Open access peer-reviewed chapter

Gene Regulation in Ruminants: A Nutritional Perspective

Written By

Johan S. Osorio and Sonia J. Moisa

Submitted: 18 April 2018 Reviewed: 23 October 2018 Published: 28 January 2019

DOI: 10.5772/intechopen.82193

From the Edited Volume

Gene Expression and Control

Edited by Fumiaki Uchiumi

Chapter metrics overview

1,803 Chapter Downloads

View Full Metrics

Abstract

This chapter will focus on cellular regulatory programs implemented by the ruminant physiology in order to respond to external stimuli such as nutrition as well as important physiological events such as parturition. The increasing adoption of “omics” technologies and bioinformatics in nutrition and physiology in ruminant research have allowed us to delineate a clearer picture on what regulates major biological process at a molecular level such as milk synthesis and meat quality and fatty acid composition as well as pathological conditions such as ketosis, mastitis, and heat stress. The assembly of such plethora of information in a blend among nutritional research, molecular biology, and novel tools to study the response of the genome to nutrition has led to emerging disciplines such as nutritional genomics or “nutrigenomics.”

Keywords

  • nutrigenomics
  • transcription factors
  • ruminants

1. Introduction

The increasing adoption of molecular biology techniques and bioinformatics in nutrition and physiology in ruminant research has provided a wealth of knowledge on regulatory mechanisms of major biological processes related to milk synthesis and meat quality and marbling at a cellular level. This body of knowledge has prompted a compelling case for a change in the paradigm in ruminant nutrition, where nutrients in ruminant diets can act as bioactive molecules and exert alterations in molecular mechanisms depending on the animal physiological state. Such alterations can be carried out through gene regulation mechanisms, also known as nutrigenomics. The continuous accumulation of nutrient-gene interactions in ruminant research will eventually lead to practical applications where nutritional interventions may be made in order to improve performance and efficiency in milk yield or skeletal muscle.

1.1. Ruminant model for nutrient-gene interactions

The adoption of advanced molecular technologies in basic nutritional research in ruminants has led to a more robust notion of how nutrients can affect the animal at the cellular level. Then, this body of knowledge has led toward a general notion in ruminant nutrition, where nutrients in the diet can no longer be considered only as: (1) the building blocks for cells, tissue, and organs or (2) energy source for cell metabolism and basic cell function, but rather a new alternative concept for nutrient is that they (3) act as bioactive molecules that can regulate fundamental molecular mechanisms depending on the animal physiological stage.

Because of the inherited gastrointestinal differences between ruminants and monogastrics, the final effect of a nutrient at the molecular level will differ primarily based on how susceptible such a nutrient is to rumen fermentation. Therefore, rumen fermentation and kinetics play an important role in the context of nutrient-gene interaction in ruminants (Figure 1). Then, from a nutrigenomic standpoint, a given nutrient in a ruminant diet will likely be fermented or bypass the rumen. If fermented in the rumen, this nutrient will become either part of the microbial biomass or an intermediate metabolite such as volatile fatty acids (VFA) which can be absorbed through the rumen wall and enter the metabolism of ruminants. In the case of nutrients bypassing the rumen, these can be converted to intermediate metabolites [1], produce a signal transduction cascade [2, 3], or directly bind and activate specialized cellular proteins called transcription factors (or nuclear receptors) [4, 5] which are responsible for carrying out the final change in gene expression by binding to specific sections in the DNA upstream of the target gene. Some transcription factors can create a secondary wave of change in gene expression by upregulating the transcription of subsequent transcription factor [6], and previously, it has been proposed that transcription factors may work in an orchestrated fashion creating a network of transcription factors that respond to dietary effects [7]. An alternative effect from intermediate metabolites is the production of DNA or histone modifications by changing the available information in the DNA [8], also known as epigenetic effects. A potential epigenetic mechanism mediated by transcription factors is the increased transcription of noncoding RNAs such as microRNAs [9], which upon transcription these small RNAs will target coding RNA prior to their translation into proteins.

Figure 1.

Proposed ruminant model for gene expression regulation of dietary nutrients through transcription factor activation and epigenetic mechanism (i.e., DNA or histone methylation and noncoding RNA).

1.2. Mediators of nutrient-gene interactions in ruminants

Nutrients and bioactive compounds in regular ruminant diets will mainly interact indirectly with the genome through mediators in the form of specialized molecular proteins such as transcription factors, DNA methyltransferases, histone methyltransferases, among others. Here, we provide a brief overview of the major mediators of nutrient-gene interactions in ruminants known to date. Based on the ruminant model for nutrient-gene interactions (Figure 1), the specific transcription factors, enzymes, or cellular mechanisms with in vivo or in vitro data in ruminants are presented in Table 1. Among the known transcription factors with nutrigenomic potential, the peroxisome proliferator-activated receptors (PPARs) have been well studied in ruminants [10]. These transcription factors belong to the ligand-dependent nuclear receptors (LdNR) family [11], and their importance for nutrigenomic interventions in ruminants relies on their ability to bind and be activated by long-chained fatty acids (LCFA) commonly present in ruminant diets. The PPAR isotypes (e.g., α, γ, and δ) play multiple roles across several tissues in mammals, for instance, PPARγ has been observed to regulate adipogenesis and insulin sensitivity [12, 13], while PPARα has a crucial role in hepatic fatty acid catabolism [14]. In contrast to PPARγ and PPARα, PPARδ has been studied to a lesser extent; however, it is known for its role in fatty acid catabolism in skeletal muscle [15] and regulation of glucose uptake [7]. Additionally, Bionaz and collaborators [10] proposed a model for the concomitant and orchestrated regulation of major physiological adaptations by the three isotypes of PPARs in dairy cows going from late pregnancy into lactation. These effects are exerted across several tissues (e.g., liver, skeletal muscle, mammary gland, adipose, immune cells, etc.) where PPARs have a strong effect, and their ability to be activated by dietary fatty acids makes them a strong candidate for nutrigenomic effects in ruminants. The PPARs exert similar effects as observed in dairy cows in other ruminants, for instance, PPARγ has been associated with adipogenic effects in beef, cows [16, 17], and goats [18] as well as fatty acid oxidation by PPARα in transition dairy goats [19].

Common name Protein symbol Gene symbol Agonist Main function Ruminant1 Reference
Transcription factors
Peroxisome proliferator-activated receptor α PPARα PPARA Fatty acids Fatty acid metabolism, inflammation, and tissue regeneration B, D, G, and S [10, 24], [126, 127]
Peroxisome proliferator-activated receptor γ PPARγ PPARG Fatty acids Adipogenesis, insulin sensitivity, and lipogenesis B, D, G, and S [10, 16, 17, 18, 24], [126]
Peroxisome proliferator-activated receptor β PPARβ PPARD Fatty acids Fatty acid metabolism, tissue regeneration, and glucose uptake in mammary tissue B, D, G, and S [10], [127–130]
Liver X receptor α LXRα NR1H3 Oxysterols/fatty acids Cholesterol homeostasis, macrophage functions, and inflammation B, D, and G [22, 62], [131]
Retinoic X receptor α RXRα RXRA 9-cis-retinoic acid Forming heterodimers with other LdNR and neutrophil differentiation B and D [132–134]
Sterol regulatory element-binding protein 1 SREBP1 SREBF1 N/A Cholesterol and fatty acid synthesis B, D, G, and S [25, 26], [135]
DNA methyltransferases
DNA methyltransferase 1 DNMT1 DNMT1 N/A Maintenance of methylation patterns B and D [72, 105]
DNA methyltransferase 3 α DNMT3a DNMT3A N/A Creates de novo methylation patterns. Present in cytoplasm and nucleus B and D [72], [136]
DNA methyltransferase 3 β DNMT3b DNMT3B N/A Creates de novo methylation patterns restricted to nucleus B and D [137, 138]
Noncoding RNA
MicroRNA 33 miR33b Regulates lipogenesis D [30]
MicroRNA 192 miR192 Regulates myogenesis S [139]

Table 1.

Important mediators associated with nutrient-gene interactions in ruminants via transcriptional regulatory factors (transcription factors) and epigenetic factors (DNA methyltransferases and noncoding RNAs).

Ruminant as B = beef cows, D = dairy cows, G = goats, and S = sheep.


Similar to the PPARs, the liver X receptor (LXR) belongs to the LdNR family and has a prominent role in controlling cholesterol synthesis [15]. The LXR is known to be activated by oxysterols and derivatives from cholesterol metabolism, and fatty acids [15]. From the two known isoforms of LXR (e.g., α and β), the LXRα presents interesting characteristics including the potential control of sterol regulatory element-binding transcription factor 1 (SREBF1) gene expression [20], which is a major transcription factor associated with the regulation of milk fat synthesis [21]. The ability of LXRα to regulate SRBEF1 expression confers this TF a strong potential to enhance milk fat synthesis in ruminants; however, most of the current data on LXRα activity have been conducted with synthetic agonist [20, 22]. Therefore, a stronger case for the nutrigenomic potential of this TF could be made by future research including its activation by common fatty acids present in ruminant diets.

Retinoids are metabolites derived from vitamin A, and they can regulate gene expression through two classes of receptors: retinoic acid receptors and retinoid X receptors (RXR). The latter can form homodimers and be activated in the presence of the retinoid 9-cis-retinoic acid and consequently activating specific target genes [23]. From the two isoforms (i.e., α and β) of RXR, the RXRα has been the most evaluated in ruminants, primarily because it can form heterodimers with most LdNR including PPAR, LXR, and VDR [24]. Although the latter confers RXRα a tremendous biological significance, there are limited data in ruminants on the potential nutrigenomic effects of vitamin A and derivative retinoids such as 9-cis-retinoic acid through RXRα.

The study of milk fat depression revealed the importance of SREBP1 in milk fat synthesis [25], which resulted in a deep understanding how t10,c12 CLA, and a milk fat-depressing diet consistently downregulate SREBF1 in bovine mammary tissue [21]. Consequently, this effect downregulates the expression of genes associated with milk fat synthesis such as fatty acid synthase (FASN), lipoprotein lipase (LPL), and insulin-induced gene 1 (INSIG1) [21]. The importance of this TF for nutrigenomics in ruminants cannot be overstated since this was the first nutrigenomic effect documented. And the importance of this TF in the regulation of fat synthesis has also been observed in beef cattle and translated in marbling and meat quality [26]. A section on this commonality between dairy and beef cattle is dedicated at the end of this chapter.

Epigenetic mechanisms play a significant role as mediators of nutrient-gene interactions in ruminants and the ramifications of these effects in ruminant nutrition and physiology are only beginning to be uncovered, and they add another layer of complexity to our model (Figure 1). From a nutrigenomic standpoint, methyl donors present in common diets fed to ruminants such as folate, vitamin B (e.g., 2, 6, 12), choline, and methionine can regulate epigenetic modifications through the one-carbon metabolism where the intermediate s-adenosylmethionine (SAM) is produced and subsequently used as the universal methyl donor for DNA and histone methylations [27]. These effects are carried out through specialized enzymes such as DNA and histone methyltransferases. While the effect of dietary methyl donors on DNA methyltransferases has been evaluated in ruminants, the effect of methyl donors on histone methyltransferases in ruminants remains unknown. Similarly, other histone modifications such as acetylation and phosphorylation have not been investigated within the context of nutrigenomics in ruminants.

Among the various epigenetic mechanisms, noncoding RNA and specifically microRNAs have received a lot of notoriety in recent years [28], and, in contrast, long noncoding RNAs (LncRNAs) are only beginning to be evaluated in ruminants [29]. Examples of microRNAs with a potential application to improving milk and meat quality are the miR33b and miR192 (Table 1). The former has been previously associated with lipogenesis in the mammary gland of dairy cows as well as having the greatest upregulation from pregnancy into lactation [30]. In the case of miR192, it has been observed to influence muscle development through myogenesis in sheep. Since the interactions between microRNAs and coding mRNAs are one-to-many, meaning that a single microRNA can regulate the translation of several coding mRNAs, special caution should be applied when interpreting this type of data. The LncRNAs are relatively new in the context of ruminant nutrition and physiology and are commonly characterized by containing >200 nt that are not translated into proteins [31]. This work provided nuances on the role of LncRNAs in the mammary gland in terms of mastitis and milk quality and production.

The importance of understanding these multiple mediators of nutrient-gene interactions cannot be overstated. The authors envision that the continuous accumulation of this wealth of knowledge will lead to accurate and consistent manipulation of the ruminant genome to access or unlock the full genetic potential with the aim to produce ruminant products more efficiently, with a targeted effect on human health, and with a lesser cost for the environment.

Advertisement

2. Gene regulation in dairy cattle

Milk is one of the most nutritious foods known to man, and milk from dairy cattle has been part of the human diet from approximately 9000 years ago [32]. And, currently, the consumption of milk and milk-derived products around the world is expected to increase, primarily due to an increase in world population and increased consumption in countries where milk has not traditionally been considered popular [33]. Until now, the demand of milk worldwide has been supplied by a large increase in milk yield per cow, which has been a product primarily from the selection and enhancement of management practices, including improved nutrition. However, because of the ever-increasing demand for milk and milk-derived products as stated above, there is a need to continue increasing milk production efficiency.

Milk and milk products are an excellent source of macronutrients such as fat, protein, and carbohydrates, and contain a variety of bioactive molecules associated with health benefits, for instance, conjugated linolenic acid (CLA). The CLA has been associated with reductions in cancer development [25]. Because of its ability to contain bioactive molecules, milk has been considered a functional food. However, our ability to understand and yet manipulate the cow genome through nutrigenomic approaches to enrich specific bioactive molecules in milk is in its infancy. This calls for a continuous development of a wealth of knowledge around the various complex nutrient-gene interactions in dairy cows as well as development of nutritional models that can account for both traditional aspects of ruminant nutrition and more novel molecular regulation of nutrient metabolism.

2.1. Gene regulation and milk biosynthesis

During the lactation, the mammary gland is in charge of the final biosynthesis of milk using preformed elements from other organs (e.g., glucose synthesized in the liver) or compounds synthesized within the mammary epithelial cells (e.g., de novo fatty acids). The biosynthesis of milk in the mammary gland is highly regulated for several factors including nutrient supply (e.g., glucose, AA, and fatty acids) and hormones primarily related to hormonal changes during the onset and at the decline of the lactation.

2.1.1. Lactogenesis and the mammary gland transcriptome

Lactogenesis is the hallmark of mammalians, and as such, this biological process conveys a tremendous impact in gene regulation with the objective to induce the mammary gland to lactate and coincides with the formation of colostrum and occurs in coordination with parturition. The strong effects of lactogenesis on the mammary gland at the cellular level have been consistently recorded through transcriptomic analysis (i.e., microarray or RNA sequencing) across several mammals including mouse [34, 35, 36], rats [37, 38], bovine [39, 40, 41], sheep [42], goat [43], human [44, 45, 46], pig [47], kangaroo [48], and seal [49]. The extreme changes in the transcriptome from pregnancy to lactation underscore the importance of such change in the transcriptome in the mammary gland to initiate and maintain milk synthesis. In the case of bovine, a closer look at such transcriptional changes due to the onset and throughout the lactation was reported previously in terms of fat [50] and protein [51] synthesis. During the lactation, the milk fat biosynthesis in the bovine mammary gland had a marked upregulation in the expression of genes associated with FA uptake from blood, intracellular transport/channeling, and key transcription factors associated with lipogenesis (i.e., PPARG and SREBF2) [50]. Then, in terms of milk protein synthesis, it was observed that cell membrane transporters, especially for AA and glucose, played an essential role along with insulin signaling through mTOR for the regulation of protein synthesis in the bovine mammary gland [51]. At a greater scale, the impact of lactogenesis in the mammary gland transcriptome has been associated with epigenetic changes that result in alterations in the DNA structure and consequently the available genetic information for transcription [39]. Bionaz [39] observed chromatin changes (i.e., euchromatin or active transcribed chromatin and heterochromatin or tightly packed and transcriptionally unavailable chromatin) associated with lactogenesis, where a decrease of euchromatin status was observed as lactation begins and followed by an increase in euchromatin status as milk yield decreases during late lactation. The latter could be associated with natural response to lactation in the mammary gland to inhibit further epigenetic effects during the onset of lactation and to maintain a consistent transcriptome until milk synthesis declines in late lactation.

2.1.2. Milk biosynthesis in the dairy cattle from a nutrigenomic approach

Nutrigenomics is a coined term to refer to the interactions between nutrients and the genome. However, the term nutrigenomics does not refer to the effect of nutrients on the sequence of DNA, but rather the nutrient-gene interactions through the intermediate action of transcription factors (TFs) in the short to medium term and epigenetic factors in the medium to long term. Bioactive compounds with potential nutrigenomic effects can be found in regular diets of dairy cattle, and such compounds can directly or indirectly activate or repress the activity of TF.

Nutrigenomics is a product of the postgenome era and its impact on human, companion animals, and livestock species has gained more attention in recent years [52, 53, 54, 55]. In dairy cattle, and overall in ruminants, the field can be considered in its infancy but, as argued by Coffey [56], holds great potential to improve health and productivity.

2.1.3. Transcription factors with nutrigenomic potential in dairy cattle

The TFs are fundamental to the study of nutrigenomics; they can act as intermediaries between dietary nutrients and the final alteration in gene expression. TFs that respond to nutrient effects can be activated directly or indirectly by nutrients, and upon activation, they translocate from the cytoplasm to the nucleus where they alter the transcription of specific target genes. Transcription factors can bind specific short DNA sequences (i.e., 6–12 nucleotides) called response elements located in the enhancer regions upstream of the actual gene sequence [57]. The ability of TF to modulate gene expression upon activation by nutrients confers these proteins a central stage in the field of nutrigenomics. Therefore, accurate identification and characterization of TF responding to specific nutrients and to what extent these TF can be manipulated through dietary effects should be the focus of future research in nutrigenomics in dairy cattle.

Although between 2000 and 3000 TFs with sequence-specific DNA-binding domains in the human genome were estimated, only ~100 have been experimentally verified for their DNA-binding and regulatory functions [58]. The AnimalTFDB website had collected information for almost 1300 TF and almost 400 transcription cofactors for Bos taurus [59]. Rather than TF working independently, a regulatory network of TF has been suggested, which is essential to coordinate the response to external stimuli and translate this into changes in gene expression [60].

Among the known TF, the nuclear receptor superfamily of TF, with 48 members in the human genome, is the most important group of nutrient sensors [61]. From this superfamily, a short list of TFs has been identified as ligand-dependent nuclear receptors (LdNR), which can bind and be activated by macro- and micronutrients [55]. Recently, the main LdNR with a potential role in nutrigenomics with an emphasis in large [62] and small [24] ruminants has been reviewed. Among the LdNR associated with macronutrients such as fatty acids are the peroxisome proliferator-activated receptors (PPAR), liver X receptors (LXR), and hepatic nuclear factor 4 (HNF4) [63]. The LdNR associated with micronutrients are primarily vitamin-specific and include retinoid X receptors (RXR) and retinoic acid receptors (RAR) activated by retinoic acids or metabolites of vitamin A [64], vitamin D receptor (VDR), and pregnane X receptor activated by vitamin E [65].

The PPAR belongs to the LdNR group of TF and requires to form heterodimers with RXR in order to be functional. The main characteristic of PPAR is to have a prominent role in controlling expression of genes involved in lipid metabolism and inflammation. The potential nutrigenomic role of PPAR in ruminants has been reviewed at length previously [10]. This review discussed the role of PPARα in controlling lipid metabolism and inflammation in liver, the potential role of PPARβ/δ in controlling glucose uptake in mammary tissue, and the potential role of PPARγ in controlling milk fat synthesis and mastitis [10]. An interesting feature of PPAR is their capacity for binding and be activated by LCFA in both monogastrics and ruminants [10, 66]. However, in the case of ruminants, data indicate that activation of the PPAR isotypes PPARα and PPARγ is more consistent with saturated LCFA, primarily palmitate and stearate, than unsaturated LCFA [10].

2.1.4. Nutriepigenomics in dairy cattle

Environment factors such as diet and ambient conditions can not only affect the short- and medium-term gene expression, but there is also a medium- to long-term regulation of genes. The latter is primarily carried out through changes in the availability of gene sequences to be transcribed into mRNA. This concept is referred to epigenetics, where “epi” is a Greek-derived term meaning “over” then, epigenetics is commonly referred as “on-top-of genetics.” The implications of epigenetics indicate that there could be a set of inherited characteristics, phenotypes, and chemical entities that are superimposed on the DNA and do not follow basic Mendelian laws. Every individual will have a set of epigenetic marks throughout the genome, which is also known as the epigenome.

Epigenetic modifications are carried out through several biological processes including DNA methylation, histone modifications (e.g., methylation and acetylation), noncoding RNA (e.g., micro- and long-RNA). And, when these biological processes respond to nutrients and compounds in the diet, it is associated with nutriepigenomic effects. In ruminants, such effects could serve important physiological adaptations during the onset of lactation including increasing the availability of gene sequences through alterations in DNA methylation for the transcription of essential proteins such as caseins in the mammary gland of dairy cows [67]. This new spinoff of nutrigenomics (i.e., nutriepigenomics) will provide essential information to our understanding of how nutrients can affect the biology of ruminants at a molecular level. However, at the same time, nutriepigenomics will add another layer of complexity to our field, where such interactions have to be fully understood, and in time, manipulated through dietary interventions.

Methylation is a major route for epigenetic modifications, through DNA and histone methylation. Therefore, methyl donors (e.g., choline, methionine, folic acid, etc.) found in the diets of dairy cattle can have a nutriepigenomic effect. These dietary methyl donors will likely increase the synthesis of SAM, which is the major biological methyl donor in the body [68]. The essential role of SAM within the context of the transition cow relies on the multiple biological processes that require this methyl donor, including transsulfuration, polyamine biosynthesis, DNA methylation [69], and histone methylation [70]. Among these, the epigenetic modifications caused by DNA and histone methylation are particularly important in order to understand the potential nutriepigenomic alterations caused by dietary methyl donor (e.g., methionine) supplementation.

DNA methylation occurs through specialized enzymes called DNA methyltransferases, which utilized the methyl group provided by SAM to methylate cytosines within a Cyt-phosphate-Gua (CpG) region (“island”) in the DNA and eventually creating methylated CpG patterns in the mammalian genome [71]. In ruminants, supplementation of rumen-protected to dairy cows resulted in a prepartal upregulation of DNMT3A, a gene that encodes for a DNA methyltransferase in charge of the de novo methylation of the DNA [72]. And, more recently, the significance of these findings was confirmed by observing alterations due to methionine supplementation in the liver of transition dairy cows in terms of global DNA methylation and specific region methylation of an important TF, the peroxisome proliferator-activated receptor alpha PPARα [8]. The characteristics and uniqueness of this TF within the context of the transition dairy cow were initially presented by Drackley [73], and since then, this nuclear receptor has become an exciting area of research in dairy cattle nutrigenomics (i.e., nutrient-gene interaction) [10]. Therefore, the connection between Met and PPARα upregulation through DNA methylation during the transition period is another suitable mechanism to explain the consistent improvements in performance (e.g., milk yield and DMI) observed in transition dairy cows supplemented with Met [74].

In the cellular nuclei, the DNA is normally packed in condensed structures called chromatins, consisting primarily of histone proteins, which serve as spools where the DNA winds around. Then, the genetic information contained in the DNA exists in two states: unavailable or wind around histone proteins, and available or unwound. Chromatin remodeling is the main mechanism by which DNA is wind or unwound from histones and these dynamic modifications occur by enzymatic modifications including acetylation, phosphorylation, ubiquitination, and methylation [75]. The latter being a potential mechanism through which Met can alter gene expression in dairy cows. Currently, the limited amount of data on histone methylation in dairy cows has been conducted in immune cells [76] primarily related to subclinical mastitis [77]. This work has provided nuances on the interactions between mastitis-related pathogens and histone methylation; however, dietary effects on histone methylation have not been investigated.

Advertisement

3. Gene regulation in beef cattle

3.1. Mutations

In order to understand how changes in a single gene could significantly alter the body structure and physiology of beef cattle, we need to focus in mutations that occur in specific areas in the bovine genome. For example, a 11-nucleotide deletion in the Myostatin gene (MSTN) determines double muscling in Belgian Blue cattle, and a single nucleotide change produces the same effect on Piedmontese cattle [78]. Furthermore, the leptin gene (LEP) presents several single nucleotide polymorphisms (SNP), like AJ236854:c.73 T > C, which induces a cysteine-to-arginine amino acid substitution that could affect protein functionality [79]. The c.73C allele of LEP is associated with higher average daily gain (ADG), lower dressing percentage, and higher marbling scores [79], which are desirable characteristics in a beef carcass. Finally, intramuscular fat deposition could also be affected by a mutation in the fatty acid synthase gene (FASN), contributing to the characteristic fatty acid composition of Japanese Black beef [80]. However, not all mutations have beneficial effects on the productivity of meat-producing animals. There are also mutations that are considered lethal, affecting, for example, the reproductive performance of females through early embryonic loss [81], or mutations that produce genetic disorders in beef cattle [82]. The Arachnomelia syndrome in Simmental cattle [83] that produce malformations of the skeleton mainly affecting the legs, the spinal column, and the skull is an example of these genetic disorders. These types of inborn errors can be prevented nowadays with techniques like genome editing [83]. Although the implementation of this technique in the animal production industry might generate controversy, it will offer tremendous potential for breeding animals with desirable traits.

Undesired mutations present in the bovine genome are difficult to avoid when they occur. Through the implementation of selection plans and genotyping the herds in order to avoid the reproduction of carrying individuals is the most commonly utilized strategy. Similarly, selection of animals that carry biomarkers in their genomes that will make them improve their meat production and marbling efficiency is the general goal of researchers passionate about beef production. Currently, identification of relevant genetic and genomic markers is ongoing, especially for tenderness—a top priority quality attribute [84].

3.2. Nutrigenomics in beef cattle

In spite of the emergence of new alternatives to beef production (e.g., cultivated meat) in order to meet the growing world population’s food demand, researchers in beef production are also focusing on techniques to regulate the bovine genome with a more natural approach (i.e., nutrigenomics). Nutrigenomics study the interactions between nutrients and genes [85], that is how the nutrients present in the diet can affect gene expression. In beef animals, nutrigenomics was widely studied [26]; following, a more detailed description of how specific nutrients regulate the expression of genes related to muscle growth and intramuscular fat deposition will be addressed.

The composition of adipose tissue produced in a meat-producing animal can be “manipulated” by diet, with some variability between breeds. For example, high silage-based diets produce less proportion of the fatty acid 18:2 n-6, with the consequent decrease in the amount of total polyunsaturated fatty acids (PUFA), as compared to low silage-based diets in subcutaneous adipose tissue [86]. This variability in fatty acids composition was attributed to greater activity of fatty acid binding protein 4 (FABP4), lipoprotein lipase (LPL), and stearoyl CoA desaturase (SCD) genes in subcutaneous adipose tissue of animals fed a low silage-based diet [86].

In the same way, we are able to regulate the fatty acid profile composition of a specific fat depot by dietary changes, researchers are trying to prioritize the growth and development of the intramuscular fat that will lead to greater marbling. Intramuscular fat starts to accumulate in the late stages of growth, as compared to other adipose tissue depots that normally develop first (i.e., visceral, intermuscular, and subcutaneous fat). The ultimate goal is to improve marbling scores that will lead to premium prices for the carcasses that classify as prime or choice according to the USDA carcass grading scale [87].

When comparing dairy and beef cattle breeds, subcutaneous fatty acid profile presents several differences in terms of fatty acids profile, probably due to the difference in the degree of fatness, which is always lower for dairy cattle [88]. The important variability in fat composition between breeds could be explained by the variability in relative SCD1 expression in subcutaneous fat [88]. SCD1 seems to have a role in a depot-specific fashion.

3.3. How management decisions can affect gene expression

The combination of early weaning and high dietary starch leads to a strong programming effect in skeletal muscle tissue, with PPARγ and CCAAT enhancer-binding protein alpha (CEBPA) as the central coordinators of this response. The implementation of early weaning (e.g., 2 months of age) in beef calves provides a different type of diet as compared to a calf weaned normally at 6–7 months of age. Therefore, the early administration of starch in a beef producing animal produces a precocious and sustained activation of the PPARG and its target genes, leading to greater intramuscular fat deposition and consequently more carcasses grading as “Choice” [89].

Castration increases intramuscular fat (IMF) deposition, improving beef quality in cattle. In a transcriptome analysis performed in longissimus muscle (LM) samples, when comparing bulls and steers, castration showed to upregulate transcription of genes for lipogenesis, fatty acid oxidation, and also genes coding for enzymes associated with the tricarboxylic acid cycle and oxidative phosphorylation [90]. Therefore, castration shifts the transcriptome of lipid/energy metabolism to favor intramuscular fat deposition in the LM following castration.

Another beef producer decision that can affect the expression of genes is the selected calving season (i.e., different temperature/humidity index = THI). For example, two groups of pregnant cows that calved during thermoneutral temperature conditions (THI = 67.3) and cows that calved in summer season (THI = 79.9) were bleed during their transition period (i.e., cows between 3 weeks before and 3 weeks after calving) for RNA extraction of white blood cells [91]. Results showed that expression of CASP-3, BCL-2, BAK, P53, and ratio of BAX/BCL-2 in white blood cells increased during summer as compared to thermoneutral conditions, suggesting the susceptibility of these cells to apoptosis or cell death [91]. Consequently, cows calving in two different calving seasons will present differences in their inflammatory response, affecting the maternal recognition of the fetus during early pregnancy [92] or also will have a negative impact in the cow’s milking ability postcalving [93].

3.4. Feedlot versus pasture

The beef industry has two main ways to produce beef: pasture-base, and grain-based or feedlot. Consuming energy above requirements helps to increase the intramuscular fat deposition in beef cattle. Feeding high concentration of cereal grains is the way to reach surplus energy that can be utilized in the rumen and the small intestine to produce volatile fatty acids for glucose and energy production. The starch present in the grain is fermented by microbes in the rumen, producing propionate (a glucose precursor), which will be the signal received by the membrane receptors present in the cell activating a cascade of events that will end up with the activation of genes related to the process of adipogenesis. This type of diet is more commonly administered during the so-called finishing or fattening phase.

When the diet is mainly based on the forage available, the rumen population consists of microbes that produce a greater proportion of acetic acid which increases the activation of 5′-prime-AMP-activated protein kinase alpha (AMPK) phosphorylation, reducing the transcriptional activity of the sterol regulatory element-binding protein 1c (SREBP1C) and the carbohydrate responsive element-binding protein (MLXIPL), which decreased the expression of lipogenic genes [94]. In beef cattle finished under a forage-based diet, the fatty acid profile varies considerably as compared to animals finished under feedlot diets. Beef finished under forage-based diets presents greater concentration of polyunsaturated fatty acids (PUFAs), especially the fatty acids with nutraceutical value (20:5 or EPA and 22:6 or DHA). These types of beef products are in the eye of consumers who care about eating healthy foods. These PUFAs mentioned above are upstream regulators of genes related to fatty acid synthesis and transport. FABP4, FASN, and PPARG are particularly activated by these additional PUFAs generated due to the administration of forage-based diets [95]. Even though we could expect a greater proportion of IMF due to the mentioned additional PUFAs, there is an overall lower fat content in a grass-fed beef product. Furthermore, grass-fed beef is known to have a different flavor and aroma as compared to grain-fed beef when cook on the grill [96].

3.5. Fetal programming in beef cattle

The fetal programming concept is related to the important physiological changes that can occur due to environmental/nutritional events during prenatal development. For example, nutrient restriction of 85% as compared to 140% of total metabolizable energy requirements during the second half of gestation can alter in fetal muscle the expression of both, myogenic and adipogenic genes, without apparent differences in fetal phenotype [97].

The canonical Wnt pathway, a β-catenin-dependent signaling pathway called the Wnt/β-catenin signaling pathway is key in establishing the fate of the undifferentiated stem cells; hence, β-catenin plays an essential role in the regulation of embryonic, postnatal, and oncogenic growth of many tissues. If the β-catenin pathway is blocked, the total number of myocytes will be reduced, and the differentiation of mesenchymal stem cells into mature adipocytes will be potentiated [98]. In the same way, adipogenesis is initiated around mid-gestation in ruminant animals; therefore, a strategic maternal nutritional plan in order to enhance the number of mesenchymal cells committed to adipogenesis will increase the number of intramuscular adipocytes in a process known as hyperplasia; therefore, this outcome will be translated as more marbling in the offspring postnatally. PPARG alone can stimulate adipocyte differentiation [99], although the continuation of this process is regulated by many PPARG target genes [100].

Bioinformatics analysis revealed a pseudoinflammatory process in early-weaned beef calves during their growing phase [101], which it is associated with the activation of the innate immune system presumably due to macrophage infiltration of intramuscular fat [101], which is a typical obesity symptom. These results could be considered as a biological sign of a precocious beginning of the adipogenic metabolic machinery in young beef calves.

3.6. Epigenetics regulations in beef cattle

Changes caused by chromatin (the complex of DNA and histone proteins that forms chromosomes within the nucleus) modification due to environmental factors is called epigenetics [102]. Another epigenetics regulation approach is through microRNAs, which are endogenous noncoding small RNA molecules (20–24 base pairs) that prevent the production of a particular protein by binding to and destroying the messenger RNA that would have produced the protein [103].

Epigenetics regulation is based on chromatin remodeling rather than alteration of the DNA code. With the aim to identify methylated genes affecting bovine growth, an elegant study provides a genome-wide landscape of DNA methylomes and their relationship with mRNA and miRNA for fetal and adult muscle of Chinese Qinchuan beef cattle [104]. Presence of SNPs in epigenetic-related genes was analyzed in different beef breeds. Interestingly, three DNA (cytosine-5)-methyltransferases (DNMTs), DNMT1, DNMT3a, and DNMT3b were found significantly associated with beef quality parameters on the carcass. In particular, DNMT3b presented five SNPs related to carcass traits, becoming a potential candidate gene for beef quality improvement [105].

In the bovine genome, no microRNAs were identified on chromosome Y, while microRNA related to adipose tissue are expressed in chromosome X [106]. This could be a reason that explains the sexual differences in fat metabolism in mammals. Furthermore, there are highly expressed microRNAs for beef adipose tissue, miR-378 which was found upregulated in steers with high levels of subcutaneous fat [107] and miR-2478 [108] which potentially targets ELOVL fatty acid elongase 6 (ELOVL6) and stearoyl CoA desaturase (SCD), is bovine specific and had higher expression in grass-fed as compared to grain-fed cattle.

A study in bovine skeletal muscle development used next-generation small RNA sequencing, a total of 512 miRNAs were identified [109]. Thirty-six miRNAs were differentially expressed between prenatal and postnatal stages of muscle development including several myomiRs (miR-1, miR-206, and let-7 families). Compared to miRNA expression between different muscle tissues, 14 miRNAs were upregulated and 22 miRNAs were downregulated in the muscle of postnatal stage [109]. Furthermore, a genome-wide landscape of DNA methylomes and their relationship with mRNA and miRNA for bovine fetal and adult muscle recently discovered will provide a solid basis for exploring the epigenetic mechanisms of muscle growth and development [104].

Muscling in cattle influenced by genetic background, ultimately affecting beef yield is of major interest to the beef industry. The best alternative to promote muscle development is through satellite cell proliferation [110]. In fact, myoblast or satellite cells are utilized for the proliferation and differentiation of cultured meat [111]. The transcription factor Sp1, an activator of myosin D (MyoD) and a suppressor of cyclin-dependent kinase inhibitor 1A (CDKN1A), plays an important role in bovine muscle cell proliferation and differentiation. This transcription factor is a target of miR-128 and, if this microRNA is overexpressed, it inhibits muscle satellite cell proliferation and differentiation [112]. Furthermore, miR-1 and miR-206 facilitate bovine skeletal muscle satellite cell myogenic differentiation by restricting the expression of their target genes, and that inhibition of miR-1 and miR-206 increased the paired box 7 (Pax7) and histone deacetylase 4 (HDAC4) protein levels enhancing satellite cell proliferation [113].

In Biceps femoris muscle of Japanese Shorthorn cattle, a grazing period up to 4 months increased the expression of miR-208b, which has a muscle fiber type-associated role. Furthermore, a target for miR-208b, MyoD, a myogenic regulatory factor associated with the shifting of muscle property to the fast type, had lower expression in the grazed cattle after 4 months of grazing, as compared to feedlot cattle. During skeletal muscle adaptation to grazing, miR-206 expression remained higher as compared to housed animals in which it decreases [114]. MiR-206 is known as the skeletal muscle-specific myomiRNA [115].

Offspring’s health depends on the maternal body condition at mid-gestation, which will make them be more predisposed to develop obesity at an early age, which in beef production is desirable. Fetal intramuscular adipogenesis was enhanced at mid-gestation due to alteration of microRNA expression; downregulation of let-7g was the main cause for this outcome [116]. This microRNA inhibits the mRNA expression of PPARG and CEBPA, both important regulators of adipogenesis [117]. Furthermore, cow plane of nutrition during the last third of gestation showed epigenetic effects on the offspring’s skeletal muscle through the downregulation of miR-34a that has a role on the activation of cell cycle arrest by suppressing SIRT1 expression, which promotes adipocyte differentiation [118].

Advertisement

4. Molecular nutrition in ruminants

The importance of ruminants to the world food security is reflected in their contribution to the demand for animal protein around the world and particularly in developing countries, and such demand is expected to increase in the future [119]. To face this demand, advancements in ruminant nutrition and physiology will require improvements on feed efficiency and development of novel functional foods from ruminants by enriching specific compounds associated with health benefits in humans. The latter will need a deep understanding and wealth of knowledge of molecular regulatory mechanisms in response to physiological conditions and nutrition. In this context, this vast amount of multilayered data in terms of mRNA, proteins, metabolites, and phenotypes can only be undertaken with powerful tools such as omics technologies and bioinformatics. In fact, these are the foundations of modern system biology, a field of study with the aim to enhance the understanding of complex biological models and interactions occurring within cells and tissues. Understanding this complexity and the outcomes of nutritional interventions and physiological conditions will allow the formulation of novel theories and ideas to enhance feed efficiency, development of new functional foods derived from ruminant products, and reduce carbon footprint.

Even though the outcome is different, there are similarities in dairy and beef cattle from a nutrigenomic perspective. For instance, both the synthesis of milk fat in dairy cows and the synthesis of intramuscular fat in beef steers are regulated by a similar network of TF. Nutrients or stimulus received with the diet (PUFAs, insulin, etc.), activates PPARα in the liver of the dairy cow and PPARγ in the intramuscular preadipocyte of a beef steer. The activated PPARs form a heterodimer with retinoic X receptor alpha (RXRA), leading to the upregulation of their lipogenesis-related target genes (Figure 2). Furthermore, in the same way, the activation of the PI3K/Akt/mTOR signaling pathway will lead to the synthesis of milk protein in dairy cows [120], the activation of the same metabolic pathway might lead to muscle hypertrophy in beef cattle, but this is a concept that has not been completely elucidated [121]. It is also worth to mention the importance of fatty acid binding proteins (FABPs) in ruminants, which bind and transport LCFA. FABP4 affects milk yield and milk protein content, both economically important traits in the dairy industry [122], and FABP4 also presents gene polymorphisms that have been associated with meat quality traits in beef cattle [123].

Figure 2.

Example of nutrigenomics linkage between beef and dairy cattle.

In a study aiming to use the fibroblast model to explore differences between a dairy breed (Holstein) and a beef breed (Angus) in their innate responses to LPS exposure, several immune-associated differentially expressed genes between breeds were found [124]. Within them, TLR4, which is the extracellular receptor responsible for recognition of LPS presented higher level of expression in Holstein cows as compared to Angus, suggests the Holstein animals will detect and respond to Gram-negative bacteria more vigorously than Angus animals.

Finally, epigenetic differences between beef and dairy cattle could also be observed mainly because of the different environments the offspring are exposed after birth. While a beef calf usually stays with the dam approximately until 6–7 months old, a dairy calf is separated from its mother as soon as it finishes consuming colostrum or earlier. Although there are studies that started to analyze the epigenetic differences between breeds [124], this is a promising area that needs to be studied in deep.

Advertisement

5. Conclusions

The general nutrigenomic model for ruminants needs to be updated based on emerging nuance information with the ever-growing pace in ruminant nutrition research with “omics” technologies. The dissection of what intermediate components or processes such as intermediate metabolites, signal transduction, TF, etc., are utilized by specific nutrients will allow for accurate predictions of the nutrigenomic outcomes of such nutrients in a practical setting. However, the multilayered and multifactorial nature of the nutrigenomic model will require the implementation of additional tools such as system biology and network theory in order to have a more holistic approach to understand how nutrients regulate milk synthesis or skeletal muscle gain and marbling.

One of the greatest challenges in ruminant nutrigenomics is to account for the final products from rumen fermentation, where several factors such as rate of passage, intake, particle size of the diet can affect rumen fermentation and kinetics. The latter can be avoided by feeding nutrients encapsulated or protected from ruminal degradation; however, this does not eliminate the need to account for the substantial impact the rumen fermentation and its products may have on the overall nutrigenomic effect from a particular diet. Because of this reason, the resurgence of the field of the microbiome in ruminant nutrition research promise to add valuable information on rumen microbes response to nutrients in the diet and correlate this with final nutrigenomic responses at the whole animal level.

Our understanding of the impact of nutrition on regulatory mechanisms at the cellular level in the ruminant animal has grown an accelerated pace over the last decades. As pointed out by Drackley 12 year ago [125], the marriage between “omics” technologies with measurements of tissue metabolism and the final performance (e.g., milk yield and skeletal muscle gain) has been enlightening and essential to identify key responses to nutritional changes and physiology. However, there is still too much to learn in the complex nutrient-gene interactions in the context of the ruminant animal. The future of nutrigenomics in ruminants is to develop technologies and algorithms to predict the final molecular outcomes of nutrients and diets fed to ruminants in a practical setting. This monumental task can only be accomplished by generating a wealth of knowledge in several orders of magnitude of what we currently have on nutrigenomics in ruminants.

References

  1. 1. Nobel S, Abrahmsen L, Oppermann U. Metabolic conversion as a pre-receptor control mechanism for lipophilic hormones. European Journal of Biochemistry. 2001;268(15):4113-4125. DOI: 10.1046/j.1432-1327.2001.02359.x
  2. 2. Clarke SD. Nutrient regulation of gene and protein expression. Current Opinion in Clinical Nutrition and Metabolic Care. 1999;2(4):287-289
  3. 3. Eastwood MA. A molecular biological basis for the nutritional and pharmacological benefits of dietary plants. Q JM. 2001;94(1):45-48
  4. 4. Dauncey MJ, White P, Burton KA, Katsumata M. Nutrition-hormone receptor-gene interactions: Implications for development and disease. The The Proceedings of the Nutrition Society 2001;60(1):63-72
  5. 5. Jacobs MN, Lewis DF. Steroid hormone receptors and dietary ligands: A selected review. The Proceedings of the Nutrition Society. 2002;61(1):105-122
  6. 6. Rakhshandehroo M, Knoch B, Muller M, Kersten S. Peroxisome proliferator-activated receptor alpha target genes. PPAR Research. 2010;2010. DOI: 10.1155/2010/612089
  7. 7. Osorio JS, Lohakare J, Bionaz M. Biosynthesis of milk fat, protein, and lactose: Roles of transcriptional and posttranscriptional regulation. Physiological Genomics. 2016;48(4):231-256. DOI: 10.1152/physiolgenomics.00016.2015
  8. 8. Osorio JS, Jacometo CB, Zhou Z, Luchini D, Cardoso FC, Loor JJ. Hepatic global DNA and peroxisome proliferator-activated receptor alpha promoter methylation are altered in peripartal dairy cows fed rumen-protected methionine. Journal of Dairy Science. 2016;99(1):234-244. DOI: 10.3168/jds.2015-10157
  9. 9. Li R, Beaudoin F, Ammah AA, Bissonnette N, Benchaar C, Zhao X, et al. Deep sequencing shows microRNA involvement in bovine mammary gland adaptation to diets supplemented with linseed oil or safflower oil. BMC Genomics. 2015;16:884. DOI: 10.1186/s12864-015-1965-7
  10. 10. Bionaz M, Chen S, Khan MJ, Loor JJ. Functional role of PPARs in ruminants: Potential targets for fine-tuning metabolism during growth and lactation. PPAR Research. 2013;2013:684159. DOI: 10.1155/2013/684159
  11. 11. Burris TP, Solt LA, Wang Y, Crumbley C, Banerjee S, Griffett K, et al. Nuclear receptors and their selective pharmacologic modulators. Pharmacological Reviews. 2013;65(2):710-778. DOI: 10.1124/pr.112.006833
  12. 12. Escher P, Wahli W. Peroxisome proliferator-activated receptors: Insight into multiple cellular functions. Mutation Research. 2000;448(2):121-138
  13. 13. Olefsky JM, Saltiel AR. PPAR gamma and the treatment of insulin resistance. Trends in Endocrinology and Metabolism. 2000;11(9):362-368
  14. 14. Desvergne B, Wahli W. Peroxisome proliferator-activated receptors: Nuclear control of metabolism. Endocrine Reviews. 1999;20(5):649-688. DOI: 10.1210/edrv.20.5.0380
  15. 15. Desvergne B, Michalik L, Wahli W. Transcriptional regulation of metabolism. Physiological Reviews. 2006;86(2):465-514. DOI: 10.1152/physrev.00025.2005
  16. 16. Sevane N, Armstrong E, Cortes O, Wiener P, Wong RP, Dunner S, et al. Association of bovine meat quality traits with genes included in the PPARG and PPARGC1A networks. Meat Science. 2013;94(3):328-335. DOI: 10.1016/j.meatsci.2013.02.014
  17. 17. Moisa SJ, Shike DW, Meteer WT, Keisler D, Faulkner DB, Loor JJ. Yin yang 1 and adipogenic gene network expression in longissimus muscle of beef cattle in response to nutritional management. Gene Regulation and Systems Biology. 2013;7:71-83. DOI: 10.4137/GRSB.S11783
  18. 18. Faulconnier Y, Bernard L, Boby C, Domagalski J, Chilliard Y, Leroux C. Extruded linseed alone or in combination with fish oil modifies mammary gene expression profiles in lactating goats. Animal: An International Journal of Animal Bioscience. 2018;12(8):1564-1575. DOI: 10.1017/S1751731117002816
  19. 19. Agazzi A, Invernizzi G, Campagnoli A, Ferroni M, Fanelli A, Cattaneo D, et al. Effect of different dietary fats on hepatic gene expression in transition dairy goats. Small Ruminant Research. 2010;93(1):31-40. DOI: 10.1016/j.smallrumres.2010.04.027
  20. 20. McFadden JW, Corl BA. Activation of liver X receptor (LXR) enhances de novo fatty acid synthesis in bovine mammary epithelial cells. Journal of Dairy Science. 2010;93(10):4651-4658. DOI: 10.3168/jds.2010-3202
  21. 21. Harvatine KJ, Bauman DE. SREBP1 and thyroid hormone responsive spot 14 (S14) are involved in the regulation of bovine mammary lipid synthesis during diet-induced milk fat depression and treatment with CLA. The Journal of Nutrition. 2006;136(10):2468-2474
  22. 22. Wang W, Luo J, Zhong Y, Lin XZ, Shi HB, Zhu JJ, et al. Goat liver X receptor alpha, molecular cloning, functional characterization and regulating fatty acid synthesis in epithelial cells of goat mammary glands. Gene. 2012;505(1):114-120. DOI: 10.1016/j.gene.2012.05.028
  23. 23. Yang YZ, Subauste JS, Koenig RJ. Retinoid X receptor alpha binds with the highest affinity to an imperfect direct repeat response element. Endocrinology. 1995;136(7):2896-2903. DOI: 10.1210/endo.136.7.7789315
  24. 24. Osorio JS, Vailati-Riboni M, Palladino A, Luo J, Loor JJ. Application of nutrigenomics in small ruminants: Lactation, growth, and beyond. Small Ruminant Research. 2017;154:29-44. DOI: 10.1016/j.smallrumres.2017.06.021
  25. 25. Bauman DE, Harvatine KJ, Lock AL. Nutrigenomics, rumen-derived bioactive fatty acids, and the regulation of milk fat synthesis. Annual Review of Nutrition. 2011;31:299-319. DOI: 10.1146/annurev.nutr.012809.104648
  26. 26. Ladeira MM, Schoonmaker JP, Gionbelli MP, Dias JC, Gionbelli TR, Carvalho JR, et al. Nutrigenomics and beef quality: A review about lipogenesis. International Journal of Molecular Sciences. 2016;17(6). DOI: 10.3390/ijms17060918
  27. 27. Xu W, Wang F, Yu Z, Xin F. Epigenetics and cellular metabolism. Genetics & Epigenetics. 2016;8:43-51. DOI: 10.4137/GEG.S32160
  28. 28. Wang X, Gu Z, Jiang H. MicroRNAs in farm animals. Animal: An International Journal of Animal Bioscience. 2013;7(10):1567-1575. DOI: 10.1017/S1751731113001183
  29. 29. Koufariotis LT, Chen YP, Chamberlain A, Vander Jagt C, Hayes BJ. A catalogue of novel bovine long noncoding RNA across 18 tissues. PLoS One. 2015;10(10):e0141225. DOI: 10.1371/journal.pone.0141225
  30. 30. Wang M, Moisa S, Khan MJ, Wang J, Bu D, Loor JJ. MicroRNA expression patterns in the bovine mammary gland are affected by stage of lactation. Journal of Dairy Science. 2012;95(11):6529-6535. DOI: 10.3168/jds.2012-5748
  31. 31. Tong C, Chen Q , Zhao L, Ma J, Ibeagha-Awemu EM, Zhao X. Identification and characterization of long intergenic noncoding RNAs in bovine mammary glands. BMC Genomics. 2017;18(1):468. DOI: 10.1186/s12864-017-3858-4
  32. 32. Evershed RP, Payne S, Sherratt AG, Copley MS, Coolidge J, Urem-Kotsu D, et al. Earliest date for milk use in the near east and southeastern Europe linked to cattle herding. Nature. 2008;455(7212):528-531. DOI: 10.1038/nature07180
  33. 33. Wiley AS. The globalization of Cow’s milk production and consumption: Biocultural perspectives. Ecology of Food and Nutrition. 2007;46(3-4):281-312. DOI: 10.1080/03670240701407657
  34. 34. Anderson SM, Rudolph MC, McManaman JL, Neville MC. Key stages in mammary gland development. Secretory activation in the mammary gland: It’s not just about milk protein synthesis! Breast Cancer Research. 2007;9(1):204. DOI: 10.1186/bcr1653
  35. 35. Lemay DG, Neville MC, Rudolph MC, Pollard KS, German JB. Gene regulatory networks in lactation: Identification of global principles using bioinformatics. BMC Systems Biology. 2007;1:56. DOI: 10.1186/1752-0509-1-56
  36. 36. Zhou Y, Gong W, Xiao J, Wu J, Pan L, Li X, et al. Transcriptomic analysis reveals key regulators of mammogenesis and the pregnancy-lactation cycle. Science China. Life Sciences. 2014;57(3):340-355. DOI: 10.1007/s11427-013-4579-9
  37. 37. Anantamongkol U, Charoenphandhu N, Wongdee K, Teerapornpuntakit J, Suthiphongchai T, Prapong S, et al. Transcriptome analysis of mammary tissues reveals complex patterns of transporter gene expression during pregnancy and lactation. Cell Biology International. 2010;34(1):67-74. DOI: 10.1042/CBI20090023
  38. 38. Rodriguez-Cruz M, Coral-Vazquez RM, Hernandez-Stengele G, Sanchez R, Salazar E, Sanchez-Munoz F, et al. Identification of putative ortholog gene blocks involved in gestant and lactating mammary gland development: A rodent cross-species microarray transcriptomics approach. International Journal of Genomics. 2013;2013:624681. DOI: 10.1155/2013/624681
  39. 39. Bionaz M, Periasamy K, Rodriguez-Zas SL, Everts RE, Lewin HA, Hurley WL, et al. Old and new stories: Revelations from functional analysis of the bovine mammary transcriptome during the lactation cycle. PLoS One. 2012;7(3):e33268. DOI: 10.1371/journal.pone.0033268
  40. 40. Finucane KA, McFadden TB, Bond JP, Kennelly JJ, Zhao FQ. Onset of lactation in the bovine mammary gland: Gene expression profiling indicates a strong inhibition of gene expression in cell proliferation. Functional & Integrative Genomics. 2008;8(3):251-264. DOI: 10.1007/s10142-008-0074-y
  41. 41. Wickramasinghe S, Rincon G, Islas-Trejo A, Medrano JF. Transcriptional profiling of bovine milk using RNA sequencing. BMC Genomics. 2012;13:45. DOI: 10.1186/1471-2164-13-45
  42. 42. Singh M, Thomson PC, Sheehy PA, Raadsma HW. Comparative transcriptome analyses reveal conserved and distinct mechanisms in ovine and bovine lactation. Functional & Integrative Genomics. 2013;13(1):115-131. DOI: 10.1007/s10142-012-0307-y
  43. 43. Faucon F, Rebours E, Bevilacqua C, Helbling JC, Aubert J, Makhzami S, et al. Terminal differentiation of goat mammary tissue during pregnancy requires the expression of genes involved in immune functions. Physiological Genomics. 2009;40(1):61-82. DOI: 10.1152/physiolgenomics.00032.2009
  44. 44. Lemay DG, Ballard OA, Hughes MA, Morrow AL, Horseman ND, Nommsen-Rivers LA. RNA sequencing of the human milk fat layer transcriptome reveals distinct gene expression profiles at three stages of lactation. PLoS One. 2013;8(7):e67531. DOI: 10.1371/journal.pone.0067531
  45. 45. Maningat PD, Sen P, Rijnkels M, Sunehag AL, Hadsell DL, Bray M, et al. Gene expression in the human mammary epithelium during lactation: The milk fat globule transcriptome. Physiological Genomics. 2009;37(1):12-22. DOI: 10.1152/physiolgenomics.90341.2008
  46. 46. Mohammad MA, Haymond MW. Regulation of lipid synthesis genes and milk fat production in human mammary epithelial cells during secretory activation. American Journal of Physiology. Endocrinology and Metabolism. 2013;305(6):E700-E716. DOI: 10.1152/ajpendo.00052.2013
  47. 47. Shu DP, Chen BL, Hong J, Liu PP, Hou DX, Huang X, et al. Global transcriptional profiling in porcine mammary glands from late pregnancy to peak lactation. OMICS. 2012;16(3):123-137. DOI: 10.1089/omi.2011.0116
  48. 48. Lefevre CM, Digby MR, Whitley JC, Strahm Y, Nicholas KR. Lactation transcriptomics in the Australian marsupial, Macropus eugenii: Transcript sequencing and quantification. BMC Genomics. 2007;8:417. DOI: 10.1186/1471-2164-8-417
  49. 49. Menzies KK, Lefevre C, Sharp JA, Macmillan KL, Sheehy PA, Nicholas KR. A novel approach identified the FOLR1 gene, a putative regulator of milk protein synthesis. Mammalian Genome. 2009;20(8):498-503. DOI: 10.1007/s00335-009-9207-4
  50. 50. Bionaz M, Loor JJ. Gene networks driving bovine milk fat synthesis during the lactation cycle. BMC Genomics. 2008;9:366. DOI: 10.1186/1471-2164-9-366
  51. 51. Bionaz M, Loor JJ. Gene networks driving bovine mammary protein synthesis during the lactation cycle. Bioinformatics and Biology Insights. 2011;5:83-98. DOI: 10.4137/bbi.s7003
  52. 52. Dawson KA. Nutrigenomics: Feeding the genes for improved fertility. Animal Reproduction Science. 2006;96(3-4):312-322. DOI: 10.1016/j.anireprosci.2006.08.009
  53. 53. Fekete SG, Brown DL. Veterinary aspects and perspectives of nutrigenomics: A critical review. Acta Veterinaria Hungarica. 2007;55(2):229-239. DOI: 10.1556/AVet.55.2007.2.9
  54. 54. de Godoy MR, Swanson KS. Companion animals symposium: Nutrigenomics: Using gene expression and molecular biology data to understand pet obesity. Journal of Animal Science. 2013;91(6):2949-2964. DOI: 10.2527/jas.2012-5860
  55. 55. Muller M, Kersten S. Nutrigenomics: Goals and strategies. Nature Reviews. Genetics. 2003;4(4):315-322. DOI: 10.1038/nrg1047
  56. 56. Coffey SG. Prospects for improving the nutritional quality of dairy and meat products. Forum of Nutrition. 2007;60:183-195. DOI: 10.1159/0000107195
  57. 57. Shlyueva D, Stampfel G, Stark A. Transcriptional enhancers: From properties to genome-wide predictions. Nature Reviews. Genetics. 2014;15(4):272-286. DOI: 10.1038/nrg3682
  58. 58. Vaquerizas JM, Kummerfeld SK, Teichmann SA, Luscombe NM. A census of human transcription factors: Function, expression and evolution. Nature Reviews. Genetics. 2009;10(4):252-263. DOI: 10.1038/nrg2538
  59. 59. Zhang HM, Liu T, Liu CJ, Song S, Zhang X, Liu W, et al. AnimalTFDB 2.0: A resource for expression, prediction and functional study of animal transcription factors. Nucleic Acids Research. 2015;43(Database issue):D76-D81. DOI: 10.1093/nar/gku887
  60. 60. Cheatle Jarvela AM, Hinman VF. Evolution of transcription factor function as a mechanism for changing metazoan developmental gene regulatory networks. EvoDevo. 2015;6(1):3. DOI: 10.1186/2041-9139-6-3
  61. 61. Chawla A, Repa JJ, Evans RM, Mangelsdorf DJ. Nuclear receptors and lipid physiology: Opening the X-files. Science. 2001;294(5548):1866-1870. DOI: 10.1126/science.294.5548.1866
  62. 62. Bionaz M, Osorio JS, Loor JJ. TRIENNIAL LACTATION SYMPOSIUM: Nutrigenomics in dairy cows: Nutrients, transcription factors, and techniques. Journal of Animal Science. 2015;93(12):5531-5553. DOI: 10.2527/jas.2015-9192
  63. 63. Khan SA, Vanden Heuvel JP. Role of nuclear receptors in the regulation of gene expression by dietary fatty acids (review). The Journal of Nutritional Biochemistry. 2003;14(10):554-567
  64. 64. Minucci S, Leid M, Toyama R, Saint-Jeannet JP, Peterson VJ, Horn V, et al. Retinoid X receptor (RXR) within the RXR-retinoic acid receptor heterodimer binds its ligand and enhances retinoid-dependent gene expression. Molecular and Cellular Biology. 1997;17(2):644-655
  65. 65. Landes N, Pfluger P, Kluth D, Birringer M, Ruhl R, Bol GF, et al. Vitamin E activates gene expression via the pregnane X receptor. Biochemical Pharmacology. 2003;65(2):269-273
  66. 66. Nakamura MT, Yudell BE, Loor JJ. Regulation of energy metabolism by long-chain fatty acids. Progress in Lipid Research. 2014;53:124-144. DOI: 10.1016/j.plipres.2013.12.001
  67. 67. Vanselow J, Yang W, Herrmann J, Zerbe H, Schuberth HJ, Petzl W, et al. DNA-remethylation around a STAT5-binding enhancer in the alphaS1-casein promoter is associated with abrupt shutdown of alphaS1-casein synthesis during acute mastitis. Journal of Molecular Endocrinology. 2006;37(3):463-477. DOI: 10.1677/jme.1.02131
  68. 68. Martinov MV, Vitvitsky VM, Banerjee R, Ataullakhanov FI. The logic of the hepatic methionine metabolic cycle. Biochimica et Biophysica Acta. 2010;1804(1):89-96. DOI: 10.1016/j.bbapap.2009.10.004
  69. 69. Lu SC, Mato JM. S-adenosylmethionine in liver health, injury, and cancer. Physiological Reviews. 2012;92(4):1515-1542. DOI: 10.1152/physrev.00047.2011
  70. 70. Shima H, Matsumoto M, Ishigami Y, Ebina M, Muto A, Sato Y, et al. S-adenosylmethionine synthesis is regulated by selective N(6)-adenosine methylation and mRNA degradation involving METTL16 and YTHDC1. Cell Reports. 2017;21(12):3354-3363. DOI: 10.1016/j.celrep.2017.11.092
  71. 71. Kass SU, Pruss D, Wolffe AP. How does DNA methylation repress transcription? Trends in Genetics. 1997;13(11):444-449
  72. 72. Osorio JS, Ji P, Drackley JK, Luchini D, Loor JJ. Smartamine M and MetaSmart supplementation during the peripartal period alter hepatic expression of gene networks in 1-carbon metabolism, inflammation, oxidative stress, and the growth hormone-insulin-like growth factor 1 axis pathways. Journal of Dairy Science. 2014;97(12):7451-7464. DOI: 10.3168/jds.2014-8680
  73. 73. Drackley JK. ADSA Foundation Scholar AwardBiology of dairy cows during the transition period: The final frontier? Journal of Dairy Science. 1999;82(11):2259-2273
  74. 74. Osorio JS, Ji P, Drackley JK, Luchini D, Loor JJ. Supplemental Smartamine M or MetaSmart during the transition period benefits postpartal cow performance and blood neutrophil function. Journal of Dairy Science. 2013;96(10):6248-6263. DOI: 10.3168/jds.2012-5790
  75. 75. Singh K, Erdman RA, Swanson KM, Molenaar AJ, Maqbool NJ, Wheeler TT, et al. Epigenetic regulation of milk production in dairy cows. Journal of Mammary Gland Biology and Neoplasia. 2010;15(1):101-112. DOI: 10.1007/s10911-010-9164-2
  76. 76. He Y, Yu Y, Zhang Y, Song J, Mitra A, Zhang Y, et al. Genome-wide bovine H3K27me3 modifications and the regulatory effects on genes expressions in peripheral blood lymphocytes. PLoS One. 2012;7(6):e39094. DOI: 10.1371/journal.pone.0039094
  77. 77. He Y, Song M, Zhang Y, Li X, Song J, Zhang Y, et al. Whole-genome regulation analysis of histone H3 lysin 27 trimethylation in subclinical mastitis cows infected by Staphylococcus aureus. BMC Genomics. 2016;17:565. DOI: 10.1186/s12864-016-2947-0
  78. 78. McPherron AC, Lee SJ. Double muscling in cattle due to mutations in the myostatin gene. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(23):12457-12461
  79. 79. Di Stasio L, Brugiapaglia A, Galloni M, Destefanis G, Lisa C. Effect of the leptin c.73T>C mutation on carcass traits in beef cattle. Animal Genetics. 2007;38(3):316-317. DOI: 10.1111/j.1365-2052.2007.01595.x
  80. 80. Abe T, Saburi J, Hasebe H, Nakagawa T, Misumi S, Nade T, et al. Novel mutations of the FASN gene and their effect on fatty acid composition in Japanese black beef. Biochemical Genetics. 2009;47(5-6):397-411. DOI: 10.1007/s10528-009-9235-5
  81. 81. Hoff JL, Decker JE, Schnabel RD, Taylor JF. Candidate lethal haplotypes and causal mutations in Angus cattle. BMC Genomics. 2017;18(1):799. DOI: 10.1186/s12864-017-4196-2
  82. 82. Ciepłoch A, Rutkowska K, Oprządek J, Poławska E. Genetic disorders in beef cattle: A review. Genes & Genomics. 2017;39:461-471
  83. 83. Bhat SA, Malik AA, Ahmad SM, Shah RA, Ganai NA, Shafi SS, et al. Advances in genome editing for improved animal breeding: A review. Veterinary World. 2017;10(11):1361-1366. DOI: 10.14202/vetworld.2017.1361-1366
  84. 84. Cassar-Malek I, Picard B. Expression marker-based strategy to improve beef quality. ScientificWorldJournal. 2016;2016:2185323. DOI: 10.1155/2016/2185323
  85. 85. Neeha VS, Kinth P. Nutrigenomics research: A review. Journal of Food Science and Technology. 2013;50(3):415-428. DOI: 10.1007/s13197-012-0775-z
  86. 86. da Costa AS, Pires VM, Fontes CM, Mestre Prates JA. Expression of genes controlling fat deposition in two genetically diverse beef cattle breeds fed high or low silage diets. BMC Veterinary Research. 2013;9:118. DOI: 10.1186/1746-6148-9-118
  87. 87. Hale DS, Goodson, K, Savell, JW. USDA Beef Quality and Yield Grades [Internet]. College Station, TX 77843-24712013. Available from: https://meat.tamu.edu/beefgrading/
  88. 88. Gamarra D, Aldai N, Arakawa A, Barron LJR, Lopez-Oceja A, de Pancorbo MM, et al. Distinct correlations between lipogenic gene expression and fatty acid composition of subcutaneous fat among cattle breeds. BMC Veterinary Research. 2018;14. DOI: 10.1186/s12917-018-1481-5
  89. 89. Moisa SJ, Shike DW, Faulkner DB, Meteer WT, Keisler D, Loor JJ. Central role of the PPARgamma gene network in coordinating beef cattle intramuscular adipogenesis in response to weaning age and nutrition. Gene Regulation and Systems Biology. 2014;8:17-32. DOI: 10.4137/GRSB.S11782
  90. 90. Jeong J, Bong J, Kim GD, Joo ST, Lee HJ, Baik M. Transcriptome changes favoring intramuscular fat deposition in the longissimus muscle following castration of bulls. Journal of Animal Science. 2013;91(10):4692-4704. DOI: 10.2527/jas.2012-6089
  91. 91. Somal A, Aggarwal A, Upadhyay RC. Effect of thermal stress on expression profile of apoptosis related genes in peripheral blood mononuclear cells of transition Sahiwal cow. Iranian Journal of Veterinary Research. 2015;16(2):137-143
  92. 92. Oliveira LJ, Barreto RS, Perecin F, Mansouri-Attia N, Pereira FT, Meirelles FV. Modulation of maternal immune system during pregnancy in the cow. Reproduction in Domestic Animals. 2012;47(Suppl 4):384-393. DOI: 10.1111/j.1439-0531.2012.02102.x
  93. 93. Froidmont E, Mayeres P, Picron P, Turlot A, Planchon V, Stilmant D. Association between age at first calving, year and season of first calving and milk production in Holstein cows. Animal. 2013;7(4):665-672. DOI: 10.1017/S1751731112001577
  94. 94. Li XW, Chen H, Guan Y, Li XB, Lei LC, Liu JX, et al. Acetic acid activates the AMP-activated protein kinase signaling pathway to regulate lipid metabolism in bovine hepatocytes. PLoS One. 2013;8(7):e67880. DOI: 10.1371/journal.pone.0067880
  95. 95. Buchanan JW, Garmyn AJ, Hilton GG, VanOverbeke DL, Duan Q , Beitz DC, et al. Comparison of gene expression and fatty acid profiles in concentrate and forage finished beef. Journal of Animal Science. 2013;91(1):1-9. DOI: 10.2527/jas.2012-5154
  96. 96. Daley CA, Abbott A, Doyle PS, Nader GA, Larson S. A review of fatty acid profiles and antioxidant content in grass-fed and grain-fed beef. Nutrition Journal. 2010;9:10. DOI: 10.1186/1475-2891-9-10
  97. 97. Paradis F, Wood KM, Swanson KC, Miller SP, McBride BW, Fitzsimmons C. Maternal nutrient restriction in mid-to-late gestation influences fetal mRNA expression in muscle tissues in beef cattle. BMC Genomics. 2017;18(1):632. DOI: 10.1186/s12864-017-4051-5
  98. 98. Du M, Tong J, Zhao J, Underwood KR, Zhu M, Ford SP, et al. Fetal programming of skeletal muscle development in ruminant animals. Journal of Animal Science. 2010;88(13 Suppl):E51-E60. DOI: 10.2527/jas.2009-2311
  99. 99. Siersbaek R, Nielsen R, Mandrup S. PPARgamma in adipocyte differentiation and metabolism--novel insights from genome-wide studies. FEBS Letters. 2010;584(15):3242-3249. DOI: 10.1016/j.febslet.2010.06.010
  100. 100. Lee JE, Ge K. Transcriptional and epigenetic regulation of PPARgamma expression during adipogenesis. Cell & Bioscience. 2014;4:29. DOI: 10.1186/2045-3701-4-29
  101. 101. Moisa SJ, Shike DW, Shoup L, Rodriguez-Zas SL, Loor JJ. Maternal plane of nutrition during late gestation and weaning age alter Angus x Simmental offspring longissimus muscle transcriptome and intramuscular fat. PLoS One. 2015;10(7):e0131478. DOI: 10.1371/journal.pone.0131478
  102. 102. Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Research. 2011;21(3):381-395. DOI: 10.1038/cr.2011.22
  103. 103. Wang H, Zheng Y, Wang G, Li H. Identification of microRNA and bioinformatics target gene analysis in beef cattle intramuscular fat and subcutaneous fat. Molecular BioSystems. 2013;9(8):2154-2162. DOI: 10.1039/c3mb70084d
  104. 104. Huang YZ, Sun JJ, Zhang LZ, Li CJ, Womack JE, Li ZJ, et al. Genome-wide DNA methylation profiles and their relationships with mRNA and the microRNA transcriptome in bovine muscle tissue (Bos taurine). Scientific Reports-UK. 2014;4:1-17. DOI: ARTN 6546/10.1038/srep06546
  105. 105. Liu X, Usman T, Wang Y, Wang Z, Xu X, Wu M, et al. Polymorphisms in epigenetic and meat quality related genes in fourteen cattle breeds and association with beef quality and carcass traits. Asian-Australasian Journal of Animal Sciences. 2015;28(4):467-475. DOI: 10.5713/ajas.13.0837
  106. 106. Romao JM, Jin W, He M, McAllister T, Guan le L. MicroRNAs in bovine adipogenesis: Genomic context, expression and function. BMC Genomics. 2014;15:137. DOI: 10.1186/1471-2164-15-137
  107. 107. Jin W, Dodson MV, Moore SS, Basarab JA, Guan LL. Characterization of microRNA expression in bovine adipose tissues: A potential regulatory mechanism of subcutaneous adipose tissue development. BMC Molecular Biology. 2010;11:29. DOI: 10.1186/1471-2199-11-29
  108. 108. Muroya S, Shibata M, Hayashi M, Oe M, Ojima K. Differences in circulating microRNAs between grazing and grain-fed wagyu cattle are associated with altered expression of intramuscular microRNA, the potential target PTEN, and lipogenic genes. PLoS One. 2016;11(9):e0162496. DOI: 10.1371/journal.pone.0162496
  109. 109. Sun J, Sonstegard TS, Li C, Huang Y, Li Z, Lan X, et al. Altered microRNA expression in bovine skeletal muscle with age. Animal Genetics. 2015;46(3):227-238. DOI: 10.1111/age.12272
  110. 110. Dayton WR, White ME. MEAT SCIENCE AND MUSCLE BIOLOGY SYMPOSIUM—Role of satellite cells in anabolic steroid-induced muscle growth in feedlot steers. Journal of Animal Science. 2014;92(1):30-38. DOI: 10.2527/jas.2013-7077
  111. 111. Sadkowski T, Ciecierska A, Oprzadek J, Balcerek E. Breed-dependent microRNA expression in the primary culture of skeletal muscle cells subjected to myogenic differentiation. BMC Genomics. 2018;19(1):109. DOI: 10.1186/s12864-018-4492-5
  112. 112. Dai Y, Zhang WR, Wang YM, Liu XF, Li X, Ding XB, et al. MicroRNA-128 regulates the proliferation and differentiation of bovine skeletal muscle satellite cells by repressing Sp1. Molecular and Cellular Biochemistry. 2016;414(1-2):37-46. DOI: 10.1007/s11010-016-2656-7
  113. 113. Dai Y, Wang YM, Zhang WR, Liu XF, Li X, Ding XB, et al. The role of microRNA-1 and microRNA-206 in the proliferation and differentiation of bovine skeletal muscle satellite cells. In Vitro Cellular & Developmental Biology. Animal. 2016;52(1):27-34. DOI: 10.1007/s11626-015-9953-4
  114. 114. Horikawa A, Ogasawara H, Okada K, Kobayashi M, Muroya S, Hojito M. Grazing-induced changes in muscle microRNA-206 and -208b expression in association with myogenic gene expression in cattle. Animal Science Journal. 2015;86(11):952-960. DOI: 10.1111/asj.12381
  115. 115. McCarthy JJ. MicroRNA-206: The skeletal muscle-specific myomiR. Biochimica et Biophysica Acta. 2008;1779(11):682-691. DOI: 10.1016/j.bbagrm.2008.03.001
  116. 116. Yan X, Huang Y, Zhao JX, Rogers CJ, Zhu MJ, Ford SP, et al. Maternal obesity downregulates microRNA let-7g expression, a possible mechanism for enhanced adipogenesis during ovine fetal skeletal muscle development. International Journal of Obesity. 2013;37(4):568-575. DOI: 10.1038/ijo.2012.69
  117. 117. Yan X, Zhu M-J, Du M. MicroRNA has-let-7g inhibits proliferation, adipogenic differentiation and inflammation in C3H10T½ cells. The FASEB Journal. 2011;25(Supplement lb):493
  118. 118. Moisa SJ, Shike DW, Shoup L, Loor JJ. Maternal plane of nutrition during late-gestation and weaning age alter steer calf longissimus muscle adipogenic microRNA and target gene expression. Lipids. 2016;51(1):123-138. DOI: 10.1007/s11745-015-4092-y
  119. 119. Wattiaux MA. Dairy sector across the world: National trends and opportunities for sustainable growth. In: Beede DK, editor. Large Dairy Herd Management. 3rd ed. Champaign, IL: American Dairy Science Association; 2017. pp. 3-19
  120. 120. Wang M, Xu B, Wang H, Bu D, Wang J, Loor JJ. Effects of arginine concentration on the in vitro expression of casein and mTOR pathway related genes in mammary epithelial cells from dairy cattle. PLoS One. 2014;9(5):e95985. DOI: 10.1371/journal.pone.0095985
  121. 121. Underwood KR, Means WJ, Zhu MJ, Ford SP, Hess BW, Du M. AMP-activated protein kinase is negatively associated with intramuscular fat content in longissimus dorsi muscle of beef cattle. Meat Science. 2008;79(2):394-402. DOI: 10.1016/j.meatsci.2007.10.025
  122. 122. Zhou H, Cheng L, Azimu W, Hodge S, Edwards GR, Hickford JG. Variation in the bovine FABP4 gene affects milk yield and milk protein content in dairy cows. Scientific Reports. 2015;5:10023. DOI: 10.1038/srep10023
  123. 123. Goszczynski DE, Papaleo-Mazzucco J, Ripoli MV, Villarreal EL, Rogberg-Munoz A, Mezzadra CA, et al. Genetic variation in FABP4 and evaluation of its effects on beef cattle fat content. Animal Biotechnology. 2017;28(3):211-219. DOI: 10.1080/10495398.2016.1262868
  124. 124. Benjamin AL, Green BB, Crooker BA, McKay SD, Kerr DE. Differential responsiveness of Holstein and Angus dermal fibroblasts to LPS challenge occurs without major differences in the methylome. BMC Genomics. 2016;17:258. DOI: 10.1186/s12864-016-2565-x
  125. 125. Drackley JK, Donkin SS, Reynolds CK. Major advances in fundamental dairy cattle nutrition. Journal of Dairy Science. 2006;89(4):1324-1336. DOI: 10.3168/jds.S0022-0302(06)72200-7
  126. 126. Gonzalez-Calvo L, Joy M, Alberti C, Ripoll G, Molino F, Serrano M, et al. Effect of finishing period length with alpha-tocopherol supplementation on the expression of vitamin E-related genes in the muscle and subcutaneous fat of light lambs. Gene. 2014;552(2):225-233. DOI: 10.1016/j.gene.2014.09.037
  127. 127. Coyne GS, Kenny DA, Childs S, Sreenan JM, Waters SM. Dietary n-3 polyunsaturated fatty acids alter the expression of genes involved in prostaglandin biosynthesis in the bovine uterus. Theriogenology. 2008;70(5):772-782. DOI: 10.1016/j.theriogenology.2008.05.048
  128. 128. Lohakare J, Osorio JS, Bionaz M. Peroxisome proliferator-activated receptor beta/delta does not regulate glucose uptake and lactose synthesis in bovine mammary epithelial cells cultivated in vitro. The Journal of Dairy Research. 2018:1-8. DOI: 10.1017/S0022029918000365
  129. 129. Shi HB, Zhang CH, Xu ZA, Lou GG, Liu JX, Luo J, et al. Peroxisome proliferator-activated receptor delta regulates lipid droplet formation and transport in goat mammary epithelial cells. Journal of Dairy Science. 2018;101(3):2641-2649. DOI: 10.3168/jds.2017-13543
  130. 130. Khan M, Couturier A, Kubens JF, Most E, Mooren FC, Kruger K, et al. Niacin supplementation induces type II to type I muscle fiber transition in skeletal muscle of sheep. Acta Veterinaria Scandinavica. 2013;55:85. DOI: 10.1186/1751-0147-55-85
  131. 131. Gruffat D, Cherfaoui M, Bonnet M, Thomas A, Bauchart D, Durand D. Breed and dietary linseed affect gene expression of enzymes and transcription factors involved in n-3 long chain polyunsaturated fatty acids synthesis in longissimus thoracis muscle of bulls. Journal of Animal Science. 2013;91(7):3059-3069. DOI: 10.2527/jas.2012-6112
  132. 132. Goszczynski DE, Mazzucco JP, Ripoli MV, Villarreal EL, Rogberg-Munoz A, Mezzadra CA, et al. Genetic characterisation of PPARG, CEBPA and RXRA, and their influence on meat quality traits in cattle. Journal of Animal Science and Technology. 2016;58:14. DOI: 10.1186/s40781-016-0095-3
  133. 133. Qu Y, Elsasser TH, Kahl S, Garcia M, Scholte CM, Connor EE, Schroeder GF, Moyes KM. The effects of feeding mixed tocopherol oil on whole-blood respiratory burst and neutrophil immunometabolic-related gene expression in lactating dairy cows. Journal of Dairy Science. 2018;101(5):4332-4342. DOI: 10.3168/jds.2017-13902
  134. 134. Akbar H, Schmitt E, Ballou MA, Correa MN, Depeters EJ, Loor JJ. Dietary lipid during late-pregnancy and early-lactation to manipulate metabolic and inflammatory gene network expression in dairy cattle liver with a focus on PPARs. Gene Regulation and Systems Biology. 2013;7:103-123. DOI: 10.4137/GRSB.S12005
  135. 135. Carreno D, Hervas G, Toral PG, Castro-Carrera T, Frutos P. Fish oil-induced milk fat depression and associated downregulation of mammary lipogenic genes in dairy ewes. Journal of Dairy Science. 2016. DOI: 10.3168/jds.2016-11019
  136. 136. Wang X, Lan X, Radunz AE, Khatib H. Maternal nutrition during pregnancy is associated with differential expression of imprinted genes and DNA methyltranfereases in muscle of beef cattle offspring. Journal of Animal Science. 2015;93(1):35-40. DOI: 10.2527/jas.2014-8148
  137. 137. Jacometo CB, Zhou Z, Luchini D, Trevisi E, Correa MN, Loor JJ. Maternal rumen-protected methionine supplementation and its effect on blood and liver biomarkers of energy metabolism, inflammation, and oxidative stress in neonatal Holstein calves. Journal of Dairy Science. 2016;99(8):6753-6763. DOI: 10.3168/jds.2016-11018
  138. 138. Liu X, Guo XY, Xu XZ, Wu M, Zhang X, Li Q, et al. Novel single nucleotide polymorphisms of the bovine methyltransferase 3b gene and their association with meat quality traits in beef cattle. Genetics and Molecular Research. 2012;11(3):2569-2577. DOI: 10.4238/2012.June.29.1
  139. 139. Zhao Q, Kang Y, Wang HY, Guan WJ, Li XC, Jiang L, et al. Expression profiling and functional characterization of miR-192 throughout sheep skeletal muscle development. Scientific Reports. 2016;6:30281. DOI: 10.1038/srep30281

Written By

Johan S. Osorio and Sonia J. Moisa

Submitted: 18 April 2018 Reviewed: 23 October 2018 Published: 28 January 2019