8):3987. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/app12083987.
Miao B-B, Dong W, Gu Y-X, Han Z-F, Luo X, Ke C-H, You W-W. OmicsSuite: a customized and pipelined suite for analysis and visualization of multi-omics big data. Horticulture Res. 2023;10(11). https://doiorg.publicaciones.saludcastillayleon.es/10.1093/hr/uhad195.
Ramos M, Schiffer L, Re A, Azhar R, Basunia A, Rodriguez C, Chan T, Chapman P, Davis SR, Gomez-Cabrero D, et al. Software for the integration of multiomics experiments in Bioconductor. Can Res. 2017;77(21):e39–42. https://doiorg.publicaciones.saludcastillayleon.es/10.1158/0008-5472.can-17-0344.
Pfau T, Galhardo M, Lin J, Sauter T. IDARE2—simultaneous visualisation of multiomics data in cytoscape. Metabolites. 2021;11(5):300. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/metabo11050300.
Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/gr.1239303.
Chen T, Abadi AJ, Lê Cao K-A, Tyagi S. Multiomics: A user-friendly multi-omics data harmonisation R pipeline. F1000Research. 2023;10:538. https://doiorg.publicaciones.saludcastillayleon.es/10.12688/f1000research.53453.2.
Andersen LK, Reading BJ. A supervised machine learning workflow for the reduction of highly dimensional biological data. Artificial Intelligence in the Life Sciences. 2024;5:100090. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.ailsci.2023.100090.
Fu G, Yuna Y. Phenotyping and phenomics in aquaculture breeding. Aquaculture and Fisheries. 2022;7(2):140–6. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aaf.2021.07.001.
Houle D, Govindaraju DR, Omholt S. Phenomics: the next challenge. Nat Rev Genet. 2010;11(12):855–66. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/nrg2897.
Pérez-Enciso M, Steibel JP. Phenomes: the current frontier in animal breeding. Genet Sel Evol. 2021;53(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12711-021-00618-1.
Fuentes R, Letelier J, Tajer B, Valdivia LE, Mullins MC. Fishing forward and reverse: Advances in zebrafish phenomics. Mech Dev. 2018;154:296–308. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.mod.2018.08.007.
Wang Z, Liu H, Zhang G, Yang X, Wen L, Zhao W. Diseased fish detection in the underwater environment using an improved YOLOV5 network for intensive aquaculture. Fishes. 2023;8(3):169. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/fishes8030169.
Babu KM, Bentall D, Ashton DT, Puklowski M, Fantham W, Lin HT, Tuckey NPL, Wellenreuther M, Jesson LK. Computer vision in aquaculture: a case study of juvenile fish counting. J R Soc N Z. 2022;53(1):52–68. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/03036758.2022.2101484.
Deng Y, Tan H, Tong M, Zhou D, Li Y, Zhu M. An automatic recognition method for fish species and length using an underwater stereo vision system. Fishes. 2022;7(6):326. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/fishes7060326.
Li D, Hao Y, Duan Y. Nonintrusive methods for biomass estimation in aquaculture with emphasis on fish: a review. Rev Aquac. 2020;12(3):1390–411. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/raq.12388.
Ranjan R, Tsukuda S, Good C. MortCam: An artificial intelligence-aided fish mortality detection and alert system for recirculating aquaculture. Aquacult Eng. 2023;102:102341. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaeng.2023.102341.
Zhao F, Hao J, Zhang H, Yu X, Yan Z, Wu F. Quality recognition method of oyster based on U-net and random forest. J Food Compos Anal. 2024;125:105746. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jfca.2023.105746.
Mayormente M. Intelligent recirculating aquaculture system of Oreochromis niloticus: A feed-conversion-ratio-based machine learning approach. Int J Intell Syst Appl Eng. 2024;12(13s):122–8.
Graham CA, Shamkhalichenar H, Browning VE, Byrd VJ, Liu Y, Gutierrez-Wing MT, Novelo N, Choi J-W, Tiersch TR. A practical evaluation of machine learning for classification of ultrasound images of ovarian development in channel catfish (Ictalurus punctatus). Aquaculture. 2022;552: 738039. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2022.738039.
Chen Y, Yue J, Li Z, Yang J, Wang W. Pacific oyster gonad identification and grayscale calculation based on unapparent object detection. In: Chinese Conference on Pattern Recognition and Computer Vision (PRCV): 2023: Springer; 2023:94–106.
Guévélou E, Allen SK Jr. Use of near infrared reflectance spectroscopy (NIRS) for the rapid compositional analysis of di-, tri-, and tetraploid eastern oysters (Crassostrea virginica). Aquaculture. 2016;459:203–9. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2016.03.022.
Rather MA, Ahmad I, Shah A, Hajam YA, Amin A, Khursheed S, Ahmad I, Rasool S. Exploring opportunities of artificial intelligence in aquaculture to meet increasing food demand. Food Chem: X. 2024:101309. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.fochx.2024.101309.
Grassi S, Benedetti S, Casiraghi E, Buratti S. E-sensing systems for shelf life evaluation: A review on applications to fresh food of animal origin. Food Packag Shelf Life. 2023;40:101221. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.fpsl.2023.101221.
Grassi S, Benedetti S, Magnani L, Pianezzola A, Buratti S. Seafood freshness: e-nose data for classification purposes. Food Control. 2022;138:108994. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.foodcont.2022.108994.
Difford GF, Boison SA, Khaw HL, Gjerde B. Validating non-invasive growth measurements on individual Atlantic salmon in sea cages using diode frames. Comput Electron Agric. 2020;173:105411. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.compag.2020.105411.
Macaulay G, Warren-Myers F, Barrett LT, Oppedal F, Føre M, Dempster T. Tag use to monitor fish behaviour in aquaculture: a review of benefits, problems and solutions. Rev Aquac. 2021;13(3):1565–82. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/raq.12534.
O’Donncha F, Stockwell CL, Planellas SR, Micallef G, Palmes P, Webb C, Filgueira R, Grant J. Data driven insight into fish behaviour and their use for precision aquaculture. Frontiers in Animal Science. 2021;2:695054. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fanim.2021.695054.
Triant DA, Walsh AT, Hartley GA, Petry B, Stegemiller MR, Nelson BM, McKendrick MM, Fuller EP, Cockett NE, Koltes JE, et al. AgAnimalGenomes: browsers for viewing and manually annotating farm animal genomes. Mamm Genome. 2023;34(3):418–36. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s00335-023-10008-1.
Field D, Amaral-Zettler L, Cochrane G, Cole JR, Dawyndt P, Garrity GM, Gilbert J, Glöckner FO, Hirschman L, Karsch-Mizrachi I, et al. The Genomic Standards Consortium. PLoS Biol. 2011;9(6):e1001088. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pbio.1001088.
Lawniczak MKN, Durbin R, Flicek P, Lindblad-Toh K, Wei X, Archibald JM, Baker WJ, Belov K, Blaxter ML, Marques Bonet T et al. Standards recommendations for the Earth BioGenome Project. Proc Nat Acad Sci. 2022;119(4).https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.2115639118.
Abascal F, Acosta R, Addleman NJ, Adrian J, Afzal V, Aken B, Ai R, Akiyama JA, Jammal OA, Amrhein H, et al. Perspectives on ENCODE. Nature. 2020;583(7818):693–8. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41586-020-2449-8.
Consortium EP. A User’s Guide to the Encyclopedia of DNA Elements (ENCODE). PLoS Biol. 2011;9(4):e1001046. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pbio.1001046.
Wilkinson MD, Dumontier M, Aalbersberg IJ, Appleton G, Axton M, Baak A, Blomberg N, Boiten J-W, da Silva Santos LB, Bourne PE, et al. The FAIR Guiding Principles for scientific data management and stewardship. Sci Data. 2016;3(1). https://doiorg.publicaciones.saludcastillayleon.es/10.1038/sdata.2016.18.
Castro LJ, Palagi PM, Beard N, Attwood TK, Brazas MD. Bioschemas training profiles: A set of specifications for standardizing training information to facilitate the discovery of training programs and resources. PLoS Comput Biol. 2023;19(6):e1011120. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pcbi.1011120.
Gaignard A, Tsueng G, Mičetić I, Castro LJ, Juty N. Improving Bioschemas creation and community adoption through process improvements, tool development, and advancing compliance to FAIR standards. In: Center for Open Science; 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.37044/osf.io/8km9j.
Tsueng G, Frika S, Gray AJG, Barbero MC, Gaignard A, Mičetić I, Castro LJ, Juty N. Enabling profile updates through the Data Discovery Engine (DDE). In: Center for Open Science; 2023. https://doiorg.publicaciones.saludcastillayleon.es/10.37044/osf.io/3b9gp.
Gray AJ, Goble CA, Jimenez RC. Bioschemas: From potato salad to protein annotation. In: ISWC (Posters, Demos & Industry Tracks): 2017; 2017.
Tuggle CK, Clarke J, Dekkers JC, Ertl D, Lawrence-Dill CJ, Lyons E, Murdoch BM, Scott NM, Schnable PS. The Agricultural Genome to Phenome Initiative (AG2PI): creating a shared vision across crop and livestock research communities. In., vol. 23: Springer; 2022. p. 1–11. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13059-021-02570-1.
Tuggle CK, Scott N, Clarke J, Murdoch BM, Dekkers JC, Ertl D, Lyons E, Lawrence-Dill CJ, Schnable PS. The AG2pi vision for resources in agricultural genomics and phenomics: How Asas can contribute. J Animal Sci. 2023;101(Supplement_3):50–1. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/jas/skad281.062.
Yang Z, Fu G, Lee M, Yeo S, Yue GH. Genes for editing to improve economic traits in aquaculture fish species. Aquac Fish. 2024. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aaf.2024.05.005.
Delomas TA, Hollenbeck CM, Matt JL, Thompson NF. Evaluating cost-effective genotyping strategies for genomic selection in oysters. Aquaculture. 2023;562(738844):738844. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2022.738844.
Delomas TA, Hollenbeck CM, Matt JL, Thompson NF. Microhaplotypes generate higher breeding value accuracy compared to SNPs for imputation-based breeding strategies. Aquaculture. 2024;586(740779):740779. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2024.740779.
Palaiokostas C, Houston RD. Genome-wide approaches to understanding and improving complex traits in aquaculture species. CAB Rev Perspect Agric Vet Sci Nutr Nat Resour. 2018;2017(055):1–10. https://doiorg.publicaciones.saludcastillayleon.es/10.1079/pavsnnr201712055.
Song H, Dong T, Yan X, Wang W, Tian Z, Sun A, Dong Y, Zhu H, Hu H. Genomic selection and its research progress in aquaculture breeding. Rev Aquac. 2023;15(1):274–91. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/raq.12716.
Boudry P, Allal F, Aslam ML, Bargelloni L, Bean TP, Brard-Fudulea S, Brieuc MSO, Calboli FCF, Gilbey J, Haffray P, et al. Current status and potential of genomic selection to improve selective breeding in the main aquaculture species of International Council for the Exploration of the Sea (ICES) member countries. Aquac Rep. 2021;20(100700):100700. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aqrep.2021.100700.
Houston RD, Kriaridou C, Robledo D. Animal board invited review: Widespread adoption of genetic technologies is key to sustainable expansion of global aquaculture. Animal. 2022;16(10):100642. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.animal.2022.100642.
Kriaridou C, Tsairidou S, Fraslin C, Gorjanc G, Looseley ME, Johnston IA, Houston RD, Robledo D. Evaluation of low-density SNP panels and imputation for cost-effective genomic selection in four aquaculture species. Front Genet. 2023;14:1194266. https://doiorg.publicaciones.saludcastillayleon.es/10.3389/fgene.2023.1194266.
Song H, Hu H. Strategies to improve the accuracy and reduce costs of genomic prediction in aquaculture species. Evol Appl. 2022;15(4):578–90. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/eva.13262.
Thompson NF, Sutherland BJ, Green T, TA D. A free lunch: microhaplotype discovery in an existing amplicon panel improves parentage assignment for the highly polymorphic Pacific oyster. G3: Genes, Genomes, Genetics. 2025. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/g3journal/jkae280.
Zhou Y, Yang L, Han X, Han J, Hu Y, Li F, Xia H, Peng L, Boschiero C, Rosen BD, et al. Assembly of a pangenome for global cattle reveals missing sequences and novel structural variations, providing new insights into their diversity and evolutionary history. Genome Res. 2022;32(8):1585–601. https://doiorg.publicaciones.saludcastillayleon.es/10.1101/gr.276550.122.
Tay Fernandez CG, Nestor BJ, Danilevicz MF, Gill M, Petereit J, Bayer PE, Finnegan PM, Batley J, Edwards D. Pangenomes as a resource to accelerate breeding of under-utilised crop species. Int J Mol Sci. 2022;23(5):2671. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms23052671.
Gong Y, Li Y, Liu X, Ma Y, Jiang L. A review of the pangenome: how it affects our understanding of genomic variation, selection and breeding in domestic animals? Journal of Animal Science and Biotechnology. 2023;14(1):73. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40104-023-00860-1.
Golicz AA, Bayer PE, Bhalla PL, Batley J, Edwards D. Pangenomics comes of age: From bacteria to plant and animal applications. Trends Genet. 2020;36(2):132–45. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.tig.2019.11.006.
Kajbaf K, Overturf K, Kumar V. Integrated alternative approaches to select feed-efficient rainbow trout families to enhance the plant protein utilization. Sci Rep. 2024;14(1):3869. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41598-024-54218-2.
Abernathy J, Brezas A, Snekvik KR, Hardy RW, Overturf K. Integrative functional analyses using rainbow trout selected for tolerance to plant diets reveal nutrigenomic signatures for soy utilization without the concurrence of enteritis. PLoS ONE. 2017;12(7):e0180972. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0180972.
Mengistu SB, Mulder HA, Bastiaansen JW, Benzie JA, Khaw HL, Trinh TQ, Komen H. Fluctuations in growth are heritable and a potential indicator of resilience in Nile tilapia (Oreochromis niloticus). Aquaculture. 2022;560:738481. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2022.738481.
Lallias D, Quillet E, Bégout M-L, Auperin B, Khaw HL, Millot S, Valotaire C, Kernéis T, Labbe L, Prunet P. Genetic variability of environmental sensitivity revealed by phenotypic variation in body weight and (its) correlations to physiological and behavioral traits. PLoS ONE. 2017;12(12):e0189943. https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pone.0189943.
McCarty AJ, Hood S, Huebert K, Cram J, McFarland K, Plough LV. Evaluating a short vs. long-term progeny test and investigating physiology associated with survival in extreme low salinity for the eastern oyster Crassostrea virginica. Aquaculture. 2023;574:739688. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2023.739688.
McCarty AJ, McFarland K, Small J, Allen S Jr, Plough L. Heritability of acute low salinity survival in the Eastern oyster (Crassostrea virginica). Aquaculture. 2020;529:735649. https://doiorg.publicaciones.saludcastillayleon.es/10.1093/g3journal/jkab368.
Thompson NF, Agnew MV, Calla B, Burge CA. Assessing selection potential for Pacific oyster (Crassostrea gigas) to a North American OsHV-1 μvar: Comparing two experimental assay methods. Aquaculture. 2024;590:741076. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2024.741076.
Vallejo RL, Pietrak MR, Milligan MM, Gao G, Tsuruta S, Fragomeni BO, Long RL, Peterson BC, Palti Y. Genetic architecture and accuracy of predicted genomic breeding values for sea lice resistance in the St John River aquaculture strain of North American Atlantic salmon. Aquaculture. 2024;586(740819): 740819. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.aquaculture.2024.740819.
Wyban J. Selective breeding of Penaeus vannamei: Impact on world aquaculture and lessons for future. J Coast Res. 2019;86(sp1):1. https://doiorg.publicaciones.saludcastillayleon.es/10.2112/si86-001.1.
Houston RD, Haley CS, Hamilton A, Guy DR, Mota-Velasco JC, Gheyas AA, Tinch AE, Taggart JB, Bron JE, Starkey WG, et al. The susceptibility of Atlantic salmon fry to freshwater infectious pancreatic necrosis is largely explained by a major QTL. Heredity (Edinb). 2010;105(3):318–27. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/hdy.2009.171.
Houston RD, Haley CS, Hamilton A, Guy DR, Tinch AE, Taggart JB, McAndrew BJ, Bishop SC. Major quantitative trait loci affect resistance to infectious pancreatic necrosis in Atlantic salmon (Salmo salar). Genetics. 2008;178(2):1109–15. https://doiorg.publicaciones.saludcastillayleon.es/10.1534/genetics.107.082974.
Moen T, Torgersen J, Santi N, Davidson WS, Baranski M, Ødegård J, Kjøglum S, Velle B, Kent M, Lubieniecki KP, et al. Epithelial cadherin determines resistance to infectious pancreatic necrosis virus in Atlantic salmon. Genetics. 2015;200(4):1313–26. https://doiorg.publicaciones.saludcastillayleon.es/10.1534/genetics.115.175406.
Vallejo RL, Silva RM, Evenhuis JP, Gao G, Liu S, Parsons JE, Martin KE, Wiens GD, Lourenco DA, Leeds TD, et al. Accurate genomic predictions for BCWD resistance in rainbow trout are achieved using low-density SNP panels: Evidence that long-range LD is a major contributing factor. J Anim Breed Genet. 2018;135(4):263–74. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jbg.12335.
Silva RMO, Evenhuis JP, Vallejo RL, Gao G, Martin KE, Leeds TD, Palti Y, Lourenco DAL. Whole-genome mapping of quantitative trait loci and accuracy of genomic predictions for resistance to columnaris disease in two rainbow trout breeding populations. Genet Sel Evol. 2019;51(1):42. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12711-019-0484-4.
Ragone Calvo LM, Calvo GW, Burreson EM. Dual disease resistance in a selectively bred eastern oyster, Crassostrea virginica, strain tested in Chesapeake Bay. Aquaculture. 2003;220(1–4):69–87. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/s0044-8486(02)00399-x.
Moss SM, Moss DR, Arce SM, Lightner DV, Lotz JM. The role of selective breeding and biosecurity in the prevention of disease in penaeid shrimp aquaculture. J Invertebr Pathol. 2012;110(2):247–50. https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jip.2012.01.013.
de Souza Iung LH, Carvalheiro R. Neves HHdR, Mulder HA: Genetics and genomics of uniformity and resilience in livestock and aquaculture species: a review. J Anim Breed Genet. 2020;137(3):263–80. https://doiorg.publicaciones.saludcastillayleon.es/10.1111/jbg.12454.
Acknowledgements
We first want to thank aquaculture producers across the United States for their tireless effort in producing nutritious food and for the collaborative efforts and continued communication that has and continues to inform and strengthen aquaculture research. We would like to acknowledge the support provided through the National Research Support Project 8 (NRSP8, National Animal Genome Research Program, NAGRP) over the last several decades and Dr. Angelica Van Goor, USDA NIFA National Program Leader-Division of Animal Systems. We thank the countless facilities personnel, technicians, students, and office staff that have facilitated these research projects and the supporting funding agencies. We are grateful to Dr. Rodolphe Barrangou (North Carolina State University, NCSU; Raleigh, NC), Dr. Xavier Lauth (Center for Aquaculture Technologies, CAT; San Diego, CA), and the NCSU Pamlico Aquaculture Field Laboratory (PAFL, Aurora, NC) staff for participating in the May 2023 Aquaculture Genomics, Genetics, and Breeding Workshop, as well as the staff assistance provided by the NCSU Libraries, Fleet Services, and College of Agriculture and Life Sciences (CALS) in hosting this workshop. We lastly thank Dr. Laura Spencer (University of Washington and National Oceanic and Atmospheric Administration, NOAA; Seattle, WA) for her technical review. The USDA is an equal opportunity provider and employer. Mention of trade names or commercial products in this publication is solely for the purpose of providing specific information and does not imply recommendation or endorsements by the U.S. Department of Agriculture.
Funding
National Research Support Project 8 (NRSP8, National Animal Genome Research Program, NAGRP) Aquaculture Species Group.
Author information
Authors and Affiliations
Consortia
Contributions
LKA and NFT conceptualized, curated, and synthesized data; wrote the original draft and prepared visual representations of the information; managed, coordinated, and planned the execution of the project; and finalized the work for submission and presentation. JWA provided oversight in planning and/or execution, reviewed, contributed additions, and provided feedback to the drafted work. CGE curated and/or synthesized data, reviewed, contributed additions, and provided feedback to the drafted work. ZL acquired funding to support the project, reviewed, contributed additions, and provided feedback to the drafted work YP was involved in management and/or coordination, reviewed, contributed additions, and provided feedback to the drafted work. SBR conceptualized overarching ideas, goals, and aims; reviewed, contributed additions, and provided feedback to the drafted work. MS Conceptualized overarching ideas, goals, and aims; curated and/or synthesized data, reviewed, contributed additions, and provided feedback to the drafted work. BJR conceptualized overarching ideas, goals, and aims; was involved in management and/or coordination; provided oversight in planning and/or execution; reviewed, contributed additions, and provided feedback to the drafted work and acquired funding to support the project. ROA, AA, RA, BHB, BC, TAD, RAD, SAF, JCG, MRG, CMH, KMJ, EK, ELL, SL, BM, JLM, SAM, CEO, KO, EJP, BCP, MPP, LVP, MPP, DAP, CMP, SMAQ, GR, CR, KLR, LAR, KS, GCW, HW, CDW reviewed, contributed additions, and provided feedback to the drafted work. Authors are listed alphabetically based on last name except for LKA and NFT (co-first author), and BJR as final author.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
The original online version of this article was revised: Rafet Al-Tobasei was affiliated with the wrong affiliation.
Supplementary Information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Andersen, L.K., Thompson, N.F., Abernathy, J.W. et al. Advancing genetic improvement in the omics era: status and priorities for United States aquaculture. BMC Genomics 26, 155 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-025-11247-z
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12864-025-11247-z