Abstract
Duck is the second-largest poultry species aside from chicken. The rate of egg production is a major determinant of the economic income of poultry farmers. Among the reproductive organs, the ovary is a major part of the female reproductive system which is highly important for egg production. Based on the importance of this organ, several studies have been carried out to identify candidate genes at the transcriptome level, and also the expression level of these genes at different tissues or egg-laying conditions, and single nucleotide polymorphism (SNPs) of genes associated with egg production in duck. In this review, expression profile and association study analyses at SNPs level of different candidate genes with egg production traits of duck were highlighted. Furthermore, different studies on transcriptome analysis, Quantitative Trait Loci (QTL) mapping, and Genome Wide Association Study (GWAS) approach used to identify potential candidate genes for egg production in ducks were reported. This review would widen our knowledge on molecular markers that are associated or have a positive correlation to improving egg production in ducks, for the increasing world populace.
Key words: egg production, candidate genes, expression profile, single nucleotide polymorphism (SNPs), transcriptome sequencing, QTL mapping, GWAS
INTRODUCTION
Muscovy duck is native to tropical areas of Central and South America (Stahl, 2008), with a strong adaptation ability, fertility, and high meat yield (Cui et al., 2019). Different varieties of Muscovy duck have been identified which include Jiaji Duck that is formerly raised at Jiaji Town, Qionghai Country, Hainan Province (Gu et al., 2020), Black Muscovy (Hu et al., 2021), and White Muscovy (Bao et al., 2020). However, in China, black and white Muscovy ducks are vastly raised.
The Domestic Muscovy duck is one of the most economically important poultry species across the globe due to its distinct meat quality and low-calorie content (Ye et al., 2019a). Aside from Muscovy duck, almost all other domesticated ducks descended from Mallard (Anas Platyrhynchos) (Veeramani et al., 2016). Moreover, Muscovy duck does not require swimming water for survival, and optimum hatchability performance due to its tolerant to hot weather is much better than Pekin duck (Zeng et al., 2013), and this has made it possible to rear them in either traditional extensive or cage system (Harun et al., 1998). However, the long incubation period (35 d) and excess broodiness have led to a decline in egg production, and low development of the Muscovy duck industry (Jiang et al., 2010). Persistent nesting, tilting head backward, clucking, and nest defense are distinct incubation of the Muscovy duck (Wu et al., 2014d). The peak of egg production of Muscovy is between 35 and 53 wk (Ye et al., 2017). Despite all these factors, these ducks have been raised on a large scale due to the high demand for duck eggs and meats by consumers couple with continuous improvement of the standard of living. More so, it has been reported by Statistics of National Waterfowl Industrial Technology System of China in 2019 that there were 187 million laying ducks in China, and the total output of duck eggs was 3.0934 million tons.
Domesticated Muscovy duck is one of the poultry species categorized as a local bird in Africa and one of the most famous waterfowl species in Nigeria (Adeola et al., 2020). Moreover, it is one of the most famous types of duck in India due to its outstanding meat quality and taste (Kameshpandian et al., 2018). Due to their high population in India, they are called by different names which include Moti, Kadna Hans, and China han in Odisha (Padhi, 2014) and Chinae haras, Cina hanh, Bor China, and Bhatt China in Northeast India (Alyethodi et al., 2010; Banerjee, 2013).
The reproductive performance which is an important economic trait of poultry species is mainly determined by egg production (Bao et al., 2020). The key objective of duck breeders is to increase egg production, although this trait is affected by many factors ranging from nutrition, environment, disease, gene expression, and others (Hu et al., 2021) as shown in Figure 1 (Although, we reviewed only about the genetic factors). Using traditional breeding techniques, reproductive traits of laying duck have been progressively improved although, an obviously significant improvement has not been recorded (Zhu et al., 2017).
Figure 1.
Approaches used to study/identify candidate genes responsible for egg production.
Egg production is a polygenic trait that is affected by both genetic and environmental factors. It involves a multifactorial interaction of genes that control energy metabolism, protein synthesis, and storage processes, this phenomenon depends on different organs involved in reproduction (Su et al., 1999). Moreover, this is regulated by several physiological processes that require the coordination of the hypothalamus-pituitary-gonadal (HPG) axis for successful procreation (Ye et al., 2019b). This axis regulates the reproductive and endocrine systems in laying hens (Padmanabhan et al., 2002), and also initiates the maturation of the ovary (Ouyang et al., 2020). In poultry species, the hypothalamus regulates reproduction activity through the secretion of gonadotropin-releasing hormone-1 (GnRH-1) that triggers the adenohypophysis (anterior pituitary gland) to enable the release of Luteinizing hormone (LH) and follicle stimulating hormone (FSH). These gonadotropins enable the gonads to stimulate gametogenesis and sex steroid hormone secretion from the specialized cell in the left ovary (Mishra et al., 2020; Figure 2). The ovary is a major part of the female reproductive system which is highly important for the regulation of oocyte development, follicle maturation, and release of sex hormones for ovulation (Eppig et al., 2002), and also an important organ that affects egg-laying (Natesampillai et al., 2008; Xu et al., 2018). Estrogen that is mainly secreted by granulosa cells in the ovary also transfers endocrine signals to other tissues (Nelson and Bulun, 2001). Based on the role of the ovary in reproduction, several studies have been carried out using this organ to identify candidate genes at the transcriptome level, expression level, and single nucleotide polymorphism (SNPs) of genes associated with egg production in ducks (Ni et al., 2007a; Asiamah Amponsah et al., 2019; Asiamah et al., 2022; Figure 1).
Figure 2.
A chart showing the processes involved in egg production along HPG axis in poultry birds.
In this review, we focus on reports of different studies on candidate genes responsible for egg production in ducks. This would widen our knowledge on molecular markers that are being used to improve egg production for the increasing world populace.
STUDIES ON EXPRESSION PROFILE AND SINGLE NUCLEOTIDE POLYMORPHISM OF CANDIDATE GENES ON EGG PRODUCTION
Having identified Muscovy ducks as one of the poultry species which has potentials for the production of eggs and meat, the low rate of egg production can be increased through genetic improvement through marker-assisted selection. Marker-assisted selection is based on the relationship that exists between DNA variation and specific genes that control a specific trait of interest. This has become an important technique in animal breeding for selecting high-laying performance ducks for genetic improvement. A candidate gene approach is useful for the understanding of the relationship that exists between various forms of a particular gene and quantitative trait loci (QTL) that are involved in the variation of economic traits of interest (Qin et al., 2015; Yuan et al., 2015).
Recently, the potential role of expressions of insulin-like growth factor-1 (IGF-1), prolactin receptor (PRLR), FSH, very low-density lipoprotein receptor (VLDLR), low-density lipoprotein receptor-related protein (LRP), estrogen receptor 1 (ESR1), luteinizing hormone receptor (LHR), and estrogen receptor 2 (ESR2) with egg traits have received serious attention (Yao et al., 2010; Wang et al., 2016; Asiamah Amponsah et al., 2019; Cui et al., 2019; Asiamah et al., 2022).
SNP is the difference that occurs among individuals at a single position in a DNA sequence. Due to their abundance and a high potential for automation, SNPs are the most commonly used molecular markers in genetic studies (Kumar et al., 2012). Moreover, SNPs have been used to identify genetic factors in different fields (Liao and Lee, 2010), including studies in animal and crop breeding (Lagudah et al., 2009; Schennink et al., 2009).
Follicle-Stimulating Hormone
FSH plays a critical role in the regulation of several reproductive activities in avian. This gene stimulates the reproductive gland function of males and females by binding to its receptor (FSHR), which is located in the testis (sertoli cells) and ovary (granulosa cells), respectively (You et al., 1996; George et al., 2011). In a recent study, there was a significantly high expression of FSH in the egg-laying period compared with the pre-laying period (Huang et al., 2015). The SNP of the FSH gene were determined to be related to the reproductive traits of chickens (Li et al., 2011; Kang et al., 2012). In a study by Xu et al. (2017), molecular characterization, expression profile, and association study of FSH on egg production in Muscovy duck were studied. From their findings, 3 different Muscovy duck FSHR transcripts (FSHRTV1, FSHRTV2, and FSHRT) were identified. Based on the expression profile of FSH mRNA in 14 different selected tissues, the ovary had the highest expression level of FSH when compared to other tissues while obvious mRNA expression of FSH was observed in lung, liver, and abdominal fat (Xu et al., 2017). Also, the study by Liu et al. (2021c) on the expression level of FSH in 3 stages of laying of Peking and Black Muscovy duck, FSH decreased in the peak laying period of Peking duck while the expression level of FSH in the Black Muscovy duck at the peak stage of laying was extremely significant compared to early and late stages of laying (Liu et al., 2021c). Sixteen SNPs were identified within 2,860 bp region of the Muscovy duck FSHR gene (Xu et al., 2017). However, 4 variations (TS59C, T409G, C424G, and C428T) at the 5’ flanking region and 8 (A112G, T125C, T13C, C72T, T231C, A54T, T42C, and A227G) in the intronic regions were detected among those identified SNPs. Moreover, 4 SNPs (C608T, A176G, C320T, and C1044G) were identified at the coding region while only one SNP (C1044G) caused a single amino acid substitution from leucine (L) to valine (V) (Xu et al., 2017). Based on the association analysis of 9 SNPs to 3 different egg production traits (age at first age [AFE], the number of the egg at 33 wk (E33W), and the number of the egg at 59 wk [E59W]), only 2 SNPs (C320T and A227G) were found in the exon and intron regions, respectively, these SNPs were significantly associated with Muscovy egg production traits (Xu et al., 2017).
Growth Hormone
Growth hormone (GH) is secreted by the hypothalamic-pituitary-gonadal (HPG) axis and released into the bloodstream and participates in several differentiation processes in different tissues which includes the stimulation of growth and development (Martínez-Moreno et al., 2011). Due to the function of GH in reproduction, Wu et al. (2014d) researched the expression, polymorphism, and association of this gene on egg production in White Muscovy duck. Only one polymorphism with genotypes AA, AG, and GG was found at intron 3 of the GH gene using Polymerase Chain reaction-Single strand conformation polymorphism (PCR-SSCP) method. Based on the expression profile using 12 different tissues (pituitary, hypothalamus, muscular stomach, glandular stomach, ovary, kidney, liver, heart, lung, duodenum, pancreas, and spleen), the mRNA level of GH in the pituitary recorded the highest expression among other tissues. Moreover, the polymorphic variants of GH had no significant difference in age at first laying (Wu et al., 2014d).
Dopamine Receptor 2 and Insulin Like Growth Factor 2
Dopamine (DA) is an important neurotransmitter that exists in the nerve center and its peripheral tissue. Dopamine receptor 2 (DRD2) could assist in the secretion of reproductive hormones through FSH and LH in chicken (Youngren et al., 1996, 1998). In a study, it was reported that IGF2 is highly expressed in the dominant follicle in mammals thereby supporting key roles in follicular development (Mao et al., 2004). Moreover, IGF2 might regulate ovarian development through stimulation of FSH (Baumgarten et al., 2015).
Ye et al. (2017) investigated the expression profile and polymorphism of IGF2 and DRD2 with laying traits of Muscovy duck. Based on their study, the highest expression levels of IGF2 were found in the kidney and ovary while DRD2 expression had the highest in the ovary. Moreover, 5 SNPs (A-1864G, C-1704G, A-584G, A-227G, and A-183G) were identified in the 5’ flanking region of IGF2 while 28 SNPs were found in DRD2. However, only 2 SNPs and 11 SNPs of IGF2 and DRD2, respectively, based on mixed pool sequencing results were considered most likely associated with egg-laying traits in their study. Moreover, 3 genotypes (AG, GG, and CC) were identified in A-18764G SNP with individuals with GG genotype had 6 to 7 eggs more than GG genotype individuals at E59W. Furthermore, the GG genotypes of C+3301G and G+3545G of the DRD2 gene were advantageous for earlier egg-laying (Ye et al., 2017).
Low-Density Lipoprotein Receptor-Related Protein
Low-density lipoprotein receptor-related protein (LRP) is one of the multitissue endogenous receptors which include LRP5, LRP6, LRP8 gene are related to ovaries and follicles, and LPR4, LPR5, LPR6, LPR8 gene can affect the reproduction of female animals through the regulation of the ovarian or follicular development (Magoori et al., 2003; Yao et al., 2010). The expression of LRP1 in Peking duck during the late laying stage was significantly higher than that in the early laying stage. However, the expression of LRP1 was extremely significantly related to the decrease of laying in the peak laying stage of Black Muscovy duck (Liu et al., 2021c). Further, the LRP8 gene is found related to duck reproductive potential and egg quality (Wang et al., 2013). Six nucleotide mutation sites in the coding region of the LRP8 gene were found linked to egg production of which c.528C >T genotype were associated with egg production, age at first laying, and body weight at maturity (Wang et al., 2013). However, the c.1371A > G genotypes were only related to egg production. Moreover, the egg production was associated with 2 haplotype sites, which include c.528C > T and c.1371A > G that were associated with egg weight and egg yolk weight (Wang et al., 2013).
Estrogen Receptor
Estrogen receptor (ESR) is an essential member of large family steroid hormone receptors which has 2 subtypes. This gene has been reported to play an important role in the ovary during the egg-laying stages and might be closely related to egg production in ducks (Wu et al., 2014b; Khristi et al., 2018). As reported by Couse et al. (2005), ESR2 is crucial in granulosa cell differentiation and the response of ovulation to gonadotropins. The expression of ESR1 was slightly higher in the peak laying stage than early and late laying stages in Black Muscovy duck (Liu et al., 2021c). However, the early laying stage had a significant expression of ESR1 than that of the peak laying stage, although, the late laying stage was extremely significantly higher than that in the peak laying stage in Peking duck (Liu et al., 2021c). Previous reports confirmed that SNPs of ESR1 (T1101C located on exon 4) gene were related to egg production in duck (Niu et al., 2017). ESR2 participated in the regulation of reproductive hormone (Zou et al., 2020). In lieu of this, the functional roles of ESR2 was investigated (Asiamah et al., 2022). Based on their study, 23 SNPs was identified and only SNPs g.56805646 T>C and exon 3-20 G>A were significant (P< 0.05) related to egg weight. It is worth knowing that the ducks with CT and AG genotypes recorded significant (P< 0.05) higher egg weight. In addition, the mRNA expression of ESR2 was significantly more (P< 0.05) in the ovary than the hypothalamus and pituitary.
Prolactin Receptor
In general, the reactions of the 2 PRLRs with prolactin (PRL) can convey information (Goffin et al., 1998; Dalrymple and Jabbour, 2000), by controlling the estrus of the corpus luteum that regulates the estrus and nest behavior in poultry species (Grosdemouge et al., 2003). PRLR is also important for the regulation of follicular development which then enables the IGF gene family to control growth, follicle maturation, and atresia, selection of dominant follicle, egg maturation, production of steroid hormones, and activity of corpus luteum (Armstrong and Hogg, 1996). Prolactin and its receptor have been reported to regulate follicle development and egg production (Wang et al., 2011b; Asiamah Amponsah et al., 2019). The expressions of PRLR were extremely significantly associated with an increase in laying at the peak laying stage which infers that PRLR might promote egg production in Black Muscovy duck (Liu et al., 2021c). Furthermore, the polymorphism of the PRL gene has shown that PRL might be a candidate gene that controls egg-laying parameters and reproduction in ducks (Wang et al., 2011b; Bai et al., 2019). Liu et al. (2012) reported that exon 6 of PRLR was significantly associated with egg production and age at first egg.
Luteinizing Hormone Receptor
LHR belongs to the uncommon members of the G protein-coupled receptor (McFarland et al., 1989), which controls the development of follicles and secrete estrogen by coordinating the role of luteinizing hormone (Zhang et al., 1997). In laying birds, there is a periodic fluctuation in the expression levels of LH and LHR with ovulation activity (Liu et al., 2021c). Despite this, there was a gradual increase in expression of LHR during follicle development before ovulation, while there is a decline in the granulosa cells of the biggest follicle before ovulation, this reduction might be mediated by the steroids (Liu et al., 2021c). In a report by Yamamura et al. (2001), the high levels of LH might inhibit the expression of LHR. Different studies have reported that high expression of LHR might lead to high egg yield (Liu and Zhang, 2008). Due to the extremely significant expression of LHR in the peak laying stage of Peking duck and Black Muscovy duck, it was inferred that the LHR gene plays an important role in the regulation of egg-laying in Pekin duck and Black Muscovy duck (Liu et al., 2021c). Moreover, expression of LHR in the ovary of Shaoxing duck increased consecutively on the first day, 30th day, 60th day, and 90th day which shows that LHR might be responsible for the regulation of egg production in Shaoxing duck (Ni et al., 2007a, b).
Melatonin Receptor Genes
In poultry species, melatonin (N-acetyl-5-methoxytrptamine) which is mainly synthesized in the pineal gland participates in reproduction activities in poultry birds (Kumar Kharwar and Haldar, 2011; Yadav and Haldar, 2013; Trivedi and Kumar, 2014). In birds, 3 melatonin receptor subtypes namely; MTNR1A, MTNR1B, and MTNR1C have been cloned (Reppert et al., 1995; Li et al., 2013). In a recent study by Feng et al. (2018), seven novel polymorphism of melatonin (MTNR1A: g. 268C>T, MTNR1B: g. 41C>T, and g. 161T>C, MTNR1C: g. 10C>T, g. 24A>G, g. 108C>T, g. 363 T>C) were detected in Shaoxing duck. Based on their findings, there was a strong association between g. 268C>T of MTNR1A gene with age at first egg, the total number of eggs at 34 wk of age, and age at first egg. Moreover, there was a significant reduction of the mRNA expression of MTNR1C in the late-matured group when compared with the early-matured group. This shows that melatonin receptors might influence the age at the first egg and, also imply their function in the sexual mature of ducks (Feng et al., 2018).
Very Low Density Lipoprotein Receptor Gene
VLDR belongs to the family of the LDLR gene (Nykjaer and Willnow, 2002). Wang et al. (2011a) reported that the VLDR gene was first isolated from the cDNA library of rabbit's heart and later cloned in chicken, human, mouse, cattle, and monkey. In similar to the LDLR gene, the VDLR gene has 5 functional domains (Willnow, 1999). This gene plays a significant role in different cellular processes which include cell proliferation, migration, and differentiation (Hussain, 2001). In chicken reproduction, VLDR gene plays a distinct role through the development of oocytes and deposition of yolk lipoprotein (Barber et al., 1991; Shen et al., 1993). Also, in the zebra finch, it was reported that expression of VLDLR mRNA has a major function in ascertaining variation among individuals in reproductive phenotype (for instance, follicle or egg size) (Han et al., 2009). Moreover, the phylogenetic tree showed that the evolutionary origin of duck VLDR proteins is closer to chicken and zebra finch than to other species (Wang et al., 2011a). Due to its role in reproduction, Wang et al. (2011a) studied its expression profile in 12 different tissues and also investigated its association with egg performance in duck using SNP. Based on their findings, it was reported that 2 VLDLR splice variants namely; VLDLR-a (with an O-linked sugar domain) and VLDLR-b (without) were present in the heart of the duck. The muscle tissue of duck has a high expression of VLDLR-a. However, there was significant expression of VLDLR-b in the ovary, pituitary gland, liver, spleen, lung, kidney, and intestine (Wang et al., 2011a). Based on their association analysis, the 2 haplotypes were significantly associated with 2 egg production traits namely, age at first egg, and body weight at first egg (Wang et al., 2011a). However, ducks with haplotype AT had a higher egg production at 210, 300, and 360 d egg production, and an earlier age for starting laying while those with haplotype CG had only higher body weight at the first (Wang et al., 2011a). In addition, a study by Liu et al. (2021c) on the expression of VLDLR in 3 different stages of egg-laying in Peking and Black Muscovy duck observed that there was an increase in the expression of VLDLR in the late laying stage of Peking duck nevertheless, it was not significantly different from the early and peak laying stages. However, this gene was extremely significantly associated with the increase of laying in the peak laying stage of black Muscovy duck (Liu et al., 2021c).
Ovoinhibitor
According to Kinoshita et al. (2004), ovoinhibitor (OIH) is the major proteinase inhibitor present in egg white. This gene is secreted in the tubular gland cell of the oviduct, which is controlled by estrogen and progesterone (Liu et al., 1971). In the reproductive trait of turkey, OIH has been reported to controls the microenvironment for the sperm in the epididymis and ductus deferens (Słowińska et al., 2014). Moreover, it was predicted that OIH might be related to the reproductive performance of birds. Based on this, Wu et al. (2018) studied the role of OIH in the ovaries of two breeds of duck namely; Jingjiang (JJ) and Shaoxing (SX) ducks at different laying stages, and also identified the SNP of the OIH gene associated with egg-laying traits. In their findings, the relative expression of OIH mRNA spontaneously increased during the laying period (from the first egg to 500 d of age). Nevertheless, there was a significant difference in the relative expression level of OIH mRNA in the ovaries between the 2 breeds from age at first egg and 180 d of age, although not at 500 d of age. Based on their SNPs study on the OIH gene, one novel mutation (G-A at the 389-nucleotide position 389G>A) was discovered at exon 5–6 using PCR-RFLP methods. Based on their association analysis, the egg production of the AG genotype ducks was significantly small at first egg compared with AA and GG counterparts in the JJ line1. However, the number of the egg at 72 wk was significantly different among AG, GG, and AA genotypes in JJ line II (Wu et al., 2018).
TRANSCRIPTOMIC STUDIES ON EGG PRODUCTION IN DUCK
Since the invention of next-generation sequencing, a powerful, highly reproducible, and cost-efficient tool for transcriptomic studies (Li et al., 2014; Morozova and Marra, 2008). This technology allows the usage of RNA-seq for examining the functions of genes by discovering the gene expression in animal samples (Yu et al., 2016). Moreover, RNA-Seq allows the detection of differences in gene expression profiles among individuals in various developmental states (Tao et al., 2017). Due to its ability to sequence the complete set of transcripts in a cell or a tissue, it provides an effective method for identifying different phenotype-associated differentially expressed genes (DEGs) (Wang et al., 2017).
The application of transcriptomic studies on the ovaries has led to the discovery of DEGs related to egg production, and this would assist in the breeding of potential egg-laying ducks to meet the need of the increasing population and to improve the economic situation of farmers (Zeng et al., 2015; Zhu et al., 2017). Different studies have been carried out to identify potential genes responsible for egg production in different types of duck which include White Muscovy (Bello et al., 2021), Black and White Muscovy (Bao et al., 2020), Black Muscovy (Hu et al., 2021), Leizhou black (Zou et al., 2020), Longyun Shan-ma (Sun et al., 2020), Peking (Ren et al., 2018), Jinding (Tao et al., 2017), and Shan Ma (Zhu et al., 2017) ducks as shown in Table 1.
Table 1.
Different transcriptomic studies to identify potential candidate genes responsible for egg production in ducks.
| Breeds of duck | Tissue used | Stage of laying considered | Group comparison | Egg production parameters considered | Number of DEGs identified | Potential candidate genes and their functions | Expression level | Transcriptome studies involved |
|---|---|---|---|---|---|---|---|---|
| White Muscovy | Hypothalamus and ovary | 59 wk | Highest producing (HP) and lowest producing (LP) | Age at first egg (AGE), number of eggs at 300 d (N300D), and number of eggs at 59 wk (N59W) | 596 (Hypothalamus) 1,502 (Ovary) |
PR2X1 Inhibition of sperm transport that might affect ovulation of eggs (White et al., 2013) LPAR2 Influences the uterine cycle during estrous cycle and early pregnancy (Seo et al., 2008) ADORA1 Plays an important role in cell proliferation and hormone secretion (Wang et al., 2015) FN1 Support of cell adhesion (Goossens et al., 2009) AKT3 Fundamental regulator of many cytokines- and hormones-driven processes(Fabi and Asselin, 2014) ADCY5 Affect the development of ovarian morphological-related (Liu et al., 2021a) ADCY8 mRNA expression of this gene control endocrine system in the HPG axis (Farmanullah et al., 2020) MAP3K8 Target gene of miRNA-509-3p that promote the secretion of estradiol (Zeng et al., 2016) PXN Involved in focal adhesions (Zeng et al., 2016) PTTG1 Key role in organ development, regulation of cell cycle (Wu et al., 2014a) |
Upregulated in hypothalamus Downregulated in hypothalamus Downregulated in hypothalamus Upregulated in ovary Upregulated in ovary Upregulated in ovary Upregulated in ovary Upregulated in ovary Upregulated in ovary Downregulated in ovary |
(Bello et al., 2021) |
| Black Muscovy | Ovary | Early, peak and late laying | Late laying (BL) vs early (BE) BL vs peak (BP), and BP vs BE |
NIL | 101 (BE vs BL) 1090 (BP vs BL) 990 (BE vs BP) |
HOXA1 Promotes the development of uterus and the adhesion of embryo HtrA3 Involved in ovarian development (Singh et al., 2011) StAR Participats in steroid hormone synthesis (Woodruff and Shea, 2007) ZP2 Play a key role in the process of oogenesis, fertilization and preimplantation development (Woodruff and Shea, 2007) TAT For the development and differentiation of chicken oviduct (Lim and Song, 2016) |
Up (BE vs BL group) BP = 6.5 by BE group Value of FKPM of BP = 5 higher than BL Up-regulated in BE group compared with BP group Relative expression significantly increased in BL group compared with BP group |
(Hu et al., 2021) |
| Black and White Muscovy | Ovary | 216–280-day-old | Black high (BH) vs White high (WH) Black low (BL) vs Black high (BH), and Black low (BL) vs White low (WL) |
Number of eggs produced | 113 (BH vs (WH) 619 (BL vs BH) 87 (BL vs WL) |
TGFβ2 Play a key role in the regulation of development of germ cells of ovaries (Hattori Ma et al., 2002) NGFR Important in regulating the growth, development and function of reproductive organs (Vera et al., 2014) CEBPD Higher proliferation rate in calf endometrium under the influence of steroid hormones (Becker et al., 2011) CPEB2 Promote meiotic maturation and embryonic development of porcine oocytes (Prochazkova et al., 2018) POSTN Follicular granulosa cell proliferation (Ożegowska et al., 2019; Kulus et al., 2019) SMOC1 It helps in reproductive trait development of fetal (Pazin and Albrecht, 2009) FGF18 Participates in follicular development (Zhang et al., 2006) EFNA5 Steroid production during follicular development (Worku et al., 2018) SDC4 Upregulation of SDC4 gene in ovulatory follicles may affect ovulation period (Lussier et al., 2017) |
Highly expressed in high-laying As above As above Ovarian tissues of high- and low-laying duck and white Muscovy ducks As above As above Expressed in the ovarian tissue of high yield black As above Ovarian tissue of HL-was significantly up-regulated |
(Bao et al., 2020) |
| Leizhou black | Ovary | High yield and low yield egg laying ducks | High yield (HG) vs low yield (LG) group | Number of eggs (NE), age at first egg (AFE) and weight at first egg, and egg weight (EW) | 1,027 (HG vs LG) |
ESR2 Allow normal follicular development (Wu et al., 2014b) PRLR (Already discussed in the first section) BMPR1B Determinant of ovulation rate (Polley et al., 2010) |
Expression was higher in HG than I have LG Not reported in this study (check Introduction) Not reported |
(Zou et al., 2020) |
| Longyan Shan-ma | Ovary | Number of egg produced and egg weight from point of lay to 71 wk | High egg number (HEN) vs low egg number (LEN) | 71 wk of laying | 343 (HEN vs LEN) |
ITGB5 Important role in ovarian; development (Herrera et al., 2005), follicular development (Hatzirodos et al., 2014), and follicular selection in cattle (Liu et al., 2009) QTL for egg albumen quality in egg layers (Antosik et al., 2016) ITGB2 mRNA level of ITGB2 is associated with ovarian follicle size in pigs (Akison et al., 2010) ITGA8 Regulating the recruitment of mesenchymal cells into epithelial structures |
Not reported Not reported Highly downregulated on ovarian follicle |
(Sun et al., 2020) |
| Peking | Ovary follicle | 60-day-old young (YD), 160-day-old first-laying (FL), and 490 day old stop-laying (OD) | YD vs FL FL vs OD |
NIL | 593 (FL vs YD) 518 (OD vs FL) |
EPOX Member of the cytochrome P450 superfamily of enzymes (Craig et al., 2011) 3β-HSD, StAR, CYP17 3β-HSD were also identified as key genes |
High expression in laying ducks |
(Ren et al., 2018) |
| Jinding | Ovary | High (HEP) and low egg producing (LEP) duck | HEP vs LEP | Number of egg produced between 169 and 310 d | 843 (HEP vs LEP) |
MC5R It plays a key role in female reproductive system (Lantang et al., 2015) APOD It might regulate remodeling of the fallopian tube (Jeong et al., 2015) ORAI1 It is involved with the calcium signaling pathway DYRK4 Important in cell differentiation, survival, proliferation and development (Papadopoulos et al., 2011) GRAMD1B It has been identified as candidates gene in female fertility (Höglund et al., 2015) |
More than 80-fold greater in the HEP than LEP Significantly greater in HEP group than LEP Upregulated (HEP vs LEP) Significant differences between HEP and LEP Not reported |
(Tao et al., 2017) |
|
Shan Ma |
Ovary |
Peak (180 d) and late stage (520 d) |
Peak vs Late |
NIL |
2,002 (peak and late) |
FSHβ, PLA2G2E, GNAQ, MAP2K4 and CALM3B are involved in GnRH signaling “an important pathway related to egg production in poultry birds (Bédécarrats et al., 2009) |
Not reported |
(Zhu et al., 2017) |
Our recent study on transcriptome analysis of hypothalami and ovaries of White Muscovy duck at 59 wk of laying revealed 10 potential genes that might be responsible for egg production (Bello et al., 2021). Based on our findings, 596 and 2,259 DEGs were found in the hypothalamus and ovary, respectively (Bello et al., 2021). It is already an established fact that the ovary of poultry birds has concourse effects of different genes that firmly control ovarian follicle and hormone secretion (Quan et al., 2019; Zou et al., 2020).
Black and White Muscovy duck are vastly raised in China. Bao et al. (2020) examined the transcriptome profiling of the ovary tissues between these 2 ducks using high and low egg production. Based on their findings, 113, 619, and 87 DEGs were identified in the Black high (BH) and White high (WH), Black low (BL) and BH, and BL and White low (WL), respectively. Based on their DEGs within the BH and BL, several genes such as TGFβ2, NGFR, CEBPD, CPEB2, POSTN, SMOC1, FGF18, EFNA5, and SDC4 that might be involved in the egg production of Muscovy duck were identified (Bao et al., 2020).
Hu et al. (2021) studied the transcriptome of the ovary of black Muscovy duck at 3 different stages of laying namely; early (BE), peak (BP), and late laying (BL) stages. Based on their findings, a total of 1683 DEGs were identified from 3 comparison groups (BE-vs-BP, BE-vs-BL, and BP-vs-BL). However, only 11 genes were co-expressed among the 3 comparison groups (Hu et al., 2021). In their study, several genes that might affect egg production in Black Muscovy duck were found, these genes include homeobox A10 (HOXA10), High-temperature requirement factor A3 (HtrA3), Steroidogenic acute regulatory protein (StAR), Zona pellucida glycoprotein 2 (ZP2), and tyrosine aminotransferase (TAT).
In a recent study by Zou et al. (2020), 1,027 DEGs of which 495 and 532 genes were upregulated and downregulated, respectively were identified between the high yield group (HG) and low yield group (LG) of Leizhou ducks. However, only 25 genes and 25 genes were related to reproduction and reproductive processes, respectively. Prolactin (PRLR), Estrogen 2 (ESR 2), and Bone morphogenetic protein receptor beta (BMPR1B) were found to be expressed in the ovarian tissues with significant expressions in HG and LG, respectively (Zou et al., 2020). The roles of PRLR and ESR in egg production have been discussed earlier in section 2 of this review.
A study by Sun et al. (2020) using the ovarian transcriptome to identify DEGs and signaling pathways responsible for egg production in higher and lower laying ducks at the late laying period (71 wk). Based on their study, a total of 343 DEGs comprising of 269 upregulated and 74 downregulated were identified. Out of the top 20 DEGs (17 upregulated and 3 downregulated expressed genes), one (LOC101793233), 4 (IL411, DUOX2, ASS1, and PTGS2), 2 (C1S and IL8), one (PRG4), and 12 (LOC101791385, F13A1, LOC101800358, LOC101795022, DDX60, TPM2, STC1, STAT1, COL1A1, LOC101790957, LOC106018024, and LOC101802453) DEGs are an essential role in cell-cell adhesion, encoding oxidases or synthases, immune response, follicle development, and encoding structural proteins or subunits, respectively (Sun et al., 2020).
Due to the importance of follicles in duck reproductive performance, Ren et al. (2018) researched the differentially expressed key genes that are related to the development of follicles in Peking ducks (Anas Platyrhynchos) at 3 different stages of life (young, laying, and old). Based on their findings, 593 and 518 coding genes were expressed between young and laying ducks and laying and old ducks, respectively (Ren et al., 2018).
In a comparative transcriptomic study of the ovaries of Jinding ducks at 2 different egg production levels (high and low), Tao et al. (2017) reported that 367 downregulated and 476 upregulated DEGs were identified in high and low egg-producing ovaries, respectively. However, among the top 24 DEGs, only 4 DEGs (MCR5R, APOD, ORAI1, and DYRK4) were more active in the ovaries of the high egg-producing ducks, which indicated that these genes might play vital roles in egg production (Tao et al., 2017).
The ovarian transcriptome in Shan Ma ducks at 2 stages of egg production was studied by Zhu et al. (2017). A total of 2,002 DEGs comprising 790 upregulated and 1,212 downregulated were detected. The annotation of the genes revealed that 1,645 genes were already annotated, of which 999 and 646 genes were downregulated and upregulated, respectively (Zhu et al., 2017). However, only the top 10 up- and downregulated DEGs in the ovaries of peak and late stages, respectively that might be related to reproductive processes were presented in their findings.
The summary of breeds of duck used, stages of laying considered, egg production parameters considered, number of DEGs, potential candidate genes and their functions, expression level of these potential genes in tissues considered are presented in Table 1.
QTL, GWAS, SELECTION SIGNATURE, OR COMBINATION OF APPROACHES TO IDENTIFY GENES
Genome wide association studies with phenotypic traits have revealed multiple QTL regions using different of markers, that is, microsatellites or SNPs, and these QTLs have narrowed down genome regions of candidate gene study (Tuiskula-Haavisto et al., 2002; Mondal et al., 2004; Hansen et al., 2005). However, there are little reports on QTLs studies related to egg production traits in ducks. Based on this, a study incorporated transcriptome, proteome and QTL analyses to identify advantageously significant genes that contribute to eggshell and albumen formation in duck (Zhang et al., 2020). In another study by Liu et al. (2021b), GWAS was utilized to reveal the genetic variations for egg internal quality in ducks. GWAS and selective sweep analysis reveal genetic loci responsible for feed conversion ratio (FCR) of egg production performances in ducks (Liu et al., 2021b). The identified SNPs at the cis-regulatory region of ABCG2 gene are related to egg color in Mallard (Liu et al., 2021a) and the 2 cis-regulatory SNPs upstream of ABCG2 can cause blue eggshell phenotype in duck (Chen et al., 2020). The summarize findings from several studies using QTL, GWAS and selection signature, and cis-regulatory are presented in Table 2.
Table 2.
List of several studies on QTL, GWAS, selection signature, and cis-regulatory on egg production traits in ducks.
| Approaches | Egg production traits | Candidate genes revealed | Location on QTL region | Expression level | References |
|---|---|---|---|---|---|
|
Transcriptome, proteome quantitative trait loci (QTL) |
Duck egg shell and albumen formation |
LPCAT3 Regulate membrane fluidity and phospholipid dependent signaling (Demeure et al., 2011) Associated with the bacterial-resistance ability of duck eggs ORM1 It is the most highly expressed gene during albumen formation in ducks key acute phase plasma protein (acute-phase reactant) (Pruitt et al., 2009) Responsible for albumen differences in duck and chickens albumen SPINK7 Function as carbohydrate-binding and involved in steroid hormone process (Compere et al., 1981) Involved in some initial phase BP1FB3 |
Albumen height/haugh unit QTL region Not reported Not reported Eggshell quality-related QTL regions (thickness, stiffness, weight, yellowness, and strength) |
Not reported Greatly increased in the active duck magnum (high expression level in the quiescent duck magnum) Not reported Greatly expressed between the two functional states |
(Zhang et al., 2020) (Zhang et al., 2020) |
| MCTP1, MDN1, TMC3, OCX-36 | Egg shell color region | ||||
|
Preteomics |
Egg shell |
CEP162, APPL1, DAG, REG4 |
Egg shell strength QTL region |
Not reported |
(Zhu et al., 2019) |
|
GWAS |
Egg internal quality |
EHF, ZDHHC13, MUC6, CHID1, CRACR2, LRR MUC6 Orthologus of ovocumin (Lang et al., 2006) The ovumucin is around 3.5% of ovalbumin (Carey, 1990). Major role in gelation of fresh eggs (Nishinari et al., 2000) PAR2, PRR5L, LDLRAD3, PAMR1, ELF5, ABTB2, SHANK2, TSPAN4, LRRC56, AKAP6, ELF5, AKAP6, ARHGAP5, and STRN3 were found close to the significant SNPs LPLRAD2 as a candidate gene associated with egg quality traits in dwarf layer (Zhang et al., 2011) All these SNPs are synonymous |
Chromosome 5 |
Notably higher in oviduct than in other tissues |
(Liu et al., 2021b) |
|
Combination of GWAS and signature of selection Combination of GWAS and signature of selection |
Feed conversion ratio (FCR) of egg production traits |
LOC106017218, CRBN, TRNT1, IL5RA,andCNTN4 MMS22L, KLHL32, LOC106017657, NDUFAF4, GPR63, FHL5, LOC106017656, FUT9, andMANEA GDF2, RBP3, ZNF488,ANTXRL, ANXA8L1, LOC106017915, LOC101795151, LOC101795347, LOC101795539, ZFAND4, ALOX5, LOC101797666, BLOC1S2, LOC101797254, PKD2L1, LOC106018867, and SCD CNTN4 TRNT1 and CRBR MANEA FHL5 GPR63, NDUFAF4, KLHL32 and MMS22L SCD Regulates lipid metabolism LOC101795347 Annotated as lncRNA ZNF488, ANTXRL, ZFAND4, ALOX5 |
Chromosome 13 Chromosome 3 Chromosome 6 Chromosome 3 Chromosome 6 |
Not reported Highly expressed in nervous tissues (CPM> 96 in the hypothalamus, CPM>29- pituitary gland) Expressed in all investigated tissues As above Highly expressed in the ovary and the follicular membrane Relatively expressed in the ovary and the follicular membrane Highest expression level in the hypothalamus Highest expression level in the oviduct (an essential organ for egg formation) Moderately expressed in the ovary, oviduct, and the follicular membrane |
(Liu et al., 2021b) |
|
Cis-regulatory |
Mallard egg color Blue eggshell |
ABCG2 ABCG2 |
Not provided Not provided |
Not provided Not provided |
(Liu et al., 2021a) (Chen et al., 2020) |
CONCLUSIONS
The number of eggs produced by ducks is below the demand of the increasing human population across the globe; this necessitates the need to improve the egg production from this waterfowl bird as the protein requirement of human beings and economic income of duck farmers might be increased through this poultry specie. In our own opinion, the following remarks would be useful to improve research on candidate genes responsible for egg production in ducks, and the highlighted remarks are summarized in Figure 3 below.
-
•
Future studies should look into the polymorphic studies of other identified genes in association with egg production traits of ducks.
-
•
Future studies should incorporate antibodies for protein expression of the candidate genes in reproductive tissues of ducks to reveal the reliability of the qRT-PCR.
-
•
More studies on QTL and GWAS could be done to identify different SNP markers of genes that might be responsible for egg production in ducks.
-
•
Proteomics study of these genes would be helpful to understand the rising need for genetic improvement of egg production in duck.
-
•
There is a need to deeply study the correlation that exists between the genes at different QTL regions in ducks.
-
•
There is need to design a SNP chips for GWAS study.
Figure 3.
Perspectives towards the improvement of egg production in ducks.
DISCLOSURES
The authors declare that they have no competing interests relevant to this publication.
ACKNOWLEDGMENTS
We would like to appreciate Dr. Balogun Adedeji Suleimon, Anthony Bassey and Hammed Ayansola for their kind criticism during the draft of this review paper. We are grateful to Oladapo Taiwo for helping us with drawing of the figures. The Local Innovative and Research Teams Project of Guangdong Province (2019BT02N630), and China Agriculture Research System (CARS-41-G03) supported this work.
Author contributions: Conceptualization: S.F., A.C.A., Q.N.; writing of draft copy of the manuscript: S.F; editing: A.C.A., Q.N, Visualization; S.F. All authors have read and agreed to the published version of the manuscript.
REFERENCES
- Adeola A.C., Ewuola M.K., Nneji L.M., Oguntunji A.O., Bello S.F., Sola-Ojo F.E., Ayoola A.O., Adesoji A.T., Sanke O.J., Ebiakpo D.L., Jonah K.N., Jia X., Wu R.-N., Peng M.-S., Zhang Y.-P. Mitochondrial DNA variation of Nigerian Muscovy duck (Cairina moschata) Anim. Genet. 2020;51:485–486. doi: 10.1111/age.12924. [DOI] [PubMed] [Google Scholar]
- Akison, L., D. Russell, and R. Robker. 2010. Progesterone receptor-regulated genes in the preovulatory ovarian follicle and oviduct. Reprod. Fertil. Dev. 22:54–54.
- Alyethodi R.R., Kumar S., Panda B.K., Singh P., Jaiswal G., Choudhary S. Molecular genetic characterization of Moti native duck using RAPD markers. J. Appl. Anim. Res. 2010;39:19–23. [Google Scholar]
- Antosik P., Jeseta M., Kranc W., Chachuła A., Bryja A., Budna J., Ciesiołka S., Wojtanowicz-Markiewicz K., Bukowska D., Brussow K.P., Bruska M., Nowicki M., Hulinska P., Machatkova M., Kempisty B. Expression of integrins and GDF9 mRNAs is associated with ovarian follicle size and donor puberty status in pigs. Med. Weter. 2016;72:750–754. [Google Scholar]
- Armstrong D., Hogg C. Insulin-like growth factor I (IGF-I), IGF-II type-I IGF receptor gene expression in the ovary of the laying hen. J. Reprod. Fertil. 1996;106:101–106. doi: 10.1530/jrf.0.1060101. [DOI] [PubMed] [Google Scholar]
- Asiamah Amponsah C., Zou K., li Lu L., Zhang S.W., Xue Y., Su Y., Zhao Z. Genetic effects of polymorphisms of candidate genes associated with ovary development and egg production traits in ducks. Anim. Reprod. Sci. 2019:106219. doi: 10.1016/j.anireprosci.2019.106219. [DOI] [PubMed] [Google Scholar]
- Asiamah C.A., Liu Y., Ye R., Pan Y., Lu L.-L., Zou K., Zhao Z., Jiang P., Su Y. Polymorphism analysis and expression profile of the estrogen receptor 2 gene in Leizhou black duck. Poult. Sci. 2022;101:101630. doi: 10.1016/j.psj.2021.101630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bai D.-P., Hu Y.-Q., Li Y.-B., Huang Z.-B., Li A. Polymorphisms analysis of the prolactin gene with egg production traits in two Chinese domestic ducks. Br. Poult. Sci. 2019;60 doi: 10.1080/00071668.2019.1567909. [DOI] [PubMed] [Google Scholar]
- Banerjee S. Morphological traits of duck and geese breeds of West Bengal, India. Anim. Genet. Resour. 2013;52 [Google Scholar]
- Bao X., Song Y., Li T., Zhang S., Huang L., Zhang S., Cao J., Liu X., Zhang J. Comparative transcriptome profiling of ovary tissue between black Muscovy duck and white Muscovy duck with high- and low-egg production. Genes (Basel) 2020;12:57. doi: 10.3390/genes12010057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Barber D.L., Sanders E.J., Aebersold R., Schneider W.J. The receptor for yolk lipoprotein deposition in the chicken oocyte. J. Biol. Chem. 1991;266:18761–18770. [PubMed] [Google Scholar]
- Baumgarten S.C., Convissar S.M., Zamah A.M., Fierro M.A., Winston N.J., Scoccia B., Stocco C. FSH regulates IGF-2 expression in human granulosa cells in an AKT-dependent manner. J. Clin. Endocrinol. Metab. 2015;100:E1046–55. doi: 10.1210/jc.2015-1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Becker C., Riedmaier I., Reiter M., Tichopad A., Groot M.J., Stolker A.A.M., Pfaffl M.W., Nielen M.F.W., Meyer H.H.D. Influence of anabolic combinations of an androgen plus an estrogen on biochemical pathways in bovine uterine endometrium and ovary. J. Steroid Biochem. Mol. Biol. 2011;125:192–201. doi: 10.1016/j.jsbmb.2011.01.007. [DOI] [PubMed] [Google Scholar]
- Bédécarrats G.Y., McFarlane H., Maddineni S.R., Ramachandran R. Gonadotropin-inhibitory hormone receptor signaling and its impact on reproduction in chickens. Gen. Comp. Endocrinol. 2009;163:7–11. doi: 10.1016/j.ygcen.2009.03.010. [DOI] [PubMed] [Google Scholar]
- Bello S.F., Xu H., Guo L., Li K., Zheng M., Xu Y., Zhang S., Bekele E.J., Bahareldin A.A., Zhu W., Zhang D., Zhang X., Ji C., Nie Q. Hypothalamic and ovarian transcriptome profiling reveals potential candidate genes in low and high egg production of white Muscovy ducks (Cairina moschata) Poult. Sci. 2021;100 doi: 10.1016/j.psj.2021.101310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carey, C.. 1990. The Avian Egg: Chemistry and Biology R. W. Burley D. V. Vadehra. Auk 107.
- Chen L., Gu X., Huang X., Liu R., Li J., Hu Y., Li G., Zeng T., Tian Y., Hu X., Lu L., Li N. Two cis-regulatory SNPs upstream of ABCG2 synergistically cause the blue eggshell phenotype in the duck. PLoS Genet. 2020;16:e1009119. doi: 10.1371/journal.pgen.1009119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Compere S.J., McKnight G.S., Palmiter R.D. Androgens regulate ovomucoid and ovalbumin gene expression independently of estrogen. J. Biol. Chem. 1981;256:6341–6347. [PubMed] [Google Scholar]
- Couse J.F., Yates M.M., Deroo B.J., Korach K.S. Estrogen receptor-β is critical to granulosa cell differentiation and the ovulatory response to gonadotropins. Endocrinology. 2005;146:3247–3262. doi: 10.1210/en.2005-0213. [DOI] [PubMed] [Google Scholar]
- Craig Z.R., Wang W., Flaws J.A. Endocrine-disrupting chemicals in ovarian function: effects on steroidogenesis, metabolism and nuclear receptor signaling. Reproduction. 2011;142:633–646. doi: 10.1530/REP-11-0136. [DOI] [PubMed] [Google Scholar]
- Cui Y.ming, Wang J., Hai-jun Z., Feng J., geng Wu S., hai Qi G. Effect of photoperiod on ovarian morphology, reproductive hormone secretion, and hormone receptor mRNA expression in layer ducks during the pullet phase. Poult. Sci. 2019;98:2439–2447. doi: 10.3382/ps/pey601. [DOI] [PubMed] [Google Scholar]
- Dalrymple A., Jabbour H.N. Localization and signaling of the prolactin receptor in the uterus of the common marmoset monkey. J. Clin. Endocrinol. Metab. 2000;85:1711–1718. doi: 10.1210/jcem.85.4.6504. [DOI] [PubMed] [Google Scholar]
- Demeure O., Lecerf F., Duby C., Desert C., Ducheix S., Guillou H., Lagarrigue S. Regulation of LPCAT3 by LXR. Gene. 2011;470:7–11. doi: 10.1016/j.gene.2010.09.002. [DOI] [PubMed] [Google Scholar]
- Eppig J.J., Wigglesworth K., Pendola F.L. The mammalian oocyte orchestrates the rate of ovarian follicular development. Proc. Natl. Acad. Sci. U. S. A. 2002;99:2890–2894. doi: 10.1073/pnas.052658699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fabi F., Asselin E. Expression, activation, and role of Akt isoforms in the uterus. Reproduction. 2014;148:R85–95. doi: 10.1530/REP-14-0270. [DOI] [PubMed] [Google Scholar]
- Farmanullah S., Hosseini M., Liang A., Hua G., Rehman Z.U., Talpur H.S., Salim M., Ahmad S., Abulaiti A., Khan M., Safdar M., Kakar I.U., Ahmad Z., Ahmad M.Z., Tingzhu Y., Schreurs N.M., Bano I., Yang L. Adaptive molecular evolution of AKT3 gene for positive diversifying selection in mammals. Biomed. Res. Int. 2020;2020:2584627. doi: 10.1155/2020/2584627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng P., Zhao W., Xie Q., Zeng T., Lu L., Yang L. Polymorphisms of melatonin receptor genes and their associations with egg production traits in Shaoxing duck. Asian-Australas. J. Anim. Sci. 2018;31 doi: 10.5713/ajas.17.0828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- George J.W., Dille E.A., Heckert L.L. Current concepts of follicle-stimulating hormone receptor gene regulation. Biol. Reprod. 2011;84:7–17. doi: 10.1095/biolreprod.110.085043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goffin V., Bouchard B., Ormandy C.J., Weimann E., Ferrag F., Touraine P., Bole-Feysot C., Maaskant R.A., Clement-Lacroix P., Edery M., Binart N., Kelly P.A. Prolactin: a hormone at the crossroads of neuroimmunoendocrinology. Ann. N. Y. Acad. Sci. 1998;840:498–509. doi: 10.1111/j.1749-6632.1998.tb09588.x. [DOI] [PubMed] [Google Scholar]
- Goossens K., Van Soom A., Van Zeveren A., Favoreel H., Peelman L.J. Quantification of Fibronectin 1 (FN1) splice variants, including two novel ones, and analysis of integrins as candidate FN1 receptors in bovine preimplantation embryos. BMC Dev. Biol. 2009;9:1. doi: 10.1186/1471-213X-9-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grosdemouge I., Bachelot A., Lucas A., Baran N., Kelly P.A., Binart N. Effects of deletion of the prolactin receptor on ovarian gene expression. Reprod. Biol. Endocrinol. 2003;1:12. doi: 10.1186/1477-7827-1-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gu L., Wang F., Lin Z., Xu T., Lin D., Xing M., Yang S., Chao Z., Ye B., Lin P., Hui C., Lu L., Hou S. Genetic characteristics of Jiaji Duck by whole genome re-sequencing. PLoS One. 2020;15:e0228964. doi: 10.1371/journal.pone.0228964. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han D., Haunerland N.H., Williams T.D. Variation in yolk precursor receptor mRNA expression is a key determinant of reproductive phenotype in the zebra finch (Taeniopygia guttata) J. Exp. Biol. 2009;212(Pt 9):1277–1283. doi: 10.1242/jeb.026906. [DOI] [PubMed] [Google Scholar]
- Hansen C., Yi N., Zhang Y.M., Xu S., Gavora J., Cheng H.H. Identification of QTL for production traits in chickens. Anim. Biotechnol. 2005;16:67–69. doi: 10.1081/abio-200055016. [DOI] [PubMed] [Google Scholar]
- Harun M.A.S., Veeneklaas R.J., Van Kampen M., Mabasso M. Breeding bology of Muscovy duck Cairina moschata in natural incubation: the effect of nesting behavior on hatchability. Poult. Sci. 1998;77:1280–1286. doi: 10.1093/ps/77.9.1280. [DOI] [PubMed] [Google Scholar]
- Hattori Ma M., Furuta H., Hiyama Y., Kato Y., Fujihara N. Prominent expression of transforming growth factor beta2 gene in the chicken embryonic gonad as revealed by suppressive subtraction cloning. Gen. Comp. Endocrinol. 2002;125:311–316. doi: 10.1006/gcen.2001.7719. [DOI] [PubMed] [Google Scholar]
- Hatzirodos N., Irving-Rodgers H.F., Hummitzsch K., Harland M.L., Morris S.E., Rodgers R.J. Transcriptome profiling of granulosa cells of bovine ovarian follicles during growth from small to large antral sizes. BMC Genomics. 2014;15:24. doi: 10.1186/1471-2164-15-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Herrera L., Ottolenghi C., Garcia-Ortiz J.E., Pellegrini M., Manini F., Ko M.S.H., Nagaraja R., Forabosco A., Schlessinger D. Mouse ovary developmental RNA and protein markers from gene expression profiling. Dev. Biol. 2005;279:271–290. doi: 10.1016/j.ydbio.2004.11.029. [DOI] [PubMed] [Google Scholar]
- Höglund J.K., Buitenhuis B., Guldbrandtsen B., Lund M.S., Sahana G. Genome-wide association study for female fertility in Nordic Red cattle. BMC Genet. 2015;16:110. doi: 10.1186/s12863-015-0269-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu Z., Liu J., Cao J., Zhang H., Liu X. Ovarian transcriptomic analysis of black Muscovy duck at the early, peak and late egg-laying stages. Gene. 2021;777:145449. doi: 10.1016/j.gene.2021.145449. [DOI] [PubMed] [Google Scholar]
- Huang Z., Li X., Li Y., Liu R., Chen Y., Wu N., Wang M., Song Y., Yuan X., Lan L., Xu Q., Chen G., Zhao W. Molecular cloning and functional analysis of the goose FSHβ gene. Br. Poult. Sci. 2015;56 doi: 10.1080/00071668.2015.1020283. [DOI] [PubMed] [Google Scholar]
- Hussain M.M. Structural, biochemical and signaling properties of the low-density lipoprotein receptor gene family. Front. Biosci. 2001;6:D417–28. doi: 10.2741/hussain1. [DOI] [PubMed] [Google Scholar]
- Jeong J., Bae H., Lim W., Bazer F.W., Song G. Diethylstilbestrol regulates expression of avian apolipoprotein D during regression and recrudescence of the oviduct and epithelial-derived ovarian carcinogenesis. Domest. Anim. Endocrinol. 2015;52:82–89. doi: 10.1016/j.domaniend.2015.03.005. [DOI] [PubMed] [Google Scholar]
- Jiang R.S., Chen X.Y., Geng Z.Y. Broodiness, egg production, and correlations between broody traits in an indigenous chicken breed. Poult. Sci. 2010;89:1094–1096. doi: 10.3382/ps.2009-00621. [DOI] [PubMed] [Google Scholar]
- Kameshpandian P., Thomas S., Nagarajan M. Genetic diversity and relationship of Indian Muscovy duck populations. Mitochondrial DNA Part A. 2018;29:165–169. doi: 10.1080/24701394.2016.1261851. [DOI] [PubMed] [Google Scholar]
- Kang L., Zhang Y., Zhang N., Zang L., Wang M., Cui X., Jiang Y. Identification of differentially expressed genes in ovaries of chicken attaining sexual maturity at different ages. Mol. Biol. Rep. 2012;39:3037–3045. doi: 10.1007/s11033-011-1066-x. [DOI] [PubMed] [Google Scholar]
- Khristi V., Chakravarthi V.P., Singh P., Ghosh S., Pramanik A., Ratri A., Borosha S., Roby K.F., Wolfe M.W., Rumi M.A.K. ESR2 regulates granulosa cell genes essential for follicle maturation and ovulation. Mol. Cell. Endocrinol. 2018;474:214–226. doi: 10.1016/j.mce.2018.03.012. [DOI] [PubMed] [Google Scholar]
- Kinoshita K., Shimogiri T., Okamoto S., Yoshizawa K., Mannen H., Ibrahim H.R., Cheng H.H., Maeda Y. Linkage mapping of chicken ovoinhibitor and ovomucoid genes to chromosome 13. Anim. Genet. 2004;35:356–358. doi: 10.1111/j.1365-2052.2004.01159.x. [DOI] [PubMed] [Google Scholar]
- Kulus M., Sujka-Kordowska P., Konwerska A., Celichowski P., Kranc W., Kulus J., Piotrowska-Kempisty H., Antosik P., Bukowska D., Iżycki D., Bruska M., Zabel M., Nowicki M., Kempisty B. New molecular markers involved in regulation of ovarian granulosa cell morphogenesis, development and differentiation during short-term primary in vitro culture-transcriptomic and histochemical study based on ovaries and individual separated follicles. Int. J. Mol. Sci. 2019;20:3966. doi: 10.3390/ijms20163966. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar S., Banks T., Cloutier S. SNP discovery through next generation sequencing and its applications. Int. J. Plant Genomics. 2012;2012:831460. doi: 10.1155/2012/831460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kumar Kharwar R., Haldar C. Anatomical and histological profile of bronchus-associated lymphoid tissue and localization of melatonin receptor types (Mel1a and Mel1b) in the lung-associated immune system of a tropical bird, Perdicula asiatica. Acta Histochem. 2011;113:333–339. doi: 10.1016/j.acthis.2010.01.003. [DOI] [PubMed] [Google Scholar]
- Lagudah E., Krattinger S., Herrera-Foessel S., Singh R., Huerta-Espino J., Spielmeyer W., Brown-Guedira G., Selter L., Keller B. Gene-specific markers for the wheat gene Lr34/Yr18/Pm38 which confers resistance to multiple fungal pathogens. Theor. Appl. Genet. 2009;119:889–898. doi: 10.1007/s00122-009-1097-z. [DOI] [PubMed] [Google Scholar]
- Lang T., Hansson G.C., Samuelsson T. An inventory of mucin genes in the chicken genome shows that the mucin domain of Muc13 is encoded by multiple exons and that ovomucin is part of a locus of related gel-forming mucins. BMC Genomics. 2006;7:197. doi: 10.1186/1471-2164-7-197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lantang A.M., Innes B.A., Gan E.H., Pearce S.H., Lash G.E. Expression of melanocortin receptors in human endometrium. Hum. Reprod. 2015;30:2404–2410. doi: 10.1093/humrep/dev188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li B., Fillmore N., Bai Y., Collins M., Thomson J.A., Stewart R., Dewey C.N. Evaluation of de novo transcriptome assemblies from RNA-Seq data. Genome Biol. 2014;15:553. doi: 10.1186/s13059-014-0553-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li D.Y., Smith D.G., Hardel R., Yang M.Y., Xu H.L., Zhang L., Yin H.D., Zhu Q. Melatonin receptor genes in vertebrates. Int. J. Mol. Sci. 2013;14:11208–11223. doi: 10.3390/ijms140611208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li G., Sun D.X., Yu Y., Liu W.J., Tang S.Q., Zhang Y., Wang Y.C., Zhang S.L., Zhang Y. Genetic effect of the follicle-stimulating hormone receptor gene on reproductive traits in Beijing You chickens. Poult. Sci. 2011;90:2487–2492. doi: 10.3382/ps.2010-01327. [DOI] [PubMed] [Google Scholar]
- Liao P.Y., Lee K.H. From SNPs to functional polymorphism: the insight into biotechnology applications. Biochem. Eng. J. 2010;49:149–158. [Google Scholar]
- Lim W., Song G. Characteristics, tissue-specific expression, and hormonal regulation of expression of tyrosine aminotransferase in the avian female reproductive tract. Domest. Anim. Endocrinol. 2016;57:10–20. doi: 10.1016/j.domaniend.2016.05.001. [DOI] [PubMed] [Google Scholar]
- Liu H., Hu J., Guo Z., Fan W., Xu Y., Liang S., Liu D., Zhang Y., Xie M., Tang J., Huang W., Zhang Q., Xi Y., Li Y., Wang L., Ma S., Jiang Y., Feng Y., Wu Y., Cao J., Zhou Z., Hou S. A single nucleotide polymorphism variant located in the cis-regulatory region of the ABCG2 gene is associated with mallard egg colour. Mol. Ecol. 2021;30:1477–1491. doi: 10.1111/mec.15785. [DOI] [PubMed] [Google Scholar]
- Liu L.Bin, Li D.Y., Zhao Z.L., Liu Y.P., Wang Y., Zhu Q. Polymorphism of prolactin receptor gene and its association with egg production traits in Erlang mountainous chicken. Asian J. Anim. Vet. Adv. 2012;7:1183–1190. [Google Scholar]
- Liu W.H., Means G.E., Feeney R.E. The inhibitory properties of avian ovoinhibitors against proteolytic enzymes. BBA - Protein Struct. 1971;229:176–185. doi: 10.1016/0005-2795(71)90331-x. [DOI] [PubMed] [Google Scholar]
- Liu H., Wang L., Guo Z., Xu Q., Fan W., Xu Y., Hu J., Zhang Y., Tang J., Xie M., Zhou Z., Hou S. Genome-wide association and selective sweep analyses reveal genetic loci for FCR of egg production traits in ducks. Genet. Sel. Evol. 2021;53:98. doi: 10.1186/s12711-021-00684-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Z., Youngquist R.S., Garverick H.A., Antoniou E. Molecular mechanisms regulating bovine ovarian follicular selection. Mol. Reprod. Dev. 2009;76:351–366. doi: 10.1002/mrd.20967. [DOI] [PubMed] [Google Scholar]
- Liu G., Zeng M., Li X., Rong Y., Hu Z., Zhang H., Liu X. Expression and analysis of ESR1, IGF-1, FSH, VLDLR, LRP, LH, PRLR genes in Pekin duck and Black Muscovy duck. Gene. 2021 doi: 10.1016/j.gene.2020.145183. [DOI] [PubMed] [Google Scholar]
- Liu H.Y., Zhang C.Q. Effects of daidzein on messenger ribonucleic acid expression of gonadotropin receptors in chicken ovarian follicles. Poult. Sci. 2008;87:541–545. doi: 10.3382/ps.2007-00274. [DOI] [PubMed] [Google Scholar]
- Lussier J.G., Diouf M.N., Lévesque V., Sirois J., Ndiaye K. Gene expression profiling of upregulated mRNAs in granulosa cells of bovine ovulatory follicles following stimulation with hCG. Reprod. Biol. Endocrinol. 2017;15:88. doi: 10.1186/s12958-017-0306-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Magoori K., Kang M.J., Ito M.R., Kakuuchi H., Ioka R.X., Kamataki A., Kim D.H., Asaba H., Iwasaki S., Takei Y.A., Sasaki M., Usui S., Okazaki M., Takahashi S., Ono M., Nose M., Sakai J., Fujino T., Yamamoto T.T. Severe hypercholesterolemia, impaired fat tolerance, and advanced atherosclerosis in mice lacking both low density lipoprotein receptor-related protein 5 and apolipoprotein E. J. Biol. Chem. 2003;278:11331–11336. doi: 10.1074/jbc.M211987200. [DOI] [PubMed] [Google Scholar]
- Mao J., Smith M.F., Rucker E.B., Wu G.M., McCauley T.C., Cantley T.C., Prather R.S., Didion B.A., Day B.N. Effect of epidermal growth factor and insulin-like growth factor I on porcine preantral follicular growth, antrum formation, and stimulation of granulosal cell proliferation and suppression of apoptosis in vitro. J. Anim. Sci. 2004;82:1967–1975. doi: 10.2527/2004.8271967x. [DOI] [PubMed] [Google Scholar]
- Martínez-Moreno C.G., Palma L., Carranza M., Harvey S., Arámburo C., Luna M. Cellular and intracellular distribution of growth hormone in the adult chicken testis. Gen. Comp. Endocrinol. 2011;172:344–357. doi: 10.1016/j.ygcen.2011.03.023. [DOI] [PubMed] [Google Scholar]
- McFarland K.C., Sprengel R., Phillips H.S., Köhler M., Rosemblit N., Nikolics K., Segaloff D.L., Seeburg P.H. Lutropin-choriogonadotropin receptor: an unusual member of the G protein-coupled receptor family. Science. 1989;245:494–499. doi: 10.1126/science.2502842. [DOI] [PubMed] [Google Scholar]
- Mishra S.K., Chen B., Zhu Q., Xu Z., Ning C., Yin H., Wang Y., Zhao X., Fan X., Yang M., Yang D., Ni Q., Li Y., Zhang M., Li D. Transcriptome analysis reveals differentially expressed genes associated with high rates of egg production in chicken hypothalamic-pituitary-ovarian axis. Sci. Rep. 2020;10:5976. doi: 10.1038/s41598-020-62886-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mondal M., Dhali A., Rajkhowa C., Prakash B.S. Secretion patterns of growth hormone in growing captive mithuns (Bos frontalis) Zoolog. Sci. 2004;21:1125–1129. doi: 10.2108/zsj.21.1125. [DOI] [PubMed] [Google Scholar]
- Morozova O., Marra M.A. Applications of next-generation sequencing technologies in functional genomics. Genomics. 2008;92:255–264. doi: 10.1016/j.ygeno.2008.07.001. [DOI] [PubMed] [Google Scholar]
- Natesampillai S., Kerkvliet J., Leung P.C.K., Veldhuis J.D. Regulation of Kruppel-like factor 4, 9, and 13 genes and the steroidogenic genes LDLR, StAR, and CYP11A in ovarian granulosa cells. Am. J. Physiol. Endocrinol. Metab. 2008;294:E385–91. doi: 10.1152/ajpendo.00480.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nelson L.R., Bulun S.E. Estrogen production and action. J. Am. Acad. Dermatol. 2001;45(Suppl 3):S116–24. doi: 10.1067/mjd.2001.117432. [DOI] [PubMed] [Google Scholar]
- Ni Y., Zhou Y., Lu L., Grossmann R., Zhao R. Developmental changes of FSH-R, LH-R, ER-β and GnRH-I expression in the ovary of prepubertal ducks (Anas platyrhynchos) Anim. Reprod. Sci. 2007 doi: 10.1016/j.anireprosci.2006.08.012. [DOI] [PubMed] [Google Scholar]
- Ni Y., Zhu Q., Zhou Z., Grossmann R., Chen J., Zhao R. Effect of dietary daidzein on egg production, shell quality, and gene expression of ER-α, GH-R, and IGF-IR in shell glands of laying hens. J. Agric. Food Chem. 2007 doi: 10.1021/jf071085r. [DOI] [PubMed] [Google Scholar]
- Nishinari K., Zhang H., Ikeda S. Hydrocolloid gels of polysaccharides and proteins. Curr. Opin. Colloid Interface Sci. 2000;5:195–201. [Google Scholar]
- Niu X., Tyasi T.L., Qin N., Liu D., Zhu H., Chen X., Zhang F., Yuan S., Xu R. Sequence variations in estrogen receptor 1 and 2 genes and their association with egg production traits in Chinese Dagu chickens. J. Vet. Med. Sci. 2017;79 doi: 10.1292/jvms.17-0014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nykjaer A., Willnow T.E. The low-density lipoprotein receptor gene family: a cellular Swiss army knife? Trends Cell Biol. 2002;12:273–280. doi: 10.1016/s0962-8924(02)02282-1. [DOI] [PubMed] [Google Scholar]
- Ouyang Q., Hu S., Wang G., Hu J., Zhang J., Li L., Hu B., He H., Liu H., Xia L., Wang J. Comparative transcriptome analysis suggests key roles for 5-hydroxytryptamlne receptors in control of goose egg production. Genes (Basel) 2020;11:455. doi: 10.3390/genes11040455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ożegowska K., Brązert M., Ciesiółka S., Nawrocki M.J., Kranc W., Celichowski P., Jankowski M., Bryja A., Jeseta M., Antosik P., Bukowska D., Skowroński M.T., Bruska M., Pawelczyk L., Zabel M., Nowicki M., Kempisty B. Genes involved in the processes of cell proliferation, migration, adhesion, and tissue development as new potential markers of porcine granulosa cellular processes in vitro: a microarray approach. DNA Cell Biol. 2019;38:549–560. doi: 10.1089/dna.2018.4467. [DOI] [PubMed] [Google Scholar]
- Padhi M.K. Evaluation of indigenous ducks of Odisha, India. Worlds. Poult. Sci. J. 2014;70:617–626. [Google Scholar]
- Padmanabhan V., Karsch F.J., Lee J.S. Hypothalamic, pituitary and gonadal regulation of FSH. Reprod Suppl. 2002;59:67–82. [PubMed] [Google Scholar]
- Papadopoulos C., Arato K., Lilienthal E., Zerweck J., Schutkowski M., Chatain N., Müller-Newen G., Becker W., De La Luna S. Splice variants of the dual specificity tyrosine phosphorylation-regulated kinase 4 (DYRK4) differ in their subcellular localization and catalytic activity. J. Biol. Chem. 2011;286:5494–5505. doi: 10.1074/jbc.M110.157909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pazin D.E., Albrecht K.H. Developmental expression of Smoc1 and Smoc2 suggests potential roles in fetal gonad and reproductive tract differentiation. Dev. Dyn. 2009;238:2877–2890. doi: 10.1002/dvdy.22124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Polley S., De S., Brahma B., Mukherjee A., V P.V., Batabyal S., Arora J.S., Pan S., Samanta A.K., Datta T.K., Goswami S.L. Polymorphism of BMPR1B, BMP15 and GDF9 fecundity genes in prolific Garole sheep. Trop. Anim. Health Prod. 2010;42:985–993. doi: 10.1007/s11250-009-9518-1. [DOI] [PubMed] [Google Scholar]
- Prochazkova B., Komrskova P., Kubelka M. CPEB2 is necessary for proper porcine meiotic maturation and embryonic development. Int. J. Mol. Sci. 2018;19:3138. doi: 10.3390/ijms19103138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pruitt K.D., Tatusova T., Klimke W., Maglott D.R. NCBI reference sequences: current status, policy and new initiatives. Nucleic Acids Res. 2009;37:D32–6. doi: 10.1093/nar/gkn721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qin N., Liu Q., Zhang Y.Y., Fan X.C., Xu X.X., Lv Z.C., Wei M.L., Jing Y., Mu F., Xu R.F. Association of novel polymorphisms of forkhead box L2 and growth differentiation factor-9 genes with egg production traits in local Chinese Dagu hens. Poult. Sci. 2015;94:88–95. doi: 10.3382/ps/peu023. [DOI] [PubMed] [Google Scholar]
- Quan Q., Zheng Q., Ling Y., Fang F., Chu M., Zhang X., Liu Y., Li W. Comparative analysis of differentially expressed genes between the ovaries from pregnant and nonpregnant goats using RNA-Seq. J. Biol. Res. (Thessalonike, Greece) 2019;26:3. doi: 10.1186/s40709-019-0095-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ren J., Sun C., Chen L., Hu J., Huang X., Liu X., Lu L. Exploring differentially expressed key genes related to development of follicle by RNA-seq in Peking ducks (Anas Platyrhynchos) PLoS One. 2018;14:e0209061. doi: 10.1371/journal.pone.0209061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Reppert S.M., Weaver D.R., Cassone V.M., Godson C., Kolakowski L.F. Melatonin receptors are for the birds: molecular analysis of two receptor subtypes differentially expressed in chick brain. Neuron. 1995;15:1003–1015. doi: 10.1016/0896-6273(95)90090-x. [DOI] [PubMed] [Google Scholar]
- Schennink A., Bovenhuis H., Kloosterziel K.M., Arendonk J., Visker M. Effect of polymorphisms in the FASN, OLR1, PPARGC1A, PRL and STAT5A genes on bovine milk-fat composition. Anim. Genet. 2009;40:909–916. doi: 10.1111/j.1365-2052.2009.01940.x. [DOI] [PubMed] [Google Scholar]
- Seo H., Kim M., Choi Y., Lee C.K., Ka H. Analysis of lysophosphatidic acid (LPA) receptor and LPA-induced endometrial prostaglandin-endoperoxide synthase 2 expression in the porcine uterus. Endocrinology. 2008;149:6166–6175. doi: 10.1210/en.2008-0354. [DOI] [PubMed] [Google Scholar]
- Shen X., Steyrer E., Retzek H., Sanders E.J., Schneider W.J. Chicken oocyte growth: receptor-mediated yolk deposition. Cell Tissue Res. 1993;272:459–471. doi: 10.1007/BF00318552. [DOI] [PubMed] [Google Scholar]
- Singh H., Endo Y., Nie G. Decidual HtrA3 negatively regulates trophoblast invasion during human placentation. Hum. Reprod. 2011;26:748–757. doi: 10.1093/humrep/der019. [DOI] [PubMed] [Google Scholar]
- Słowińska M., Liszewska E., Nynca J., Bukowska J., Hejmej A., Bilińska B., Szubstarski J., Kozłowski K., Jankowski J., Ciereszko A. Isolation and characterization of an ovoinhibitor, a multidomain Kazal-like inhibitor from turkey (Meleagris gallopavo) seminal plasma. Biol. Reprod. 2014;91:108. doi: 10.1095/biolreprod.114.118836. [DOI] [PubMed] [Google Scholar]
- Stahl, P. W. 2008. Animal domestication in South America.in The Handbook of South American Archaeology.
- Su H., Silversides F.G., Villeneuve P. Ovarian follicular growth and maturity and follicular production of progesterone and oestradiol in response to porcine luteinising hormone and porcine follicle stimulating hormone in albino (S*AS) hens in vivo and in vitro. Br. Poult. Sci. 1999;40:545–551. doi: 10.1080/00071669987340. [DOI] [PubMed] [Google Scholar]
- Sun Y., Wu Q., Pan J., Li T., Liu L., Chen D., Zhang X., Chen H., Li Y., Lin R. Identification of differentially expressed genes and signalling pathways in the ovary of higher and lower laying ducks. Br. Poult. Sci. 2020;61:1–6. doi: 10.1080/00071668.2020.1792834. [DOI] [PubMed] [Google Scholar]
- Tao Z., Song W., Zhu C., Xu W., Liu H., Zhang S., Huifang L. Comparative transcriptomic analysis of high and low egg-producing duck ovaries. Poult. Sci. 2017;96:4378–4388. doi: 10.3382/ps/pex229. [DOI] [PubMed] [Google Scholar]
- Trivedi A.K., Kumar V. Melatonin: an internal signal for daily and seasonal timing. Indian J. Exp. Biol. 2014;52:425–437. [PubMed] [Google Scholar]
- Tuiskula-Haavisto M., Honkatukia M., Vilkki J., De Koning D.J., Schulman N.F., Mäki-Tanila A. Mapping of quantitative trait loci affecting quality and production traits in egg layers. Poult. Sci. 2002;81:919–927. doi: 10.1093/ps/81.7.919. [DOI] [PubMed] [Google Scholar]
- Veeramani P., Prabakaran R., S N S., Sivakumar T., Shanmugam S., Smk K. Phylogenetic analysis of six duck populations. Indian J. Anim. Res. 2016;50 [Google Scholar]
- Vera C., Tapia V., Vega M., Romero C. Role of nerve growth factor and its TRKA receptor in normal ovarian and epithelial ovarian cancer angiogenesis. J. Ovarian Res. 2014;7:82. doi: 10.1186/s13048-014-0082-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Z., Li Q., Chamba Y., Zhang B., Shang P., Zhang H., Wu C. Identification of genes related to growth and lipid deposition from transcriptome profiles of pig muscle tissue. PLoS One. 2015 doi: 10.1371/journal.pone.0141138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C., Li S.J., Li C., Yu G.H., Feng Y.P., Peng X.L., Gong Y.Z. Molecular cloning, expression and association study with reproductive traits of the duck LRP8 gene. Br. Poult. Sci. 2013;54:567–574. doi: 10.1080/00071668.2013.819488. [DOI] [PubMed] [Google Scholar]
- Wang C., Li S.J., Yu W.H., Xin Q.W., Li C., Feng Y.P., Peng X.L., Gong Y.Z. Cloning and expression profiling of the VLDLR gene associated with egg performance in duck (Anas platyrhynchos) Genet. Sel. Evol. 2011 doi: 10.1186/1297-9686-43-29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang C., Liang Z., Yu W., Feng Y., Peng X., Gong Y., Li S. Polymorphism of the prolactin gene and its association with egg production traits in native Chinese ducks. South Afr. J. Anim. Sci. 2011 [Google Scholar]
- Wang Z., Meng G., Bai Y., Liu R., Du Y., Su L. Comparative transcriptome analysis provides clues to molecular mechanisms underlying blue-green eggshell color in the Jinding duck (Anas platyrhynchos) BMC Genomics. 2017 doi: 10.1186/s12864-017-4135-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Z.P., Meng G.H., Li N., Yu M.F., Liang X.W., Min Y.N., Liu F.Z., Gao Y.P. The association of very low-density lipoprotein receptor (VLDLR) haplotypes with egg production indicates VLDLR is a candidate gene for modulating egg production. Genet. Mol. Biol. 2016;39:380–391. doi: 10.1590/1678-4685-GMB-2015-0206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White C.W., Choong Y.T., Short J.L., Exintaris B., Malone D.T., Allen A.M., Evans R.J., Ventura S. Male contraception via simultaneous knockout of α1A-adrenoceptors and P2X1-purinoceptors in mice. Proc. Natl. Acad. Sci. U. S. A. 2013;110:20825–20830. doi: 10.1073/pnas.1318624110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Willnow T.E. The low-density lipoprotein receptor gene family: multiple roles in lipid metabolism. J. Mol. Med. 1999;77:306–315. doi: 10.1007/s001090050356. [DOI] [PubMed] [Google Scholar]
- Woodruff T.K., Shea L.D. The role of the extracellular matrix in ovarian follicle development. Reprod. Sci. 2007;14(Suppl 8):6–10. doi: 10.1177/1933719107309818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Worku T., Wang K., Ayers D., Wu D., Ur Rehman Z., Zhou H., Yang L. Regulatory roles of ephrinA5 and its novel signaling pathway in mouse primary granulosa cell apoptosis and proliferation. Cell Cycle. 2018;17:892–902. doi: 10.1080/15384101.2018.1456297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu Y., Liang H., Zhang H., Pi J.S., Pan A.L., Shen J., Pu Y.J., Du J.P. The differential expression and snp analysis of the ovoinhibitor gene in the ovaries of laying duck breeds (Anas Platyrhynchos) Braz. J. Poult. Sci. 2018;20:281–286. [Google Scholar]
- Wu J.Y., Sun Y.X., Wang A.B., Che G.Y., Hu T.J., Zhang X.M. Effect of newborn bovine serum on cryopreservation of adult bovine testicular tissue. Andrologia. 2014 doi: 10.1111/and.12084. [DOI] [PubMed] [Google Scholar]
- Wu Y., Xiao H.W., Liang Z.H., Pan A.L., Shen J., Pi J.S., Pu Y.J., Du J.P., Chen Z.H. Differential expression profiling of estrogen receptor in the ovaries of two egg duck (Anas platyrhynchos) breeds. Czech J. Anim. Sci. 2014;59:238–243. [Google Scholar]
- Wu X., Yan M., Lian S., Liu X., Li A. GH gene polymorphisms and expression associated with egg laying in muscovy ducks (Cairina moschata) Hereditas. 2014;151 doi: 10.1111/j.1601-5223.2013.00016.x. [DOI] [PubMed] [Google Scholar]
- Xu J., Gao X., Li X., Ye Q., Bekele E., Abdalla B., Nie Q. Molecular characterization, expression profile of the FSHR gene and its association with egg production traits in muscovy duck. J. Genet. 2017;96:341–351. doi: 10.1007/s12041-017-0783-x. [DOI] [PubMed] [Google Scholar]
- Xu R., Qin N., Xu X., Sun X., Chen X., Zhao J. Inhibitory effect of SLIT2 on granulosa cell proliferation mediated by the CDC42-PAKs-ERK1/2 MAPK pathway in the prehierarchical follicles of the chicken ovary. Sci. Rep. 2018;8:9168. doi: 10.1038/s41598-018-27601-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yadav S.K., Haldar C. Reciprocal interaction between melatonin receptors (Mel1a, Mel1b, and Mel1c) and androgen receptor (AR) expression in immunoregulation of a seasonally breeding bird, Perdicula asiatica: role of photoperiod. J. Photochem. Photobiol. B Biol. 2013;122:52–60. doi: 10.1016/j.jphotobiol.2013.03.005. [DOI] [PubMed] [Google Scholar]
- Yamamura N., Takeishi M., Goto H., Tagami M., Mizutani T., Miyamoto K., Doi O., Kamiyoshi M. Expression of messenger RNA for gonadotropin receptor in the granulosa layer during the ovulatory cycle of hens. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2001;129:327–337. doi: 10.1016/s1095-6433(00)00350-0. [DOI] [PubMed] [Google Scholar]
- Yao J.F., Chen Z.X., Xu G.Y., Wang X.L., Ning Z.H., Zheng J.X., Qu L.J., Yang N. Low-density lipoprotein receptor-related protein 8 gene association with egg traits in dwarf chickens. Poult. Sci. 2010;89:883–886. doi: 10.3382/ps.2009-00613. [DOI] [PubMed] [Google Scholar]
- Ye P., Ge K., Li M., Yang L., Jin S., Zhang C., Chen X., Geng Z. Egg-laying and brooding stage-specific hormonal response and transcriptional regulation in pituitary of Muscovy duck (Cairina moschata) Poult. Sci. 2019;98 doi: 10.3382/ps/pez433. [DOI] [PubMed] [Google Scholar]
- Ye P., Li M., Liao W., Ge K., Jin S., Zhang C., Chen X., Geng Z. Hypothalamic transcriptome analysis reveals the neuroendocrine mechanisms in controlling broodiness of Muscovy duck (Cairina moschata) PLoS One. 2019 doi: 10.1371/journal.pone.0207050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ye Q., Xu J., Gao X., Ouyang H., Luo W., Nie Q. Associations of IGF2 and DRD2 polymorphisms with laying traits in Muscovy duck. Peer J. 2017;5:e4083. doi: 10.7717/peerj.4083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- You S., Bridgham J.T., Foster D.N., Johnson A.L. Characterization of the chicken follicle-stimulating hormone receptor (cFSH-R) complementary deoxyribonucleic acid, and expression of cFSH-R messenger ribonucleic acid in the ovary. Biol. Reprod. 1996;55:1055–1062. doi: 10.1095/biolreprod55.5.1055. [DOI] [PubMed] [Google Scholar]
- Youngren O.M., Chaiseha Y., El Halawani M.E. Serotonergic stimulation of avian prolactin secretion requires an intact dopaminergic system. Gen. Comp. Endocrinol. 1998;112:63–68. doi: 10.1006/gcen.1998.7130. [DOI] [PubMed] [Google Scholar]
- Youngren O.M., Pitts G.R., Phillips R.E., El Halawani M.E. Dopaminergic control of prolactin secretion in the Turkey. Gen. Comp. Endocrinol. 1996;104:225–230. doi: 10.1006/gcen.1996.0165. [DOI] [PubMed] [Google Scholar]
- Yu J., Lou Y., Zhao A. Transcriptome analysis of follicles reveals the importance of autophagy and hormones in regulating broodiness of Zhedong white goose. Sci. Rep. 2016;6:36877. doi: 10.1038/srep36877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuan J., Sun C., Dou T., Yi G., Qu L.J., Qu L., Wang K., Yang N. Identification of promising mutants associated with egg production traits revealed by genome-wide association study. PLoS One. 2015;10:e0140615. doi: 10.1371/journal.pone.0140615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeng T., Chen L., Du X., Lai S.J., Huang S.P., Liu Y.L., Lu L.Z. Association analysis between feed efficiency studies and expression of hypothalamic neuropeptide genes in laying ducks. Anim. Genet. 2016;47:606–669. doi: 10.1111/age.12457. [DOI] [PubMed] [Google Scholar]
- Zeng T., Jiang X., Li J., Wang D., Li G., Lu L., Wang G. Comparative proteomic analysis of the hepatic response to heat stress in Muscovy and Pekin ducks: insight into thermal tolerance related to energy metabolism. PLoS One. 2013;8:e76917. doi: 10.1371/journal.pone.0076917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zeng T., Zhang L., Li J., Wang D., Tian Y., Lu L. De novo assembly and characterization of Muscovy duck liver transcriptome and analysis of differentially regulated genes in response to heat stress. Cell Stress Chaperones. 2015;20:483–493. doi: 10.1007/s12192-015-0573-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang Y., Hou Z., Chen Z., Zheng J., Chen S., Qu L., Li J., Xu G., Yang N. Low-density lipoprotein receptor-related protein 2 gene is associated with egg-quality traits in dwarf layers. Poult. Sci. 2011;90:2718–2722. doi: 10.3382/ps.2011-01751. [DOI] [PubMed] [Google Scholar]
- Zhang X., Ibrahimi O.A., Olsen S.K., Umemori H., Mohammadi M., Ornitz D.M. Receptor specificity of the fibroblast growth factor family: the complete mammalian FGF family. J. Biol. Chem. 2006;281:15694–15700. doi: 10.1074/jbc.M601252200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang C., Shimada K., Saito N., Kansaku N. Expression of messenger ribonucleic acids of luteinizing hormone and follicle-stimulating hormone receptors in granulosa and theca layers of chicken preovulatory follicles. Gen. Comp. Endocrinol. 1997;105:402–409. doi: 10.1006/gcen.1996.6843. [DOI] [PubMed] [Google Scholar]
- Zhang F., Yin Z.-T., Zhang J.-F., Zhu F., Hincke M., Yang N., Hou Z.-C. Integrating transcriptome, proteome and QTL data to discover functionally important genes for duck eggshell and albumen formation. Genomics. 2020;112:3687–3695. doi: 10.1016/j.ygeno.2020.04.015. [DOI] [PubMed] [Google Scholar]
- Zhu Z.M., Miao Z.W., Chen H.P., Xin Q.W., Li L., Lin R.L., Huang Q.Lou, Zheng N.Z. Ovarian transcriptomic analysis of Shan Ma ducks at peak and late stages of egg production. Asian-Australas. J. Anim. Sci. 2017;30:1215–1224. doi: 10.5713/ajas.16.0470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhu F., Zhang F., Hincke M., Yin Z.T., Chen S.R., Yang N., Hou Z.C. iTRAQ-based quantitative proteomic analysis of duck eggshell during biomineralization. Proteomics. 2019;19:e1900011. doi: 10.1002/pmic.201900011. [DOI] [PubMed] [Google Scholar]
- Zou K., Asiamah C.A., li Lu L., Liu Y., Pan Y., Chen T., Zhao Z., Su Y. Ovarian transcriptomic analysis and follicular development of Leizhou black duck. Poult. Sci. 2020;99:6173–6187. doi: 10.1016/j.psj.2020.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]



