Вы находитесь на странице: 1из 13

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/326887674

Feedomics: Promises for food security with sustainable food animal


production

Article  in  TrAC Trends in Analytical Chemistry · August 2018


DOI: 10.1016/j.trac.2018.07.025

CITATION READS

1 94

2 authors, including:

Huizeng Sun
University of Alberta
8 PUBLICATIONS   4 CITATIONS   

SEE PROFILE

All content following this page was uploaded by Huizeng Sun on 18 February 2019.

The user has requested enhancement of the downloaded file.


Trends in Analytical Chemistry 107 (2018) 130e141

Contents lists available at ScienceDirect

Trends in Analytical Chemistry


journal homepage: www.elsevier.com/locate/trac

Feedomics: Promises for food security with sustainable food animal


production
Hui-Zeng Sun, Le Luo Guan*
Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, T6G 2P5, AB, Canada

a r t i c l e i n f o a b s t r a c t

Article history: The production of adequate and nutritious animal proteins for the increasing human population is an
Available online 7 August 2018 urgent global task. Therefore, enhancing the efficiency and sustainability of food animal production
requires advanced analytical techniques. We propose the concept of “feedomics” for food animal
Keywords: research, an emerging field using omics technologies, to understand and uncover the mechanisms
Animal production and health involved in many biological processes that determine animal productivity, product quality, and health as
Feed science
a result of the interactions among feed, environment, animal genetics, physiology, and its symbiotic
Genomics
microbiota. In this review, we summarize the findings to date based on the omics approaches including
Epigenomics
Proteomics
(meta)genomics, epigenomics, (meta)transcriptomics, proteomics, and metabolomics in food animal
Metabolomics species and consider how these can be used to understand the processes from the “gate” to “plate”. We
(Meta)genomics also highlight future directions for applying feedomics in fundamental and practical studies to improve
(Meta)transcriptomics the quantity, quality, safety and functional properties of food animal products.
microRNAs © 2018 Elsevier B.V. All rights reserved.
Feedomics

1. Introduction which directly affect the animal's performance and welfare. In


addition, food animal production has been considered to have a
By 2050, the world human population is projected to reach 9.4 negative environmental impact due to the emission of greenhouse
billion [1]. Animal protein (milk, egg, fish, and meat) is an integral gases (methane, CO2) and the release of nitrous compounds (NO2,
component of the human diet globally, and it has contributed to the ammonia) and phosphate [6]. Moreover, the recent attention on
development and elimination of poverty in human society. To meet reducing the antimicrobial growth promotants in food animal
the demand of the increasing human population, the global food production has driven the need to seek alternative feeding strate-
animal production is expected to increase 2.3% annually to 2050 [2]. gies to maintain sustainable production [7]. Therefore, improving
Currently, food animal production consumes more than 32% of the sustainability of food animal production is urgent and vital to
global human edible cereal grains (e.g., corns, barley) and occupies promote animals' welfare, to prevent further expansion of occupied
approximately 40% of all arable land [3]. Over the past several de- arable land, and to reduce the negative environmental impact.
cades, many technologies have been applied to improve food ani- Even with the rapid development of feed science, vast hard-to-
mal production efficiency. First, the modern commercial food decipher biological mechanisms still limit the regulatory strategies
animal sector has adapted to a highly intensive management sys- of sustainable food animal production. The “Foodomics” was first
tem with using the high proportion of cereal grains in the diets. proposed as a discipline for studying the food and nutrition do-
Second, since the 1960s, as one such “innovation”, antimicrobial mains through the application and integration of advanced omics
growth promotants have been widely applied to improve the pro- technologies to improve human well-being, health, and confidence
duction efficiency of food animals. Third, recent advanced research by Cifuentes in 2009 [8]. Although the word feedomics can be
in molecular genetics has shown some promise for breeding more found in the titles of a PhD thesis [9] and a project (https://cphpig.
efficient animals [4]. However, the high grain-fed animals are more ku.dk/english/news/2015/haja/), a systematic definition and
susceptible to metabolic dysfunction, disorders, and diseases [5], detailed introduction are still lacking. Similar to “foodomics”, we
propose the term “feedomics” for food animal research, which
devises a systematic omics approach to understand the process
from “gate” to “plate” for animal productivity and health as well as
* Corresponding author.
the product nutritional value for the human. In this review, we
E-mail address: lguan@ualberta.ca (L.L. Guan).

https://doi.org/10.1016/j.trac.2018.07.025
0165-9936/© 2018 Elsevier B.V. All rights reserved.
H.-Z. Sun, L.L. Guan / Trends in Analytical Chemistry 107 (2018) 130e141 131

integrate the knowledge of feed science, animal nutrition, animal


Abbreviations genetics and genomics, animal physiology and microbiology, plant
science and biochemistry, analytical chemistry, bioinformatics,
DE differential expressed engineering, and so on, which push forward the development and
ESI electrospray ionization inter-application of related disciplines.
GC-TOF/MS gas chromatography-time of flight/mass
spectrometry 2.2. The omics technologies applied in feedomics
GC-Q-MS gas chromatography-quadrupole-mass
spectrometry In feedomics, genomics is the study of all genetic information
GIT gastrointestinal tract (the genome) about a food animal organism mainly through gen-
GWAS genome-wide association study otyping and/or whole genome sequencing [10]. In transcriptomics,
HPLC high-performance liquid chromatography the expression patterns of all genes in the specific cell and/or tissue
ICAT isotope-coded affinity tag type under certain nutritional, physiological and environmental
LC-MS liquid chromatographyemass spectrometry conditions are studied [11]. Although genomics and tran-
MALDI-TOF/MS matrix-assisted laser desorption ionization- scriptomics provide the functional potential and the activity of the
time of flight/mass spectrometry functional coding genes, only ~3% of the genome encodes proteins,
MeDIP-Seq methylated DNA immunoprecipitation- with the rest of the genome being non-coding regions, and up to
sequencing 80% of the genome including non-coding regions is transcribed
MiRNA microRNA [12]. The non-coding RNAs are likely to contribute more in func-
NGS next-generation sequencing tional transcriptomes, which has led to an increasing emphasis on
NMR nuclear magnetic resonance studying the food animal non-coding small RNAs (especially
RFI residual feed intake microRNAomes) [13]. Chemical changes on the genome including
RNA-Seq RNA-Sequencing chromatin remodeling, histone protein modification and DNA
SILAC stable isotope labeling with amino acids in cell methylation have been considered to alter the phenotypic out-
culture comes in food animals, which may be passed on to the offspring
SNP single nucleotide polymorphisms and can be investigated by epigenomics [14]. Proteomics refers to
UHPLC-QQQ-MS ultra-high-performance liquid all of the proteins derived from coding-gene expression and
chromatography-triple quadrupole-mass translation and post-translational modifications, which serve as the
spectrometry main components of the proteome of food animal [15]. The large set
UPLC-Q-TOF-MS/MS ultra-performance liquid of downstream metabolites represents the final products of the
chromatography-quadrupole-time of genome, transcriptome, and proteome and reflects the external
flight-mass spectrometry/mass phenotype most directly, which can be qualitatively and quantita-
spectrometry tively analyzed using untargeted and targeted metabolomics in
feedomics [16]. The study of microbiomics provides information on
all the microbes of a given microbiota community (referring mainly
to epithelia and digesta of the gastrointestinal tract (GIT) in food
review the main omics approaches recently used in different food animals) in response to feeding intake [17]. Metagenomics and
animal species and their potential roles in helping achieve sus- metatranscriptomics are the main feedomics approaches that are
tainable developed food animal production. currently used to explore the genes and transcripts of the micro-
biome, and they provide a comprehensive understanding of the
2. The definition and analytical technologies of feedomics microorganisms’ composition, function, and undergoing activities
[18].
2.1. Definition and concept of feedomics The powerful force and holistic perspectives of feedomics are
mainly attributed to the advanced omics analytical systems, which
The recent advanced analytical technologies, especially with can potentially capture all molecules at different biological levels
high-throughput characteristics, unbiased, and untargeted “omics”, and generate their corresponding big datasets (which can be
have created the new era of feed science and animal nutrition/ defined as the feedome). The high-throughput next-generation
health and made it possible to identify complex findings at a system sequencing (NGS) techniques are widely applied to generate fee-
biological level. Here, we define feedomics as an emerging research dome datasets. To be specific, the untargeted whole genome, epi-
field that studies feed science, animal nutrition and animal pro- genome, transcriptome, microbial metagenome and
duction/health in a global molecular and systematic scale by using metatranscriptome datasets can be generated using NGS platforms
high sensitivity and resolution omics technologies. These omics such as Illumina NextSeq, MiSeq, HiSeq 2000, HiSeq 3000, HiSeq
methodologies include genomics, epigenomics, transcriptomics, 4000 with different depths [19], and even third-generation
proteomics, metabolomics, metagenomics, and metatran- sequencing methods such as 3-D genomic Hi-C [20]. Further, pro-
scriptomics. Recent efforts have been made to apply omics in food teomes and metabolomes can be identified by spectrometry- and
animal field (focused mainly on cattle, sheep, swine, poultry, and chromatography-based technologies, including liquid
fish) to address research questions and industrial problems, but chromatography-mass spectrometry (LC-MS) and gas
most published studies focus on only a single “omic” approach and chromatography-time of flight/MS (GC-TOF/MS) [21].
lack a linkage between the feed input and the end products. Proteomics and metabolomics are more relied-upon analytical
Therefore, we propose a general workflow of feedomics that in- techniques compared with other omics methods [22]. Proteomics is
cludes defining scientific questions, determining experimental of great importance to assess food animal productivity (especially
design, sampling collection, molecular (DNA, RNA, proteins, or for meat and milk safety and quality), health, and welfare [23]. MS-
metabolites) extraction, identification using high-throughput based proteomic techniques usually consist of an ion source
platforms, data analysis, mechanism interpretation, and valida- (nebulize the peptides into ionized droplets), a mass analyzer
tion. These steps contribute to a series of systematic processes and (measures the mass-to-charge ratio (m/z) of the ionized analyte)
132 H.-Z. Sun, L.L. Guan / Trends in Analytical Chemistry 107 (2018) 130e141

and a detector (records the ion numbers of each m/z value) [24]. polar compounds volatile enough for GC detection. Untargeted
Electrospray ionization (ESI) and matrix-assisted laser desorption/ metabolomics usually use one or several of various high-
ionization (MALDI) are widely used in MS-based proteomics to throughput technologies, especially mass spectrometry-based
produce ions; the former is normally combined with advanced platforms (GC-MS, GC-TOF/MS, GC  GC-TOF/MS, GC-Q-MS, LC-
separation tools such as GC and high-performance liquid chroma- MS, ultra-performance liquid chromatography (UPLC)-Q-TOF-MS/
tography (HPLC) to analyze complex peptides, and the latter is MS) to get a full scan of all metabolites (what metabolites are
commonly harnessed to analyze relatively simple samples [25]. present and the relative abundance of these metabolites) that play
With the rapid advances in analytical chemistry techniques and an important role in capturing unknown or novel metabolites,
data analysis methods, metabolomics has become more accessible subtle pathways and key mechanisms. Furthermore, the metabo-
to broad research areas in food animal-related disciplines, such as lites of interest can be examined using targeted metabolomics (the
animal health and disease, animal nutrition, human health and real concentration), with the commonly used techniques including
nutrition, and feed analysis [26]. MS-based analytical technologies GC-TOF/MS, GC-Q-MS, UPLC-Q-TOF-MS/MS, and UHPLC-QQQ-MS.
are most commonly applied in metabolite identification because of For metabolomics studies, it is important to determine which
the high selectivity, sensitivity and structural information of the samples should be used for analysis due to the different charac-
detected metabolites [27]. TOF or quadrupole (Q) is usually com- teristic of samples, various compound extraction methods, high
bined with MS as a mass analyzer to strengthen the accuracy and costs and lack of databases.
resolution of mass measurement [28]. Prior to MS detection, a GC or These approaches vary in their resolution, sensitivity, accuracy,
LC separation tool is required based on the chemical properties of coverage, stability, run time, sample loading amount, cost per
the metabolites in the sample. Typically, LC is more suitable for sample and so on, so it is important to choose suitable techniques
detecting non-volatile metabolites such as hydroxy-carboxylic based on different research objectives and comparable information
acids, and GC is easier to capture volatile metabolites such as including the applications, advantages, and limitations of major
xylene [29]. Meanwhile, derivatization can be utilized to make the analytical technologies applied in feedomics (Table 1).

Table 1
The applications, advantages, and limitations of major analytical technologies applied in feedomics.

Analytical technologies Applications in feedomics Advantages Limitations

NextSeq Whole (meta)genomics and (meta) High output range; High coverage Costly
transcriptomics;
Epigenomics
Ion Torrent PGM Targeted DNA, RNA and microbial sequencing Short run time; High output per hour Low sequencing quality; High error rate
Hi-C Genomes conformation [20] Can detect chromatin interactions Low resolution; High noise
BeadChip DNA methylation; Genotyping [38e43] Cost-effective for large samples; Not suitable for low frequency sites
Microarray Genotyping; DNA methylation; Gene Simple workflow Low dynamic range; Need probe design
expression [58,60], sequences
microRNA [66,68]
HiSeq Metagenomics [116,119,120]; High-throughput; Broader dynamic range Short reads; High cost; High computation
Transcriptomics [35,52e55,61]; needs
Metatranscriptomics
[18,117,119]; MicroRNAome [67,70,71];
MeDIP-Seq based epigenomics [78,79];
Bisulfite-Seq based epigenomics [83]
MiSeq 16S metagenomics [116,121] Long reads Low output
Methylated DNA DNA methylation rates [75] Easy and quick to perform; High accuracy Lack specific sites information
qualification kit
Pyrosequencing Methylation and genomic variation, microbial Long reads Low accuracy; Costly
diversity
[101,115,118]
ChIP-on-chip Histone modifications in epigenomics Aim at broad binding Lack whole genome scan; Require
highly specific antibodies
ChIP-Seq Histone modifications in epigenomics High resolution; High coverage; Low Costly; Relative slow
sample loading
LC-MS/MS Separation and qualitative analysis in Less sample loading; High coverage; Easy to be affected by the MS stability
HPLC-TOF/MS proteomics High accuracy
[86,88,90]and metabolomics [96,104]
MALDI-TOF/MS Identification and quantitative analysis of High throughput; High sensitivity Low accuracy
HPLC/ESI-MS proteomics Long analysis time
[85,114]
i-TRAQ Proteome quantitative analysis [86,87,92,114] High throughput; Time saving Costly
SILAC High label efficiency Only used for live cell
ICAT Broad compatibility Only label peptides containing cysteine,
and can only mark two samples
NMR Untargeted metabolomics [95,105] High reproducibility; Simple sample Low sensitivity; Less feature metabolites
preparation observation; Poor database
GC-MS Identification and relative quantitative analysis High throughput and sensitivity Complex extraction and derivatization
of process
GC-TOF/MS untargeted and targeted metabolomics Wide m/z range; High sensitivity Relatively low qualitative ability
GC  GC-TOF/MS [94,97e102,105,108,110,113] Four dimensions of analytical resolution Low robustness of instruments
GC-Q-TOF/MS Untargeted and targeted metabolome High quantitative stability; High selection; Costly
quantitative High scan speed
UPLC-Q-TOF-MS/MS analysis [109] Good quantitative reproducibility; Low resolution; Costly
High accuracy; High sensitivity
H.-Z. Sun, L.L. Guan / Trends in Analytical Chemistry 107 (2018) 130e141 133

3. The applications of different feedomics technologies for food animal species such as cattle [32], chicken [33], sheep [34], pig
sustainable food animal production [35] and a number of aquatic animals [36] have promoted the
application of high-throughput genetic SNP markers and accelerated
3.1. Genomics and nutrigenetics in food animal production genetic improvement for food animal production and efficiency. For
efficiency and quality example, a genome-wide association study (GWAS) was successfully
used to identify genetic variants, regions, and candidate genes
Genotyping aims to identify trait-associated genetic variations associated with feed efficiency. In total, 25, 66, and 12 SNPs have
(such as a single nucleotide polymorphism (SNP), a variation in a been found to be associated with residual feed intake (RFI, one of the
single nucleotide that occurs at a specific position on the genome) measures for feed efficiency) in hybrid bulls [37], Angus steers [38]
that can be potentially be used for genomic breeding and/or targeted and Yorkshire pigs [39] using the BeadChip measurement. The ge-
section [30]. High coverage SNP datasets through the whole genome netic variation identified by BeadChips is further identified which
not only help detect low abundance SNPs but also enable the efficient candidate genes are associated with animal feed efficiency for Nelore
screening and tracking of genetic markers that could transfer from cattle [40], hybrid bulls [37], pig [41], and chicken [42] (all genes and
parents to offspring [31]. Released whole genome sequences of major their functions are summarized in Table 2). Similarly, Sanchez

Table 2
The information of different phenotypes related genes in feedomics.

Name Full name or location Tissues Traits Animals

HRH4 Histamine Receptor H4 Blood Feed efficiency Nelore cattle [40]


ALDH7A1 Aldehyde Dehydrogenase 7 Family Member A1
APOA2 Apolipoprotein A2
LIN7C Lin-7 Homolog C, Crumbs Cell Polarity Complex
Component
CXADR CXADR, Ig-Like Cell Adhesion Molecule
ADAM12 ADAM Metallopeptidase Domain 12
MAP7 Microtubule Associated Protein 7
GHR Growth Hormone Receptor Liver Hybrid bulls [37]
CAST Calpastatin
ACAD11 Acyl-CoA Dehydrogenase Family Member 11
UGT3A1 UDP Glycosyltransferase Family 3 Member A1
XIRP2 Xin Actin Binding Repeat Containing 2 Backfat Pig [41]
TTC29 Tetratricopeptide Repeat Domain 29
SOGA1 Suppressor Of Glucose, Autophagy Associated 1
MAS1 MAS1 Proto-Oncogene
GPCR G Protein-Coupled Receptor
GPK5 G Protein-Coupled Receptor Kinase 5
GPR155 G Protein-Coupled Receptor 155
ZFYVE26 Zinc Finger FYVE-Type Containing 26
HTR1B 5-Hydroxytryptamine Receptor 1B Blood Chicken [42]
RGS3 Regulator Of G Protein Signaling 3
ABCG2 ATP-binding cassette sub-family G member 2 Milk Milk protein Dairy cow [43]
AGPAT6 1-acylglycerol-3-phosphate O-acyltransferase 6
DGAT1 diacylglycerol O-acyltransferase 1
DHX15 DEAH-box helicase 15
MROH1 maestro heat like repeat family member 1
SLC37A1 solute carrier family 37 member 1
CSN3 casein kappa
PKD2 polycystin 2, transient receptor potential cation
channel
HERC3 HECT and RLD domain containing E3 ubiquitin
protein ligase 3
ALPL alkaline phosphatase, liver/bone/kidney
AHKH ANKH inorganic pyrophosphate transport regulator
SEL1L3 SEL1L family member 3
MGST1 microsomal glutathione S-transferase 1
CSN1S2 casein alpha-S2
MEPE matrix extracellular phosphoglycoprotein
PICALM phosphatidylinositol binding clathrin assembly protein
SEPSECS Sep (O-phosphoserine) tRNA:Sec (selenocysteine) tRNA
synthase
BOP1 block of proliferation 1
CSN1S1 casein alpha s1
CSN2 casein beta
RECQL4 RecQ like helicase 4
PAEP progestagen-associated endometrial protein
GPR120 G protein-coupled receptors Adipose, skeletal muscle, ileum, Fat deposition Pig [48] [49],
jejunum, duodenum, kidney, lung,
spleen, liver, and heart; blood,
muscle and ear
CYP19A1A cytochrome P450, family 19, subfamily A, polypeptide 1 Ovary Ovarian development Japanese flounder [75]
FOXL2 forkhead box L2
SMAD1 SMAD Family Member 1 Blood and muscle Body size Sheep [78]
TSC1 Tuberous Sclerosis 1
AKT1 AKT Serine/Threonine Kinase 1
134 H.-Z. Sun, L.L. Guan / Trends in Analytical Chemistry 107 (2018) 130e141

reported the 22 most plausible candidate genes associated with milk respectively, using RNA-Sequencing (RNA-Seq)-based tran-
protein composition in dairy cattle using GWAS [43] (Table 2). Thus, scriptomics with the HiSeq 2500 machine. These genes were
whole genome- and genotyping-based GWAS studies will allow re- involved in steroid hormone biosynthesis, lipid metabolism, and
searchers to discover gene markers that can be exploited in future drug metabolism cytochrome P450 [52]. A similar approach
breeding programs to achieve high feed efficiency and high-quality sequenced by the HiSeq 2000 system was used to study rumen
products farm animals. epithelial tissues of cattle with varied feed efficiency and found 122
Although genetic information can indicate the performance DE genes involved in the function of acetylation, remodeling of
potential of the animals, nutrients act as an important intermediate adherens junctions, cytoskeletal dynamics, cell migration, and cell
to affect the biological and metabolic processes that determine the turnover [53]. Based on these findings, transcriptomics can deter-
overall performance [44]. It is known that diet can alter the mine the underlying regulating functions of these two digestive
expression of genes, signaling, and metabolic pathways in the cells, organs and can help identify the biological mechanism underlying
which subsequently changes the physiological status and finally feed efficiency. In addition, RNA-Seq based transcriptomic studies
leads to different phenotypes [45]. Additionally, different genetics provide a fundamental understanding of functional aspects of tis-
may drive cellular responses and host metabolism to the nutrients, sues. A recent study revealed transcriptomes of digestive tissues in
which has been defined as “nutrigenetics” in human-related sheep, showing the GIT transcriptomes are mainly driven by
studies. Nutrigenetics is used to reveal the relationship between epidermal differentiation complex genes [54]. Such work can pro-
diet and the animal genome and aims to understand whether and vide novel insights into fundamental issues concerning mammalian
how the individual genetic components drive the responses to di- digestive and gastrointestinal systems, which can directly impact
etary variation [46]. The concept of identifying the genetic markers animal growth and performance through feed digestion and
related to dietary responses has been applied in swine production. nutrient absorption.
Using the widely studied G protein-coupled receptor (GPR120) gene In addition to the understanding of altered molecular mecha-
as an example, this gene encodes the omega-3 fatty acid receptor nisms related to animal production, the transcriptomic-based
which is involved in various physiological homeostasis processes approach can be applied to study the molecular mechanisms of
such as fat deposition, regulation of appetite and food preference animal health. For example, the whole blood transcriptome has
[47]. The variants in GPR120 have been significantly associated with been proven to be a potential molecular phenotype diagnostic tool
fat deposition and growth in pigs [48], and a recent study has for food animal health. A recent study using the RNA-Seq based
identified 3 SNPs (g.114743623G>C; g.114764865G>A; and whole blood transcriptomics with the HiSeq 2000 instrument
g.114765469C>T) among different breeds, including Italian Large identified 246 DE genes in sick pigs involved in a variety of systemic
White, Italian Duroc, Italian Landrace, Casertana, Pietrain, Meishan, immune and inflammatory responses to many physiological pro-
and wild boars, which are associated with average daily gain [49]. cesses, thus suggesting their gene marker functions are associated
Ichimura et al. demonstrated and verified that GPR120 had a key with health status in commercial pigs [55]. Similarly, blood tran-
role in sensing dietary fat and controlling energy balance in both scripts can be applied as biomarkers for selecting pigs with reduced
humans and rodents [50], suggesting that different breeds of pigs susceptibility to Salmonella shedding [56]. Increasing dietary DHA
may have different ways to exact energy from diet for growth. levels were associated with the upregulation of hepatic immune
However, no study has been performed on how the varied GPR120 pathways, especially chemokine signaling, FC epsilon RI signaling
genotypes could lead to altered dietary responses within the same and natural killer cell-mediated cytotoxicity pathways in Atlantic
breed and whether it relates to performance. To date, the linkage of salmon, which expanded our understanding of how functional di-
“genetics” and “nutrition” is scarce in food animal production. etary nutrients elicit signaling molecules and a physiological
Genome-wide nutrigenetics could be a powerful tool for defining impact [57]. Moreover, transcriptomic analysis of circulating leu-
appropriate feeding practices to raise more sustainable food ani- kocytes in blood using a microarray identified novel aspects of the
mals based on an animal's genetic potentials. The variability of host systemic inflammatory response to sheep scab mites, which
marker genes (such as GPR120 gene) should be considered by using provided new insights into the mechanisms of local allergen-
nutrigenetics approaches to define the appropriate feeding prac- induced inflammatory responses [58]. However, transcriptomics
tices, to obtain more efficient feed in heavy pigs, and to evaluate research in animal health is still in its infancy due to the high cost,
host gene-feeding interactions. lack of large phenotype data collection, and challenges in sample
collection as well as the complex causal relationship. Broader ap-
3.2. Transcriptomics and nutrigenomics in food animal production plications of animal health transcriptomics may be expected in the
and health near future with the dropping cost of NGS technologies and suc-
cessful experience from human studies.
3.2.1. Transcriptomics
As mentioned above, having the greatest genetic potential does 3.2.2. Nutrigenomics
not guarantee the best performance of the animals since the As mentioned in section 3.1, diet can influence the physiological
function of genes can be affected by many factors, with the tran- status of the host, which could be caused by the altered gene
scription process from DNA to mRNA being one of the first deter- expression at the cellular, tissue and organ levels. To study such
minant factors. The emerging studies of RNA molecules (also changes in response to diet, nutrigenomics has been widely
defined as transcriptomics) can therefore provide valuable infor- accepted as a concept for investigating the effects of feed or nu-
mation on whether “important/key genes” are being “transcribed trients on gene expression, which provides understanding on how
or not”. As one of the outcomes of transcriptomic studies, differ- nutrition affects metabolic pathways and diet-gene interactions
ential expressed (DE) gene analysis is used to elucidate the changes [59]. Research on nutrigenomics provide the promise to uncover
in biological functions and metabolic pathways that are related to key biological responses to nutrients in food animal species,
production trait variation. Using feed efficiency in cattle as an especially when considering the dietary supplementations. Recent
example, this trait is complex, and its biology has been proposed research based on a microarray revealed how Zn supplementation
but remains largely unidentified [51]. A recent study identified 70 altered the expression of genes in jugular venous blood of lactating
and 19 total genes that were differentially expressed in the liver of sheep, which is related to immunity and cardiac contraction pat-
Holstein and Jersey dairy cows with different feed efficiency, terns [60]. Similarly, the dietary nutritional level can alter the genes
H.-Z. Sun, L.L. Guan / Trends in Analytical Chemistry 107 (2018) 130e141 135

involved in spermatogenesis or apoptosis in testis of sheep, thus miRNA genes were identified, and the cattle had the most poly-
providing a cellular and molecular understanding of the testis morphic miRNA genes, followed by human, fruit fly, mouse,
response to nutrition and potential nutrition strategies to improve chicken, pig, horse, and sheep (ranged from 3.8% in tetraodon to
reproduction [61]. Using nutrigenomics, the impact of dietary 91.7% in cattle) [65].
manipulations on the immune system in fish is becoming clearer We summarized the miRNAs reported to be associated with
[62]. Nutrigenomics make it possible to formulate individualized nitrogen efficiency, heat stress, early pregnancy, lipid metabolism
diet from alternative feed resources, to further develop precision immune response, fat depot and so on in food animals, and this
feeding strategies in terms of productivity enhancement and/or to summary also showed the predicted targets of certain miRNAs and
prevent diet-induced diseases. In summary, the transcriptomics their specificity to different tissues and species in Table 3. Such
and nutrigenomics studies can help develop the targeted diet findings highlight the potential regulatory correlations between
modifications for highly efficient food animal production and low miRNAs and different phenotypes. Our lab is among one of the first
diet-related disease risk in the defined farm population. The food to study bovine miRNAs and has established methods for studying
animal-based nutrigenomics has just started and focused on only bovine tissue [66] and circulating miRNAs [67] using the micro-
limited number of animals, so it lacks specific, convincing evidence array and HiSeq 2000 system, respectively. We have identified
about diet-gene interactions. There is still a long way to make real miRNAs from many tissues and body fluids (whole blood, serum,
personalized nutrition using food animal nutrigenomics even with and milk) and have discovered bovine miRNAs that are associated
high hope. with dairy cow nitrogen utilization efficiency, gut tissue develop-
ment, fat adipogenesis and foodborne pathogen shedding in pigs
3.3. MicroRNAomes in food animal production and health [68e70]. One of our recent efforts revealed the features of miRNAs
in bovine sera and exosomes and found that miRNAs from sera may
MicroRNAs (miRNAs) have been identified as important post- be preferable for detecting inflammation while miRNAs from exo-
transcriptional regulators of gene expression and have been somes should be better for monitoring muscle development and
proven to affect stress, tissue development, and reproduction in lipid metabolism status in cattle [67]. In addition, other studies
food animal species [63]. To date, most of the studies on miRNA have shown that the circulating miRNAs from plasma and serum
discovery are based on RNA-Seq, although miRNA microarrays can can be used as informative biomarkers for food animal health and
also be effective for screening the known miRNAs. The currently well-being (also summarized in Table 3), such as miR-26a for cattle
known miRNA genes in animals including human, orangutan, early pregnancy [71] and miR-19a and miR-19b for cattle heat stress
cattle, horse, pig, sheep, dog, mouse, rat, chicken, platypus, [72]. The miR-3570 could target and inhibit the expression of the
zebrafish, tetraodon, zebra finch and fruit fly were 1881, 642, 808, mitochondrial antiviral signaling protein, thereby inhibiting NF-kB
715, 383, 106, 502, 1193, 495, 740, 396, 346, 132, 247 and 256, and IRF3 signaling-mediated immunity in teleost fish [73], which
respectively [64]. Similar to coding genes, the genetic variability of was identified as a novel mechanism for virus evasion in fish. The

Table 3
The information of different phenotypes related microRNAs in feedomics.

Name Predicted targetsa Tissues Traits Animals

miR-99b ST6GALNAC4, HS3ST2, KBTBD8 Rumen Nitrogen efficiency Holstein cows [70]
miR-2336 C6ORF50, COLCA2, CD28, DOK4, LIN52, EVA1B, MATR3 Duodenum
miR-652 ISL1 Jejunum
miR-1 GJA1, POGK, ARCN1, SH3BGRL3, PGRMC1, TAGLN2, Liver
TMSB4X, SLC44A1, TWF1
miR-181a ZNF781, ZNF283, ZNF788, ZNF791, ZNF136, ZNF594, Mammary gland
ZNF788, ZNF302, ZFP30, RBAK, CTD-2228K2.5, ZNF268, Serum Heat stress Holstein cows [72]
ZFP82, ZNF527, CTD-2140B24.4, ZNF140, CDON, UBN2,
ZNF470, ZNF256, ZNF23
miR-26a STRADB, CHORDC1, KLHL42, SLC25A16, HMGA2, TET2, Blood Early pregnancy Holstein-Friesian heifers [71]
MAB21L1, TAF13, PIH1D3, CKS2, OSBPL11, THAP2, CREBZF, UBN2
miR-19a KBTBD8, ZMYND11, EXOC6B, C11orf96, FOXD4L2, UBL3, ENPP5, Serum Heat stress Holstein cows [72]
miR-19b FGF6, CNOT6, CHIC1, PMEPA1, SGK1, SLC26A7, MDM4, LONRF1,
ADRB1, RAP2C, PIK3CB, MPPED2, PTEN, HBP1, PFN2
miR-27b GORASP1, KIAA1109, AKIRIN1, NOVA1, ONECUT2, FAM65B, EBI3,
PLK2, NGFRAP1, HOXA5, CCNC
miR-30a-5p TNXB, AC002451.1, MKRN3, EED, DLGAP1, B3GNT5, LHX8, INO80D,
RP11-160N1.10, ANKRA2, KLHL20, STX2, PIP4K2A, SRSF7, DESI2
miR-181b Same as miR-181a
miR-345-3p DAZ4, C4orf6, ZNF616, ZNF189, PRR26, ATF7IP2, LINC00998, HMGCS1
miR-1246 CDR1as, TMPRSS11A, GSG1L, FUT9, ZNF23, ORC6, AL355390.1, LCE2D
miR-196 HOXC8, HOXA7, HOXB8, RP1-170O19.20, NR6A1, HOXA9, HMGA2, Adipose tissue Fat depot Hereford  Aberdeen Angus [66]
HOXB7, HAND1
miR-2454 e
miR-33a e Lipid metabolism Beef cattle [122]
miR-1281 HIST2H3A, AL592284.1, C22orf46, SLC11A1, GAB2, LTB, MSANTD1, LIF,
TEX22, CTD-3214H19.16, FAM98A, ADAMTS13
miR-214 ANKRD65 Blood Immune responses to Pig [69]
miR-331-3p TSPAN18, ZNF513 Salmonella infection
miR-3570 MAVS Cell line Immune Response to Teleost fish [73]
Rhabdovirus
miR-378 e Cell line Osteoblast differentiation [122]
a
Predicted targets, only selected the target gene with cumulative weighted contextþþ score less than 0.7. The target genes of selected miRNAs were commonly predicted
by TargetScan Relase 6.0 (http://www.targetscan.org/) [123] and miRanda (http://www.microrna.org/microrna/home.do).
136 H.-Z. Sun, L.L. Guan / Trends in Analytical Chemistry 107 (2018) 130e141

current progress in the miRNAome area has provided influential during dietary changes, it is speculated that different feeds could
evidence of miRNAs associated with various diseases and the po- lead to shifts in the methylation pattern, which could be related to
tential biomarkers applied for disease prevention and diagnosis. phenotypic changes. Studies are increasingly starting to reveal the
These miRNAs offer potential roles in assisting industry with evidence of epigenetic-regulated gene expression under various
management strategies to improve cattle production, health, and environmental conditions, but most of them have focused only on
welfare. DNA methylation. Studies on other epigenetic mechanisms such as
In addition, many miRNAs have been identified from crops and histone modification and how to determine the heritability of
observed in some public animal miRNA datasets; for example, one epigenetic-driven traits are also needed. Specifically, how the feed
study verified that miR-168a in rice can bind to the low-density composition could alter the epigenomes and whether the altered
lipoprotein receptor adapter protein 1 mRNA of human/mouse, epigenetic variations can be passed to the next generation are
with the inhibited expression of this mRNA in liver and a decrease largely unknown. For meat animals, the epigenetics should more
in low-density lipoprotein removal from mouse plasma based on contribute to the unexplained phenotypic variation in food animal
in vitro and in vivo experiments [74]. Such findings significantly production traits, but for milk production animals, the epigenetic
extend our understanding of the role of miRNAs from food/feed on variations that persist throughout life and be transmitted to the
animals. For food animals, the feed miRNAs may serve as “bioac- next generation are worth being studied well. Therefore, feed se-
tive” components that can pass through the food animal GIT, enter lection or management strategies should take the epigenetic marks
the circulatory system, and regulate the expression of target genes into consideration, which is needed to achieve the targeted out-
in different various organs. This suggests that developing miRNA- comes for growth, physiology and performance.
based feed additives may be a lighting direction for the future
biotechnology and food animal commodity to alter animal gene 3.5. Proteomics in food animal production, product quality, and
expression for improving productivity and health. In ruminants, the health
feed miRNAs may be degraded in the rumen, but they may be able
to escape the destructive conditions in monogastric animals. To The number of proteomics and metabolomics research in food
date, no research has been done to see whether feed-derived animal species has rapidly increased, indicating another advanced
miRNAs can enter and regulate the physiological functions in research era for furthering our understanding of animal biology.
food animal conditions, which has a great potential for developing Based on the HPLC/ESI-MS proteomics method, cow milk adulter-
miRNAs into a novel nutrient component. ation in goat milk was quantified [85], and LC-MS/MS-based whey
proteomics revealed that the milk quality was genetically and
3.4. Epigenomics in food animal phenotypic variations biologically diverse among ruminants. The milk could be grouped
into three clusters: (1) cow, buffalo, and yak milk; (2) goat, cow,
Epigenetic regulation is another factor that can affect gene buffalo, and yak milk; and (3) camel milk [86]. It is known that
expression that may alter the genetic potential. As part of the different feeding strategies are applied for milk-producing rumi-
epigenetic machinery, DNA methylation-driven epigenetic changes nants, which contribute many of the components in the milk. This
in response to dietary variation have been largely observed in study provided information on nutritional values for human intake
different food animal species. For example, the variation trend of could also help to develop better strategies in the non-cow rumi-
the DNA methylation level in promoter and exon 1 of CYP19A1A and nants for productivity and milk quality. In addition, the proteomes
coding region of FOXL2 was negatively associated with their have been reported to be associated with meat quality in beef, pork,
expression levels during ovarian development in Japanese flounder and chicken, such as beef muscle color, meat tenderness, fresh,
using the methylated DNA kit [75], which clarified the functions of lean-to-fat ratio, intramuscular fat content and quality in different
these 2 genes in ovary development from an epigenetic perspec- breeds using the iTRAQ labeling-based proteomics [87]. The dietary
tive. The changed gene expression and methylation of non- yeast cell wall extracts caused a change in the proteomic profile of
structural maintenance of chromosomes subunits of condensin I Atlantic salmon skin mucus, and the calreticulin-like protein was
in the liver and skeletal muscle of offspring was observed using identified as a putative biomarker for yeast-derived functional
Bisulfite sequencing (Bisulfite-Seq) with QIAGEN sequencing ser- feeds using LC-MS/MS-based proteomics [88]. Meanwhile, the
vices when high and low feed protein level applied in the pregnant targeted feeding and management proteomics have shown positive
pig diet, which indicated an involvement of DNA methylation by outcomes to improve the water-holding capacity in pork loin meat
maternal protein supply [76]. It has also been reported that DNA (using feed additive) and meat quality in swine production [89],
methylation differences had a highly significant effect on muscle lamb meat tenderness studied by the HPLC-Q-TOF/MS [90], meat
fiber density and drip loss in a Chinese native chicken breed [77]. In quality and yield, color, and oxidative stability, and woody broiler
addition, the DNA methylation of SMAD1, TSC1, and AKT1 genes breast in poultry production identified by the ESI-Q-TOF/MS [91].
showed significant differences across different sheep breeds, and 6 These results suggest that proteomics is a powerful tool that can
CpG islands were significantly correlated with RNA expression and guide the production of more customer-traits products. Although
body size using the methylated DNA immunoprecipitation- many studies have accelerated proteomics research in food animal
sequencing (MeDIP-Seq) with the HiSeq 2000 system [78]. More- product traits, their linkage to feed biochemistry, genomics, and
over, Genome-wide DNA methylation profiles have been reported microbiomics should be investigate urgently, especially the mech-
in bovine muscle tissue [79] and placentas [80] using the MeDIP- anism of certain nutrients on production-related biology and
Seq with the HiSeq 2000 system; pig neocortex, spleen, liver, nutrient pathways from feed to product at the molecular level.
femoral muscle, olfactory epithelium and pulmonary alveolar Proteomics can also be a valuable tool for evaluating food animal
macrophages cell lines [81]; sheep muscle [82] using the reduced welfare and coping with unavoidable stress challenges. It is re-
representation Bisulfite-Seq with the HiSeq 2000 analyzer; and ported that the proteome changed in normal milk exosomes, milk
chicken pectoral muscle tissues [77], retina, cornea and brain using fat globule membranes and whey proteomes from cows infected
the Bisulfite-Seq with the HiSeq 2500 platform [83]. Moreover, it with mastitis using iTRAQ labeling-based proteomics [92]. Further,
has been suggested that DNA methylation may play a regulatory a combined omics study with proteomics, transcriptomics, and
role in silencing of gene expression related to milk protein in the metabolomics showed that the carbohydrate metabolism and
dairy cow [84]. Although such relationships have not been studied cytoskeleton stabilization cellular mechanisms in muscle tissue
H.-Z. Sun, L.L. Guan / Trends in Analytical Chemistry 107 (2018) 130e141 137

were affected by the restraint and transport stress in chickens [93], of nutrients partition, specific function, output analyzed by
suggesting that omics can be applied for the potential diagnosis of Electrophoresis-TOF/MS [103], and metabolism evaluation (insulin
animal health. Based on these findings, food animal proteomics has neutralization and fasting in chicken [104] and potential metabolic
revealed the physiological, pathological, and putative diagnostic disorder of cattle using NMR, GC-MS and direct flow injection-MS/
information through hundreds or even thousands of proteins under MS [105]).
different diets and environmental and nutritional statuses. This Since the metabolites are located at the end of biological path-
information can therefore help optimize feed at both the quanti- ways, transfer the information of DNA, RNA, and protein and reflect
tative and qualitative levels to improve an animal's metabolic ac- the real metabolic profiling under internal and external stimulates,
tivity to produce high quantity and desirable quality meat and milk metabolomics generates lots of internal phenotypes (also called
and to balance animal production and animal health and welfare. “metabotype”), which can link well to external phenotypes [106].
Accumulating evidence identified by NMR suggests that metab-
3.6. Metabolomics to drive sustainable food animal production otypes can benefit the sustainable food animal industry in many
aspects. For instance, plasma metabolites are associated with var-
We summarized the study level, type, and topic in the applica- iations in production traits in beef cattle and varied abundance of
tion of metabolomics on food animals in Fig. 1 to help select the creatine, hippurate and carnitine accounted for 32% of the pheno-
suitable platform when performing food animal metabolomics typic variation in RFI [107]. The accuracy of prediction using these
studies. The food animal untargeted and targeted metabolomics are metabolites associated with feed efficiency was 95%, which
typically classified into overall and partial levels based on the strongly indicated plasma metabolites could be used as markers for
sample characteristics, which represent the comprehensive meta- selection for these traits [107]. In addition, Melzer et al. proposed
bolism profiles of the whole organism and unique metabolism that metabolites can be used to predict milk traits based on their
patterns of a certain tissue or organ, respectively (Fig. 1). Blood (as research on the 190 milk metabolites identified from 1305 Holstein
well as plasma and serum) and urine metabolomics in food animals cows over 18 commercial farms using GC-MS [108]. In addition, the
are widely assessed to address well-being (beef cattle welfare, rumen fluid metabolomics showed a high association between
hormone abuse in cattle, dairy cow insulin resistance with cinna- rumen metabolites and feed efficiency on crossbreed steers using
mon using GC-MS and UPLC-MS/MS [94], asphyxia, resuscitation UPLC-Q-TOF-MS/MS [109]. Moreover, Sharifi et al. detected signif-
and weaning stress in pig using NMR [95]), health (dairy cows icant associations between egg yolk metabolites and hatchability
hepatic lipidosis, ketosis, milk fever, displaced abomasum, lame- traits in laying hens using GC-TOF/MS [110]. Using direct analysis in
ness using LC-MS/MS [96], cold stress response in the discus fish real time-TOF/MS-based metabolomics, the differentiation of
using GC-TOF/MS [97]), comprehensive metabolism with GC-TOF/ common carp muscle in response to dietary supplementation was
MS [98], and biomarker (infeed antibiotics in piglets using GC-MS feasible [111]. These findings suggest that metabolomics is a
[99], forage type induced different milk protein productions using powerful tool for evaluating diet on food animal health and product
GC-TOF/MS [100]) related problems. Digesta, fecal and saliva quality, which would promote sustainable food animal production
metabolomics have also been studied, which are assigned to the and reduce animal welfare concerns. However, the metabolites for
partial level (Fig. 1) and provide information about feed utilization, food animal species lack a known coverage database. Dr. Wishart's
nutrient digest and interface with the microbiome (rumen micro- lab at the University of Alberta has built an open-access and
biome in goat using GC-TOF/MS [101] and hindgut microbiome in comprehensive livestock metabolome database (LMDB, http://
pig using GC-MS [102]). The tissue (major about liver, muscle, ad- www.lmdb.ca) with 1070 metabolites from different tissues and
ipose, mammary gland, rumen, and gut), milk and single cell biofluids of 5 major livestock species (bovine, ovine, caprine,
metabolomics also belong to a partial level, with the research topic equine, and porcine) in 149 peer-reviewed papers, which could

Fig. 1. Study level, type, and topic in the application of metabolomics in food animal.
138 H.-Z. Sun, L.L. Guan / Trends in Analytical Chemistry 107 (2018) 130e141

serve as a hub for food animal researchers and the food animal 3.8. Microbiomics with metagenomics and metatranscriptomics in
industry to further advance the field of food animal metabolomics food animal production
[106]. This is currently the only database for the food animal
metabolome, but it only compiles data from published papers Tremendous research has shown that symbiotic and commensal
without validation or in-depth investigation. microbiota play vital roles in feed digestion and in shaping host
functions. Food animal microbes play important roles in extracting
3.7. Proteomics and metabolomics to understand the utilization of nutrients and energy harvesting from the feed, regulating methane
alternative feed source emissions, relating inflammatory responses, affecting fat storage,
and promoting gut epithelium renewal, as revealed by the pyro-
Proteomics and metabolomics were also successfully applied sequencing using the HiSeq 2500 system [115]. For this review, we
in the mechanical characterization of alternative feed source. Crop use the rumen microbiome as an example to highlight the recent
by-products is one type of alternative feed sources, especially for research on its role in cattle feed efficiency and methane emission.
ruminants. Enhancing the utilization of crop by-products to pro- Ruminants harbor a complex rumen microbiome that can convert
duce high-quality food animal product is a promising direction to human indigestible feed materials into volatile fatty acids and other
achieve sustainable food animal production. Corn stover (CS) and nutrients so the host can produce high-quality protein meat and
rice straw (RS), as the most abundant crop by-products, can milk. The recent metagenomics research on rumen microbiome of
significantly lower both the milk production and milk protein dairy cows revealed that microbial diversity, microbial taxa, and
content when replacing 23% of alfalfa hay (AH, current practice in genes are associated with different feed efficiency using HiSeq
the modern dairy farm) in dairy cow diets [112]. The rumen fluid, 2500 and MiSeq systems [116]. Similarly, the rumen metatran-
serum, milk and urine metabolomics showed that the gluconeo- scriptomic profiling of beef cattle rumen showed active microbial
genesis, pyruvate metabolism, tricarboxylic acid cycle, glycer- taxa and functions associated with feed efficiency beef cattle using
olipid metabolism, and aspartate metabolism pathways were the the HiSeq 2000 system [117]. Based on rumen metagenomics and
most enriched in AH [98]; CS-fed cows had significantly down- metatranscriptomics, methanogenesis pathway transcription pro-
regulated glycine, serine, and threonine metabolism, tyrosine files were identified to be correlated with methane yields in sheep
metabolism, and phenylalanine metabolism compared with AH- [118], and Kamke et al. further revealed that low-methane-yield
fed cows [113]; and hippuric acid and N-methyl-glutamic in the sheep had a Sharpea-enriched microbiome with a more lactic acid
urine were further identified as potential biomarkers for formation and utilization using rumen identified with meta-
discriminating CS and AH diets [100]. The proteome results genomics and metatranscriptomics using the HiSeq 2000 system
showed 231 up-regulated and 286 down-regulated proteins and [119]. These findings provide a foundation for developing new
inhibited energy and fatty acid metabolism in the mammary nutritional and management strategies to mitigate methane
gland tissue in the RS group [114]. The above information pro- emission and improve feed efficiency by manipulating the micro-
vided a new understanding of and applicable candidates for the biota composition and functions. In other food animal species,
mechanisms of crop by-product utilization. However, the lack of metagenomics analysis showed that microbiota and functional
interactions with feed metabolomics and proteomics is the com- capacities in the cecum have also been reported to be associated
mon limitation of these studies. with feed efficiency in finishing pigs identified by HiSeq 2500

Fig. 2. Future directions of feedomics. The four compositions of feedomics (feed, food animal, products, and human) were illustrated with grey color. The grey arrow represents the
currently studied contents in feedomics. The blue arrow represents the future research directions in feedomics. The corresponding targets and omics tools are also described in the
figure.
H.-Z. Sun, L.L. Guan / Trends in Analytical Chemistry 107 (2018) 130e141 139

platform [120] and in chickens using the MiSeq system [121], Acknowledgments
suggesting that the regulation of microorganism composition could
be applied to the pig production industry. The above ecological and This work was supported by China Opportunity Fund (spon-
mechanistic understanding of the GIT microbiome could lead to an sored by University of Alberta, RES0031665) and NSERC Discovery
increase in productivity and an environmentally friendly food an- grant.
imal industry. The limitations of these studies are that they do not
consider the potential roles and interactions with feed and envi-
ronment microbiomes. References

[1] W. Lutz, W.P. Butz, K.E. Samir, World Population & Human Capital in the
4. Conclusions and future outlooks Twenty-first Century: an Overview, Oxford University Press, 2017.
[2] L. Bouwman, K.K. Goldewijk, K.W. Van Der Hoek, A.H. Beusen, D.P. Van
Vuuren, J. Willems, M.C. Rufino, E. Stehfest, Exploring global changes in ni-
In this paper, we proposed the concept of “feedomics” in feed
trogen and phosphorus cycles in agriculture induced by livestock production
science and animal nutrition area and provided a detailed review over the 1900e2050 period, P. Natl. Acad. Scd. USA 110 (2013)
of the recent studies applying genomics, epigenomics, tran- 20882e20887.
scriptomics, proteomics, metabolomics, metagenomics and met- [3] A. Mottet, C. de Haan, A. Falcucci, G. Tempio, C. Opio, P. Gerber, Livestock: on
our plates or eating at our table? A new analysis of the feed/food debate,
atranscriptomics strategies to study the production and health Glob. Food Secur. 14 (2017) 1e8.
issues of food animals. Feedomics provides a new understanding [4] D. Tilman, K.G. Cassman, P.A. Matson, R. Naylor, S. Polasky, Agricultural
of the complex biochemical mechanisms involved in the inter- sustainability and intensive production practices, Nature 418 (2002) 671.
[5] C. Phillips, Animal Welfare, Nutrition, and Economics: Past, Present, and
action of diet, environment, host physiology, microbial functions Future, Nutrition and the Welfare of Farm Animals, Springer, 2016,
and phenotypes and has the promise to be well developed into a pp. 229e240.
targeted method for improving the quantity, quality, safety and [6] C. Li, W. Salas, R. Zhang, C. Krauter, A. Rotz, F. Mitloehner, Manure-DNDC: a
biogeochemical process model for quantifying greenhouse gas and ammonia
functional properties of food animal products while reducing the emissions from livestock manure systems, Nutr. Cycl. Agroecosys. 93 (2012)
use of antimicrobial growth promotants and mitigating green- 163e200.
house gas emissions from the food animal sectors using regular [7] T.P. Van Boeckel, E.E. Glennon, D. Chen, M. Gilbert, T.P. Robinson,
B.T. Grenfell, S.A. Levin, S. Bonhoeffer, R. Laxminarayan, Reducing antimi-
or alternative feed resource or novel feed additives (e.g., miRNAs) crobial use in food animals, Science 357 (2017) 1350e1352.
to meet the increasing human consumption demand in the [8] A. Cifuentes, Food analysis and foodomics, J. Chromatogr. A 1216 (2009)
future. These omics-based strategies provide important insights 7109.
[9] S.K. Kar, FeedOmics, an Approach to Evaluate the Functional Properties of
into animal biological information and omics-based nutrition,
Protein Containing Feed Ingredients, Wageningen University, 2017.
which are far beyond but also tightly associated with the mea- [10] S. Goodwin, J.D. McPherson, W.R. McCombie, Coming of age: ten years of
surement results from traditional techniques. The crucial power next-generation sequencing technologies, Nat. Rev. Genet. 17 (2016)
of these omics methods is revealing true causal internal pheno- 333e351.
[11] B. Tasic, V. Menon, T.N. Nguyen, T.K. Kim, T. Jarsky, Z. Yao, B. Levi, L.T. Gray,
types (gene, transcripts, protein, and metabolites), metabolic S.A. Sorensen, T. Dolbeare, Adult mouse cortical cell taxonomy by single cell
functions, regulatory pathways leading to improved feed effi- transcriptomics, Nat. Neurosci. 19 (2016) 335.
ciency, well animal welfare, and sustainable development in the [12] E.P. Consortium, An integrated encyclopedia of DNA elements in the human
genome, Nature 489 (2012) 57e74.
food animal industry. The outcomes of omics should be applied [13] Y. Hasin, M. Seldin, A. Lusis, Multi-omics approaches to disease, Genome Biol.
in nutritional regulation, the breeding program (especially for 18 (2017) 83.
the local breeds in extremely regions) and management [14] E.M. Ibeagha-Awemu, X. Zhao, Epigenetic marks: regulators of livestock
phenotypes and conceivable sources of missing variation in livestock
improvement practice, which would be great benefit to animals improvement programs, Front. Genet. 6 (2015).
in the future. [15] M. Larance, A.I. Lamond, Multidimensional proteomics for cell biology, Nat.
Except for the currently studied contents of improving food Rev. Mol. Cell Biol. 16 (2015) 269.
[16] C.H. Johnson, J. Ivanisevic, G. Siuzdak, Metabolomics: beyond biomarkers and
animal sectors’ efficiency and sustainability from input (feed) to
towards mechanisms, Nat. Rev. Mol. Cell Biol. 17 (2016) 451e459.
output (product) in feedomics, several other directions such as [17] C. Quince, A.W. Walker, J.T. Simpson, N.J. Loman, N. Segata, Shotgun meta-
feed quality assessment, interactions with plant omics and genomics, from sampling to sequencing and analysis, Nat. Biotechnol 35
(2017) 833e844.
biochemistry, food animal impact on human health and future
[18] L. Moitinho-Silva, C. Díez-Vives, G. Batani, A.I. Esteves, M.T. Jahn, T. Thomas,
customer-traits evaluation, should be paid attention to in future Integrated metabolism in spongeemicrobe symbiosis revealed by genome-
work (Fig. 2). Using plant genomics, proteomics, metabolomics centered metatranscriptomics, ISME J 11 (2017) 1651e1666.
and miRNAs to develop a species-specific flavor feed, contami- [19] J.A. Reuter, D.V. Spacek, M.P. Snyder, High-throughput sequencing technol-
ogies, Mol. Cell. 58 (2015) 586e597.
nation reduction feed, and less-emission (methane, CO2) feed [20] L. Andersson, A.L. Archibald, C.D. Bottema, R. Brauning, S.C. Burgess,
source will be novel investigations in the feed quality assessment D.W. Burt, E. Casas, H.H. Cheng, L. Clarke, C. Couldrey, Coordinated inter-
of feedomics (Fig. 2). Feedomics could also be a powerful tool to national action to accelerate genome-to-phenome with FAANG, the
functional annotation of animal genomes project, Genome Biol. 16 (2015)
reveal the underlying mechanism of food animal on human 57.
health, such as antimicrobial resistance and zoonosis. Mean- [21] M. Zampieri, K. Sekar, N. Zamboni, U. Sauer, Frontiers of high-throughput
while, evaluating future customer traits such as being rich in metabolomics, Curr. Opin. Chem. Biol. 36 (2017) 15e23.
[22] X. Zhan, Y. Long, M. Lu, Exploration of variations in proteome and metab-
natural healthy molecules and live ingredients will require olome for predictive diagnostics and personalized treatment algorithms:
tremendous efforts of feedomics, especially by proteomics, innovative approach and examples for potential clinical application,
metabolomics, and microbiomics. The action of different levels of J. Proteomics (2017). https://doi.org/10.1016/j.jprot.2017.08.020 in press.
[23] A. Almeida, A. Bassols, E. Bendixen, M. Bhide, F. Ceciliani, S. Cristobal,
biomolecules (e.g., DNA, mRNA, miRNA, protein, lipid, metabo- P. Eckersall, K. Hollung, F. Lisacek, G. Mazzucchelli, Animal board invited
lite) will play a vital role and provide clues for feed, food animal, review: advances in proteomics for animal and food sciences, Animal 9
product and human in future feedomics studies. The integrated (2015) 1e17.
[24] R. Aebersold, M. Mann, Mass spectrometry-based proteomics, Nature 422
application of multi-omics, especially for the entire pool of
(2003) 198.
different levels of biomolecules, to understand the information [25] L. Monaci, A. Visconti, Mass spectrometry-based proteomics methods for
flow will be a new trend in feedomics. Feedomics is still in its analysis of food allergens, TrAC e Trend. Anal. Chem. 28 (2009) 581e591.
infancy period, so further enhancing its analytical sensitivity, [26] S. Kim, J. Kim, E.J. Yun, K.H. Kim, Food metabolomics: from farm to human,
Curr. Opin. Biotechnol. 37 (2016) 16e23.
accuracy, coverage depth and matched databases is still a big [27] J.F. Xiao, B. Zhou, H.W. Ressom, Metabolite identification and quantitation in
challenge for future work. LC-MS/MS-based metabolomics, TrAC e Trend. Anal. Chem. 32 (2012) 1e14.
140 H.-Z. Sun, L.L. Guan / Trends in Analytical Chemistry 107 (2018) 130e141

[28] G.P. Danezis, A.S. Tsagkaris, F. Camin, V. Brusic, C.A. Georgiou, Food [54] R. Xiang, V.H. Oddy, A.L. Archibald, P.E. Vercoe, B.P. Dalrymple, Epithelial,
authentication: techniques, trends & emerging approaches, TrAC e Trend. metabolic and innate immunity transcriptomic signatures differentiating the
Anal. Chem. 85 (2016) 123e132. rumen from other sheep and mammalian gastrointestinal tract tissues, PeerJ
[29] M. Vinaixa, E.L. Schymanski, S. Neumann, M. Navarro, R.M. Salek, O. Yanes, 4 (2016) e1762.
Mass spectral databases for LC/MS-and GC/MS-based metabolomics: state of [55] M. Ye, H. Bao, Y. Meng, L. Guan, P. Stothard, G. Plastow, Comparative tran-
the field and future prospects, TrAC e Trend. Anal. Chem. 78 (2016) 23e35. scriptomic analysis of porcine peripheral blood reveals differentially
[30] A.-C. Syv€ anen, Toward genome-wide SNP genotyping, Nat. Genet. 37 (2005) expressed genes from the cytokineecytokine receptor interaction pathway
S5eS10. related to health status, Genome 60 (12) (2017) 1e8.
[31] G. Gorjanc, J.-F. Dumasy, S. Gonen, R.C. Gaynor, R. Antolin, J.M. Hickey, Potential [56] A. Kommadath, H. Bao, A.S. Arantes, G.S. Plastow, C.K. Tuggle, S.M. Bearson,
of low-coverage genotyping-by-sequencing and imputation for cost-effective P. Stothard, Gene co-expression network analysis identifies porcine genes
genomic selection in biparental segregating populations, Crop Sci. 57 (2017) associated with variation in Salmonella shedding, BMC Genom. 15 (2014)
1404e1420. 452.
[32] C.G. Elsik, R.L. Tellam, K.C. Worley, The genome sequence of taurine cattle: a [57] B.D. Glencross, C. De Santis, B. Bicskei, J.B. Taggart, J.E. Bron, M.B. Betancor,
window to ruminant biology and evolution, Science 324 (2009) 522e528. D.R. Tocher, A comparative analysis of the response of the hepatic tran-
[33] C.-J. Rubin, M.C. Zody, J. Eriksson, J.R. Meadows, E. Sherwood, M.T. Webster, scriptome to dietary docosahexaenoic acid in Atlantic salmon (Salmo salar)
L. Jiang, M. Ingman, T. Sharpe, S. Ka, Whole-genome resequencing reveals post-smolts, BMC Genom. 16 (2015) 684.
loci under selection during chicken domestication, Nature 464 (2010) [58] S.T. Burgess, A. Greer, D. Frew, B. Wells, E.J. Marr, A.J. Nisbet, J.F. Huntley,
587e591. Transcriptomic analysis of circulating leukocytes reveals novel aspects of the
[34] A. Archibald, N. Cockett, B. Dalrymple, T. Faraut, J. Kijas, J. Maddox, host systemic inflammatory response to sheep scab mites, PLoS One 7 (2012)
J. McEwan, V. Hutton Oddy, H. Raadsma, C. Wade, The sheep genome e42778.
reference sequence: a work in progress, Anim. Genet. 41 (2010) 449e453. [59] M. Kussmann, P.J. Stover, Nutrigenomics and Proteomics in Health and
[35] M.A. Groenen, A.L. Archibald, H. Uenishi, C.K. Tuggle, Y. Takeuchi, Disease: towards a Systems-level Understanding of Gene-diet Interactions,
M.F. Rothschild, C. Rogel-Gaillard, C. Park, D. Milan, H.-J. Megens, Analyses of John Wiley & Sons, 2017.
pig genomes provide insight into porcine demography and evolution, Nature [60] R. Elgendy, F. Palazzo, F. Castellani, M. Giantin, L. Grotta, L. Cerretani,
491 (2012) 393e398. M. Dacasto, G. Martino, Transcriptome profiling and functional analysis of
[36] P.M. Rodrigues, S.A. Martin, T.S. Silva, S. Boonanuntanasarn, D. Schrama, sheep fed with high zinc-supplemented diet: a nutrigenomic approach,
M. Moreira, C. Raposo, Proteomics in Fish and Aquaculture Research, Pro- Anim. Feed Sci. Technol. 234 (2017) 195e204.
teomics in Domestic Animals: from Farm to Systems Biology, Springer, 2018, [61] Y. Guan, G. Liang, G.B. Martin, Functional changes in mRNA expression and
pp. 311e338. alternative pre-mRNA splicing associated with the effects of nutrition on
[37] B. Karisa, J. Thomson, Z. Wang, P. Stothard, S. Moore, G. Plastow, Candidate apoptosis and spermatogenesis in the adult testis, BMC Genom. 18 (2017)
genes and single nucleotide polymorphisms associated with variation in 64.
residual feed intake in beef cattle, J. Anim. Sci. 91 (2013) 3502e3513. [62] S.A. Martin, E. Kro l, Nutrigenomics and immune function in fish: new in-
[38] M. Rolf, J. Taylor, R. Schnabel, S. McKay, M. McClure, S. Northcutt, M. Kerley, sights from omics technologies, Dev. Comp. Immunol. 75 (2017) 86e98.
R. Weaber, Genome-wide association analysis for feed efficiency in Angus [63] J. Ioannidis, J. Risse, F.X. Donadeu, Profiling of MicroRNAs in the Biofluids of
cattle, Anim. Genet. 43 (2012) 367e374. Livestock Species, MicroRNA Protocols, Springer, 2018, pp. 65e77.
[39] D.N. Do, T. Ostersen, A.B. Strathe, T. Mark, J. Jensen, H.N. Kadarmideen, [64] M. Zorc, D.J. Skok, I. Godnic, G.A. Calin, S. Horvat, Z. Jiang, P. Dovc, T. Kunej,
Genome-wide association and systems genetic analyses of residual feed Catalog of microRNA seed polymorphisms in vertebrates, PLoS One 7 (2012)
intake, daily feed consumption, backfat and weight gain in pigs, BMC Genet. e30737.
15 (2014) 27. [65] M. Zorc, J. Obsteter, P. Dovc, T. Kunej, Genetic variability of microRNA genes
[40] P.S. de Oliveira, A.S. Cesar, M.L. do Nascimento, A.S. Chaves, P.C. Tizioto, in 15 animal species, J. Genom. 3 (2015) 51.
R.R. Tullio, D.P. Lanna, A.N. Rosa, T.S. Sonstegard, G.B. Mourao, Identification [66] J.M. Romao, W. Jin, M. He, T. McAllister, Altered microRNA expression in
of genomic regions associated with feed efficiency in Nelore cattle, BMC bovine subcutaneous and visceral adipose tissues from cattle under different
Genet. 15 (2014) 100. diet, PLoS One 7 (2012) e40605.
[41] D.N. Do, A.B. Strathe, T. Ostersen, S.D. Pant, H.N. Kadarmideen, Genome-wide [67] K. Zhao, G. Liang, X. Sun, Comparative miRNAome analysis revealed different
association and pathway analysis of feed efficiency in pigs reveal candidate miRNA expression profiles in bovine sera and exosomes, BMC Genom. 17
genes and pathways for residual feed intake, Front. Genet. 5 (2014) 307. (2016) 630.
[42] J. Yuan, K. Wang, G. Yi, M. Ma, T. Dou, C. Sun, L.-J. Qu, M. Shen, L. Qu, N. Yang, [68] J.M. Romao, W. Jin, M. He, T. McAllister, MicroRNAs in bovine adipogenesis:
Genome-wide association studies for feed intake and efficiency in two laying genomic context, expression and function, BMC Genom. 15 (2014) 137.
periods of chickens, Genet. Sel. Evol. 47 (2015) 82. [69] H. Bao, A. Kommadath, G. Liang, X. Sun, A.S. Arantes, C.K. Tuggle,
[43] M.-P. Sanchez, A. Govignon-Gion, P. Croiseau, S. Fritz, C. Hoze , G. Miranda, S.M. Bearson, G.S. Plastow, P. Stothard, L.L. Guan, Genome-wide whole blood
P. Martin, A. Barbat-Leterrier, R. Letaïef, D. Rocha, Within-breed and multi- microRNAome and transcriptome analyses reveal miRNA-mRNA regulated
breed GWAS on imputed whole-genome sequence variants reveal candi- host response to foodborne pathogen Salmonella infection in swine, Sci. Rep.
date mutations affecting milk protein composition in dairy cattle, Genet. Sel. 5 (2015).
Evol. 49 (2017) 68. [70] D. Wang, G. Liang, B. Wang, H. Sun, J. Liu, L.L. Guan, Systematic micro-
[44] A. Ribani, Towards the Development of Nutrigenetic Concepts in Pigs by RNAome profiling reveals the roles of microRNAs in milk protein metabolism
Merging Genomics, Transcriptomics and Metabolomics, Alma, 2017. and quality: insights on low-quality forage utilization, Sci. Rep. 6 (2016).
[45] R. Garg, N. Sharma, S.K. Jain, Nutrigenomics and Nutrigenetics: Concepts and [71] J. Ioannidis, F.X. Donadeu, Circulating miRNA signatures of early pregnancy
Applications in Nutrition Research and Practice, Acta Med. Int. 1 (2014) in cattle, BMC Genom. 17 (2016) 184.
124e130. [72] Y. Zheng, K.-l. Chen, X.-m. Zheng, H.-x. Li, G.-l. Wang, Identification and
[46] S. Calsamiglia, A. Siurana, Nutrigenics and nutrigenomics: the potential bioinformatics analysis of microRNAs associated with stress and immune
application to animal nutrition: feed science, AFMA Matrix 25 (2016) response in serum of heat-stressed and normal Holstein cows, Cell Stress
34e35. Chaperon 19 (2014) 973e981.
[47] S. Miyauchi, A. Hirasawa, A. Ichimura, T. Hara, G. Tsujimoto, New frontiers in [73] T. Xu, Q. Chu, J. Cui, D. Bi, Inducible MicroRNA-3570 feedback inhibits the
gut nutrient sensor research: free fatty acid sensing in the gastrointestinal RIG-I-Dependent innate immune response to rhabdovirus in teleost fish by
tract, J. Pharmacol. Sci. 112 (2010) 19e24. targeting MAVS/IPS-1, J. Virol. 92 (2018) e01594-01517.
[48] T. Song, J. Peng, J. Ren, H.-k. Wei, J. Peng, Cloning and characterization of [74] L. Zhang, D. Hou, X. Chen, D. Li, L. Zhu, Y. Zhang, J. Li, Z. Bian, X. Liang,
spliced variants of the porcine G protein coupled receptor 120, BioMed Res. X. Cai, Exogenous plant MIR168a specifically targets mammalian
Int. 2015 (2015). LDLRAP1: evidence of cross-kingdom regulation by microRNA, Cell Res.
[49] L. Fontanesi, F. Bertolini, E. Scotti, G. Schiavo, M. Colombo, P. Trevisi, 22 (2012) 107.
A. Ribani, L. Buttazzoni, V. Russo, S. Dall'Olio, Next generation semi- [75] Y. Si, Y. Ding, F. He, H. Wen, J. Li, J. Zhao, Z. Huang, DNA methylation level of
conductor based-sequencing of a nutrigenetics target gene (GPR120) and cyp19a1a and Foxl2 gene related to their expression patterns and repro-
association with growth rate in Italian Large White pigs, Anim. Biotechnol. duction traits during ovary development stages of Japanese flounder (Para-
26 (2015) 92e97. lichthys olivaceus), Gene 575 (2016) 321e330.
[50] A. Ichimura, A. Hirasawa, O. Poulain-Godefroy, A. Bonnefond, T. Hara, L. Yengo, [76] S. Altmann, E. Murani, M. Schwerin, C.C. Metges, K. Wimmers, S. Ponsuksili,
I. Kimura, A. Leloire, N. Liu, K. Iida, Dysfunction of lipid sensor GPR120 leads to Maternal dietary protein restriction and excess affects offspring gene
obesity in both mouse and human, Nature 483 (2012) 350e354. expression and methylation of non-SMC subunits of condensin I in liver and
[51] D. Berry, J. Crowley, Cell Biology Symposium: genetics of feed efficiency in skeletal muscle, Epigenetics 7 (2012) 239e252.
dairy and beef cattle, J. Anim. Sci. 91 (2013) 1594e1613. [77] Y. Zhang, J. Guo, Y. Gao, S. Niu, C. Yang, C. Bai, X. Yu, Z. Zhao, Genome-wide
[52] M. Salleh, G. Mazzoni, J. Ho € glund, D. Olijhoek, P. Lund, P. Løvendahl, methylation changes are associated with muscle fiber density and drip loss
H. Kadarmideen, RNA-Seq transcriptomics and pathway analyses reveal in male three-yellow chickens, Mol. Biol. Rep. 41 (2014) 3509e3516.
potential regulatory genes and molecular mechanisms in high-and low- [78] J. Cao, C. Wei, D. Liu, H. Wang, M. Wu, Z. Xie, T.D. Capellini, L. Zhang, F. Zhao,
residual feed intake in Nordic dairy cattle, BMC Genom. 18 (2017) 258. L. Li, DNA methylation Landscape of body size variation in sheep, Sci. Rep. 5
[53] R.S. Kong, G. Liang, Y. Chen, P. Stothard, Transcriptome profiling of the rumen (2015).
epithelium of beef cattle differing in residual feed intake, BMC Genom. 17 [79] Y.-Z. Huang, J.-J. Sun, L.-Z. Zhang, C.-J. Li, J.E. Womack, Z.-J. Li, X.-Y. Lan, C.-
(2016) 592. Z. Lei, C.-L. Zhang, X. Zhao, Genome-wide DNA methylation profiles and their
H.-Z. Sun, L.L. Guan / Trends in Analytical Chemistry 107 (2018) 130e141 141

relationships with mRNA and the microRNA transcriptome in bovine muscle microbiota with increasing dietary grain in a goat model, Environ. Microbiol.
tissue (Bos taurine), Sci. Rep. 4 (2014). 18 (2016) 525e541.
[80] J. Su, Y. Wang, X. Xing, J. Liu, Y. Zhang, Genome-wide analysis of DNA [102] L. Zhou, L. Fang, Y. Sun, Y. Su, W. Zhu, Effects of the dietary protein level on
methylation in bovine placentas, BMC Genom. 15 (2014) 12. the microbial composition and metabolomic profile in the hindgut of the pig,
[81] M. Choi, J. Lee, M.T. Le, D.T. Nguyen, S. Park, N. Soundrarajan, Anaerobe 38 (2016) 61e69.
K.M. Schachtschneider, J. Kim, J.-K. Park, J.-H. Kim, Genome-wide analysis of [103] M. El-Sabagh, D. Taniguchi, T. Sugino, T. Obitsu, Metabolomic profiling re-
DNA methylation in pigs using reduced representation bisulfite sequencing, veals differential effects of glucagon-like peptide-1 and insulin on nutrient
DNA Res. 22 (2015) 343e355. partitioning in ovine liver, Anim. Sci. J. 87 (2016) 1480e1489.
[82] C. Couldrey, R. Brauning, J. Bracegirdle, P. Maclean, H.V. Henderson, [104] B. Ji, B. Ernest, J.R. Gooding, S. Das, A.M. Saxton, J. Simon, J. Dupont,
J.C. McEwan, Genome-wide DNA methylation patterns and transcription S. Me tayer-Coustard, S.R. Campagna, B.H. Voy, Transcriptomic and metab-
analysis in sheep muscle, PLoS One 9 (2014) e101853. olomic profiling of chicken adipose tissue in response to insulin neutrali-
[83] I. Lee, B.A. Rasoul, A.S. Holub, A. Lejeune, R.A. Enke, W. Timp, Whole genome zation and fasting, BMC Genom. 13 (2012) 441.
DNA methylation sequencing of the chicken retina, cornea and brain, Sci. [105] F. Saleem, B. Ametaj, S. Bouatra, R. Mandal, Q. Zebeli, S. Dunn, D. Wishart,
Data 4 (2017). A metabolomics approach to uncover the effects of grain diets on rumen
[84] K. Singh, A. Molenaar, K. Swanson, B. Gudex, J. Arias, R. Erdman, health in dairy cows, J. Dairy Sci. 95 (2012) 6606e6623.
K. Stelwagen, Epigenetics: a possible role in acute and transgenerational [106] S.A. Goldansaz, A.C. Guo, T. Sajed, M.A. Steele, G.S. Plastow, D.S. Wishart,
regulation of dairy cow milk production, Animal 6 (2011) 375e381. Livestock metabolomics and the livestock metabolome: a systematic review,
[85] R.K. Chen, L.W. Chang, Y.Y. Chung, M.H. Lee, Y.C. Ling, Quantification of cow PLoS One 12 (2017) e0177675.
milk adulteration in goat milk using high-performance liquid chromatog- [107] B. Karisa, J. Thomson, Z. Wang, C. Li, Y. Montanholi, S. Miller, S. Moore, G. Plastow,
raphy with electrospray ionization mass spectrometry, Rapid Commun. Mass Plasma metabolites associated with residual feed intake and other productivity
Sp. 18 (2004) 1167e1171. performance traits in beef cattle, Livest. Sci. 165 (2014) 200e211.
[86] Y. Yang, D. Bu, X. Zhao, P. Sun, J. Wang, L. Zhou, Proteomic analysis of cow, [108] N. Melzer, D. Wittenburg, S. Hartwig, S. Jakubowski, U. Kesting, L. Willmitzer,
yak, buffalo, goat and camel milk whey proteins: quantitative differential J. Lisec, N. Reinsch, D. Repsilber, Investigating associations between milk
expression patterns, J. Proteome Res. 12 (2013) 1660e1667. metabolite profiles and milk traits of Holstein cows, J. Dairy Sci. 96 (2013)
[87] Y. Mao, D.L. Hopkins, Y. Zhang, P. Li, L. Zhu, P. Dong, R. Liang, J. Dai, X. Wang, 1521e1534.
X. Luo, Beef quality with different intramuscular fat content and proteomic [109] V.M. Artegoitia, A.P. Foote, R.M. Lewis, H.C. Freetly, Rumen fluid metab-
analysis using isobaric tag for relative and absolute quantitation of differ- olomics analysis associated with feed efficiency on crossbred steers, Sci. Rep.
entially expressed proteins, Meat Sci. 118 (2016) 96e102. 7 (2017).
[88] G. Micallef, P. Cash, J.M. Fernandes, B. Rajan, J.W. Tinsley, R. Bickerdike, [110] A. Sharifi, Y. Zhang, S. Weigend, A. Weigend, R. Preisinger, M. Schmutz,
S.A. Martin, A.S. Bowman, Dietary yeast cell wall extract alters the proteome L. Ro€misch-Margl, R. Jonczyk, M. Gülerso €nmez, H. Piepho, Association be-
of the skin mucous barrier in Atlantic Salmon (Salmo salar): increased tween metabolites in egg yolk and hatchability traits in laying hens, in: 10th
abundance and expression of a calreticulin-like protein, PLoS One 12 (2017) World Congress on Genetics Applied to Livestock Production: Proceedings;
e0169075. Vancouver, BC, Canada; August 17e22, 2014, 2014.
[89] R. Delles, A. Naylor, A. Kocher, K. Dawson, R. Samuel, 142 Diets with organic [111] T. Cajka, H. Danhelova, A. Vavrecka, K. Riddellova, V. Kocourek, F. Vacha,
trace minerals (Bioplex®) and yeast Protein (NuPro®) improved the water- J. Hajslova, Evaluation of direct analysis in real time ionizationemass spec-
holding capacity of pork loin meat, J. Anim. Sci. 94 (2016), 66. trometry (DARTeMS) in fish metabolomics aimed to assess the response to
[90] A. della Malva, R. Marino, A. Santillo, G. Annicchiarico, M. Caroprese, A. Sevi, dietary supplementation, Talanta 115 (2013) 263e270.
M. Albenzio, Proteomic approach to investigate the impact of different dietary [112] B. Wang, S. Mao, H. Yang, Y. Wu, J. Wang, S. Li, Z. Shen, J. Liu, Effects of
supplementation on lamb meat tenderness, Meat Sci. 131 (2017) 74e81. alfalfa and cereal straw as a forage source on nutrient digestibility and
[91] A. Zheng, J. Luo, K. Meng, J. Li, S. Zhang, K. Li, G. Liu, H. Cai, W.L. Bryden, lactation performance in lactating dairy cows, J. Dairy Sci. 97 (2014)
B. Yao, Proteome changes underpin improved meat quality and yield of 7706e7715.
chickens (Gallus gallus) fed the probiotic Enterococcus faecium, BMC Genom. [113] H.-Z. Sun, D.-M. Wang, B. Wang, J.-K. Wang, H.-Y. Liu, L.L. Guan, J.-X. Liu,
15 (2014) 1167. Metabolomics of four biofluids from dairy cows: potential biomarkers for
[92] T.A. Reinhardt, R.E. Sacco, B.J. Nonnecke, J.D. Lippolis, Bovine milk proteome: milk production and quality, J. Proteome Res. 14 (2015) 1287e1298.
quantitative changes in normal milk exosomes, milk fat globule membranes [114] W. Dai, Q. Chen, Q. Wang, R.R. White, J. Liu, H. Liu, Complementary transcriptomic
and whey proteomes resulting from Staphylococcus aureus mastitis, and proteomic analyses reveal regulatory mechanisms of milk protein produc-
J. Proteomics 82 (2013) 141e154. tion in dairy cows consuming different forages, Sci. Rep. 7 (2017).
[93] D. Hazard, X. Fernandez, J. Pinguet, C. Chambon, F. Letisse, J.-C. Portais, [115] S. Jiao, H. Cao, Y. Dai, J. Wu, J. Lv, R. Du, B. Han, Effect of high-fat diet and
Z. Wadih-Moussa, H. Re mignon, C. Molette, Functional genomics of the growth stage on the diversity and composition of intestinal microbiota in
muscle response to restraint and transport in chickens, J. Anim. Sci. 89 (2011) healthy bovine livestock, J. Sci. Food Agr. 10 (2017) 2958e2972.
2717e2730. [116] S.K.B. Shabat, G. Sasson, A. Doron-Faigenboim, T. Durman, S. Yaacoby,
[94] J.A. Carrillo, Y. He, Y. Li, J. Liu, R.A. Erdman, T.S. Sonstegard, J. Song, Integrated M.E.B. Miller, B.A. White, N. Shterzer, I. Mizrahi, Specific microbiome-
metabolomic and transcriptome analyses reveal finishing forage affects dependent mechanisms underlie the energy harvest efficiency of rumi-
metabolic pathways related to beef quality and animal welfare, Sci. Rep. 6 nants, ISME J. 10 (2016) 2958e2972.
(2016) 25948. [117] F. Li, Metatranscriptomic profiling reveals linkages between the active
[95] Q. He, H. Tang, P. Ren, X. Kong, G. Wu, Y. Yin, Y. Wang, Dietary supple- rumen microbiome and feed efficiency in beef cattle, Appl. Environ. Micro-
mentation with l-arginine partially counteracts serum metabonome induced biol. 83 (2017) e00061-00017.
by weaning stress in piglets, J. Proteome Res. 10 (2011) 5214e5221. [118] W. Shi, C.D. Moon, S.C. Leahy, D. Kang, J. Froula, S. Kittelmann, C. Fan,
[96] B. Ametaj, G. Zhang, E. Dervishi, S. Dunn, R. Mandal, D. Wishart, 0150 Tar- S. Deutsch, D. Gagic, H. Seedorf, Methane yield phenotypes linked to dif-
geted metabolomics reveals multiple metabolite alterations in the urine of ferential gene expression in the sheep rumen microbiome, Genome Res. 24
transition dairy cows preceding the incidence of lameness, J. Anim. Sci. 94 (2014) 1517e1525.
(2016) 72e73. [119] J. Kamke, S. Kittelmann, P. Soni, Y. Li, M. Tavendale, S. Ganesh, P.H. Janssen, W. Shi,
[97] B. Wen, S.-R. Jin, Z.-Z. Chen, J.-Z. Gao, Physiological responses to cold stress in J. Froula, E.M. Rubin, Rumen metagenome and metatranscriptome analyses of
the gills of discus fish (Symphysodon aequifasciatus) revealed by conven- low methane yield sheep reveals a Sharpea-enriched microbiome characterised
tional biochemical assays and GC-TOF-MS metabolomics, Sci. Total Environ. by lactic acid formation and utilisation, Microbiome 4 (2016) 56.
640 (2018) 1372e1381. [120] Z. Tan, T. Yang, Y. Wang, K. Xing, F. Zhang, X. Zhao, H. Ao, S. Chen, J. Liu,
[98] H.-Z. Sun, K. Shi, X.-H. Wu, M.-Y. Xue, Z.-H. Wei, J.-X. Liu, H.-Y. Liu, Lactation- C. Wang, Metagenomic analysis of cecal microbiome identified microbiota
related metabolic mechanism investigated based on mammary gland and functional capacities associated with feed efficiency in Landrace fin-
metabolomics and 4 biofluids' metabolomics relationships in dairy cows, ishing pigs, Front. Microbiol. 8 (2017) 1546.
BMC Genom. 18 (2017) 936. [121] W. Yan, C. Sun, J. Yuan, N. Yang, Gut metagenomic analysis reveals promi-
[99] C. Mu, Y. Yang, K. Yu, M. Yu, C. Zhang, Y. Su, W. Zhu, Alteration of metab- nent roles of Lactobacillus and cecal microbiota in chicken feed efficiency,
olomic markers of amino-acid metabolism in piglets with in-feed antibiotics, Sci. Rep. 7 (2017) 45308.
Amino Acids 49 (2017) 771e781. [122] S. Kahai, S.-C. Lee, D.Y. Lee, J. Yang, M. Li, C.-H. Wang, Z. Jiang, Y. Zhang,
[100] H. Sun, B. Wang, J. Wang, H. Liu, J. Liu, Biomarker and pathway analyses of C. Peng, B.B. Yang, MicroRNA miR-378 regulates nephronectin expression
urine metabolomics in dairy cows when corn stover replaces alfalfa hay, modulating osteoblast differentiation by targeting GalNT-7, PLoS One 4
J. Anim. Sci. Biotechnol. 7 (2016) 49. (2009) e7535.
[101] S.Y. Mao, W.J. Huo, W.Y. Zhu, Microbiomeemetabolome analysis reveals [123] A.J. Enright, B. John, U. Gaul, T. Tuschl, C. Sander, D.S. Marks, MicroRNA
unhealthy alterations in the composition and metabolism of ruminal targets in Drosophila, Genome Biol. 5 (2003) R1.

View publication stats

Вам также может понравиться