Advances in Food Technology and Nutritional Sciences

Open journal

ISSN 2377-8350

Avianomics: The Beginning at the End

Mary Komp, Rebecca Bowie, Jean-Rémi Teyssier, Brooke C. Bodle, Alison Ferver, Richard A. Mudarra, Bin Zuo, Travis Tabler, Derrell T. Lee, Craig W. Maynard, Garrett Mullenix and Sami Dridi

Received: September 25th, 2020; Revised: November 13th, 2020 Accepted: Noveber 16th, 2020; Published: November 25th, 2020

AVIAN TRANSCRIPTOMICS

The transcriptome of an organism, cell, or tissue refers to the complete set of ribonucleic acid (RNA) species or gene transcripts that are being transcribed at a certain time point. Transcriptomic research aims to compare cells or tissues under defined conditions or disease states to identify changes in gene expression; therefore, giving us a better idea, of which functions different coding or non-coding genes might have, and a better understanding of how they contribute to the phenotype of individuals and their functional adaptations during their lifetime.1,2

There are two main techniques in the field of transcriptomics: microarrays and RNA sequencing. Microarrays are used to measure predetermined sequences, whereas ribonucleic acid (RNA) sequencing captures all sequences through high-throughput sequencing (whole-transcriptome shotgun sequencing).3 These techniques have allowed for a greater understanding of how gene expression changes and identifies trends that cannot be detected by more targeted assays.4

In avian research, transcriptomics has had major implications for our understanding of avian diseases, nutrition, growth, and well-being. The use of transcriptomics is also paving the way for future research opportunities and technology developments that will bridge the gap between traditional avian research and genomics and stimulate more interdisciplinary research.

Transcriptomics in Health and Disease

By using transcriptomics to compare gene expression profiles in healthy and diseased individuals we can attain a greater knowledge of what parts of the genome are or are not being regulated during disease and observe structural alterations, such as mutations. A particular interest in understanding what makes a species resistant or susceptible to a certain infectious disease has further led to several comparative immuned bological studies, which has provided great advancement in the field of immunology. For instance, the use of dual RNA-seq of host and pathogen hasled to studies of host and parasite co-evolution. Transcriptomic analysis of chicken lungs characterized the transcriptomic profile of chicken lungs infected with the parasite, Pasteurella multocida, and provided valuable information to understand host responses. The study identified an avian P. multocidaserogroup A strain (PmQ) showing high lethality to chickens and a bovine P. multocidasero group A strain (PmCQ2) with no lethality to chickens.5 Analyzing the harderian gland, trachea, and lung tissues, Lamont’s group identified several markers associated with Newcastle disease.6 Similarly, by using RNA-seq, several differentially expressed genes were identified in woodybreast affected chickens when compared with unaffected counterparts.7 Marchesi and co-workers identified several genes associated with white striping in broiler chickens.8 Berri’s group performed a muscle transcriptome analysis and revealed several biomarkers associated with meat defect in chickens.9 By using RNA-seq, several osteochondral genes have been shown to be altered in bacterial chondronecrosis with osteomyelitis (BCO) in chickens.10

Transcriptomics in Well-Being

Transcriptomics has the ability to play a key role in many aspects of avian welfare. For example, a transcriptome analysis reveals that stocking density affects the expression of genes involved in several metabolic pathways such as glycolysis, proteolysis, immune stress, muscle development, cell adhesion, matrix, and collagen.11 Several studies showed different transcriptome profiles in various chicken tissues in response to high ambient temperature.12,13,14 They identified Angiopoietin-like-4 (ANGPTL4) gene as a candidate for the improvement of heat tolerance in chickens.

Transcriptomics in Growth, Development and Feed Efficiency

Feed efficiency is a key agricultural and economic trait. Transcriptomic technology could allow for a deeper understanding of residual feed intake among other nutrition related components. A recent study suggested that using transcriptome architectures based on high-quality RNA-Seq data reveals 41 differentially expressed genes associated with residual feed intake. These genes play a role in digestibility, metabolism, stress response, and energy homeostasis.15 Recently, Yang Lei and co-workers identified key genes and pathways associated with feed efficiency in native chickens.16 Using high and low feed efficient birds, Kong et al17 identified several key genes in breast muscle that are involved in anabolic pathways. By comparing the transcriptome between pigeon ovaries before and after ovulation, Xu et al18 found that the expression of several genes is modulated during different stages of ovulation. By studying six avian species, Wright et al19 found higher gene divergence on the Z chromosome compared to autosomal chromosomes.

AVIAN PROTEOMICS

Proteomics, in its simplest definition, is the study of the proteome, which encompasses the expression of whole protein profiles in a biological system. Marc Wilkins, in 1994, is credited for developing the concept and coining the term “proteome”. Its application today in regard to poultry is, but not limited to, studying the cell, tissue, or organ proteins that are expressed (measured and compared), have protein-protein interactions, or are structural components that may impact bird health, well-being, and growth. In addition to improving bird welfare, its application has extended to improving meat quality to the consumer.

Several techniques are frequently used in proteomics including 2-hybrid systems, 2-dimensional gel electrophoresis, and mass spectrometry. Systems biology is a scientific approach that takes into account the complex relationships among and between genes and proteins and determines how all of these interactions come together to form a functional organism. Proteomic tools can simultaneously probe the properties of numerous proteins and thus are a great aid to the emerging field of systems biology, in which the functional interactions of numerous proteins are studied instead of studying individual proteins as isolated entities. In the field of avian biology, proteomics has been used to study different processes from the development, growth, and function of organs and systems to the interactions of infectious agents and the altered states that they induce in their hosts.

Proteomics in Health, Disease and Meat Quality

The following studies have helped elucidate protein changes involving poultry disease and stress such as ammonia stress,20 ascites,21 bacterial, protozoa, and viral infection,22,23,24 femoral head necrosis,25 woody breast,26 and sudden death syndrome.27 The selection of broilers for high growth performance has resulted in an increase in the occurrence of muscle myopathies. The most common are white striping and woody breast myopathies, which have been estimated to cost the U.S. industry $200 million or more.28 Work by Kuttappan et al28 helped elucidate that protein metabolism and protein translocation (141 involved proteins) are important contributors to these myopathies. Another incidence is the pale, soft, and exudative (PSE) syndrome that reduces color, water holding capacity, and quality of poultry meat, which deters consumers. Xing et al29 identified proteins related to the glycolysis pathway, calcium signaling, and molecular chaperones that are integral in PSE syndrome. Tenderness is an important – possibly the most important – driving force for consumer purchase decisions. Mekchay et al30 showed three protein spots that were correlated with shear force, play a role in the glycolytic pathway, and are closely related to tenderness. Other meat quality factors like water holding capacity31 and flavor32,33 have been evaluated for protein biomarkers to improve these areas. These studies not only helped to better understand the protein mechanisms but identified biomarkers for subsequent development of target strategies to reduce or alleviate these issues.

A popular strategy in regulating the host immune system is through feeding probiotics. Luo et al34 offered Enterococcus faecium to broilers and identified 42 intestinal mucosal proteins related to metabolism, immune and antioxidant systems.

Proteomics in Well-Being

Heat load is one of the most challenging stressors to poultry from its adverse effects on growth, gut health, well-being, and mortality. Intestinal health and function are paramount to achieving efficient feed utilization, growth, and welfare. O’Reilly et al35 determined the effects of microbial challenge on broiler intestinal proteome.35 They found that the levels of cytoskeleton35 proteins increased over time in the small intestine. Microbial challenges, however, reduced the expression of Villin-1.35 Tu et al36 and coworkers performed proteomic analysis on the hypothalamus of heat-stressed broilers and identified 114 proteins that are mainly involved in cellular processes, metabolism, transport, and cellular defensive response against oxidative stress and toxin export and delivery.

Proteomics in Growth, Development and Feed Efficiency

Proteomics has not only allowed a better understanding of changes in protein processes but has introduced a new era of selecting broiler genetic lines for superior performance. Kong et al37 selected higher feed efficiency birds from a standard flock and demonstrated that birds having a higher feed efficiency had greater breast muscle mitochondria protein expression caused by higher mitochondrial function and oxidative phosphorylation. PhongpaNgan et al31 separated broilers that had a fast or slow growth rate from the same flock and found broilers with a faster growth rate had a unique protein profile in muscle tissue. In the past decade, the selection of broilers for improved performance has been accompanied by excessive body fat. Wang et al38 identified 33 different proteins that are related to biological oxidation, cell proliferation, cytoskeleton, lipid metabolism, chaperones, protein synthesis, and signal transduction between fat and lean broilers.

AVIAN KINOMICS

Kinomics is mainly used to study the abundance, activity, substrate specificity, phosphorylation status, and other characteristics of kinases and phosphatases in cells or tissues. Protein kinase refers to a class of enzymes that modifies other proteins by chemically adding phosphate groups to them. The protein phosphorylation is one of the important protein post-translational modifications, which plays key roles in the metabolism, transcription, cell cycle regulation, apoptosis, movement, and other cellular processes of most eukaryotic cells.39 Studies have shown that more than 30% of proteins are phosphorylated at any time in cells.40 In the human kinome, 518 protein kinases were identified, and 244 kinases were found to be related to the occurrence of diseases through chromosome mapping and other means.41 Phosphorylation and dephosphorylation are like a molecular switch that could regulate the activation and inactivation of certain proteins. Peptide or protein array screen coupled with mass spectrometry (MS) is widely applied to determine phosphorylation because of the convenience and sensitivity, and proteomics has high-throughput potential and requires no knowledge of sequence preference of the kinase.42,43

Although kinomics is an emerging powerful technology at large scale, very limited studies have been conducted in avian species.

Kinomics in Health and Disease

By using a chicken-specific kinome array, Kogut and colleagues showed that Salmonella modulates host immune signaling pathways in the cecum to establish a persistent infection.44 Specifically, they showed that the response is characterized by alterations in the activation of T-cell signaling mediated by the dephosphorylation of phospholipase c-γ1 (PLCG1) that inhibits NF-κB signaling and activates nuclear factor of activated T-cells (NFAT) signaling and blockage of interferon-γ (IFN-γ) production through the disruption of the JAK-STAT signaling pathway.44 The same group identified that AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) are key sensors and regulators of immunometabolic changes during Salmonella infection in chickens.45

Kinomics in Well-Being

Napper and co-workers used the global cellular kinase approach to characterize kinome response within the breast and thigh muscles of heat- and cold-stressed chickens.46 In the breast, both cold and heat stress activated calcium-dependent metabolic adaptations. Also within the breast, but specific to cold stress, the activation of ErbB signaling as well as dynamic patterns of phosphorylation of AMPK were observed. In the thigh, cold stress activates innate immune signaling and transforming growth factor-beta (TGFβ), however, heat stress-activated pathways are associated with protein and fat metabolism through adipocytokine and mTOR signaling.

EPIGENOMICS

Epigenomics is the study of the complete set of epigenetic (modifications that alter gene expression that cannot be attributed to the modification of the primary deoxyribonucleic acid (DNA) sequence) modifications on the genetic material of a cell, known as the epigenome. DNA methylation and histone modification are among the best-characterized epigenetic processes.47 Several approaches, including histone modification assays, Chip-Chip and Chip-Seq, DNA methylation assays, bisulfite sequencing, and chromatin accessibility assays, are used.

Epigenomics in Health and Disease

Deoxyribonucleic acid methylation has been investigated in relation to disease states in chicken, specifically Marek’s disease, comparing two lines, Fayoumi and Leghorn. This study confirmed that variation in DNA methylation leading to changes in gene regulation likely confers disease resistance and it was observed that DNA methylation was highest in internal exons.48 Immune related genes, such as TLR4, was found to be both differentially methylated and expressed in the two lines and further demonstrates the potential regulatory role of DNA methylation in the control of gene expression. Marek’s disease infection of two lines, with differing resistance, was also analyzed for histone post-translational modifications (HPTMs). Differentially enriched regions were seen between the susceptible and resistant lines, with the HPTM profile being modulated after infection around immune response genes.49,50,51 Epigenetic modifications were also observed in the immune underlying mechanisms in chicken susceptible to Salmonella enteritidis.52

Epigenomics in Well-Being

Epigenetic studies have been conducted in heat-stressed chickens and showed alterations in the methylation level of CpG site in the promoter of the brain-derived neurotrophic factor (BNDF) gene. Modifications of histone H3 lysine 9 (H3K9) and methylation of histone H3 lysine residue 27 (H3K27) in the promoter of hypothalamic brain-derived neurotrophic factor (BDNF) were also observed during thermotolerance acquisition on day 3 posthatch.9,53 In addition to postnatal experience and genetics, it has been reported that chicken behavior can be modulated by epigenetic mechanisms.54

Epigenomics in Growth, Development and Feed Efficiency

It has been shown that environmental and nutritional factors can influence embryonic development and the adult phenotype, at least partially, via epigenetic mechanisms and adaptation. For instance, Tzschentke and Basta have shown that temperature variation during the prenatal period influences hypothalamic neuronal thermosensitivity during the postnatal stage.55 Similarly, nutritional programming affects offspring phenotypes. Rao et al56 showed that 4 week-old chicks from mothers fed low-protein diets had a higher body and breast muscle weights confirming that maternal nutrition can affect offspring phenotypes.

METABOLOMICS

Metabolomics is the emerging field of metabolome analysis that characterize, identify, and quantify metabolites in biological (cell, tissues fluids) samples. As metabolites play a crucial role in the maintenance ad growth of organisms, they are considered as the “canaries” of the genome and the intermediate phenotypes. Their application in livestock in general and poultry, in particular, might become a cornerstone for the “next-generation phenotyping” approaches. An increasing number of studies have integrated metabolomics in poultry.

Metabolomics in Health and Disease

Le Roy et al57 established a high-resolution proton nuclear magnetic resonance-based metabolic atlas and identified several metabolites in different tissues in chickens. By using ultra-performance liquid chromatography/tandem mass spectrometry, Aggrey’s group identified several metabolites associated with fatty acids metabolism and beta-oxidation in Eimeria acervulina-infected chickens.58 Ma and coworkers have shown that Salmonella enterica infection altered the intestinal metabolome in chickens.59 Dridi’s group identified a total of 108 known metabolites in chicken breast, with 60 having significant mean intensity (42 higher and 18 lower) in woody breast-affected compared to unaffected muscles.60 These differentially abundant-metabolites were associated with purine nucleotide degradation and de novo biosynthesis, sirtuinsignaling pathway, citrulline-nitric oxide cycle, salvage pathways of pyrimidine DNA, IL-1 signaling, iNOS, Angiogenesis, PI3K/AKT signaling, and oxidative phosphorylation.60

Metabolomics in Growth, Development and Feed Efficiency

Mignon-Gasteau group showed that ileal, caecal, and serum metabolome explains approximately 75-76% of the variability of the apparent metabolizable energy and digestive efficiency in chickens.61 Analyzing serum metabolome, Metzler-Zebeli et al62 observed a distinct profile between two divergently selected chicken lines for residual feed intake and feed efficiency. Supplementation with Magnolia Bark extract or Bacillus subtilis modulates the intensity of several intestinal metabolites associated with amino acids, fatty acids, peptides, ad nucleosides.63 Peng and co-workers have shown that 39 and 68 metabolites were significantly changed from E14 to E19 and from E19 to hatch, respectively in chicken embryos, indicating a potential role of the metabolome in chicken development.64 Similarly, Wu et al65 studied the hepatic metabolic changes during postnatal liver maturation in breeder roosters and identified several differentially abundant metabolites involved in carbohydrate, protein, amino acid, lipid, cholesterol, nucleic acid, and vitamin metabolisms.

MICROBIOMICS

The microbiome is defined as a characteristic microbial community in a reasonably well-defined habitat, which has distinct physiochemical properties as their theater of activity.66 The study of this microbial community is known as microbiomics. Until early 2000, the study of the microbiome was only relying on the culturable microorganism, which represents a very low portion of the bacterial population as low as 1%. In the microbial research field, the two last decades have been marked by the development of highthroughput DNA-based sequencing methods, new bio-informatics developments, and analysis methods. That methodological progress has allowed the detection and quantification of unculturable bacteria and access to an important amount of data on genome diversity. The study of the microbiome has revealed bacteria to be implicated in numerous functions for the human organism, but the microbiome research has also a wide range of applications in food science, biotechnologies, and agriculture.66

Microbiomics in Health and Disease

As described above, gut health, function, and integrity are essential for the overall well-being and health of birds. Consequently, the gut microbiota, which contains tremendous numbers of bacterial species, has recently been extensively characterized in some avian species including chickens, turkeys, and ducks.67,68,69 The influence of viral and bacterial infections on gut health, integrity, and homeostasis have been also studied. For instance, Experimental infection of young chickens with H9N2 virus caused alterations in the intestinal microbiota composition with increased phylum Proteobacteria.70 In a study conducted by Li et al,71 in addition to increased phylum proteobacteria, Firmicutes was decreased in H9N2 virus-infected chickens. Hird and colleagues reported that the cloacal microbiome in wild ducks is specie-dependent and varies by influenza A virus infection status.72 Similarly, the effect of Newcastle and bursal disease viruses on avian microbiota has been investigated.73,74

Microbiomics in Growth, Development and Feed Efficiency

Several studies investigated the effect of diet on growth and gut microbiome in regard to diversity and composition. The inclusion of xylanase and β-glucanase enzymes in diet enhance the growth of lactic acid bacteria, which adhere to the gut epithelium and compete with pathogens for its colonization while decreasing lumen viscosity.75 Supplementation with phytase promotes the abundance of Aeromonadaceae and Flavobacteriaceae in the crop, and Lactobacillus sp., Clostridium leptum and Enterococcus sp. in the ileum and enhances growth performances in chickens.76 Costa et al77 have shown that the administration of different antibiotic growth promoters induces specific changes in the cecal microbiota in broilers. Wang et al78 showed that probiotics modulate gut microbiota and improves meat flavor in chickens. By performing 16S ribosomal ribonucleic acid (rRNA) sequencing, Jurburg et al79 monitored the chicken fecal microbiome at different ages and identified three successional stages characterized by dominant Streptococcus and Escherichia taxa from 1 to 3 d post-hatch, followed by Lachnospiraceae and Ruminococcus-like species from 4 to 10 d post-hatch, and a final stage with dominant Candidatus Arthrobacter and Romboustia from day 10 (Figure 1).

Figure 1. Graphical Abstract

AFTNSOJ-6-171Fig1

CONCLUSION

Challenges and Perspectives

From ontogenesis to homeostasis, the phenotypes are shaped by multi-complex interactions and networks between genes, proteins, and microbiome and their interaction with their surrounding environment (nutrition, environment, stress, etc.).

In poultry, multi-omics analysis revolutionized avian researches and revealed unprecedented interesting data. However, exploring the full extent of the abovementioned interactions requires careful considerations of study design to efficiently generate optimal data integration. For instance, past omics studies used a variety of tissues and mixed cell populations to attain the differential expression profile, which might mask the cellular specific information. In the future, integrative single-cell omics is warranted.

Development of portable technology would help omics studies in an on-site field, which would minimize the time between harvesting and sequencing, but can also facilitate the workflow in countries where sequencing facilities are scarce or where export of tissues is a hampering factor. If the technology of such is incorporated, rapid identification of the virus or pathogens could be achieved, which in turn could help to minimize the spread and outbreak of infection.

Despite their descriptive nature due to lack of mechanistic studies, the multi-omics studies have the potential to change the future of poultry production and sustainability from dietary, environmental, management, and breeder guidelines by identifying key markers and their networks (gene, protein, metabolite, etc.) for precision selection and personalized nutrition approach under appropriate conditions and adaptations.

CONFLICTS OF INTEREST

All authors declare that there is no conflict of interest in conducting this review or in publishing its results.

1. Chambers DC, Lukowski SW, Powell JE. Transcriptomics and single-cell RNA-sequencing. Respirology. 2019; 24(1): 29-36.
doi: 10.1111/resp.13412

2. Jax E, Wink M, Kraus RHS. Avian transcriptomics: Opportunities and challenges. J Ornithol. 2018; 159: 599-629. doi: 10.1007/s10336-018-1532-5

3. Wolf JB. Principles of transcriptome analysis and gene expression quantification: An RNA-seq tutorial. Mol Ecol Resour. 2013; 13: 559-572.
doi: 10.1111/1755-0998.12109

4. Lowe R, Shirley N, Bleackley M, Dolan S, Shafee T. Transcriptomics technologies. PLoS Comput Biol. 2017; 13: e1005457.
doi: 10.1371/journal.pcbi.1005457

5. Li P, He F, Wu C, Zhao G, Hardwidge PR, Li N, et al. Transcriptomic analysis of chicken lungs infected with avian and bovine pasteurella multocida serotype A. Front Vet Sci. 2020; 7: 452. doi: 10.3389/fvets.2020.00452

6. Deist MS, Gallardo RA, Dekkers JCM, Zhou H, Lamont SJ. Novel combined tissue transcriptome analysis after lentogenic newcastle disease virus challenge in inbred chicken lines of differential resistance. Front Genet. 2020; 11: 11. doi: 10.3389/fgene.2020.00011

7. Mutryn MF, Brannick EM, Fu W, Lee WR, Abasht B. Characterization of a novel chicken muscle disorder through differential gene expression and pathway analysis using RNA-sequencing. BMC Genomics. 2015; 16: 399. doi: 10.1186/s12864-015-1623-0

8. Marchesi JAP, Ibelli AMG, Peixoto JO, Cantão ME, Pandolfi JRC, Marciano CMM, et al. Whole transcriptome analysis of the pectoralis major muscle reveals molecular mechanisms involved with white striping in broiler chickens. Poult Sci. 2019; 98: 590-601. doi: 10.3382/ps/pey429

9. Beauclercq S, Hennequet-Antier C, Praud C, Godet E, Collin A, Tesseraud S, et al. Muscle transcriptome analysis reveals molecular pathways and biomarkers involved in extreme ultimate pH and meat defect occurrence in chicken. Sci Rep. 2017; 7: 6447. doi: 10.1038/s41598-017-06511-6

10. de Oliveira HC, Ibellim AMG, Guimarães SEF, Cantão ME, de Oliveira Peixoto J, Coutinho LL, et al. RNA-seq reveals downregulated osteochondral genes potentially related to tibia bacterial chondronecrosis with osteomyelitis in broilers. BMC Genet. 2020; 21: 58.
doi: 10.1186/s12863-020-00862-2

11. Wu Y, Wang Y, Yin D, Mahmood T, Yuan J. Transcriptome analysis reveals a molecular understanding of nicotinamide and butyrate sodium on meat quality of broilers under high stocking density. BMC Genomics21, 412 (2020). doi: 10.21203/rs.2.16378/v2

12. Coble DJ, Fleming D, Persia ME, Ashwell CM, Rothschild MF, Schmidt CJ, et al. RNA-seq analysis of broiler liver transcriptome reveals novel responses to high ambient temperature. BMC Genomics. 2014; 15: 1084. doi: 10.1186/1471-2164-15-1084

13. Monson MS, Van Goor AG, Persia ME, Rothschild MF, Schmidt CJ, Lamont SJ. Genetic lines respond uniquely within the chicken thymic transcriptome to acute heat stress and low dose lipopolysaccharide. Sci Rep. 2019; 9: 13649. doi: 10.1038/s41598-019-50051-0

14. Monson MS, Van Goor AG, Ashwell CM, Persia ME, Rothschild MF, Schmidt CJ, et al. Immunomodulatory effects of heat stress and lipopolysaccharide on the bursal transcriptome in two distinct chicken lines. BMC Genomics. 2018; 19: 643. doi: 10.1186/s12864-018-5033-y

15. Yi G, Yuan J, Bi H, Yan W, Yang N, Qu L. In-depth duodenal transcriptome survey in chickens with divergent feed efficiency using RNA-Seq. PLoS One. 2015; 10: e0136765. doi: 10.1371/journal.pone.0136765

16. Yang L, He T, Xiong F, Chen X, Fan X, Jin S, et al. Identification of key genes and pathways associated with feed efficiency of native chickens based on transcriptome data via bioinformatics analysis. BMC Genomics. 2020; 21: 292. doi: 10.1186/s12864-020-6713-y

17. Kong BW, Song JJ, Lee JY, Hargis BM, Wing T, Lassiter K, Bottje W. Gene expression in breast muscle associated with feed efficiency in a single male broiler line using a chicken 44K oligo microarray. I. Top differentially expressed genes. Poult Sci. 2011; 90: 2535-2547.
doi: 10.3382/ps.2011-01435

18. Xu X, Zhao X, Lu L, Duan X, Qin H, Du X, et al. Transcriptomic analysis of different stages of pigeon ovaries by RNA-sequencing. Mol Reprod Dev. 2016; 83: 640-648. doi: 10.1002/mrd.22670

19. Wright AE, Harrison PW, Zimmer F, Montgomery SH, Pointer MA, Mank JE. Variation in promiscuity and sexual selection drives avian rate of Faster-Z evolution. Mol Ecol. 2015; 24: 1218-1235. doi: 10.1111/mec.13113

20. Lu M, Bai J, Xu B, Sun QY, Wei FX, Tang XF, et al. Effect of alpha-lipoic acid on relieving ammonia stress and hepatic proteomic analyses of broilers. Poult Sci. 2017; 96: 88-97. doi: 10.3382/ps/pew285

21. Wang Y, Guo Y, Ning D, Peng Y, Cai H, Tan J, et al. Changes of hepatic biochemical parameters and proteomics in broilers with cold-induced ascites. J Anim Sci Biotechnol. 2012; 3: 41. doi: 10.1186/2049-1891-3-41

22. Cumberbatch JA, Brewer D, Vidavsky I, Sharif S. Chicken major histocompatibility complex class II molecules of the B haplotype present self and foreign peptides. Anim Genet. 2006; 37: 393-396. doi: 10.1111/j.1365-2052.2006.01459.x

23. Polansky O, Seidlerova Z, Faldynova M, Sisak F, Rychlik I. Protein expression in the liver and blood serum in chickens in response to Salmonella Enteritidis infection. Vet Immunol Immunopathol. 2018; 205: 10-16. doi: 10.1016/j.vetimm.2018.10.006

24. Zhao Z, Zhao Q, Zhu S, Huang B, Lv L, Chen T, et al. iTRAQ-based comparative proteomic analysis of cells infected with Eimeria tenella sporozoites. Parasite. 2019; 26: 7. doi: 10.1051/parasite/2019009

25. Packialakshmi B, Liyanage R, Lay JO, Okimoto R, Rath NC. Proteomic changes in the plasma of broiler chickens with femoral head necrosis. Biomark Insights. 2016; 11: 55-62. doi: 10.4137/BMI.S38291

26. Cai K, Shao W, Chen X, Campbell YL, Nair MN, Suman SP,et al. Meat quality traits and proteome profile of woody broiler breast (pectoralis major) meat. Poult Sci. 2018; 97: 337-346. doi: 10.3382/ps/pex284

27. Ning H, Cui Y, Song X, Chen L, Yin Z, Hua L, et al. iTRAQbased proteomic analysis reveals key proteins affecting cardiac function in broilers that died of sudden death syndrome. Poult Sci. 2019; 98: 6472-6482. doi: 10.3382/ps/pez532

28. Kuttappan VA, Bottje W, Ramnathan R, Hartson SD, Coon CN, Kong B-W, et al. Proteomic analysis reveals changes in carbohydrate and protein metabolism associated with broiler breast myopathy. Poult Sci. 2017; 96: 2992-2999. doi: 10.3382/ps/pex069

29. Xing T, Wang C, Zhao X, Dai C, Zhou G, Xu X. Proteome analysis using isobaric tags for relative and absolute analysis quantitation (iTRAQ) reveals alterations in stress-induced dysfunctional chicken muscle. J Agric Food Chem. 2017; 65: 2913-2922. doi: 10.1021/acs.jafc.6b05835

30. Mekchay S, Teltathum T, Nakasathien S, Pongpaichan P. Proteomic analysis of tenderness trait in Thai native and commerical broiler chicken muscles. Japan Poult Sci Assoc. 2010; 8: 8-12. doi: 10.2141/jpsa.009033

31. Phongpa-Ngan P, Grider A, Mulligan JH, Aggrey SE, Wicker L. Proteomic analysis and differential expression in protein extracted from chicken with a varying growth rate and water-holding capacity. J Agric Food Chem. 2011; 59: 13181-13187. doi: 10.1021/jf202622n

32. Liu XD, Jayasena DD, Jung Y, Jung S, Kang BS, Heo KN,et al. Differential proteome analysis of breast and thigh muscles between Korean native chickens and commercial broilers. Asian-Australasian J Anim Sci. 2012; 25: 895-902. doi: 10.5713/ajas.2011.11374

33. Zheng A, Chang W, Liu G, Yue Y, Li J, Zhang S, et al. Molecular differences in hepatic metabolism between AA broiler and big bone chickens: A proteomic study. PLoS One. 2016; 11: e0164702. doi: 10.1371/journal.pone.0164702

34. Luo J, Zheng A, Meng K, Chang W, Bai Y, et al. Proteome changes in the intestinal mucosa of broiler (Gallus gallus) activated by probiotic Enterococcus faecium. J Proteomics. 2013; 91: 226-241. doi: 10.1016/j.jprot.2013.07.017

35. O’Reilly EL, Burchmore RJ, Sparks NH, Eckersall PD. The effect of microbial challenge on the intestinal proteome of broiler chickens. Proteome Sci. 2016; 15: 10. doi: 10.1186/s12953-017-0118-0

36. Tu WL, Cheng C-Y, Chen C-J, Chan H-L, Wang S-H, Tang P-C, et al. Proteomic analysis of the hypothalamus of broiler-type Taiwan country chickens in response to acute heat stress. Anim Sci J. 2018; 89: 1475-1485. doi: 10.1111/asj.13060

37. Kong BW, Lassiter K, Piekarski-Welsher A, Dridi S, Reverter-Gomez A, Hudson NJ, et al. Proteomics of breast muscle tissue associated with the phenotypic expression of feed efficiency within a pedigree male broiler line: I. Highlight on mitochondria. PLoS One. 2016; 11: e0155679.
doi: 10.1371/journal. pone.0155679

38. Wang L, Cheng B, Li H, Wang Y. Proteomics analysis of preadipocytes between fat and lean broilers. Br Poult Sci. 2019; 60: 522-529.
doi: 10.1080/00071668.2019.1621989

39. Ubersax JA, Ferrell JE. Mechanisms of specificity in protein phosphorylation. Nat Rev Mol Cell Biol. 2007; 8: 530-541. doi: 10.1038/nrm2203

40. Cohen P. The regulation of protein function by multisite phosphorylation–a 25 year update. Trends Biochem Sci. 2000; 25: 596-601.
doi: 10.1016/s0968-0004(00)01712-6

41. Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S. The protein kinase complement of the human genome. Science. 2002; 298: 1912-1934. doi: 10.1126/science.1075762

42. Zhu H, Bilgin M, Bangham R, Hall D, Casamayor A, Bertone P, et al. Global analysis of protein activities using proteome chips. Science. 2001; 293: 2101-2105. doi: 10.1126/science.1062191

43. Reimer U, Reineke U, Schneider-Mergener J. Peptide arrays: From macro to micro. Curr Opin Biotechnol. 2002; 13: 315-320.
doi: 10.1016/s0958-1669(02)00339-7

44. Kogut MH, Swaggerty CL, Byrd JA, Selvaraj R, Arsenault RJ. Chicken-specific kinome array reveals that salmonella enterica serovar enteritidis modulates host immune signaling pathways in the cecum to establish a persistence infection. Int J Mol Sci. 2016; 17: 1207.
doi: 10.3390/ijms17081207

45. Kogut MH. Genovese KJ, He H, Arsenault RJ. AMPK and mTOR: Sensors and regulators of immunometabolic changes during Salmonella infection in the chicken. Poult Sci. 2016; 95: 345-353. doi: 10.3382/ps/pev349

46. Napper S, Dadgar S, Arsenault RJ, Trost B, Scruten E, Kusalik A, et al. Induction of tissue- and stressor-specific kinomic responses in chickens exposed to hot and cold stresses. Poult Sci. 2015; 94: 1333-1345. doi: 10.3382/ps/pev046

47. Alabert C, Groth A. Chromatin replication and epigenome maintenance. Nat Rev Mol Cell Biol. 2012; 13: 153-167. doi: 10.1038/nrm3288

48. Li J, Li R, Wang Y, Hu X, Zhao Y, Li L, et al. Genome-wide DNA methylome variation in two genetically distinct chicken lines using MethylC-seq. BMC Genomics. 2015; 16: 851. doi: 10.1186/s12864-015-2098-8

49. Luo J, Mitra A, Tian F, Chang S, Zhang H, Cui K, et al. Histone methylation analysis and pathway predictions in chickens after MDV infection. PLoS One 2012; 7: e41849. doi: 10.1371/journal.pone.0041849

50. Mitra A, Luo J, Zhang H, Cui K, Zhao K, Song J. Marek’s disease virus infection induces widespread differential chromatin marks in inbred chicken lines. BMC Genomics. 2012; 13: 557. doi: 10.1186/1471-2164-13-557

51. Mitra A, Luo J, He Y, Gu Y, Zhang H, Zhao K, et al. Histone modifications induced by MDV infection at early cytolytic and latency phases. BMC Genomics. 2015; 16: 311. doi: 10.1186/s12864-015-1492-6

52. Gou Z, Liu R, Zhao G, Zheng M, Lim P, Wang H, et al. Epigenetic modification of TLRs in leukocytes is associated with increased susceptibility to Salmonella enteritidis in chickens. PLoS One. 2012; 7: e33627. doi: 10.1371/journal.pone.0033627

53. Kisliouk T, Yosefi S, Meiri N. MiR-138 inhibits EZH2 methyltransferase expression and methylation of histone H3 at lysine 27, and affects thermotolerance acquisition. Eur J Neurosci. 2011; 33: 224-235. doi: 10.1111/j.1460-9568.2010.07493.x

54. Jensen P. Behaviour epigenetics – The connection between environment, stress and welfare. Applied Animal Behaviour Science. 2014; 157: 1-7. doi: 10.1016/j.applanim.2014.02.009

55. Tzschentke B, Basta D. Early development of neuronal hypothalamic thermosensitivity in birds: influence of epigenetic temperature adaptation. Comp Biochem Physiol A Mol Integr Physiol. 2002; 131: 825-832. doi: 10.1016/s1095-6433(02)00020-x

56. Rao K, Xiem J, Yang X, Chen L, Grossmann R. Zhao R. Maternal low-protein diet programmes offspring growth in association with alterations in yolk leptin deposition and gene expressionin yolk-sa c membrane, hypothalamus and muscle of developing Langshan chicken embryos. Br J Nutr. 2009; 102: 848-857. doi: 10.1017/S0007114509276434

57. Le Roy CI, Mappley LJ, La Ragione RM, Woodward MJ, Claus SP. NMR-based metabolic characterization of chicken tissues and biofluids: a model for avian research. Metabolomics. 2016; 12: 157. doi: 10.1007/s11306-016-1105-7

58. Aggrey SE, Milfort MC, Fuller AL, Yuan J, Rekaya R. Effect of host genotype and Eimeria acervulina infection on the metabolome of meat-type chickens. PLoS One. 2019; 14: e0223417. doi: 10.1371/journal.pone.0223417

59. Ma B, Mei X, Lei C, Li C, Gao Y-F, Kong LH, et al. Enrofloxacin shifts intestinal microbiota and metabolic profiling andhi nders recovery from salmonella enterica subsp. enterica Serovar Typhimurium Infection in Neonatal Chickens. mSphere. 2020; 5:1-16. doi: 10.1128/mSphere.00725-20

60. Greene E, Cauble R, Dhamad AE, Kidd MT, Kong B, Howard SM, et al. Muscle Metabolome profiles in woody breast-(un) affected broilers: Effects of quantum blue phytase-enriched diet. Front Vet Sci. 2020; 7: 458. doi: 10.3389/fvets.2020.00458

61. Beauclercq S, Nadal-Desbarats L, Hennequet-Antier C, Gabriel I, Tesseraud S, Calenge F, et al. Relationships between digestive efficiency and metabolomic profiles of serum and intestinal contents in chickens. Sci Rep. 2018; 8: 6678. doi: 10.1038/s41598-018-24978-9

62. Metzler-Zebeli BU, Siegerstetter S-C, Magowan E, Lawlor PG, O’Connell NE, Zebeli Q. Feed restriction reveals distinct serum metabolome profiles in chickens divergent in feed efficiency traits. Metabolites. 2019; 9: 38. doi: 10.3390/metabo9020038

63. Park I, Oh S, Lillehoj EP, Lillehoj HS. Dietary supplementation with magnolia bark extract alters chicken intestinal metabolite levels. Front Vet Sci. 2020; 7: 157. doi: 10.3389/fvets.2020.00157

64. Peng ML, He QQ, Zhao JL, Li LL, Ma HT. Based serum metabolomics analysis reveals simultaneous interconnecting changes during chicken embryonic development. J Anim Physiol Anim Nutr (Berl). 2018; 102: 1210-1219. doi: 10.1111/jpn.12925

65. Wu S, Liu Y, Zhu L, Han D, Bello M, Yang X, et al. Hepatic metabolomic profiling changes along with postnatal liver maturation in breeder roosters. Biol Open. 2018; 7: bio.028944. doi: 10.1242/doi: 10.1242/bio.028944

66. Berg G, Rybakova D, Fischer D, Cernava T, M-C C Vergès, et al. Microbiome definition re-visited: Old concepts and new challenges. Microbiome. 2020; 8: 103. doi: 10.1186/s40168-020-00875-0

67. Choi JH, Kim GB, Cha CJ. Spatial heterogeneity and stability of bacterial community in the gastrointestinal tracts of broiler chickens. Poult Sci. 2014; 93: 1942-1950. doi: 10.3382/ps.2014- 03974

68. Wilkinson TJ, Cowan AA, Vallin HE, Onime LA, Oyama LB, Cameron SJ, et al. Characterization of the microbiome along  thegastrointestinal tract of growing turkeys. Front Microbiol. 2017; 8:1089. doi: 10.3389/fmicb.2017.01089

69. Best AA, Porter AL, Fraley SM, Fraley GS. Characterization of gut microbiome dynamics in developing pekin ducks and impact of management system. Front Microbiol. 2016; 7: 2125. doi: 10.3389/fmicb.2016.02125

70. Yitbarek A, Weese JS, Alkie TN, Parkinson J, Sharif S. Influenza a virus subtype H9N2 infection disrupts the composition of intestinal microbiota of chickens. FEMS Microbiol Ecol. 2018; 94. doi: 10.1093/femsec/fix165

71. Li H, Liu X, Chen F, Zuo K, Wu C, Yan Y, et al. Avian influenza virus subtype H9N2 affects intestinal microbiota, barrier structure injury, and inflammatory intestinal disease in the chicken ileum. Viruses. 2018; 10: 270. doi: 10.3390/v10050270

72. Hird SM, Ganz H, Eisen JA, Boyce WM. The cloacal microbiome of five wild duck species varies by species and influenza A virus infection status. mSphere. 2018; 3: doi: 10.1128/mSphere.00382-18

73. Cui N, Huang X, Kong Z, Huang Y, Huang Q, Yang S, et al. Newcastle disease virus infection interferes with the formation of intestinal microflora in newly hatched specific-pathogen-free chicks. Front Microbiol. 2018; 9: 900. doi: 10.3389/fmicb.2018.00900

74. Li L, Kubasova T, Rychlik I, Hoerr FJ, Rautenschlein S. Infectious bursal disease virus infection leads to changes in the gut associated-lymphoid tissue and the microbiota composition. PLoS One. 2018; 13: e0192066. doi: 10.1371/journal.pone.0192066

75. Rodriguez ML, Rebolé A, Velasco S, Ortiz LT, Treviño J, Alzueta C. Wheat- and barley-based diets with or without additives influence broiler chicken performance, nutrient digestibility and intestinal microflora. J Sci Food Agric. 2012; 92: 184-190. doi: 10.1002/jsfa.4561

76. Ptak A, Bedford MR, Swiatkiewicz S, Zyla K, Jozefiak D. Phytase modulates ileal microbiota and enhances growth performance of the broiler chickens. PLoS One. 2015; 10: e0119770. doi: 10.1371/journal.pone.0119770

77. Costa MC, Bessegatto JA, Alfieri AA, Weese JS, Filho JAB, Oba A, et al. Different antibiotic growth promoters induce specific changes in the cecal microbiota membership of broiler chicken. PLoS One. 2017; 12: e0171642. doi: 10.1371/journal.pone.0171642

78. Wang Y, Sun J, Zhong H, Li N, Xu H, Zhu Q, et al. Effect of probiotics on the meat flavour and gut microbiota of chicken. Sci Rep. 2017; 7: 6400. doi: 10.1038/s41598-017-06677-z

79. Jurburg SD, Brouwer MSM, Ceccarelli D, van der Goot J, Jansman AJM, Bossers A. Patterns of community assembly in the developing chicken microbiome reveal rapid primary succession. Microbiologyopen. 2019; 8: e00821. doi: 10.1002/mbo3.821

LATEST ARTICLES

Use of Black Soldier Fly (Hermetia illucens) Prepupae Reared on Organic Waste

review

2024 Mar

Isayas A. Kebede*, Haben F. Gebremeskel and Gelan D. Dahesa,
Figure 11. Risk Map for the Introduction of Ruminant Diseases at Borders

Original Research, peer reviewed

2024 Mar

Yassir Lezaar*, Mehdi Boumalik, Youssef Lhor, Moha El-Ayachi, Abelilah Araba and Mohammed Bouslikhane

Commentary, End-of-Life, Family Dynamics, palliative Care, peer reviewed

2024 Mar

Neil A. Nijhawan*, Rasha Mustafa and Aqeela Sheikh

Retrospective Study

2024 Mar

Shrenik J. Shah*, Abhishek Jha, Chirag Davara, Rushi Mistry and Kapil Kachhadiya

Attention, Disaster, Mindfulness, Original Research, peer reviewed

2024 Mar

Christina Hagen*, Lars Lien, Edvard Hauff and Trond Heir

review

2024 Mar

Isayas A. Kebede* and Gelan D. Dahesa

review

2024 Feb

Bekiyad S. Daro*; Yared A. Deneke, Jiregna G. Negasa and Ishetu N. Dibaba