You are on page 1of 18

Received: 30 March 2022 Revised: 14 July 2022 Accepted: 17 July 2022

DOI: 10.1111/raq.12716

REVIEW

Genomic selection and its research progress in aquaculture


breeding

Hailiang Song | Tian Dong | Xiaoyu Yan | Wei Wang | Zhaohui Tian |
Ai Sun | Ying Dong | Hua Zhu | Hongxia Hu

Fisheries Science Institute, Beijing Academy of


Agriculture and Forestry Sciences & Beijing Abstract
Key Laboratory of Fisheries Biotechnology,
Since its introduction in 2001, genomic selection (GS) has progressed rapidly. As a
Beijing, China
research and application hot topic, GS has led to a revolution in the field of animal
Correspondence
and plant breeding. Thanks to its ability to overcome the shortcomings of traditional
Hongxia Hu, Fisheries Science Institute, Beijing
Academy of Agriculture and Forestry breeding methods, GS has garnered increasing attention. Both theoretical and practi-
Sciences & Beijing Key Laboratory of Fisheries
cal breeding studies have revealed the higher accuracy of GS than that of traditional
Biotechnology, Beijing 100068, China.
Email: huhongxia@bjfishery.com breeding, which can accelerate genetic gain. In recent years, many GS studies have
been conducted on aquaculture species, which have shown that GS produces higher
Funding information
Beijing Natural Science Foundation, Grant/ prediction accuracy than traditional pedigree-based method. The present study
Award Number: 6222014; China Agriculture
reviews the principles and processes, preconditions, advantages, analytical methods
Research System of MOF and MARA, Grant/
Award Number: CARS-45-32; Beijing Joint and factors influencing GS as well as the progress of research in aquaculture into
Research Program for Germplasm Innovation
these aspects. Furthermore, future directions of GS in aquaculture are also discussed,
and New Variety Breeding, Grant/Award
Number: G20220628008; Youth Foundation which should expand its application to more aquaculture species.
of Beijing Academy of Agriculture and Forestry
Sciences, Grant/Award Number: QNJJ202105 KEYWORDS
aquaculture, breeding, future direction, genomic selection, method, research progress

1 | I N T RO DU CT I O N gene but less effective for the selection of quantitative traits that are
greatly affected by the environment. With advances in the theory of
In 2018, the estimated global fish production reached about 179 mil- quantitative genetics, breeders have successively proposed best linear
lion tons. Of this, 156 million tons were used for human consumption, unbiased prediction (BLUP),6 which combines pedigree and pheno-
which is equivalent to an average person eating about 20.5 kg of fish type records using all relevant information to estimate the breeding
every year.1 However, at present less than 10% of aquaculture pro- value (EBV) of individuals. Furthermore, with the development of
duction is based on genetically improved animals, despite the rapid computational technologies, BLUP (selective breeding) has been used
2
growth in annual genetic gain of aquaculture species. Furthermore, to improve various economic traits in many aquaculture species, such
with continuous population growth, pressure on global fisheries is as Vibrio alginolyticus resistance in Pacific oyster (Crassostrea gigas),7
3
expected to rise. Although conventional breeding programmes have carotenoid traits in Chinese mitten crab (Eriocheir sinensis),8 gill-
played a critical role in the genetic improvement of 50 important associated virus resistance in black tiger shrimp (Penaeus monodon),9
4,5
aquaculture species, the low efficiency of this approach can not tilapia lake virus resistance in Nile tilapia (Oreochromis niloticus),10
meet human needs. Therefore, modern breeding technologies must growth and survival in rohu carp (Labeo rohita)11 and growth and egg-
be urgently applied to accelerate the genetic improvement of aquacul- related traits in Russian sturgeon (Acipenser gueldenstaedtii).12 How-
ture species. ever, conventional BLUP yields low genetic gain for certain traits, such
In conventional breeding, highest genetically ranked individuals as traits with low heritability (e.g., disease resistance traits), traits that
are selected based primarily on their phenotypes. This approach is are difficult to measure (e.g., disease resistance and behavioural traits),
more effective for the selection of quality traits controlled by a single sex-restricted traits (e.g., reproductive and oviposition traits) and traits

274 © 2022 John Wiley & Sons Australia, Ltd. wileyonlinelibrary.com/journal/raq Rev Aquac. 2023;15:274–291.
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SONG ET AL. 275

that require slaughter for measurement (e.g., meat quality and carcass affects quantitative traits is in linkage disequilibrium (LD) with at least
composition traits), necessitating breakthroughs using novel breeding one molecular marker (e.g., SNP marker) among genome-wide markers
methods. (Figure 1). Therefore, GS can simultaneously fit all markers in the
With advances in genome sequencing and reduction in the cost model, regardless of whether they are statistically significant or not,
of related technologies, massive molecular marker (e.g., single- and can be traced back to all QTLs that affect traits, which explains
nucleotide polymorphism [SNP] marker) data have been accumulated. the “polygene hypothesis” of quantitative genetics from a molecular
Breeders have started exploring the use of genomic information to perspective and achieves accurate prediction of GEBVs.
improve the accuracy of individual selection. Currently, there are two The basic process of GS is as follows. First, reference and candi-
major breeding methods that apply genomic information, namely, date populations are established. Individuals in the reference popula-
marker-assisted selection (MAS)13 and genomic selection (GS).14 tion possess the genotypic and phenotypic information, and the
Marker-assisted selection has only proven to be useful when there reference population is representative of the whole population.
are genes with a relatively large effect on a trait, for example, for Meanwhile, individuals in the candidate population only require geno-
infectious pancreatic necrosis resistance in Atlantic salmon (Salmo typic information. The statistical model used for GS is:
salar), where a single gene explains 80%–100% of the genetic varia- y ¼ 1μ þ W 1 X1 þ W 2 X 2 þ … þ W n X n þ e, where W 1 is the coded geno-
tion for the trait.15,16 However, the application of MAS to improve type of SNP j for j = 1,…, n, X 1 is the allele substitution effect of SNP j
complex traits controlled by many genes with a small effect is limited. for j = 1,…, n and n is the number of SNPs. X1 as random effects with
This is because (1) quantitative trait loci (QTL) detection is difficult, prior distribution N(0, σ 2j ). Different statistical models are used to esti-
and most effort is spent on finding major genes. Moreover, (2) few mate the SNP effect (x1, x2, x3…) of the reference population. Next,
large-effect QTLs are known, and even those found still only partly according to the SNP effect and SNP genotype (w1, w2, w3…, coded
explain the genetic variation in given traits. Therefore, despite sub- as 0, 1 and 2 based on the number or copy of one allele) of the candi-
stantial efforts devoted to detect QTLs in aquaculture species, pro- date population, GEBV (GEBV = w1x1 + w2x2 + w3x3…) of the can-
moting MAS application in aquaculture breeding remains a didate population is calculated. Alternatively, GEBV of the candidate
challenge.17 To overcome the deficiencies of MAS, Meuwissen et al.14 population can be calculated directly by solving the mixed model
suggested a different approach, known as GS. Unlike MAS, GS equations. Finally, highest genetically ranked individuals are selected
enables the estimation of breeding values by estimating the effects of based on GEBV (Figure 2).
markers at a high density (HD) covering the whole genome, thus being
able to explain greater genetic variations in given traits. Furthermore,
GS gives a better estimate of true genetic relationships among individ- 2.2 | Precondition for GS
uals than pedigree data and achieves higher prediction accuracy of
the EBV, for example, without considering inbreeding, the kinship The precondition for GS is high-throughput genotyping technology of
coefficient between full siblings estimated using pedigree information genome-wide markers. In general, phenotypic records of traits are
is 0.5, whereas the true kinship coefficient obtained using genomic available in conventional breeding systems; however, HD marker
information may be less or greater than 0.5. genotypes require rapid detection of markers covering the whole
To this end, the present review summarises the technology of GS, genome. Currently, SNPs, which are DNA sequence polymorphisms
outlines research progress related to the application of GS in aquacul- caused by a single nucleotide variation in the genome, are considered
ture breeding, and discusses future directions of GS in aquaculture. the ideal markers due to their high abundance, high reproducibility
The present review aims to provide aquaculture scientists and breed- and relatively easy high-throughput detection.18 In recent years,
ing companies a general overview of the potential and gaps of GS in genomic sequencing and reference genome assembly of many aqua-
aquaculture breeding. culture species, including Atlantic salmon,19 grass carp (Ctenopharyn-
godon idella),20 Pacific white shrimp (Litopenaeus vannamei)21 and
Manila clam (Ruditapes philippinarum),22 have been completed. For
2 | TEC HNOLOGY OF GS specific species, the large number of SNP markers generated through
genome sequencing have laid the foundation for the application of
2.1 | Principle and process of GS GS. Low-coverage resequencing can reduce the cost of genotyping
and has broad application prospects. Studies have shown that when
Genomic selection is a molecular breeding method proposed by Meu- sequencing depth is less than or equal to 1X, the use of genotype
14
wissen et al. The principle of this method is to use whole genome imputation can produce genomic prediction accuracy similar to high-
marker and phenotypic data to estimate the effect value of each coverage resequencing in pig (Sus scrofa)23 and dairy cattle (Bos tau-
molecular marker or chromosome segment and obtain the individual rus).24 Zhang et al.25 compared the effect of different sequencing
EBV by adding the effect values of all markers.14 The EBV obtained depths on the accuracy of genome prediction in large yellow croaker
thus is called the genomic EBV (GEBV). Also, by fitting all markers (Larimichthys crocea) populations and noted that the genomic predic-
simultaneously, GS potentially uses all of the total genetic variance tion accuracy obtained using 0.5X was similar to that obtained using
available. A basic assumption of GS is that every QTL or gene that 8X. In addition, genotyping by sequencing (GBS) has allowed for rapid
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
276 SONG ET AL.

F I G U R E 1 Linkage disequilibrium between SNP markers and QTLs/genes in the whole genome. Quantitative traits (disease resistance and
growth) are usually affected by many genes with large or small effects (the peak of black bands), but these genes are as unknown as the black
box, and consequently the benefit from marker-assisted selection is limited by the proportion of the genetic variance explained by the
quantitative trait loci (QTL). However, a large number of single-nucleotide polymorphisms (SNPs) (genotype coded as 0, 1 and 2) in genome can
be obtained by SNP genotyping technology. Given the large number of SNPs, it is expected that every QTL or gene that affects quantitative traits
is in linkage disequilibrium (LD) with at least one SNP marker. Therefore, genomic selection can be traced back to all QTLs that affect traits

F I G U R E 2 Basic process of genomic selection in aquaculture. Reference population and candidate population are established from a number
of families. These individuals in reference population can be phenotyped for growth or disease resistance traits using phenotyping technology
(e.g., machine vision). Both candidate population and reference population are genotyped by genotyping technology (e.g., re-sequencing), and the
phenotype of candidate population is unknown. The genotypes can be represented by a variable (w), which takes the values 0 or 1 or
2 corresponding to one of the homozygotes, the heterozygote or the other homozygote. The statistical analysis of the reference population
estimates effects for each marker (x) using statistical method (e.g., GBLUP), and hence a prediction equation can be generated that combines all
the marker genotypes with their effects to predict the breeding value (GEBV) of each individual. This prediction equation can then be applied to
candidate population and excellent breeders are selected based on GEBV

advances in aquaculture genetics and breeding to date, as reviewed species with high-quality reference genomes, even species without
26
by Robledo et al. In particular, restriction site-associated DNA reference genomes can be assembled by de novo, and the assembled
sequencing (RAD-Seq) has been widely used to generate population- fragments can be used as reference sequences to develop markers.
level SNP data. Genotyping by sequencing techniques are suitable for Therefore, these methods have been widely used by researchers and
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SONG ET AL. 277

companies in relatively understudied non-model aquaculture species. between SNPs and QTLs (major sources of genetic variation), which
Vallejo et al.27 compared the prediction accuracy of RAD-Seq and can explain all genetic variants in the genome, thereby improving the
SNP chips for the bacterial cold water disease resistance of rainbow accuracy of selection. Third, GS can negate potential limiting effects
trout (Oncorhynchus mykiss) and found that although the marker den- of genotype by environment (G  E) interactions. Mulder49 reported
sity of the SNP chip was higher (40K for SNP chip vs. 10K for RAD- that GS was much better able to increase resilience and reduce envi-
Seq), the selection accuracy of the two technologies was comparable. ronmental sensitivity than traditional breeding schemes. Finally, GS is
In addition, they found that compared with the resequencing technol- expected to reduce inbreeding50 and increase in rate of genetic gain
ogy, the SNP chip technology required a lower cost and provided a at a set level of inbreeding.51 This is mainly because GS gives a more
faster detection speed. SNP chips have been widely used in livestock accurate prediction of the true genetic similarity between individuals
GS. Table 1 lists commercial SNP chips commonly used in aquaculture than pedigree data, which increases the difference between full-sib-
species, such as low-density Pacific Oyster Illumina GoldenGate 1K lings, thereby reducing the probability of their co-selection for seeding
chip (marker number 1K, commonly called the 1K chip), medium- and ultimately preventing inbreeding. Genetic progress (G) can be cal-
irσ g
density Atlantic salmon Affymetrix Axiom 50K chip and HD catfish culated using the following formula: G ¼ L , where i is the intensity
(Silurus spp.) Affymetrix Axiom 690 K chip. of selection, r is the accuracy of selection, σ g is the genetic standard
deviation and L is the generation interval. Based on the formula, GS
can accelerate genetic progress. Genomic selection offers diverse
2.3 | Advantages of GS advantages in different species. In dairy cattle, GS significantly
enhanced genetic progress mainly by reducing the generation interval
The advantages of GS are mainly reflected in the following four and improving the GEBV accuracy of candidate individuals.52 In pigs,
aspects. First, it does not rely on phenotypic information for candidate however, GS did not significantly shorten the generation interval due
individuals and candidate individuals are selected based on the GEBV, to the short breeding cycle, which improved GEBV accuracy and
which can distinguish the differences between individuals. Genomic accelerated genetic progress. Genomic selection is particularly suitable
selection can be performed as soon as DNA is available, which allows for selecting traits that are difficult to measure (disease resistance,
accurate selection in early stage of life, shortens the generation feed utilisation, reproductive traits, health traits and meat quality
interval,47 offers greater advantages in the selection of sex-restricted traits),53 for example, Lillehammer et al.54 demonstrates large
traits47 and low-heritability traits.48 Second, GS can explain all genetic potential for further genetic improvement of White spot syndrome
variations in the genome. A basic premise of GS is to exploit the LD virus resistance in the evaluated L. vannamei population using

TABLE 1 SNP chips commonly used in aquaculture species

Species Number of SNPs Platform technology Reference


Salmo salar 286,021 Affymetrix Axiom 28
200,000 Affymetrix Axiom 29
55,000 Affymetrix Axiom 30
5919 Illumina Infinium 31
Silurus spp. 250,113 Affymetrix Axiom 32
693,567 Affymetrix Axiom 33
Oncorhynchus kisutch 220,001 Affymetrix Axiom 34
Cyprinus caprio 250,000 Affymetrix Axiom 35
Crassostrea gigas and Ostrea edulis 14,950 Affymetrix Axiom 36
Penaeus monodon 6000 Illumina Infinium 37
Larimichthys crocea 579,400 Affymetrix Axiom 38
Paralichthys olivaceus 48,697 Affymetrix Axiom 39
Oreochromis niloticus 58,466 Affymetrix Axiom 40
65,000 ThermoFisher Axiom 41
Crassostrea gigas 1536 Illumina GoldenGate 42
190,420 Affymetrix Axiom 43
40,625 Affymetrix Axiom 36
Litopenaeus vannamei 8967 Illumina Infinium 44
Oncorhynchus mykiss 57,501 Affymetrix Axiom 45
Pinctada maxima 2782 Illumina Infinium 46
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
278 SONG ET AL.

GS. Compared with livestock, aquatic animals exhibit higher fecundity produce no effect, while the other fraction (1  π) produces an effect
and larger family size, which offers two major advantages. First, the drawn from a t-distribution.14 BayesCπ is similar to BayesB, except
closer relationship between reference and candidate populations that SNPs with an effect are assumed to show a normal distribution
leads to a higher prediction accuracy even at a lower SNP marker den- instead of a t-distribution.72 BayesCπ is computationally simpler to
sity, because related individuals share longer genome fragments. Sec- solve than BayesB, although it still yields almost the same accuracy.72
ond, traditional pedigree records are strictly not needed, because the BayesR is an extension of BayesCπ with multiple normal distributions,
relationships between individual fish are calculated based on the which allows reliable prediction under a range of genetic architec-
genetic marker information only. In addition, some aquatic animals, tures, including traits whose genetic variants produce a moderate
such as sturgeons, show a long breeding cycle and late sexual matu- effect.73 The advantage of the Bayesian method is that it assumes
rity. Specifically, cultured Russian sturgeons require 5–6 years to that markers have different variances, which is more consistent with
mature. The application of GS in sturgeons can improve the prediction the biological basis of complex traits; however, the disadvantage is
accuracy, especially for the traits that are difficult to measure, for that the calculation time is rather long. Recent studies have proven
example, caviar yield, so as to improve the genetic gain. that the Bayesian method is better than the GBLUP method for simu-
lated data74; however, for real data, Bayesian methods perform simi-
larly or slightly better than GBLUP method.18,47 MacLeod et al.75
2.4 | Methods of GEBV calculation proposed the BayesRC method using omics information, in which the
markers are divided into several groups based on prior information
GEBV calculation methods can be classified into the following three (≥2). For instance, markers within or close to the candidate gene are
categories. The first category is based on the BLUP theory. As one of assigned to the first group, and the other markers are assigned to the
the earliest proposed methods, ridge regression BLUP (RRBLUP)14 second group. Analysis is performed using BayesR for each group. In
first estimates the SNP marker effect values and then calculates the addition, Fernando et al.76 proposed a single-step Bayesian method,
GEBV of the candidate population. VanRaden55 proposed genomic which can simultaneously analyse both individuals with and without
BLUP (GBLUP) that can directly calculate the GEBV of the candidate genotype information. Compared with the conventional Bayesian
population and produces the same result as RRBLUP. Genomic BLUP method, which uses only animals with genotype information, this
replaces the conventional pedigree-based relationship matrix (A) method shows a higher prediction accuracy.
matrix of BLUP by constructing a genomic relationship matrix (G). The third category is based on machine-learning algorithms.
56
Misztal et al. proposed a method that combines phenotype, pedi- Thanks to rapid developments in this field, machine learning algo-
gree and genotype data into a single step evaluation. This method is rithms have found applications in genomic prediction.77 Commonly
called single-step GBLUP (ssGBLUP) and involves replacing the A in used algorithms include support vector machine (SVM),78 random for-
conventional BLUP with a realised relationship matrix (H), which com- est (RF),79 reproducing kernel Hilbert space (RKHS)80 and deep learn-
bines pedigree and genomic relationships. As a modification of ing (DL).81 These methods offer the flexibility to model multiple types
GBLUP, the superiority of ssGBLUP over GBLUP has been proven in of genetic effects, including additive and dominant effects as well as
several studies.57–64 Furthermore, the algorithm for proven and young epistasis effects. Machine learning methods primarily differ from con-
animals,65 which was proposed to make getting the inverse for mil- ventional statistical methods (based on BLUP and Bayesian theory) in
lions of animals simple and improve the computational efficiency of that they are nonparametric models providing tremendous flexibility
GBLUP and ssGBLUP. In addition, many other improved methods to adapt to the complex associations between data and output.82
based on GBLUP have been proposed. For instance, TABLUP pro- Overall, no current method offers absolute advantages in all situa-
posed by Zhang et al.66 weights G for different traits and offers obvi- tions, in practical breeding, it is necessary to evaluate the performance
ous advantages for traits with major genes. Moreover, the BLUPjGA of various statistical methods and then select the most suitable
method proposed by Zhang et al.67 divides the genomic relationship method. Genomic BLUP and Bayesian methods are widely used in
matrix into two parts, namely one marker with large effect and aquaculture species (Table 2). Table 3 summarises common GS
another with small effect, and assigns different weights to each part. methods.
68
The GFBLUP method proposed by Edwards et al. divides additive
variance in the mixed linear model into a genomic feature part and the
remaining part, providing high accuracy on simulation data,68 dairy 2.5 | Factors affecting the accuracy of genomic
cattle69 and pigs.70,71 prediction
The second category is based on the Bayesian theory. According
to variance distribution of the SNP effect, BayesA, BayesB, BayesCπ The accuracy of genomic prediction is measured using correlation
and BayesR models have been developed. In the BayesA model, the coefficient between the GEBV and true breeding values (TBV). How-
SNP effect is assumed to present a t-distribution,14 under which some ever, in actual breeding, TBV cannot be obtained. Typically, individuals
SNPs produce a larger effect than under the assumption of normality, with desired phenotypic performance and high reliability in terms of
because a t-distribution presents broader tails than a normal distribu- conventional breeding values are selected as candidates to form a val-
tion. In the BayesB model, a fraction (π) of the SNPs is assumed to idation population, and the accuracy of genomic prediction is
TABLE 2 Studies of genomic selection in aquaculture species

Number of
genotyped Accuracy of genomic
SONG ET AL.

Species individuals SNPs Traits Method prediction Reference

Atlantic Salmon (Salmo 622 111,908 Growth GBLUP 0.700 83


salar)
531/588 33,000 Sea lice resistance GBLUP 0.340–0.610 84
624 78,362 Sea lice resistance, body weight GBLUP 0.580–0.690 85
2404 36,616 Sea lice resistance GBLUP, BayesC, BayesLASSO 0.490–0.500 86
2392 49,684 Salmon rickettsial syndrome resistance GBLUP, RRBLUP, BayesC, BayesLASSO 0.368–0.424 87
1430 7168 Amoebic gill disease (AGD) resistance GBLUP 0.620–0.700 88
1333 53,109 AGD resistance GBLUP 0.490–0.720 89
610 78,362 Sea lice resistance GBLUP 0.530 90
563 49,726 Omega-3 fatty acid content GBLUP 0.321–0.563 91
1385 745,998 Sea lice resistance GBLUP, BayesC 0.605–0.676 92
2258/2345 35,479/32,579 Sea lice resistance, body weight GBLUP 0.390–0.780 93
Rainbow trout 636 10,052 Bacterial cold water disease (BCWD) resistance BayesB, BayesC, WssGBLUP 0.410–0.500 27
(Oncorhynchus mykiss) 1473 35,636 BCWD resistance BayesB, ssGBLUP, WssGBLUP 0.630–0.710 94
930 35,636 BCWD resistance BayesB, ssGBLUP, WssGBLUP 0.500–0.720 95
1934 27,490 Salmon rickettsial syndrome resistance GBLUP, ssGBLUP, BayesC, BayesLASSO 0.470–0.798 96
768 38,292 Infectious pancreatic necrosis virus disease resistance ssGBLUP 0.530–0.560 97
1044 35,397 Infectious haematopoietic necrosis virus disease ssGBLUP, WssGBLUP 0.300–0.390 98
resistance
1185/1137 35,900/33980 Columnaris disease resistance WssGBLUP 0.770–0.810 99
1624 34,640 Infectious haematopoietic necrosis virus disease ssGBLUP, WssGBLUP, BayesB, BayesC 0.330–0.380 100
resistance
1343 57,000 Female reproduction traits GBLUP 0.500–0.600 101
1568 35,303 Fillet yield and firmness ssGBLUP 0.260–0.380 102
1382 29,652 Fatty acid composition GBLUP 0.360–0.700 103
Large yellow croaker 500 29,748 Eviscerated weight, whole body weight GBLUP, BayesB 0.050–0.450 104
(Larimichthys crocea) 222 5,391,554 Visceral white spot disease resistance GBLUP 0.380 25
811 16,515 Parasitic ciliate Cryptocaryon irritans resistance BayesLASSO 0.200–0.600 105
326/565 34,994 Adapting to high plant protein diet GBLUP, BayesB 0.064–0.214 106
Gilthead sea bream 825 12,085 Pasteurellosis resistance RRBLUP, BayesA, BayesB, BayesC 0.380–0.460 107
(Sparus aurata)
1296 28,330 Pasteurellosis resistance GBLUP, BayesB, BayesC, BayesLASSO 0.545–0.569 108
841 21,773 Sparicotyle chrysophrii resistance GBLUP 0.550 109
1026 43,618 Pasteurellosis resistance GBLUP 0.610–0.640 110

(Continues)
279

17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
TABLE 2 (Continued)
280

Number of
genotyped Accuracy of genomic
Species individuals SNPs Traits Method prediction Reference

Nile Tilapia (Oreochromis 1238 32,306 Growth and fillet weight and fillet yield ssGBLUP 0.601–0.621 111
niloticus)
1444 18,960 Growth and fillet yield GBLUP 0.290–0.540 112
2684 35,745 Streptococcus agalactiae resistance GBLUP 0.730–0.840 113
755 48,431 Feed-efficiency traits GBLUP, ssGBLUP 0.400–0.500 114
1780 45,595 Francisellosis resistance GBLUP, BayesB, BayesC, BayesS 0.310–0.630 115
2472 50,690 Streptococcus agalactiae resistance BayesB, BayesC, BayesR, BayesS, GBLUP 0.560–0.780 116
Pacific white shrimp 205 6359 Growth RRBLUP, BayesA, BayesLASSO 0.284–0.413 117
(Litopenaeus vannamei)
200 23,049 Growth RRBLUP, BayesA, BayesLASSO 0.499–0.619 118
200 23,049 Acute hepatopancreatic necrosis disease resistance GBLUP 0.199–0.500 119
474 35,869 Feed efficiency traits GBLUP, ssGBLUP 0.566–0.706 120
Yellowtail kingfish 752 14,448 Growth GBLUP 0.440–0.830 121
(Seriola lalandi)
752 14,448 Skim fluke disease resistance GBLUP, KAML 0.165–0.432 122
Common carp (Cyprinus 1425 12,311 Growth GBLUP 0.660–0.710 123
carpio) 1425 15,615 Koi herpesvirus disease resistance RRBLUP, BayesA, BayesB, BayesC 0.510–0.550 124
Pacific oyster 820 23,388 Growth GBLUP 0.540–0.670 125
(Crassostrea gigas)
762 17,919 Ostreid herpesvirus resistance GBLUP 0.758 126
Japanese flounder 931 1,934,475 Edwardsiella tarda disease resistance GBLUP, BayesCπ 0.603–0.604 127
(Paralichthys olivaceus)
1099 4,978,724 Edwardsiella tarda disease resistance BayesB, ssGBLUP, WssGBLUP 0.600–0.660 128
European seabass 1538 9195 Viral nervous necrosis disease resistance RRBLUP, BayesA, BayesB, BayesC 0.660–0.710 129
(Dicentrarchus labrax)
972 50,136 Stress response, Vibrio anguillarum resistance, body GBLUP 0.310–0.600 130
weight
Yesso scallop 349 2364 Growth GBLUP, RRBLUP, BayesA, BayesB, LASSO 0.150–0.400 131
(Patinopecten
yessoensis)
Zhikong scallop (Chlamys 509 31,361 Growth GBLUP, BayesB, RKHS, SNN 0.370–0.580 132
farreri)
Channel catfish (Ictalurus 2911 54,837 Harvest and carcass weight ssGBLUP 0.240–0.380 133
punctatus)
Coho salmon 764 9389 Piscirickettsia salmonis disease resistance GBLUP, BayesC, ssGBLUP, WssGBLUP 0.299–0.807 134
(Oncorhynchus kisutch)
Yellow drum (Nibea 371 53,677 Body size-related traits BayesA, BayesB, BayesC, MMixp, GBLUP, CNN 0.145–0.412 135
albiflora)
Banana shrimp 562 9472 Hepatopancreatic parvovirus resistance and growth, GBLUP, BayesC, BayesCπ 0.420–0.760 136
(Fenneropenaeus carcass and quality traits
merguiensis)
SONG ET AL.

17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SONG ET AL. 281

evaluated using the correlation coefficient between GEBV and response

Reference

Abbreviations: Bayes-Alphabet (e.g., BayesA, BayesB, BayesC, BayesS, BayesCπ, BayesR, BayesLASSO), Bayesian methods with different assumptions of SNP effect distribution; CNN, convolutional neural network; GBLUP,
variables. Common response variables include the original phenotype,88

genomic best linear unbiased prediction; KAML, machine learning-based method incorporating cross-validation, multiple regression, grid search and bisection algorithms; MMixp, Bayesian using markov chain Monte Carlo-
corrected phenotype value59 and de-regressed proof.140,141

137

138

139

based Pareto principle; RKHS, reproducing kernel Hilbert space; RRBLUP, ridge regression best linear unbiased prediction; SNN, sparse neural networks; ssGBLUP, single-step GBLUP; WssGBLUP, weighted ssGBLUP.
The accuracy of genomic prediction is affected by several factors,
which are divided into two major types: uncontrollable and controlla-
ble factors. Uncontrollable factors include the average length of chro-
Accuracy of genomic

mosomes, number of genes affecting traits and heritability of traits.


For instance, Daetwyler et al.142 found that the accuracy of GEBV
0.230–0.310

0.400–0.550
prediction

gradually decreased with increase in the number of QTLs but


0.460

increased with increase in the heritability of traits. These uncontrolla-


ble factors lead to significant differences in the implementation
effects of GS in different species, populations and traits. Controllable
factors include reference population size, marker density, GEBV esti-
mation method and the relationship between the reference and candi-
GBLUP, BayesLASSO, RRBLUP, BayesA,

date populations. These factors can be controlled via artificial means


to improve the accuracy of GEBV, which is another key consideration
and work direction in GS.
BayesB, BayesC
GBLUP, BayesB

2.5.1 | Reference population size


Method

GBLUP

Phenotypic information for the reference population is essential for


estimating the GEBV; therefore, the size of the reference population
plays a crucial role in the accuracy of GS. The larger the reference
population, the richer the phenotypic information, the more pheno-
typic records corresponding to each marker, and the higher the accu-
racy of GS.48,143 Conversely, if the reference population is small,
marker effect value or chromosome fragment effect value cannot be
accurately estimated. Regardless of the heritability of traits, the accu-
Note: Before and after the slash (/) represent two different populations that were analysed separately.

racy of GS increases with increase in the size of the reference popula-


tion. For small reference populations, combining populations of the
Scuticociliatosis resistance

same breed or populations of related breeds could increase the accu-


racy of genomic prediction, as observed for Holstein populations in
EuroGenomics144 as well as North American consortia145 and pig
Heat tolerance

populations in China.71,143 Similar study has also been reported for


disease resistance in European sea bass (Dicentrarchus labrax) and the
Growth
Traits

gilthead sea bream (Sparus aurata), in which with the increase of refer-
ence population size, the accuracy of genome prediction is
improved.110 Our previous study has also clarified that as the refer-
ence population size decreased, the accuracy of genomic prediction
18,125

16,162

64,788

decreased in four aquaculture species (Atlantic salmon, common carp


SNPs

[Cyprinus carpio], sea bream and rainbow trout).146


Number of
genotyped
individuals

1349

1122

2.5.2 | Marker density


455
(Continued)

GS uses a whole-genome panel of dense markers simultaneously so


(Oplegnathus fasciatus)
Pacific abalone (Haliotis
Turbot (Scophthalmus

that all QTLs are in LD with at least one marker. Increase in marker
density increases the LD between markers and QTLs, and the resul-
discus hannai)

tant increase in the estimation accuracy of marker effect value or


Rock Bream
maximus)
TABLE 2

chromosome segment effect value improves the accuracy of


Species

GS. Generally, the higher the marker density, the higher the accuracy
of GS.147,148 Occasionally, however, the increase in marker density
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
282 SONG ET AL.

TABLE 3 Methods of genomic selection

Category Method Feature Reference


Based on the BLUP RRBLUP All SNPs have an effect, and effects follow a normal distribution 14
theory GBLUP Equivalent to RRBLUP, but constructs a genomic relationship matrix (G) 55
ssGBLUP Constructs a combined genotype–pedigree relationship matrix (H) 56
TABLUP Constructs a trait-specific relationship matrix (TA) based on marker 66
genotypes and their weights relative to the trait of interest
BLUPjGA Constructs a trait-specific relationship matrix (GA), which puts an optimal 67
weight on a subset of SNPs with the strongest effects
GFBLUP Divides additive variance in the mixed linear model into a genomic 68
feature part and the remaining part
Based on the BayesA All SNP effects follow Student's t-distribution 14
Bayesian theory BayesB A fraction (π) of the SNPs with zero effect and remainder (1  π) with a 14
t-distribution
BayesC/Cπ A fraction (π) of SNPs with zero effect and the remainder (1  π) with a 72
normal distribution, π was estimated from the data (BayesCπ)
BayesR SNP effects are assumed to follow a mixture of normal distributions 73
BayesRC Allocates a priori all genotyped variants into (≥2) classes according to 75
their genome sites; all variants within or close to the candidate genes
are allocated to Class I and all other variants to Class II
Single-step Uses individuals with and without genotype information at the same time 76
Bayesian method
Based on machine RF Uses multiple decision trees and bootstrap aggregation to perform 79
learning classification or regression tasks
algorithms SVM Relies on the construction of multidimensional hyperplanes that separate 78
similarly labelled objects into linearly separable sets
RKHS Uses a nonlinear kernel function to map the data to a higher dimensional 80
kernel space
DL Uses neural networks with a large number of simple computational units, 81
growing into more complex modelling systems

Abbreviations: Bayes-Alphabet (e.g., BayesA, BayesB and BayesCπ), Bayesian methods with different assumptions of SNP effect distribution; BLUPjGA,
BLUP-given genetic architecture; DL, deep learning; RF, random forest; GBLUP, genomic best linear unbiased prediction; GFBLUP, genomic feature BLUP;
RKHS, reproducing kernel Hilbert space; RRBLUP, ridge regression best linear unbiased prediction; ssGBLUP, single-step GBLUP; SVM, support vector
machine; TABLUP, BLUP including a trait-specific relationship matrix.

induces some noise markers, which interfere with the accuracy of the the different assumptions of various calculation methods, their scope
GEBV estimation. For example, Pérez-Enciso et al.149 stated that using of application varies. In addition, species type, population size and
whole-genome sequencing data did not increase prediction accuracy traits lead to differences in evaluation methods. Therefore, the evalu-
150
compared to HD array data. van Binsbergen et al. reported that GP ation method must be selected according to the desired scenario and
with imputed whole-genome sequencing data did not lead to a higher its effects should be verified to maximise the advantages of GS.
prediction accuracy, compared to the HD array data from more than
5000 Holstein–Friesian bulls. Similar results were also reported by
Gong et al.,138 in which models with 1000 SNPs will provide predic- 2.5.4 | Relationship between reference and
tive ability equivalent to that by using all the available SNPs for com- candidate populations
plex growth-related traits in Rock Bream (Oplegnathus fasciatus). In
addition, the cost of chips with different densities and sequencing Generally, the closer the relationship between the reference and can-
must be considered to balance efficiency and cost. didate populations, the higher the accuracy of GS.58,151,152 In aquatic
animals, the close relationship between the reference and candidate
populations yields a high GS accuracy. Saura et al.153 reported that
2.5.3 | GEBV estimation methods the current effective population size is small (equal to or less than
50 fish) for turbot (Scophthalmus maximus), gilthead sea bream,
The GEBV estimation method is the core of GS. The commonly used European sea bass and common carp, which indicates the often close
GEBV estimation methods are introduced above (Table 3). Owing to genetic relationship between populations of most aquaculture species.
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SONG ET AL. 283

Fraslin et al.93 confirmed the importance of close genetic relationships genetic gain for amoebic gill disease resistance and harvest weight
between training and selection populations in salmon breeding pro- attributed to the implementation of GS in Tasmanian Atlantic salmon,
grammes. The accuracy of GS decreased with increase in the number whose value was estimated at AU$1 and AU$5 million per year,
of generations between the candidate and reference populations.152 respectively. Jerry et al.159 also indicated that higher levels of genetic
This is mainly because the degree of LD between molecular markers gain for barramundi (Lates calcarifer) growth-related traits would result
and QTLs affecting the traits decreases with increase in the number from the application of GS (GBLUP) than pedigree-based selection
of generations, which in turn affects the accuracy of GEBV estima- (BLUP). These studies suggest that GS has great potential to improve
tion.152 Therefore, the reference population should be updated regu- the rate of genetic gain in aquaculture species.
larly rather than statically. Most GS studies in aquaculture species used the GBLUP and
Bayesian methods to estimate the GEBV (Table 2), for example, Joshi
et al.115 reported that GBLUP and Bayesian methods (BayesB and
3 | P R O G R E S S O F R E S EA RC H O N G S I N BayesC) could increase the predictive ability for resistance to Franci-
A Q U A C U L T U RE B RE E D I N G sellosis in commercial Nile tilapia population, compared to pedigree-
based models. Furthermore, incorporating preselected potential causal
GS proposed almost 20 years ago has revolutionised design and markers into the GS model is an effective strategy to improve predic-
implementation of plant and animal breeding programmes, and has tion accuracy. For instance, Dong et al.160 reported that the accuracy
become a promising tool for plant and animal breeding programmes of genomic prediction for body weight-related traits in large yellow
to accelerate the rate of genetic gain.154,155 Since the implementation croaker could be improved by preselecting SNPs based on SNP
of GS in the United States, the rates of genetic gain per year have effects. Yoshida and Yáñez161 reported that in rainbow trout, the
increased from 50% to 100% for medium heritability traits (milk yield, accuracy of genomic prediction for growth under chronic thermal
milk fat and milk protein) and 300% to 400% for low heritability traits stress could be improved using preselected variants from GWAS.
(somatic cell score, productive life and daughter pregnancy rate) in Mapping and understanding the causative variants that have an
154
dairy cattle. To date, GS has been conducted in about 20 aquacul- impact on complex traits has potential additional benefits for increas-
ture species, among these aquaculture species, GS was first studied in ing rates of genetic gain in breeding through improved selection accu-
Atlantic salmon, and its application was realised thanks to the devel- racy, for example, Ni et al.162 reported that using only SNPs in or
opment of the first HD SNP array and demonstration of its utility and around a gene from whole-genome sequencing data increased the
accuracy in predicting breeding values in a typical salmon breeding accuracy of GS in laying chickens. However, identifying causative vari-
programme.28,156 With the development of whole-genome sequenc- ants of target traits is a knowledge gap to help bridge relationships
ing technology and reduction in the cost of re-sequencing, research between phenotypes and genotypes in most aquaculture species. In
on GS in aquaculture species has gradually advanced. Genomic selec- addition, several studies have reported the use of machine learning
tion has been applied to the complex traits of several important aqua- methods (e.g., SVM, RF and DL) for genomic prediction of disease
culture species (Table 2), including Atlantic salmon,83,84,90 rainbow resistance and growth traits of aquaculture species, and these
trout,27,95,101 large yellow croaker,105 Nile tilapia,41,112 Pacific white methods have been shown to improve the accuracy of genomic
shrimp,54 Japanese flounder (Paralichthys olivaceus)128 and rock prediction.163–165 In general, as assumptions and information used
138
bream, and so forth. Studies in aquaculture species have shown vary with models, no model performs best in all situations and GBLUP
that the accuracy of genomic predictions varies from 0.165 to 0.840 and Bayesian models are more commonly used in aquaculture species.
for traits associated with disease resistance, from 0.145 to 0.830 for
traits associated with growth and from 0.064 to 0.780 for traits asso-
ciated with feed efficiency, production yield and female reproduction, 4 | FU T U R E D I R E C T I O N S
the number of genotyped individuals and SNPs used in GS ranged
from 200 to 2911 and 2364 to 5,391,554, respectively (Table 2). In 4.1 | Developing accurate phenotyping techniques
addition, Houston et al.17 reviewed several cases in which the accu-
racy of genomic prediction was higher than that of conventional pre- It is crucial to obtain accurate phenotypes in breeding programme
diction based on pedigree information in aquaculture species. On because the accuracy of trait measurements directly affects the accu-
average, the prediction accuracy increased by 22% for disease resis- racy of GS and thus the genetic gain per generation. Traditional man-
tance traits and by 24% for growth-related traits. Moreover, Boudry ual measurement methods are easily affected by subjective factors
157
et al. reviewed the current status of and developments in GS in such as experience, habits, preferences, and external environmental
several major aquaculture species, including Atlantic salmon, rainbow interference, which make the measurement process time-consuming
trout, Atlantic cod (Gadus morhua), American catfish, Pacific oyster, and labour-intensive. In addition, aquatic animals are often highly sen-
European sea bass and gilthead sea bream, in member countries of sitive and easily coerced animals, and manual measurement can easily
the International Council for the Exploration of the Sea, which pointed cause harm to them, even lead to disease and death, affecting the nor-
out that the potential of GS is clear. For the practical application value mal growth of aquatic animals. Therefore, collecting such data directly
of GS, Verbyla et al.158 reported significant increases in the rate of on selected candidates in the breeding nucleus and their relatives in
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
284 SONG ET AL.

the test or production environments can limit genetic progress in 4.3 | Considering genotype by environment
breeding programmes. Yang et al.166 and Zenger et al.18 reviewed the (G  E) interactions in GS
application of machine learning and artificial intelligence in phenotyp-
ing of aquaculture species. Machine vision systems have been exten- Typically, economically important traits are complex and influenced by
sively explored, and high concordance (>97%) between aquaculture genes, the environment and G  E interaction. Different production
animal and image analysis and phenotypic measurements has been systems and climates can induce G  E interactions on populations,
167–169
demonstrated. Other emerging aquaculture phenotyping tech- and consideration of these interactions in breeding decisions is imper-
niques include hyperspectral imaging and near-infrared spectroscopy, ative. A G  E interaction is defined as different genotypes reacting
which can quantify the physical (e.g., size, weight and colour) and unequally to environment changes.173 Ignoring possible G  E inter-
chemical (e.g., protein, fat and unsaturated fatty acids) attributes of actions may reduce genetic gains. In aquaculture production systems,
169,170
aquatic animals with high prediction accuracy (>80%). Combina- breeding companies typically supply fish to various farms worldwide,
tion of high-throughput and high-resolution phenotyping in breeding either directly or via hatcheries. The environment of breeding farms
programme with GS is expected to allow for effective genetic may be rather different from that of commercial production farms
improvement of aquaculture species. (e.g., altitude, temperature, daylight, water quality, salinity and
farm management systems). Many studies have shown moderate or
strong G  E interactions in fish.174–181 In addition, Mulder49
4.2 | Developing strategies to reduce GS cost expounded that GS offers great opportunities to exploit G  E to
increase resilience of animals. Jerry et al.159 reported that GBLUP pro-
The main challenge for further implementation of GS is associated vided greater accuracy for EBV prediction, higher expected genetic
with the costs and benefits of genotyping. In aquaculture breeding gains and more effectively accounted for G  E interactions for the
programmes, genotyping of thousands of animals in each generation cohort of fish reared in the two different commercial environments.
is essential, which is expensive. Therefore, cost-effective strategies Therefore, G  E interactions must be considered in GS models for
must be developed to translate the benefits of GS into most aquacul- aquaculture species. Multi-trait and reaction norm models are the two
ture species. Different strategies of selecting SNPs for low-density common methods for handling G  E interactions in genetic evalua-
panels have been reported to reduce the cost of GS. For instance, tions. Falconer et al.173 detected G  E interactions using a multi-trait
Robledo et al.88 evaluated the effect of reduced SNP density based model, in which the same trait in different environments was regarded
on their minor allele frequency and their even position in the genome as different traits and genetic correlation between traits in different
on prediction accuracy in Atlantic salmon, and a reduction in marker environments was a measure of G  E. The multi-trait model could
density to 2000 SNPs was sufficient to obtain high accuracy. Kriari- capture all the G  E interactions between these environments, while
dou et al.171 investigated the accuracy of genomic prediction by ran- its computational requirements will increase with the number of envi-
domly selecting markers from SNP chips in four aquaculture species and ronments. The application scenario of multi-trait model is when the
found that SNP densities between 1000 and 2000 provided selection environment can be split into several discrete classes.182,183 For reac-
accuracies close to those obtained with HD genotyping. Our previous tion norm model, trait expression is on a continuous environmental
study on four species (Atlantic salmon, sea bream, common carp and scale and breeding value and genetic parameters change gradually
rainbow trout) showed that reducing the density of SNP panels to 3 K along the environmental scale. At different locations on the environ-
was sufficient to obtain accuracies similar to those obtained using the mental scale, traits may have different means and (co)variances, and
whole dataset, and the cost of GS with reduced SNP density was esti- there is a genetic correlation between trait expression at different
mated to be 50% lower than that of genotyping of all animals with HD points of the environment scale.184 However, reaction norm model
panels. In addition, when the reference population size was reduced by captures only part of the G  E interactions in contrast to the multi-
10% uniformly from a full-sib family, the accuracy of genomic prediction trait model because it needs to accommodate a continuous range of
remained nearly unchanged, and the cost was reduced by 8%.146 environmental values and cannot select excellent individuals using the
In addition, genotype imputation, GBS and low-coverage unique estimated breeding value in actual breeding.185,186 The main
sequencing have also been used to reduce GS costs. First, genotype application of reaction norm model is when the environmental vari-
imputation, which utilises LD information from haplotypes of a known ables explaining the G  E interactions are continuous.187 Therefore,
reference panel to predict missing genotypes and is achieved by in aquatic animal breeding, the corresponding model should be
imputing low- to high-density SNP markers, and many studies have selected for G  E analysis according to the environment.
reported that the use of imputation-based HD SNP markers could
improve the accuracy of GS without increasing the cost.90,94,172 Sec-
ond, as mentioned above, GBS and low-coverage sequencing are also 4.4 | Considering allele dosage in genomic
likely to help reduce GS costs by reducing genotyping costs. There- prediction for polyploid fish
fore, methods of low-cost genotyping and low-density panels com-
bined with genotype imputation hold potential and will help transfer Among vertebrates, fish have abundant polyploidization. The most dis-
the benefits to aquaculture industries. tinctive and recognised autopolyploid fish include Salmonidae188,189 and
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SONG ET AL. 285

Acipenseridae.190–192 In particular, within Acipenseridae, different stur- rate of genetic gain after a few and many generations of selection.214
geon species show diverse ploidy levels. While sterlets (Acipenser ruthe- However, the application of genome editing for improving complex traits
nus) are tetraploid, and most sturgeons are octoploid (e.g., Russian requires the discovery of quantitative trait nucleotides associated with
sturgeon and Siberian sturgeon [Acipenser baerii]).190–192 A previous traits, which in turn requires larger datasets than currently available. Many
study showed that polyploidy affected phenotypes through allelic dosage SNPs associated with traits obtained from GWASs, family-based QTL
(additive effect of multiple copies of the same alleles).193 Therefore, sim- mapping and gene function annotation can be rapidly fixed in breeding
plifying a polyploid genotype as a diploid genotype may induce bias in populations through genome editing. Based on this, the implementation
genomic predictions due to higher-order interactions and allele substitu- of GS can further accelerate genetic improvement in aquaculture species.
tion effects. In this context, GS methods must be modified for the
improvement of tetraploid or octoploid performance, since available
studies and theories are largely based on diploids. There are few studies 5 | CONC LU SION
on genomic prediction for autopolyploid species using allele dosage,
although majority of these are related to plant breeding.194–196 In a Genomic selection, a technology that uses markers covering the
previous study, we compared the impact of ploidy parametrization on whole genome to predict genomic estimated breeding values of indi-
prediction accuracy in a simulated polyploid sturgeon population using viduals, has revolutionised livestock breeding. In recent years, many
the GBLUP and RKHS methods. Our results showed that a higher GS studies have been conducted on aquaculture species, which have
accuracy of genomic prediction could be obtained by considering allele shown that GS produces higher prediction accuracy than traditional
dosage in autopolyploidy.197 Currently, with the development of pedigree-based method. However, as assumptions and information
sequencing technologies, genotyping of polyploid aquaculture species is used vary with models, no model performs best in all situations and
possible. Thus, allele dosage should be considered in genomic prediction GBLUP and Bayesian models are more commonly used in aquaculture
for polyploid, particularly hyperploid species (e.g., Russian sturgeon and species. In the future, novel phenotyping (e.g., machine vision) and
Siberian sturgeon).197 genomics (e.g., low-coverage re-sequencing) technologies, coupled
with related advances in statistical methods and computational effi-
ciency, will further facilitate the use of GS in aquaculture. In addition,
4.5 | Integrating multi-omics data and genome GS will become more widespread as phenotyping and genomics tech-
editing into GS nologies become more affordable, coupled with tailor-made
approaches to apply low-cost methods to aquaculture species. Due to
Currently, GS strategies mainly rely on direct association analysis the large differences in the production environments, it is possible
between phenotypes and SNPs in the genome or prior biological that G  E interaction is important for the breeding of aquaculture
information in public databases.66,68,71,198,199 However, the genetic species, thus G  E should be considered in GS model, which can
links between phenotype and genome variants are too complex to effectively improve fish performance across multiple environments
determine directly at the genome sequencing level. At present, multi- will aid farmers in diverse environmental conditions to produce food
omics data (e.g., genomics, transcriptomics, proteomics and metabolo- for the growing human population. Moreover, with the development
mics) can be obtained for genomic predictions, allowing to uncover of sequencing technologies, allele dosage should be considered in GS
genotype–phenotype relationships and provide a ridge between model to further improve the accuracy of GS for polyploid species. In
organismal phenotype and genome variation that cannot be readily addition, integrating multi-omics data (e.g., genomics, transcriptomics,
captured at the genome sequence level. Previous studies have used proteomics and metabolomics) and genome editing into GS can fur-
omics data for genomic prediction of complex traits in humans,200,201 ther accelerate genetic improvement in aquaculture species in the
202–205 206,207
plants and model animals. Therefore, with the accumula- future. With continuous progress of related technologies, GS is
tion of data, application of multi-omics data in GS is anticipated to expected to become widespread in aquaculture breeding and produce
more comprehensively and reliably understand the physiological a far-reaching impact in this field.
mechanisms and genetic bases of character formation.
Moreover, gene and genome editing technologies, including AUTHOR CONTRIBU TIONS
ZFNs, TALLENS and CRISPR/Cas9, are promising tools for accelerat- Hailiang Song: Writing – original draft. Tian Dong: Writing – review
ing genetic improvement.208,209 Genome editing is widely used to and editing. Xiaoyu Yan: Writing – review and editing. Wei Wang:
analyse the functions of candidate genes in aquaculture species.210,211 Writing – review and editing. Zhaohui Tian: Writing – review and
By the knocking out and knocking in of different alleles of candidate editing. Ai Sun: Writing – review and editing. Ying Dong: Writing –
genes, the favourite alleles for important traits can be identified.212 In review and editing. Hua Zhu: Writing – review and editing. Hongxia
addition, gene editing can be integrated with GS in breeding. Genomic Hu: Writing – review and editing.
selection of gene-edited animals can further increase selection accu-
racy and accelerate genetic progress.213 A previous simulation study AC KNOW LEDG EME NT S
on Holstein cattle showed that in comparison to GS only, editing all This work was supported by Beijing Natural Science Foundation
sires for 20 edited quantitative trait nucleotides per sire doubled the (6222014), Beijing Joint Research Program for Germplasm
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
286 SONG ET AL.

Innovation and New Variety Breeding; (G20220628008), the 16. Moen T, Torgersen J, Santi N, et al. Epithelial cadherin determines
Youth Foundation of Beijing Academy of Agriculture and Forestry resistance to infectious pancreatic necrosis virus in Atlantic salmon.
Genetics. 2015;200(4):1313-1326.
Sciences (QNJJ202105) and China Agriculture Research System of
17. Houston RD, Bean TP, Macqueen DJ, et al. Harnessing genomics to
MOF and MARA (CARS-45-32). fast-track genetic improvement in aquaculture. Nat Rev Genet. 2020;
21(7):389-409.
CONF LICT OF IN TE RE ST 18. Zenger KR, Khatkar MS, Jones DB, Khalilisamani N, Jerry DR,
Raadsma HW. Genomic selection in aquaculture: application, limita-
The authors declare no conflict of interest.
tions and opportunities with special reference to marine shrimp and
pearl oysters. Front Genet. 2019;9:693.
DATA AVAI LAB ILITY S TATEMENT 19. Lien S, Koop BF, Sandve SR, et al. The Atlantic salmon genome pro-
Since this is a review paper, there is no data available. All information vides insights into rediploidization. Nature. 2016;533(7602):
200-205.
can be found in the cited references.
20. Wang Y, Lu Y, Zhang Y, et al. The draft genome of the grass carp
(Ctenopharyngodon idellus) provides insights into its evolution and
ORCID vegetarian adaptation. Nat Genet. 2015;47(6):625-631.
Hailiang Song https://orcid.org/0000-0003-4915-0002 21. Zhang X, Yuan J, Sun Y, et al. Penaeid shrimp genome provides
insights into benthic adaptation and frequent molting. Nat Commun.
2019;10:356.
RE FE R ENC E S
22. Yan X, Nie H, Huo Z, et al. Clam genome sequence clarifies the
1. FAO. The State of World Fisheries and Aquaculture: Sustainability in molecular basis of its benthic adaptation and extraordinary Shell
Action. FAO; 2020. color diversity. iScience. 2019;19:1225-1237.
2. Gjedrem T, Robinson N, Rye M. The importance of selective breed- 23. Yang R, Guo X, Zhu D, et al. Accelerated deciphering of the genetic
ing in aquaculture to meet future demands for animal protein: a architecture of agricultural economic traits in pigs using a low-
review. Aquaculture. 2012;350:117-129. coverage whole-genome sequencing strategy. Gigascience. 2021;
3. Clavelle T, Lester SE, Gentry R, Froehlich HE. Interactions and man- 10(7):giab048.
agement for the future of marine aquaculture and capture fisheries. 24. Teng J, Zhao C, Wang D, et al. Assessment of the performance of
Fish Fish. 2019;20(2):368-388. different imputation methods for low-coverage sequencing in Hol-
4. Gjedrem T, Rye M. Selection response in fish and shellfish: a review. stein cattle. J Dairy Sci. 2022;105(4):3355-3366.
Rev Aquac. 2018;10(1):168-179. 25. Zhang WJ, Li WB, Liu GJ, et al. Evaluation for the effect of low-
5. Shen Y, Yue G. Current status of research on aquaculture genetics coverage sequencing on genomic selection in large yellow croaker.
and genomics-information from ISGA 2018. Aquac Fish. 2019;4(2): Aquaculture. 2021;534:736323.
43-47. 26. Robledo D, Palaiokostas C, Bargelloni L, Martinez P, Houston R.
6. Henderson CR. Best linear unbiased estimation and prediction under Applications of genotyping by sequencing in aquaculture breeding
a selection model. Biometrics. 1975;31(2):423-447. and genetics. Rev Aquac. 2018;10(3):670-682.
7. Zhai S, Yang B, Zhang F, Li Q, Liu S. Estimation of genetic parameters 27. Vallejo RL, Leeds TD, Fragomeni B, et al. Evaluation of genome-
for resistance to Vibrio alginolyticus infection in the Pacific oyster enabled selection for bacterial cold water disease resistance using
(Crassostrea gigas). Aquaculture. 2021;538:736545. progeny performance data in rainbow trout: insights on genotyping
8. Li QQ, Sun QF, Liu Q, Cheng YX, Wu XG. Estimation of genetic methods and genomic prediction models. Front Genet. 2016;7:96.
parameters for carotenoid traits in Chinese mitten crab, Eriocheir 28. Houston RD, Taggart JB, Cezard T, et al. Development and valida-
sinensis, females. Aquaculture. 2021;532:735990. tion of a high density SNP genotyping array for Atlantic salmon
9. Noble TH, Coman GJ, Wade NM, et al. Genetic parameters of gill- (Salmo salar). BMC Genomics. 2014;15:90.
associated virus infection and body weight under commercial condi- 29. Yanez JM, Naswa S, Lopez ME, et al. Genomewide single nucleotide
tions in black tiger shrimp, Penaeus monodon. Aquaculture. 2020;528: polymorphism discovery in Atlantic salmon (Salmo salar): validation
735580. in wild and farmed American and European populations. Mol Ecol
10. Barria A, Trinh TQ, Mahmuddin M, Benzie JAH, Chadag VM, Resour. 2016;16(4):1002-1011.
Houston RD. Genetic parameters for resistance to Tilapia Lake Virus 30. Bangera R, Thorland I, Hillestad B, et al. Accuracy of genomic predic-
(TiLV) in Nile tilapia (Oreochromis niloticus). Aquaculture. 2020;522: tions for yield traits, flesh colour and fillet fat in Atlantic salmon
735126. (Salmo salar). Paper presented at: Proceedings of the World Con-
11. Gjerde B, Mahapatra KD, Reddy PVGK, et al. Genetic parameters for gress on Genetics Applied to Livestock Production; 2018.
growth and survival in rohu carp (Labeo rohita). Aquaculture. 2019; 31. Lien S, Gidskehaug L, Moen T, et al. A dense SNP-based linkage map
503:381-388. for Atlantic salmon (Salmo salar) reveals extended chromosome
12. Song H, Xu S, Luo K, et al. Estimation of genetic parameters for homeologies and striking differences in sex-specific recombination
growth and egg related traits in Russian sturgeon (Acipenser guelden- patterns. BMC Genomics. 2011;12:615.
staedtii). Aquaculture. 2022;546:737299. 32. Liu S, Sun L, Li Y, et al. Development of the catfish 250K SNP array
13. Soller M, Beckmann JS. Genetic polymorphism in varietal identifica- for genome-wide association studies. BMC Res Notes. 2014;7:135.
tion and genetic improvement. Theor Appl Genet. 1983;67(1):25-33. 33. Zeng QF, Fu Q, Li Y, et al. Development of a 690 K SNP array in cat-
14. Meuwissen THE, Hayes BJ, Goddard ME. Prediction of total genetic fish and its application for genetic mapping and validation of the ref-
value using genome-wide dense marker maps. Genetics. 2001; erence genome sequence. Sci Rep. 2017;7:40347.
157(4):1819-1829. 34. Martine V, Naish K, Bravo C, et al. Development of a high-density
15. Moen T, Baranski M, Sonesson AK, Kjøglum S. Confirmation and genomic platform for Coho salmon and its use in population assess-
fine-mapping of a major QTL for resistance to infectious pancreatic ment in Chile and North America. Paper presented at: Proceedings
necrosis in Atlantic salmon (Salmo salar): population-level associa- of the World Congress on Genetics Applied to Livestock Production;
tions between markers and trait. BMC Genomics. 2009;10(1):368. 2018.
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SONG ET AL. 287

35. Xu J, Zhao ZX, Zhang XF, et al. Development and evaluation of the 54. Lillehammer M, Bangera R, Salazar M, et al. Genomic selection for
first high-throughput SNP array for common carp (Cyprinus carpio). white spot syndrome virus resistance in whiteleg shrimp boosts survival
BMC Genomics. 2014;15:307. under an experimental challenge test. Sci Rep. 2020;10:20571.
36. Gutierrez AP, Turner F, Gharbi K, et al. Development of a 55. VanRaden PM. Efficient methods to compute genomic predictions.
medium density combined-species SNP array for Pacific and J Dairy Sci. 2008;91(11):4414-4423.
European oysters (Crassostrea gigas and Ostrea edulis). G3. 2017; 56. Misztal I, Legarra A, Aguilar I. Computing procedures for genetic
7(7):2209-2218. evaluation including phenotypic, full pedigree, and genomic informa-
37. Baranski M, Gopikrishna G, Robinson NA, et al. The development of tion. J Dairy Sci. 2009;92(9):4648-4655.
a high density linkage map for black Tiger shrimp (Penaeus monodon) 57. Christensen OF, Madsen P, Nielsen B, Ostersen T, Su G. Single-step
based on cSNPs. PloS One. 2014;9(1):e85413. methods for genomic evaluation in pigs. Animal. 2012;6(10):1565-
38. Zhou T, Chen B, Ke Q, et al. Development and evaluation of a high- 1571.
throughput single-nucleotide polymorphism array for large yellow 58. Gao HD, Christensen OF, Madsen P, et al. Comparison on genomic
croaker (Larimichthys crocea). Front Genet. 2020;11:571751. predictions using three GBLUP methods and two single-step blending
39. Zhou Q, Chen YD, Lu S, Liu Y, Chen SL. Development of a 50K SNP methods in the Nordic Holstein population. Genet Sel Evol. 2012;44:8.
array for japanese flounder and its application in genomic selection 59. Guo X, Christensen OF, Ostersen T, Wang Y, Lund MS, Su G.
for disease resistance. Engineering. 2020;7:406-411. Improving genetic evaluation of litter size and piglet mortality for
40. Joshi R, Arnyasi M, Lien S, Gjøen HM, Alvarez AT, Kent MP. A high both genotyped and nongenotyped individuals using a single-step
density linkage map for Nile tilapia. Paper presented at: Proceedings method. J Anim Sci. 2015;93(2):503-512.
of the World Congress on Genetics Applied to Livestock Production; 60. Li XJ, Wang S, Huang J, Li LY, Zhang Q, Ding XD. Improving the
2018. accuracy of genomic prediction in Chinese Holstein cattle by using
41. Penaloza C, Robledo D, Barria A, et al. Development and validation one-step blending. Genet Sel Evol. 2014;46:66.
of an open access SNP Array for Nile tilapia (Oreochromis niloticus). 61. Su G, Madsen P, Nielsen US, et al. Genomic prediction for Nordic
G3. 2020;10(8):2777-2785. Red Cattle using one-step and selection index blending. J Dairy Sci.
42. Hedgecock D, Shin G, Gracey AY, Van Den Berg D, Samanta MP. 2012;95(2):909-917.
Second-generation linkage maps for the Pacific oyster Crassostrea 62. Song HL, Zhang JX, Zhang Q, Ding XD. Using different single-step
gigas reveal errors in assembly of genome scaffolds. G3. 2015;5(10): strategies to improve the efficiency of genomic prediction on body
2007-2019. measurement traits in pig. Front Genet. 2019;9:730.
43. Qi HG, Song K, Li CY, et al. Construction and evaluation of a high- 63. Aguilar I, Misztal I, Johnson DL, Legarra A, Tsuruta S, Lawlor TJ. Hot
density SNP array for the Pacific oyster (Crassostrea gigas). PloS One. topic: a unified approach to utilize phenotypic, full pedigree, and
2017;12(3):e0174007. genomic information for genetic evaluation of Holstein final score.
44. Jones DB, Jerry DR, Khatkar MS, et al. A comparative integrated J Dairy Sci. 2010;93(2):743-752.
gene-based linkage and locus ordering by linkage disequilibrium map 64. Christensen OF, Lund MS. Genomic prediction when some animals
for the Pacific white shrimp, Litopenaeus vannamei. Sci Rep. 2017;7: are not genotyped. Genet Sel Evol. 2010;42:2.
10360. 65. Misztal I, Legarra A, Aguilar I. Using recursion to compute the
45. Palti Y, Gao G, Liu S, et al. The development and characterization of inverse of the genomic relationship matrix. J Dairy Sci. 2014;97(6):
a 57K single nucleotide polymorphism array for rainbow trout. Mol 3943-3952.
Ecol Resour. 2015;15(3):662-672. 66. Zhang Z, Liu JF, Ding XD, Bijma P, de Koning DJ, Zhang Q. Best lin-
46. Jones DB, Jerry DR, Foret S, Konovalov DA, Zenger KR. Genome- ear unbiased prediction of genomic breeding values using a trait-
wide SNP validation and mantle tissue transcriptome analysis in the specific marker-derived relationship matrix. Plos One. 2010;5(9):
silver-lipped pearl oyster, Pinctada maxima. Mar Biotechnol. 2013; e12648.
15(6):647-658. 67. Zhang Z, Erbe M, He J, et al. Accuracy of whole-genome prediction
47. Hayes BJ, Bowman PJ, Chamberlain AJ, Goddard ME. Invited using a genetic architecture-enhanced variance-covariance matrix.
review: Genomic selection in dairy cattle: progress and challenges. G3. 2015;5(4):615-627.
J Dairy Sci. 2009;92(2):433-443. 68. Edwards SM, Sorensen IF, Sarup P, Mackay TFC, Sorensen P. Geno-
48. Goddard ME, Hayes BJ. Mapping genes for complex traits in domes- mic prediction for quantitative traits is improved by mapping vari-
tic animals and their use in breeding programmes. Nat Rev Genet. ants to gene ontology categories in Drosophila melanogaster.
2009;10(6):381-391. Genetics. 2016;203(4):1871-1883.
49. Mulder HA. Genomic selection improves response to selection in 69. Fang LZ, Sahana G, Ma PP, et al. Use of biological priors enhances
resilience by exploiting genotype by environment interactions. Front understanding of genetic architecture and genomic prediction of
Genet. 2016;7:178. complex traits within and between dairy cattle breeds. BMC Geno-
50. de Roos APW, Schrooten C, Veerkamp RF, van Arendonk JAM. mics. 2017;18:604.
Effects of genomic selection on genetic improvement, inbreeding, 70. Sarup P, Jensen J, Ostersen T, Henryon M, Sorensen P. Increased
and merit of young versus proven bulls. J Dairy Sci. 2011;94(3): prediction accuracy using a genomic feature model including prior
1559-1567. information on quantitative trait locus regions in purebred Danish
51. Scott BA, Haile-Mariam M, Cocks BG, Pryce JE. How genomic selec- Duroc pigs. BMC Genet. 2016;17:11.
tion has increased rates of genetic gain and inbreeding in the 71. Song H, Ye S, Jiang Y, Zhang Z, Zhang Q, Ding X. Using imputation-
Australian national herd, genomic information nucleus, and bulls. based whole-genome sequencing data to improve the accuracy of
J Dairy Sci. 2021;104(11):11832-11849. genomic prediction for combined populations in pigs. Genet Sel Evol.
52. García-Ruiz A, Cole JB, VanRaden PM, Wiggans GR, Ruiz-Lo  pez FJ, 2019;51(1):58.
Van Tassell CP. Changes in genetic selection differentials and gener- 72. Habier D, Fernando RL, Kizilkaya K, Garrick DJ. Extension of the
ation intervals in US Holstein dairy cattle as a result of genomic bayesian alphabet for genomic selection. BMC Bioinformatics. 2011;
selection. Proc Natl Acad Sci U S A. 2016;113(28):E3995-E4004. 12:186.
53. Samore AB, Fontanesi L. Genomic selection in pigs: state of the art 73. Erbe M, Hayes BJ, Matukumalli LK, et al. Improving accuracy of
and perspectives. Ital J Anim Sci. 2016;15(2):211-232. genomic predictions within and between dairy cattle breeds with
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
288 SONG ET AL.

imputed high-density single nucleotide polymorphism panels. J Dairy 95. Vallejo RL, Silva RMO, Evenhuis JP, et al. Accurate genomic predic-
Sci. 2012;95(7):4114-4129. tions for BCWD resistance in rainbow trout are achieved using low-
74. Lund MS, Sahana G, de Koning DJ, Su G, Carlborg O. Comparison of density SNP panels: evidence that long-range LD is a major contrib-
analyses of the QTLMAS XII common dataset. I: Genomic selection. uting factor. J Anim Breed Genet. 2018;135(4):263-274.
BMC Proc. 2009;3(suppl 1):S1. 96. Yoshida GM, Bangera R, Carvalheiro R, et al. Genomic prediction
75. MacLeod IM, Bowman PJ, Vander Jagt CJ, et al. Exploiting biological accuracy for resistance against Piscirickettsia salmonis in farmed rain-
priors and sequence variants enhances QTL discovery and genomic bow trout. G3. 2018;8(2):719-726.
prediction of complex traits. BMC Genomics. 2016;17:144. 97. Yoshida GM, Carvalheiro R, Rodriguez FH, Lhorente JP, Yanez JM.
76. Fernando RL, Dekkers JC, Garrick DJ. A class of Bayesian methods Single-step genomic evaluation improves accuracy of breeding value
to combine large numbers of genotyped and non-genotyped animals predictions for resistance to infectious pancreatic necrosis virus in
for whole-genome analyses. Genet Sel Evol. 2014;46:50. rainbow trout. Genomics. 2019;111(2):127-132.
77. Nayeri S, Sargolzaei M, Tulpan D. A review of traditional and 98. Vallejo RL, Cheng H, Fragomeni BO, et al. Genome-wide association
machine learning methods applied to animal breeding. Anim Health analysis and accuracy of genome-enabled breeding value predictions
Res Rev. 2019;20(1):31-46. for resistance to infectious hematopoietic necrosis virus in a com-
78. Ben-Hur A, Horn D, Siegelmann HT, Vapnik V. Support vector clus- mercial rainbow trout breeding population. Genet Sel Evol. 2019;
tering. J Mach Learn Res. 2002;2(2):125-137. 51(1):47.
79. Breiman L. Random forests. Mach Learn. 2001;45(1):5-32. 99. Silva RMO, Evenhuis JP, Vallejo RL, et al. Whole-genome mapping
80. Gianola D, van Kaam JBCHM. Reproducing kernel Hilbert spaces of quantitative trait loci and accuracy of genomic predictions for
regression methods for genomic assisted prediction of quantitative resistance to columnaris disease in two rainbow trout breeding
traits. Genetics. 2008;178(4):2289-2303. populations. Genet Sel Evol. 2019;51(1):42.
81. LeCun Y, Bengio Y, Hinton G. Deep learning. Nature. 2015; 100. Vallejo RL, Fragomeni BO, Cheng H, et al. Assessing accuracy of
521(7553):436-444. genomic predictions for resistance to infectious hematopoietic
82. Montesinos-Lopez OA, Montesinos-Lopez A, Perez-Rodriguez P, necrosis virus with progeny testing of selection candidates in a com-
et al. A review of deep learning applications for genomic selection. mercial rainbow trout breeding population. Front Vet Sci. 2020;7:
BMC Genomics. 2021;22:19. 590048.
83. Tsai HY, Hamilton A, Tinch AE, et al. Genome wide association and 101. D'Ambrosio J, Morvezen R, Brard-Fudulea S, et al. Genetic architec-
genomic prediction for growth traits in juvenile farmed Atlantic ture and genomic selection of female reproduction traits in rainbow
salmon using a high density SNP array. BMC Genomics. 2015;16:969. trout. BMC Genomics. 2020;21(1):558.
84. Tsai HY, Hamilton A, Tinch AE, et al. Genomic prediction of host 102. Al-Tobasei R, Ali A, Garcia ALS, Lourenco D, Leeds T, Salem M.
resistance to sea lice in farmed Atlantic salmon populations. Genet Genomic predictions for fillet yield and firmness in rainbow trout
Sel Evol. 2016;48:47. using reduced-density SNP panels. BMC Genomics. 2021;22(1):92.
85. Tsai HY, Matika O, Edwards SM, et al. Genotype imputation to 103. Blay C, Haffray P, D'Ambrosio J, et al. Genetic architecture and
improve the cost-efficiency of genomic selection in farmed Atlantic genomic selection of fatty acid composition predicted by Raman
salmon. G3. 2017;7(4):1377-1383. spectroscopy in rainbow trout. BMC Genomics. 2021;22(1):788.
86. Correa K, Bangera R, Figueroa R, Lhorente JP, Yanez JM. The use of 104. Dong LS, Xiao SJ, Wang QR, Wang ZY. Comparative analysis of the
genomic information increases the accuracy of breeding value pre- GBLUP, emBayesB, and GWAS algorithms to predict genetic values
dictions for sea louse (Caligus rogercresseyi) resistance in Atlantic in large yellow croaker (Larimichthys crocea). BMC Genomics. 2016;
salmon (Salmo salar). Genet Sel Evol. 2017;49:15. 17:460.
87. Bangera R, Correa K, Lhorente JP, Figueroa R, Yanez JM. Genomic 105. Zhao J, Bai HQ, Ke QZ, et al. Genomic selection for parasitic ciliate
predictions can accelerate selection for resistance against Piscirick- Cryptocaryon irritans resistance in large yellow croaker. Aquaculture.
ettsia salmonis in Atlantic salmon (Salmo salar). BMC Genomics. 2017; 2021;531:735786.
18:121. 106. Ke QZ, Wang JY, Bai YL, et al. GWAS and genomic prediction
88. Robledo D, Matika O, Hamilton A, Houston RD. Genome-wide asso- revealed potential for genetic improvement of large yellow croaker
ciation and genomic selection for resistance to amoebic gill disease adapting to high plant protein diet. Aquaculture. 2022;553:738090.
in Atlantic salmon. G3. 2018;8(4):1195-1203. 107. Palaiokostas C, Ferraresso S, Franch R, Houston RD, Bargelloni L.
89. Boison SA, Gjerde B, Hillestad B, Makvandi-Nejad S, Moghadam HK. Genomic prediction of resistance to pasteurellosis in gilthead sea
Genomic and transcriptomic analysis of amoebic gill disease resis- bream (Sparus aurata) using 2b-RAD sequencing. G3. 2016;6(11):
tance in Atlantic salmon (Salmo salar L.). Front Genet. 2019;10:68. 3693-3700.
90. Tsairidou S, Hamilton A, Robledo D, Bron JE, Houston RD. Optimiz- 108. Aslam ML, Carraro R, Bestin A, et al. Genetics of resistance to
ing low-cost genotyping and imputation strategies for genomic photobacteriosis in gilthead sea bream (Sparus aurata) using 2b-RAD
selection in Atlantic salmon. G3. 2020;10(2):581-590. sequencing. BMC Genet. 2018;19:43.
91. Horn SS, Meuwissen THE, Moghadam H, Hillestad B, Sonesson AK. 109. Aslam ML, Carraro R, Sonesson AK, et al. Genetic variation, GWAS
Accuracy of selection for omega-3 fatty acid content in Atlantic and accuracy of prediction for host resistance to Sparicotyle chryso-
salmon fillets. Aquaculture. 2020;519:734767. phrii in farmed gilthead Sea bream (Sparus aurata). Front Genet.
92. Kjetsa MH, Odegard J, Meuwissen THE. Accuracy of genomic pre- 2020;11:594770.
diction of host resistance to salmon lice in Atlantic salmon (Salmo 110. Griot R, Allal F, Phocas F, et al. Optimization of genomic selection to
salar) using imputed high-density genotypes. Aquaculture. 2020;526: improve disease resistance in two marine fishes, the European sea
735415. bass (Dicentrarchus labrax) and the gilthead sea bream (Sparus aur-
93. Fraslin C, Yanez JM, Robledo D, Houston RD. The impact of genetic ata). Front Genet. 2021;12:665920.
relationship between training and validation populations on genomic 111. Yoshida GM, Lhorente JP, Correa K, Soto J, Salas D, Yanez JM.
prediction accuracy in Atlantic salmon. Aquac Rep. 2022;23:101033. Genome-wide association study and cost-efficient genomic predic-
94. Vallejo RL, Leeds TD, Gao GT, et al. Genomic selection models dou- tions for growth and fillet yield in Nile tilapia (Oreochromis niloticus).
ble the accuracy of predicted breeding values for bacterial cold G3. 2019;9(8):2597-2607.
water disease resistance compared to a traditional pedigree-based 112. Joshi R, Skaarud A, de Vera M, Alvarez AT, Odegard J. Genomic pre-
model in rainbow trout aquaculture. Genet Sel Evol. 2017;49:17. diction for commercial traits using univariate and multivariate
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SONG ET AL. 289

approaches in Nile tilapia (Oreochromis niloticus). Aquaculture. 2020; 130. Oikonomou S, Samaras A, Tekeoglou M, et al. Genomic selection
516:734641. and genome-wide association analysis for stress response, disease
113. Joshi R, Skaaurd A, Alvarez AT. Experimental validation of genetic resistance and body weight in European seabass. Animals. 2022;
selection for resistance against Streptococcus agalactiae via different 12(3):277.
routes of infection in the commercial Nile tilapia breeding pro- 131. Dou J, Li X, Fu Q, et al. Evaluation of the 2b-RAD method for geno-
gramme. J Anim Breed Genet. 2021;138(3):338-348. mic selection in scallop breeding. Sci Rep. 2016;6:19244.
114. Barria A, Benzie JAH, Houston RD, De Koning DJ, de Verdal H. 132. Wang YF, Sun GD, Zeng QF, et al. Predicting growth traits with
Genomic selection and genome-wide association study for feed- genomic selection methods in Zhikong scallop (Chlamys farreri).
efficiency traits in a farmed Nile tilapia (Oreochromis niloticus) popu- Marine Biotechnol. 2018;20(6):769-779.
lation. Front Genet. 2021;12:737906. 133. Garcia ALS, Bosworth B, Waldbieser G, Misztal I, Tsuruta S,
115. Joshi R, Almeida DB, da Costa AR, et al. Genomic selection for resis- Lourenco DAL. Development of genomic predictions for harvest
tance to Francisellosis in commercial Nile tilapia population: genetic and carcass weight in channel catfish. Genet Sel Evol. 2018;50:66.
and genomic parameters, correlation with growth rate and predic- 134. Barria A, Christensen KA, Yoshida GM, et al. Genomic predictions
tive ability. Aquaculture. 2021;537:736515. and genome-wide association study of resistance against Piscirick-
116. Joshi R, Skaarud A, Alvarez AT, Moen T, Odegard J. Bayesian geno- ettsia salmonis in Coho Salmon (Oncorhynchus kisutch) using ddRAD
mic models boost prediction accuracy for survival to Streptococcus sequencing. G3. 2018;8(4):1183-1194.
agalactiae infection in Nile tilapia (Oreochromus nilioticus). Genet Sel 135. Liu GJ, Dong LS, Gu LL, et al. Evaluation of genomic selection for
Evol. 2021;53(1):37. seven economic traits in yellow drum (Nibea albiflora). Mar Biotech-
117. Wang QC, Yu Y, Li FH, Zhang XJ, Xiang JH. Predictive ability of nol. 2019;21(6):806-812.
genomic selection models for breeding value estimation on growth 136. Nguyen NH, Phuthaworn C, Knibb W. Genomic prediction for dis-
traits of Pacific white shrimp Litopenaeus vannamei. Chin J Oceanol ease resistance to Hepatopancreatic parvovirus and growth, carcass
Limnol. 2017;35(5):1221-1229. and quality traits in Banana shrimp Fenneropenaeus merguiensis.
118. Wang QC, Yu Y, Yuan JB, et al. Effects of marker density and popu- Genomics. 2020;112(2):2021-2027.
lation structure on the genomic prediction accuracy for growth trait 137. Saura M, Carabano MJ, Fernandez A, et al. Disentangling genetic
in Pacific white shrimp Litopenaeus vannamei. BMC Genet. 2017; variation for resistance and endurance to Scuticociliatosis in turbot
18:45. using pedigree and genomic information. Front Genet. 2019;10:539.
119. Wang QC, Yu Y, Zhang Q, et al. Evaluation on the genomic selection 138. Gong J, Zhao J, Ke QZ, et al. First genomic prediction and genome-
in Litopenaeus vannamei for the resistance against Vibrio parahaemo- wide association for complex growth-related traits in Rock Bream
lyticus. Aquaculture. 2019;505:212-216. (Oplegnathus fasciatus). Evol Appl. 2021;15(4):523-536.
120. Dai P, Kong J, Liu J, et al. Evaluation of the utility of genomic infor- 139. Liu J, Peng W, Yu F, et al. Genomic selection applications can
mation to improve genetic evaluation of feed efficiency traits of the improve the environmental performance of aquatics: a case study
Pacific white shrimp Litopenaeus vannamei. Aquaculture. 2020;527: on the heat tolerance of abalone. Evol Appl. 2022;15:992-1001.
735421. 140. Garrick DJ, Taylor JF, Fernando RL. Deregressing estimated breed-
121. Nguyen NH, Premachandra HKA, Kilian A, Knibb W. Genomic pre- ing values and weighting information for genomic regression ana-
diction using DArT-Seq technology for yellowtail kingfish Seriola lyses. Genet Sel Evol. 2009;41:55.
lalandi. BMC Genomics. 2018;19:107. 141. Song H, Li L, Zhang Q, Zhang S, Ding X. Accuracy and bias of geno-
122. Nguyen NH, Vu NT. Threshold models using Gibbs sampling and mic prediction with different de-regression methods. Animal. 2018;
machine learning genomic predictions for skin fluke disease 12(6):1111-1117.
recorded under field environment in yellowtail kingfish Seriola 142. Daetwyler HD, Pong-Wong R, Villanueva B, Woolliams JA. The
lalandi. Aquaculture. 2022;547:737513. impact of genetic architecture on genome-wide evaluation methods.
123. Palaiokostas C, Kocour M, Prchal M, Houston RD. Accuracy of Genetics. 2010;185(3):1021-1031.
genomic evaluations of juvenile growth rate in common carp (Cypri- 143. Song H, Zhang J, Jiang Y, et al. Genomic prediction for growth and
nus carpio) using genotyping by sequencing. Front Genet. 2018;9:82. reproduction traits in pig using an admixed reference population.
124. Palaiokostas C, Vesely T, Kocour M, et al. Optimizing genomic pre- J Anim Sci. 2017;95(8):3415-3424.
diction of host resistance to koi herpesvirus disease in carp. Front 144. Lund MS, de Roos AP, de Vries AG, et al. A common reference pop-
Genet. 2019;10:543. ulation from four European Holstein populations increases reliability
125. Gutierrez AP, Matika O, Bean TP, Houston RD. Genomic selection of genomic predictions. Genet Sel Evol. 2011;43:43.
for growth traits in Pacific oyster (Crassostrea gigas): potential of 145. VanRaden PM, Van Tassell CP, Wiggans GR, et al. Reliability of
low-density marker panels for breeding value prediction. Front genomic predictions for North American Holstein bulls. J Dairy Sci.
Genet. 2018;9:391. 2009;92(1):16-24.
126. Gutierrez AP, Symonds J, King N, Steiner K, Bean TP, Houston RD. 146. Song HL, Hu HX. Strategies to improve the accuracy and reduce
Potential of genomic selection for improvement of resistance to costs of genomic prediction in aquaculture species. Evol Appl. 2022;
ostreid herpesvirus in Pacific oyster (Crassostrea gigas). Anim Genet. 15(4):578-590.
2020;51(2):249-257. 147. Meuwissen T, Goddard M. Accurate prediction of genetic values for
127. Liu Y, Lu S, Liu F, et al. Genomic selection using BayesCπ and complex traits by whole-genome resequencing. Genetics. 2010;
GBLUP for resistance against Edwardsiella tarda in Japanese floun- 185(2):623-631.
der (Paralichthys olivaceus). Mar Biotechnol. 2018;20(5):559-565. 148. Hayes BJ, Macleod IM, Daetwyler HD, et al. Genomic prediction
128. Lu S, Liu Y, Yu XJ, et al. Prediction of genomic breeding values based from whole genome sequence in livestock: the 1000 Bull Genomes
on pre-selected SNPs using ssGBLUP, WssGBLUP and BayesB for Project. Paper presented at: World Congress on Genetics Applied to
Edwardsiellosis resistance in Japanese flounder. Genet Sel Evol. Livestock Production; 2014.
2020;52(1):49. 149. Pérez-Enciso M, Rinco  n JC, Legarra A. Sequence- vs. chip-assisted
129. Palaiokostas C, Cariou S, Bestin A, et al. Genome-wide association genomic selection: accurate biological information is advised. Genet
and genomic prediction of resistance to viral nervous necrosis in Sel Evol. 2015;47(1):43.
European sea bass (Dicentrarchus labrax) using RAD sequencing. 150. van Binsbergen R, Calus MPL, Bink MCAM, van Eeuwijk FA,
Genet Sel Evol. 2018;50:30. Schrooten C, Veerkamp RF. Genomic prediction using imputed
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
290 SONG ET AL.

whole-genome sequence data in Holstein Friesian cattle. Genet Sel 170. Liu D, Zeng XA, Sun DW. NIR spectroscopy and imaging techniques
Evol. 2015;47:71. for evaluation of fish quality—a review. Appl Spectrosc Rev. 2013;
151. Pszczola M, Strabel T, Mulder HA, Calus MPL. Reliability of direct 48(8):609-628.
genomic values for animals with different relationships within and 171. Kriaridou C, Tsairidou S, Houston RD, Robledo D. Genomic predic-
to the reference population. J Dairy Sci. 2012;95(1):389-400. tion using low density marker panels in aquaculture: performance
152. Ma PP, Lund MS, Aamand GP, Su GS. Use of a Bayesian model across species, traits, and genotyping platforms. Front Genet. 2020;
including QTL markers increases prediction reliability when test ani- 11:124.
mals are distant from the reference population. J Dairy Sci. 2019; 172. Yoshida GM, Carvalheiro R, Lhorente JP, et al. Accuracy of genotype
102(8):7237-7247. imputation and genomic predictions in a two-generation farmed
153. Saura M, Caballero A, Santiago E, et al. Estimates of recent and his- Atlantic salmon population using high-density and low-density SNP
torical effective population size in turbot, seabream, seabass and panels. Aquaculture. 2018;491:147-154.
carp selective breeding programmes. Genet Sel Evol. 2021;53(1):85. 173. Falconer DS, Mackay TF, Frankham R. Introduction to quantitative
154. Correction for Garcia-Ruiz et al., Changes in genetic selection differ- genetics (4th edn). Trends Genet. 1996;12(7):280.
entials and generation intervals in US Holstein dairy cattle as a result 174. Fishback AG, Danzmann RG, Ferguson MM, Gibson JP. Estimates
of genomic selection. Proc Natl Acad Sci U S A 2016;113(33):E4928. of genetic parameters and genotype by environment interactions
155. Hickey JM, Chiurugwi T, Mackay I, Powell W, Cgi IGS. Genomic pre- for growth traits of rainbow trout (Oncorhynchus mykiss) as
diction unifies animal and plant breeding programs to form platforms inferred using molecular pedigrees. Aquaculture. 2002;206(3–4):
for biological discovery. Nat Genet. 2017;49(9):1297-1303. 137-150.
156. Odegard J, Moen T, Santi N, Korsvoll SA, Kjoglum S, Meuwissen TH. 175. Kause A, Ritola O, Paananen T, Wahlroos H, Mantysaari EA. Genetic
Genomic prediction in an admixed population of Atlantic salmon trends in growth, sexual maturity and skeletal deformations, and
(Salmo salar). Front Genet. 2014;5:402. rate of inbreeding in a breeding programme for rainbow trout
157. Boudry P, Allal F, Aslam ML, et al. Current status and potential of (Oncorhynchus mykiss). Aquaculture. 2005;247(1-4):177-187.
genomic selection to improve selective breeding in the main aqua- 176. Dupont-Nivet M, Karahan-Nomm B, Vergnet A, et al. Genotype by
culture species of International Council for the Exploration of the environment interactions for growth in European seabass (Dicen-
Sea (ICES) member countries. Aquac Rep. 2021;20:100700. trarchus labrax) are large when growth rate rather than weight is
158. Verbyla KL, Kube PD, Evans BS. Commercial implementation of considered. Aquaculture. 2010;306(1-4):365-368.
genomic selection in Tasmanian Atlantic salmon: scheme evolution 177. Sae-Lim P, Kause A, Mulder HA, et al. Genotype-by-environment
and validation. Evol Appl. 2021;15(4):631-644. interaction of growth traits in rainbow trout (Oncorhynchus mykiss):
159. Jerry DR, Jones DB, Lillehammer M, et al. Predicted strong genetic a continental scale study. J Anim Sci. 2013;91(12):5572-5581.
gains from the application of genomic selection to improve growth 178. Li YZ, Yang YM, Zheng WW, Cheng JY. Genetic parameters and
related traits in barramundi (Lates calcarifer). Aquaculture. 2022;549: genotype by environment interactions for growth traits and survival
737761. of olive flounder (Paralichthys olivaceus) in recirculating aquaculture
160. Dong LS, Xiao SJ, Chen JW, Wan L, Wang ZY. Genomic selection system and flow-through system. Aquaculture. 2019;510:56-60.
using extreme phenotypes and pre-selection of SNPs in large yellow 179. Mengistu SB, Mulder HA, Benzie JAH, et al. Genotype by environ-
croaker (Larimichthys crocea). Mar Biotechnol. 2016;18(5):575-583. ment interaction between aerated and non-aerated ponds and the
161. Yoshida GM, Yáñez JM. Increased accuracy of genomic predictions impact of aeration on genetic parameters in Nile tilapia (Oreochromis
for growth under chronic thermal stress in rainbow trout by priori- niloticus). Aquaculture. 2020;529:735704.
tizing variants from GWAS using imputed sequence data. Evol Appl. 180. Sae-Lim P, Gjerde B, Nielsen HM, Mulder H, Kause A. A review of
2021;15(4):537-552. genotype-by-environment interaction and micro-environmental sen-
162. Ni GY, Cavero D, Fangmann A, Erbe M, Simianer H. Whole-genome sitivity in aquaculture species. Rev Aquac. 2016;8(4):369-393.
sequence-based genomic prediction in laying chickens with different 181. Folio DM, Gil J, Caudron A, Labonne J. Genotype-by-environment
genomic relationship matrices to account for genetic architecture. interactions drive the maintenance of genetic variation in a Salmo
Genet Sel Evol. 2017;49:8. trutta L. hybrid zone. Evol Appl. 2021;14(11):2698-2711.
163. Bargelloni L, Tassiello O, Babbucci M, et al. Data imputation and 182. Li XJ, Lund MS, Janss L, et al. The patterns of genomic variances and
machine learning improve association analysis and genomic predic- covariances across genome for milk production traits between Chi-
tion for resistance to fish photobacteriosis in the gilthead sea bream. nese and Nordic Holstein populations. BMC Genet. 2017;18:26.
Aquac Rep. 2021;20:100661. 183. Li X, Lund MS, Zhang Q, Costa CN, Ducrocq V, Su G. Short commu-
164. Palaiokostas C. Predicting for disease resistance in aquaculture spe- nication: improving accuracy of predicting breeding values in
cies using machine learning models. Aquac Rep. 2021;20:100660. Brazilian Holstein population by adding data from Nordic and
165. Zhu XH, Ni P, Xing Q, et al. Genomic prediction of growth traits in French Holstein populations. J Dairy Sci. 2016;99(6):4574-4579.
scallops using convolutional neural networks. Aquaculture. 2021; 184. Kolmodin R, Strandberg E, Madsen P, Jensen J, Jorjani H. Genotype
545:737171. by environment interaction in Nordic dairy cattle studied using reac-
166. Yang XT, Zhang S, Liu JT, Gao QF, Dong SL, Zhou C. Deep learning tion norms. Acta Agric Scand A Anim Sci. 2002;52(1):11-24.
for smart fish farming: applications, opportunities and challenges. 185. Song HL, Zhang Q, Misztal I, Ding XD. Genomic prediction of
Rev Aquac. 2021;13(1):66-90. growth traits for pigs in the presence of genotype by environment
167. Zhang D, Lillywhite KD, Lee DJ, Tippetts BJ. Automatic shrimp interactions using single-step genomic reaction norm model. J Anim
shape grading using evolution constructed features. Comput Electron Breed Genet. 2020;137(6):523-534.
Agric. 2014;100:116-122. 186. Liu A, Su G, Hoglund J, et al. Genotype by environment interaction
168. Sture O, Oye ER, Skavhaug A, Mathiassen JR. A 3D machine vision for female fertility traits under conventional and organic production
system for quality grading of Atlantic salmon. Comput Electron Agric. systems in Danish Holsteins. J Dairy Sci. 2019;102(9):8134-8147.
2016;123:142-148. 187. Song HL, Zhang Q, Ding XD. The superiority of multi-trait models
169. Saberioon M, Gholizadeh A, Cisar P, Pautsina A, Urban J. Application with genotype-by-environment interactions in a limited number of
of machine vision systems in aquaculture with emphasis on fish: environments for genomic prediction in pigs. J Anim Sci Biotechnol.
state-of-the-art and key issues. Rev Aquac. 2017;9(4):369-387. 2020;11:88.
17535131, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1111/raq.12716 by Hec Montreal, Wiley Online Library on [21/11/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SONG ET AL. 291

188. Allendorf FW, Thorgaard GH. Tetraploidy and the evolution of sal- 202. Azodi CB, Pardo J, VanBuren R, de los Campos G, Shiu SH. Tran-
monid fishes. In: Turner BJ, ed. Evolutionary Genetics of Fishes. scriptome-based prediction of complex traits in maize. Plant Cell.
Springer; 1984:1-53. 2020;32(1):139-151.
189. Phillips R, Ráb P. Chromosome evolution in the Salmonidae (Pisces): 203. Xu Y, Xu C, Xu S. Prediction and association mapping of agronomic
an update. Biol Rev. 2001;76(1):1-25. traits in maize using multiple omic data. Heredity. 2017;119(3):174-184.
190. Du K, Stock M, Kneitz S, et al. The sterlet sturgeon genome 204. Wang SB, Wei JL, Li RD, et al. Identification of optimal prediction
sequence and the mechanisms of segmental rediploidization. Nat models using multi-omic data for selecting hybrid rice. Heredity.
Ecol Evol. 2020;4(6):841-852. 2019;123(3):395-406.
191. Gille DA, Famula TR, May BP, Schreier AD. Evidence for a maternal 205. Hu XH, Xie WB, Wu CC, Xu SZ. A directed learning strategy inte-
origin of spontaneous autopolyploidy in cultured white sturgeon grating multiple omic data improves genomic prediction. Plant Bio-
(Acipenser transmontanus). Aquaculture. 2015;435:467-474. technol J. 2019;17(10):2011-2020.
192. Havelka M, Hulak M, Rab P, et al. Fertility of a spontaneous hexa- 206. Morgante F, Huang W, Sorensen P, Maltecca C, Mackay TFC.
ploid male Siberian sturgeon, Acipenser baerii. BMC Genet. 2014; Leveraging multiple layers of data to predict Drosophila complex
15:5. traits. G3. 2020;10(12):4599-4613.
193. Osborn TC, Pires JC, Birchler JA, et al. Understanding mechanisms 207. Li ZC, Gao N, Martini JWR, Simianer H. Integrating gene expression
of novel gene expression in polyploids. Trends Genet. 2003;19(3): data into genomic prediction. Front Genet. 2019;10:126.
141-147. 208. Gupta RM, Musunuru K. Expanding the genetic editing tool kit: ZFNs,
194. Lara LAD, Santos MF, Jank L, et al. Genomic selection with allele TALENs, and CRISPR-Cas9. J Clin Invest. 2014;124(10):4154-4161.
dosage in Panicum maximum Jacq. G3. 2019;9(8):2463-2475. 209. Yang ZT, Yu YP, Tay YX, Yue GH. Genome editing and its applica-
195. Oliveira ID, Amadeu RR, Ferrao LFV, Munoz PR. Optimizing whole- tions in genetic improvement in aquaculture. Rev Aquac. 2022;14:
genomic prediction for autotetraploid blueberry breeding. Heredity. 178-191.
2020;125(6):437-448. 210. Aishi K, Sinnasamy S, MacRae TH, et al. Hsp70 knockdown reduced
196. Amadeu RR, Cellon C, Olmstead JW, Garcia AAF, Resende MFR, the tolerance of Litopenaeus vannamei post larvae to low pH and
Munoz PR. AGHmatrix: R package to construct relationship matrices salinity. Aquaculture. 2019;512:734346.
for autotetraploid and diploid species: a blueberry example. Plant 211. Gratacap RL, Regan T, Dehler CE, et al. Efficient CRISPR/Cas9
Genome. 2016;9(3);1-10. genome editing in a salmonid fish cell line using a lentivirus delivery
197. Song H, Hu H. Impact of accounting for allele dosage on genomic system. BMC Biotechnol. 2020;20(1):35.
prediction in a simulated autopolyploid sturgeon population. Aqua- 212. Blix TB, Dalmo RA, Wargelius A, Myhr AI. Genome editing on finfish:
culture. 2022;548:737626. current status and implications for sustainability. Rev Aquac. 2021;
198. Veerkamp RF, Bouwman AC, Schrooten C, Calus MPL. Genomic 13(4):2344-2363.
prediction using preselected DNA variants from a GWAS with 213. Gratacap RL, Wargelius A, Edvardsen RB, Houston RD. Potential of
whole-genome sequence data in Holstein-Friesian cattle. Genet Sel genome editing to improve aquaculture breeding and production.
Evol. 2016;48:95. Trends Genet. 2019;35(9):672-684.
199. Song H, Li L, Ma P, et al. Short communication: improving the accu- 214. Jenko J, Gorjanc G, Cleveland MA, et al. Potential of promotion of
racy of genomic prediction of body conformation traits in Chinese alleles by genome editing to improve quantitative traits in livestock
Holsteins using markers derived from high-density marker panels. breeding programs. Genet Sel Evol. 2015;47:55.
J Dairy Sci. 2018;101(6):5250-5254.
200. Vazquez AI, Veturi Y, Behring M, et al. Increased proportion of vari-
ance explained and prediction accuracy of survival of breast cancer
patients with use of whole-genome multiomic profiles. Genetics. How to cite this article: Song H, Dong T, Yan X, et al.
2016;203(3):1425-1438.
Genomic selection and its research progress in aquaculture
201. Dimitrakopoulos L, Prassas I, Diamandis EP, Charames GS. Onco-
proteogenomics: multi-omics level data integration for accurate phe- breeding. Rev Aquac. 2023;15(1):274‐291. doi:10.1111/raq.
notype prediction. Crit Rev Clin Lab Sci. 2017;54(6):414-432. 12716

You might also like