Next Article in Journal
Evaluating the Impact of Arginine-to-Lysine Ratios on Growth Performance, Antioxidant Defense, and Immune Modulation in Juvenile Largemouth Bass (Micropterus salmoides)
Previous Article in Journal
Amoxicillin Resistance: An In Vivo Study on the Effects of an Approved Formulation on Antibiotic Resistance in Broiler Chickens
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Overview of Advancements in Proteomic Approaches to Enhance Livestock Production and Aquaculture

by
Jakree Jitjumnong
1,2,
Anukul Taweechaipaisankul
3,
Jou-Ching Lin
4,
Supatirada Wongchanla
5,
Schwann Chuwatthanakhajorn
6,
Chih-Jen Lin
7,
Luu Tang Phuc Khang
1,
Nguyen Vu Linh
1,
Papungkorn Sangsawad
8,
Nguyen Dinh-Hung
9,
Pin-Chi Tang
4,10 and
Tossapol Moonmanee
1,2,*
1
Department of Animal and Aquatic Sciences, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
2
Functional Feed Innovation Center, Faculty of Agriculture, Chiang Mai University, Chiang Mai 50200, Thailand
3
Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
4
Department of Animal Science, College of Agriculture and Natural Resources, National Chung Hsing University, Taichung 40227, Taiwan
5
Department of Animal Science, University of California, Davis, CA 95616, USA
6
Faculty of Veterinary Science, Mahidol University, Salaya, Nakhon Pathom 73170, Thailand
7
Centre for Reproductive health, Institute for Regeneration and Repair (IRR), University of Edinburgh, Edinburgh EH16 4 UU, UK
8
School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
9
Aquaculture Pathology Laboratory, School of Animal & Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ 85721, USA
10
The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan
*
Author to whom correspondence should be addressed.
Animals 2025, 15(13), 1946; https://doi.org/10.3390/ani15131946
Submission received: 13 March 2025 / Revised: 28 June 2025 / Accepted: 29 June 2025 / Published: 2 July 2025
(This article belongs to the Section Animal System and Management)

Simple Summary

Proteomics is a powerful tool transforming both livestock production and aquaculture by providing molecular insights into animal health and productivity. In livestock, it enhances breeding by identifying the biomarkers for reproduction, disease resistance, and productivity. It also improves meat quality, fertility, and feed efficiency, while aiding stress management through biomarkers like heat shock proteins. In aquaculture, proteomics optimizes fish growth, immune responses, and stress adaptation through non-invasive techniques like blood and mucus analysis. It supports disease management by identifying immune markers and disease mechanisms, while also improving reproductive biology and hatchery management through protein markers for egg and sperm quality. Overall, proteomics contributes to more sustainable and efficient practices in both sectors, improving breeding, feed efficiency, welfare, disease control, and environmental adaptation.

Abstract

Proteomics, the large-scale study of proteins and their functions, is an essential tool in biological research, particularly in livestock production and aquaculture. This review explores the significance of proteomic techniques and technologies in enhancing agricultural practices. Key methods, including mass spectrometry, two-dimensional gel electrophoresis, and protein microarrays, enable researchers to analyze protein complexity in biological systems. In livestock production, proteomics improves animal health, growth, reproduction, and disease resistance, contributing to more efficient and sustainable practices. In aquaculture, it optimizes fish health, breeding strategies, and feed efficiency, promoting sustainable farming. Despite its potential, proteomics faces challenges, such as complexity, the need for standardized methods, and difficulties in data interpretation. However, emerging advances—including multi-omics integration, real-time monitoring, and improved understanding of protein functions under varying environmental conditions—offer promising solutions. In conclusion, proteomics is poised to transform livestock production and aquaculture, addressing key challenges in food security and sustainable agriculture.

1. Introduction

Proteomics, which involves the large-scale analysis of proteins within biological systems, emerged in the mid-1990s as a complementary field to genomics [1]. Its primary objective is to comprehensively map the proteome—the complete set of proteins in cells, tissues, or organisms—including their interactions and post-translational modifications [2]. Unlike approaches that focus on individual proteins, proteomics enable high-throughput analysis, providing a holistic understanding of biological systems [2]. The proteome is highly dynamic, influenced by physiological conditions and environmental factors, necessitating advanced analytical methodologies [1]. Standard proteomic workflows typically involve protein separation, mass spectrometry analysis, and bioinformatic-driven data interpretation [3]. The complexity of the proteome is further compounded by factors such as tissue-specific gene expression, alternative splicing, and post-translational modifications, which together result in a proteome that exceeds the number of genes in the genome [4]. Proteomics has become an invaluable tool in biomedical research, particularly for comparative analyses of protein expression between healthy and diseased states, offering critical insights into disease mechanisms [1]. Moreover, its application in pharmaceutical research and novel drug discovery has significantly expanded in recent years [5]. Advances in targeted liquid chromatography–mass spectrometry (LC-MS) have enhanced the accuracy and sensitivity of protein identification and quantification, facilitating progress in various areas of systems biology, such as proteogenomic, interactomics, and kinomics [6]. Despite its promising potential, proteomics faces several challenges, including complexities in sample preparation, protein fractionation, and data interpretation [7]. Mass spectrometry remains the cornerstone of proteomic investigations, with liquid chromatography–tandem mass spectrometry (LC-MS-MS) and matrix-assisted laser desorption/ionization time-of-flight/time-of-flight (MALDI-TOF/TOF) being widely employed [8]. The inherent complexity of proteomes, coupled with the dynamic nature of protein expression and the high costs associated with proteomics infrastructure and specialized personnel, however, limits broader adoption, especially in resource-limited settings [8]. Nevertheless, proteomics continues to drive advances in biomedical sciences by elucidating protein structure, functions, and signaling pathways relevant to various diseases [8].
Beyond human health, proteomics has become an essential tool in livestock and aquaculture research, addressing critical challenges in these rapidly expanding sectors. In aquaculture, proteomics analyses provide a deeper understanding of fish biology, disease diagnosis, stress management, and food security [9,10]. Proteomics facilitate the identification of biomarkers for species differentiation, disease surveillance, and animal welfare assessment [9]. In dairy production, proteomics research supports sustainable practices by improving knowledge of animal health, disease prevention, and management systems [11]. Notably, proteomics plays a crucial role in investigating pathogen–host interactions, making it a promising tool for animal disease research and diagnostics [10].
This review aims to explore the role of proteomics in advancing our understanding of biological systems, with a particular focus on its applications in livestock and aquaculture. This review will discuss proteomics techniques, their specific applications in animal sciences, and the challenges and limitations faced by the field. Additionally, it will highlight the increasing significance of proteomics in enhancing productivity, sustainability, and disease management in these industries while discussing future directions for the continued advancement of proteomic research.

2. Applications of Proteomics in Livestock Production

The rapid evolution of molecular biology has significantly transformed livestock breeding, shifting from traditional phenotypic selection to advanced genetic and molecular approaches. Key innovations such as genomic selection and marker-assisted selection (MAS) have revolutionized breeding programs by enhancing genetic improvement and providing deeper insights into economically important traits. High-throughput omics technologies, particularly in genomics and proteomics, have further refined these methods, allowing breeders to make more precise and informed decisions. In animal breeding, genomic selection and MAS have accelerated the identification of genes and quantitative trait loci (QTL) related to production and reproductive traits, facilitating more targeted breeding strategies. Meanwhile, proteomic analysis helps in understanding the molecular mechanisms that govern these traits, offering new avenues for optimizing fertility, meat quality, and overall health. By identifying protein biomarkers associated with reproduction, immunity, and stress response, proteomics has become an indispensable tool for improving livestock management. This chapter explores the impact of genetic improvement and breeding programs, with a particular focus on the role of proteomics in advancing livestock productivity. From enhancing fertility and disease resistance to optimizing meat quality and feed efficiency, molecular tools are shaping the future of sustainable and profitable livestock production.

2.1. Genetic Improvement and Breeding Programs

These methods enhance genetic improvement, providing insights into economically relevant traits and underlying biological processes [12]. High-throughput omics technologies have advanced genomic selection, making it a precise and widely accepted approach in animal breeding [13]. Numerous genes and quantitative trait loci (QTL) associated with production and reproductive traits have been identified, facilitating selection and breeding decisions [14]. Proteomics has become integral to understanding the molecular mechanisms underlying key livestock traits, including reproduction, disease resistance, and stress tolerance [15,16,17]. Proteomics supports genetic improvement programs by analyzing protein functions, structures, and interactions [12,18]. Proteomic studies of reproductive processes provide insights into fertility regulation, enabling the early detection of fertility issues and optimization of breeding strategies in vitro [19,20]. Differential protein expression in high- and low-fertility sperm affects capacitation, acrosomal reaction, and sperm–oocyte communication [21]. For instance, the upregulation of metabolic proteins such as pyruvate kinase M2 (PKM2) and ATP5B enhances ATP production essential for sperm motility [18]. Additionally, the increased expression of casein kinase 2 (CKII) and A-kinase anchor protein-4 (AKAP4) in high-fertility sperm improves chromatin decondensation and motility [18].
Proteomic analysis of reproductive fluids provides insights into hormonal regulation, maternal–gamete interactions, and embryonic development [19,22]. Oviductal fluid proteins vary across estrous cycle stages, with heat shock proteins (HSPA5, HSPA8, and HSP90B1) upregulated post-ovulation, influencing embryonic development [23]. NF-κB-related proteins in uterine fluid post-embryo transfer support trophoblast invasion and placental formation [24]. Proteomic analysis of bovine follicular fluid has identified fertility-related proteins, including TIMP2 and complement component C8 alpha chain [25].
Proteomics also improves meat quality by identifying the biomarkers linked to flavor, tenderness, and juiciness [26] (Figure 1). Protein biomarkers relate to pathways such as energy metabolism, oxidative stress, and structural integrity [27,28,29]. Enzymes like MDH1 and ENO3 are more abundant in tender meat, while ALDH1A1 is associated with toughness [29,30]. Heat shock proteins (HSP1A1, CRYAB, HSP20, and HSP27) are prevalent in tougher meat, whereas MYL1 and MYH1 are linked to tenderness. Additionally, TRIM72 is abundant in tender meat, while FHL1 is higher in tough samples [27].
In dairy science, proteomic analysis identifies biomarkers influencing milk quality, fat content, and lactation efficiency [31,32]. Proteomic variations between colostrum and mature milk reveal immunoglobulin dominance in colostrum, while antigen-specific proteins such as CLU and MFGE8 are prevalent in mature milk [33]. Furthermore, proteomics assesses protein modifications in dairy processing, including lactosylation changes in casein and β-lactoglobulin due to ultra-heat treatment and pasteurization [34].

2.2. Feed Efficiency and Nutritional Optimization

Feed accounts for approximately half of livestock production costs, making feed efficiency a key factor in reducing expenses and improving profit margins [35]. Residual feed intake (RFI) is a widely used measure that identifies animals consuming less dry matter than expected relative to their body weight and growth, thereby reducing feed costs [36,37]. However, RFI studies are complex due to their dependence on intricate biological mechanisms regulated by multiple genes affecting physiological and metabolic functions [37]. Instead, omics technologies provide insights into feed efficiency by analyzing metabolic and proteomic changes [38,39].
Feed efficiency is influenced by dietary composition, management, and metabolic states [40]. Studying RFI often requires invasive techniques, such as liver biopsy, due to the liver’s central role in metabolism [41,42]. Metabolomic and proteomic liver analyses reveal that high-forage diets in dairy heifers significantly alter protein abundance, affecting fatty acid oxidation, ketone synthesis, amino acid metabolism, gluconeogenesis, and oxidative stress defense [39]. Lipid metabolism is crucial for energy balance, as lipids provide an alternative energy source during metabolic stress [43]. High-forage diets upregulate the mitochondrial enzymes (ACSS3, ECHS1) and peroxisomal enzymes (ACOX1) involved in fatty acid breakdown [39,44]. Increased fatty acid oxidation also enhances ketone body synthesis, providing alternative energy [45]. Additionally, oxidative defense proteins (CYP450s, SOD1, GSTMs, TXN) increase in high-forage-fed heifers, likely due to reactive oxygen species from metabolic processes, contributing to reduced energy efficiency [46,47].
Beyond invasive sampling, few studies have explored the semi-invasive and non-invasive methods for proteomic analysis of RFI traits. Notably, high-RFI cattle showed increased TGF-β levels, a key regulator of feed efficiency [48]. Additionally, low-RFI cattle exhibited higher levels of metabolic hormones (KNG1, FBN1, ADIPOQ), which influence feed intake, energy metabolism, and lipid accumulation [49,50]. Non-invasive saliva proteomics offers insights into both host and microbial proteins, improving understanding of animal–microbiota interactions [51]. Salivary proteomics may also provide reliable health biomarkers, serving as an alternative to blood-based analysis [52,53,54].

2.3. Stress and Welfare

The study of stress and welfare in livestock is of paramount importance due to its direct impact on animal health, productivity, and ethical considerations in animal husbandry. Proteomics, as a cutting-edge approach, plays a pivotal role in this field by enabling the identification of protein biomarkers that reflect the physiological responses of livestock to stress and welfare conditions (Table 1). Stress in livestock, whether physiological or environmental, can lead to adverse effects on immune function, growth, reproduction, and overall well-being, ultimately affecting the quality and efficiency of animal production systems. Recent findings have reported the application of proteomics on animal welfare and stress detection in various species of livestock. For instance, a study by Marco-Ramell and colleagues identified cytoplasmic β-actin as a key protein associated with stocking density stress in pigs. Elevated serum actin levels are suggested to serve as a marker of cell or tissue damage associated with overcrowding conditions [55]. Likewise, a multi-omics analysis of chicken thigh muscle has revealed elevated corticosterone levels and reduced hexose phosphate abundance in response to road transport-induced stress, concluding that road transport impacts cytoskeletal structure and carbohydrate metabolism [56]. Heat stress is a major challenge to livestock welfare, particularly in regions with extreme temperatures. In pigs experiencing acute heat stress, proteins associated with glycolysis, glycogenesis, and glycogenolysis were markedly upregulated. In addition, elevated levels of manganese superoxide dismutase (SOD) and reduced abundance of peroxiredoxin 2 were identified as significant biomarkers for stress assessment [57]. A study investigating the effects of heat stress on the liver proteome of Peking ducks (Anas platyrhynchos) and Muscovy ducks (Cairina moschata) revealed the overexpression of heat shock proteins 70 and 10 in both species as a response to heat stress. Moreover, proteins such as α-enolase and S-adenosylmethionine synthetase exhibited species-specific expression patterns, underscoring distinct physiological responses to heat stress [58]. Heat stress in dairy cows has been linked to alterations in serum protein profiles, including increased levels of heat shock protein 70 (HSP70) and reduced concentrations of metabolic proteins such as albumin. These changes serve as biomarkers for heat stress, helping farmers implement management strategies like cooling systems and dietary adjustments [59,60,61]. Monitoring nutritional stress is crucial for maintaining animals in optimal conditions that support their health and prevent unnecessary suffering. Negative energy balance (NEB) is a common metabolic challenge in early lactating cows, arising from insufficient nutrient and energy intake to support the high demands of milk production. This imbalance triggers excessive lipolysis, resulting in the release of free fatty acids into the circulatory system, ultimately contributing to metabolic disorders such as ketosis and fatty liver [62]. Plasma proteomic profile have indicated that increases in acute phase proteins (APPs) such as haptoglobin, a-1-acid glycoprotein (AGP), and a-chymotrypsin and a decrease in Apolipoprotein-AIV and a-2-HS-glycoprotein were highly related to ketosis in cows [63,64]. Furthermore, stomatin and galactose-1-phosphate were reported as key indicators of NEB in milk [65,66]. These insights allow for the development of evidence-based management strategies aimed at improving animal welfare, enhancing disease resilience, and fostering sustainable livestock production.
Table 1. Proteomics applications for stress and welfare in livestock.
Table 1. Proteomics applications for stress and welfare in livestock.
IndicatorsProteomics Techniques/MethodSpeciesFindingsReferences
Stress responseLC-MS/MSSwineLabel-free quantitative proteomics identified 66 differentially abundant proteins between non-stressed and stressed pigs, with 30 increased and 36 decreased in the non-stressed group.[67]
Pre-slaughter stressOFFGEL, SDS-PAGE and LC-MSBovineFive protein bands showed significant differences between normal and DFD meat samples, containing actin, phosphoglucomutase-1, alpha-crystallin B, heat shock protein beta-6, and heat shock protein beta-1.[68]
Chronic lamenessLC-MS/MS and SDS-PAGEBovineChronic inflammatory lameness in dairy cows is associated with increased expression of stress proteins with chaperone, metabolism, redox, and structural functions in the dorsal horn of the spinal cord.[69]
Heat stressLC-MS/MSSwineOverexpression of HSP70 in intestinal epithelial cells led to changes in the expression of many proteins involved in cell–extracellular matrix interactions, cell adhesion, and apoptosis.[70]
Environmental enrichmentiTRAQ and LC-MS/MSSwinePigs in enriched environments had lower plasma cortisol and lactate levels, indicating a reduced stress response.
Pigs in enriched environments showed changes in neurotransmitter levels in the brain, including decreased noradrenaline and dopamine, and increased serotonin, also suggesting a lower stress response.
[71]
Slaughter methodsMALDI-TOF MSPoultryRitual slaughter without stunning resulted in significantly elevated stress indicators like cortisol and triiodothyronine compared to commercial slaughter with electrical stunning.[72]
Electrical stunning2-DE and MALDI-TOF/TOF MSSheepThe study found 243 proteins that were significantly differentially expressed between stunned and non-stunned (halal) slaughter, with 119 being upregulated and 124 being downregulated.[73]
Chronic circadian disruptionLC-MS/MSBovineDairy cows exposed to circadian rhythm disruption during late gestation showed increased markers of oxidative stress and metabolism in their muscle tissue.[74]
Response of bovine granulosa cells to acute heat stressLC-MS/MSBovineHeat stress triggered oxidative stress-mediated apoptosis in bovine granulosa cells (bGCs), but the cells exhibited a time-dependent recovery of proliferation potential by 48 h.
The study identified 37 differentially regulated metabolites in bGCs in response to acute heat stress, which were involved in bioenergetics support mechanisms and cellular adaptations.
[75]

2.4. Disease Diagnosis and Health Management

Proteomics has emerged as an indispensable tool in the diagnosis and management of diseases in livestock production (Table 2). This field focuses on the large-scale study of proteins, which are crucial for understanding the biological processes and disease mechanisms. The application of proteomics in veterinary medicine is particularly relevant for identifying biomarkers that can facilitate early disease detection, monitor health status, and improve overall livestock management practices. This approach offers immense potential for disease detection by enabling the identification of biomarkers critical for diagnosing and monitoring diseases in domestic animals, by utilizing panels of disease-related proteins rather than relying solely on single-protein assessments [76,77]. The analysis of protein profiles from tissues or bodily fluids allows for the precise identification of disease-specific protein signatures. Moreover, this method serves as a cornerstone for livestock health monitoring, providing a comprehensive understanding of protein expression patterns linked to both normal physiological states and pathological conditions. In cattle, acute-phase proteins such as serpin A3-1, vitronectin-like protein, and complement factor H have been identified as early indicators of subclinical mastitis. Similarly, haptoglobin and apolipoprotein A-I have been recognized as key biomarkers of clinical mastitis, a critical disease affecting animal health and milk production [78,79]. In addition, the promising application of proteomics for mastitis detection was highlighted, demonstrating differential protein expression between the E. coli and S. aureus infection groups [80]. Furthermore, variations in serum Interleukin 8 (IL-8), Interferon-gamma (IFN-γ), transforming growth factor β1 (TGF-β1), and overall haptoglobin concentration patterns have been identified among dairy cows infected with different S. aureus strains, enhancing the capacity to identify pathogenic strains at the strain level in mastitis cases [81]. Proteomic approaches for disease detection have also been successfully applied in swine. Elevated levels of alpha-1-acid glycoprotein and complement factors have been identified as key biomarkers for the early detection of porcine reproductive and respiratory syndrome virus (PRRSV) [82]. Comparatively, it has been reported that the appearance of apolipoprotein A-IV precursor, haptoglobin, and the probable chemoreceptor glutamine deamidase cheD were found after foot and mouth disease virus (FMDV) infection in piglets [83]. Additionally, studies have underscored protein expression patterns associated with diseases in pigs including classical swine fever and peritonitis-induced sepsis using proteomic approaches [84,85]. In poultry, proteomic profiling has identified elevated levels of heat-shock proteins (HSP70 and HSP90) in blood plasma as early indicators of Salmonella infection in chickens [86]. Beyond disease detection, proteomic profiling is also applied in gut health monitoring [87]. A study on broiler chickens found that serum citrulline, IFN-ɤ, and cloacal IgA are key candidate biomarkers for identifying intestinal inflammation, which significantly affects feed efficiency, growth, and intestinal barrier function [88]. Likewise, several proteins associated with inflammation, serum leakage, and tissue damage have been identified as critical markers for intestinal barrier damage [89]. These findings highlight the utility of proteomics in early disease detection, offering livestock producers the ability to implement targeted treatments and biosecurity measures.
Proteomics also contributes to the assessment of feed efficiency by identifying biomarkers linked to metabolic and proteomic changes, thereby reducing feed costs. In slow-growing Korat chickens, the label-free quantitative proteomic analysis of duodenal tissue revealed differentially abundant proteins associated with feed efficiency. The study identified 40 proteins involved in pathways such as glycolysis/gluconeogenesis and oxidative phosphorylation, suggesting their potential as biomarkers for selecting chickens with higher feed utilization efficiency [90]. Similarly, research on Nellore cattle using liver proteomics identified 42 proteins with differential abundance between high and low feed efficiency groups, highlighting pathways like glycolysis/glucogenesis and lipid metabolism. These findings demonstrate the potential of proteomics in developing biomarkers for early identification of efficient animals, ultimately optimizing beef production [91]. Collectively, these studies show that proteomic and metabolomic analyses are powerful tools for identifying biomarkers associated with feed efficiency, contributing to reduced feed costs and improved sustainability in livestock production.
Table 2. Proteomics applications for disease diagnosis and health management in livestock.
Table 2. Proteomics applications for disease diagnosis and health management in livestock.
IndicatorsProteomics Techniques/MethodSpeciesFindingsReferences
NeosporosisLC-MS/MSBovineNeospora caninum infection primarily impacts the host cell’s mitochondrial processes and metabolism.
The low-virulence isolate Nc-Spain1H had a greater influence on the host cell proteome compared to the high-virulence isolate Nc-Spain7.
[92]
Salmonella infectionLC-MS/MSPoultrySalmonella infection increased the abundance of proteins involved in the host’s response to oxidative stress, amino acid metabolism, and lysosomal activity in the spleen of broiler chickens.
Salmonella infection decreased the abundance of proteins involved in cell cycle progression, RNA binding, and cytoskeletal development in the spleen of broiler chickens.
[93]
Pneumonia and mastitis2-DE and MALDI-TOF mass spectrometryBovineIdentified 60 secreted proteins from Mycoplasma bovis, a pathogen that causes pneumonia and mastitis in cattle, and 8 of these proteins were predicted to be virulence-related factors.[94]
Mycobacterium avium paratuberculosis infectionlabel-free LC-MS/MSBovineCows resistant to MAP infection showed higher abundance of TLR2 and MHC class II proteins in their PBMCs, indicating a successful defensive immune response.
Cows persistently infected with MAP showed higher abundance of ITGA2B and KCNMA1 in their PBMCs, suggesting an unsuccessful immune response.
[95]
KetosisLC-MSBovineThe metabolomic analysis showed that cows with clinical ketosis had significant alterations in pathways related to amino acid, carbohydrate, and nucleotide metabolism compared to healthy controls, and these changes were consistent across the transition from prepartum to postpartum.[96]
Toxoplasma gondii infectioniTRAQ labeling, and LC-MS/MSPigOverexpression of two potential anti-T. gondii proteins, HSP70.2 and PDIA3, in swine macrophage cells enhanced resistance to T. gondii infection.[97]
MastitisMALDI-TOF mass spectrometryGoatIdentified the Staphylococcus species present in 19 isolates from subclinical caprine mastitis, with S. epidermidis being the most common at 47.36%.
Henotypic resistance testing showed high resistance to penicillin G (58%), but lower resistance to cefoxitin and oxacillin (both 26.31%). All strains were susceptible to amoxicillin + clavulanic acid.
[98]
Coinfection with Marek’s disease virus and reticuloendotheliosis virusTandem mass tag (TMT) labeling and LC-MS/MSChickenMDV and REV coinfection increased viral replication compared to single infections.
Coinfection led to differential expression of 98 proteins, with 38 upregulated and 60 downregulated.
[99]
Rotavirus infectioniTRAQ and LC-MS/MSPigIdentified 223 differentially accumulated proteins (DAPs) in porcine rotavirus (PoRV)-infected IPEC-J2 cells compared to mock-infected cells, with 125 being up-accumulated and 98 being down-accumulated.[100]
DeltacoronavirusiTRAQ and LC-MS/MSPigA total of 78 differentially expressed proteins (DEPs) were identified in IPEC-J2 cells infected with porcine deltacoronavirus (PDCoV), with 23 being upregulated and 55 being downregulated.[101]
MastitisMALDI-TOF MSBovineMALDI-TOF MS fingerprinting was superior to 16S rRNA gene sequencing for discriminating between different streptococcal species and subspecies involved in bovine mastitis.
MALDI-TOF MS analysis identified three specific protein biomarkers characteristic of the Streptococcus genus and showed variability at both the species and subspecies level.
[102]

3. Applications of Proteomics in Aquaculture

Proteomics, the large-scale study of proteins, has become an indispensable tool in advancing our understanding of fish physiology, particularly in the context of aquaculture. This technology allows researchers to identify key biomarkers associated with various critical traits in aquatic animals, including growth performance, muscle development, immune response, and stress adaptation. By providing insights into the molecular mechanisms underlying these traits, proteomics supports key areas of aquaculture, such as nutrition optimization, farm management, disease control, and food security. The ability to analyze proteins from non-invasive samples, such as mucus and blood, enables researchers to study immune responses, metabolic functions, and reproductive processes without sacrificing fish, making it an ethical and sustainable approach for monitoring fish health and development. Furthermore, proteomic techniques like LC-MS/MS, 2D-PAGE, and MALDI-TOF/TOF offer high-resolution mapping of protein dynamics across different tissues and conditions, enhancing our ability to understand how environmental factors, diets, and diseases affect fish. As aquaculture continues to evolve, proteomics plays a vital role in improving both the quality of farmed fish and the sustainability of aquaculture practices by providing molecular insights that drive improvements in fish health, welfare, and production efficiency.

3.1. Proteomic Insights into Fish Physiology

Proteomics has been employed to identify key biomarkers associated with growth performance, muscle development, immune response, and other critical traits that enhance health and production in aquatic animals [9]. It plays a significant role in various aspects of aquaculture, including nutrition and diet optimization, quality control, farm management, and food security [103,104,105] (Figure 2). Proteomics can be applied non-invasively, using samples such as mucus and blood, to identify proteins involved in immune responses, reproduction, stress adaptation, and metabolic functions without sacrificing fish (Table 3). For example, mucus proteomics stimulate innate immune responses and stress indicators such as galectin-1 and heat shock proteins [106], whereas body fluids (e.g., blood and seminal plasma) reveal proteins linked to reproduction and oxidative stress [107]. Meanwhile, analyses of internal organs, including muscles and lymphoid tissues, provide insights into biochemical responses to environmental and dietary variations [108,109].
Table 3. Proteomics applications in monitoring fish health.
Table 3. Proteomics applications in monitoring fish health.
IndicatorsTechnique UsedSpeciesFindingsReferences
Immune and stress biomarkers in skin mucus2D-PAGE, LC-MS/MSGadus morhua, Cyclopterus
lumpus
Identified immune-related proteins such as galectin-1, cystatin B, heat shock proteins, and peroxiredoxin1.[106,110]
Innate immunity and physiological statusLC-MS/MS, 2-DESparus aurataKey proteins (e.g., actins, glycolytic enzymes, heat shock proteins) linked to immune defense, metabolism, and stress responses.[111,112]
Antimicrobial and proteolytic activityNano-LC-MS/MSScorpaena plumieriIdentified 391 proteins with antimicrobial and venom-related activity, including proteolytic enzymes.[113]
Feeding mechanisms of bloodsucking fishLC-MS/MS, 1D SDS-PAGELampetra moriiNovel proteins involved in blood coagulation suppression and host immune evasion during feeding were identified.[114]
Reproductive processes in seminal plasmaHPLC-ESI-MS/MS, 2D-DIGEOncorhynchus mykiss, Cyprinus carpioIdentified proteins regulating sperm motility, membrane stability, antioxidative defense, and inflammation responses.[107,115]
Growth and muscle developmentDIGE, MALDI-TOF/TOFSparus aurataProteins such as parvalbumins and Wap65 were linked to growth, stress adaptation, and dietary influences.[116,117]
Gill proteome analysisData-independent acquisitionGasterosteus aculeatusExplored molecular differences in gill proteins among ecotypes, revealing adaptation to environmental and morphological variations.[118]
Lymphoid organ functionShotgun proteomicsOncorhynchus mykissProfiling of head kidney and spleen proteins provided insights into immune mechanisms and DNA repair processes.[119]
Confirmation of specific proteinsLC-MS/MSGadus morhuaVerified the presence of natterin-like proteins in skin, liver, and kidney, linked to immune responses.[120]

3.2. Disease Resistance and Immunity

Diseases significantly influence fish health, a critical component of aquaculture welfare, and can impact fish production as well as human health through the food chain [121]. Maintaining fish health is essential for ensuring the quality and safety of aquaculture products. Proteomics plays a pivotal role in understanding the disease pathogenesis and immune responses in fish by identifying protein markers involved in immune regulation [10]. For instance, a proteomic analysis of skin mucus from common carp (Cyprinus carpio) infected with Ichthyophthirius multifiliis revealed the overexpression of metabolic proteins and the downregulation of cytoskeletal proteins, indicating impaired wound healing during the parasitic infection [122]. In Streptococcus agalactiae-infected tilapia, proteomic and transcriptomic studies highlighted the pathogen’s activation of toll-like receptor pathways to evade the immune system, while the host relied on NOD receptor signaling to combat the infection [123]. Similarly, the proteomic profiling of rainbow trout (Oncorhynchus mykiss) infected with Aeromonas salmonicida demonstrated the upregulation of acute-phase response proteins and complement system components [124], while phosphoproteomic analysis identified fibrinogen alpha chain (FGA) proteins as significant in the infection response [125].
Proteomics also aids in understanding skeletal deformities in fish. For example, proteins such as myosin light chain (MYL), apolipoproteins, creatine kinase M-type (CKM), and calmodulin have been associated with skeletal deformities in rainbow trout larvae [126]. Furthermore, immunoproteomics has been instrumental in vaccine development, particularly in identifying immunogenic outer membrane proteins (OMPs). For instance, OMPs such as YaeT, GroEL, and GldJ were identified as potential antigen candidates for protecting fish against Edwardsiella and Flavobacterium infections [127]. A study of the Chinese vaccine strain (J-1) of Aeromonas hydrophila found that Omp38 is a key antigen that enhances both specific and nonspecific immunity in Chinese breams, suggesting its potential as a vaccine candidate [128]. By integrating proteomics and immunoblotting, researchers have identified virulence-related and antigenic proteins in fish and bacterial pathogens. For example, an analysis of outer membrane proteins from Vibrio harveyi identified 11 specific and 4 nonspecific immunogenic proteins in orange-spotted grouper (Epinephelus coioides) sera [129]. Similarly, studies on Francisella noatunensis in Nile tilapia (Oreochromis niloticus) provided insights into host–pathogen interactions [130]. These proteomic approaches offer a molecular-level understanding of disease mechanisms, facilitating targeted disease control strategies and advances in aquaculture health management.

3.3. Environmental Adaptation and Stress Response

Stresses, similar to that experienced by higher vertebrates, significantly impact fish growth and overall health. Climate change exacerbates stress in fish, with contributing factors including fluctuations in temperature, pH, oxygen and carbon dioxide levels, handling methods, overcrowding, and osmotic conditions [131]. Enhancing welfare standards in aquaculture requires effective stress monitoring and optimization of fish stress responses [132]. Understanding stress responses is critical for aquaculture sustainability, fisheries management, and fish welfare [131]. Effective stress management and improved disease resistance are essential for sustainable aquaculture development. Advances in genetic methodologies, including transcriptomics and genomics, provide insights into stress responses, physiological adaptations, and disease resistance mechanisms [133]. Stress management strategies in aquaculture involve optimizing environmental conditions, adopting appropriate handling practices, and utilizing genetically improved stocks [131]. Proteomics plays a crucial role in assessing stress at the molecular level by analyzing protein expression and metabolic processes, to develop strategies for mitigating or preventing adverse effects (Table 4). Proteomics studies have identified proteins associated with energy metabolism, immune function, and signaling pathways that exhibit differential expression under physical and chemical stress. These findings highlight the interplay between the immune and osmoregulatory systems in facilitating fish adaptation to environmental fluctuations [134,135].
Table 4. Application of proteomics in physical and chemical stress agents in aquaculture.
Table 4. Application of proteomics in physical and chemical stress agents in aquaculture.
StressorsTechnique UsedSpeciesFindingsReferences
Physical stress
OvercrowdingLC-MS/MS, 2-DESparus aurataLiver and immune proteins showed significant changes under overcrowding compared to optimized rearing conditions.[136,137]
2-DESalmo salarProteins in muscle and plasma revealed secondary and tertiary stress responses.[138]
X-ray irradiationLC-MSSalmo salarPersistent alterations in gill proteins observed in early-stage rainbow trout.[139]
Freezing conditions2-DE, MALDI-TOF/TOF MSCyprinus carpioSperm proteins supported antioxidative defense, membrane stability, and motility control.[140,141]
High temperatureLTQ-Orbitrap XLSalmo salarProteomic changes in the liver indicated reduced energy expenditure to cope with oxidative stress.[142]
Low temperatureiTRAQTakifugu fasciatusEnhanced oxidative stress response and mitochondrial enzyme activity were linked to cold tolerance.[143]
HypersalinityLC-MSUnspecifiedDifferentially expressed proteins supported osmoregulation, digestion, and mineral regulation in response to salinity changes.[144]
High CO2 and temperature2-DE, Nanoflow LC-MS/MSHippoglossus hippoglossusEnergy metabolism proteins in gills and immune proteins in blood plasma were significantly affected.[145]
Chemical stress
Copper2D-DIGE, iTRAQOncorhynchus mykiss, Cyprinus carpio, Carassius auratus gibelioOxidative stress markers, such as superoxide dismutase and cytochrome c, were identified in gills.[146]
Arsenic2D, MALDI-TOF/TOF MSLabeo rohitaIdentified biomarkers (Apo-A1, A2ML, Wap65) indicate arsenic-induced liver toxicity.[147]
Benzotriazole2-DE, TOF/TOF MS/MSGobiocypris rarusNeurological and liver alterations differed between male and female fish.[148,149]
PAHs (Polycyclic Aromatics)LC-MS/MSGadus morhuaAn albumin-like protein in plasma was linked to PAH-induced stress.[150]
PCB (Polychlorinated Biphenyl)MALDI-TOF MS, MS/MSGadus morhuaProteins linked to neurotoxicity and stress pathways (e.g., Notch signaling) were identified.[151]
Bisphenol ALC-MS/MSDanio rerioProteomic changes in brain tissue suggested complex toxicity mechanisms involving metabolism and transport.[152]
Pesticides (Permethrin, Terbufos)LC-MS/MSPimephales promelasEnrichment of proteins associated with proteasome systems and glycolysis was observed.[153]
Pesticide (Dieldrin)LC-MS/MS, iTRAQDanio rerioMitochondrial proteins showed links to diseases like Parkinson’s and Huntington’s.[154]
Herbicide (Ametryn)SDS-PAGEDanio rerioInduced proteins were linked to glycolysis and lipid transport, while suppressed proteins were associated with oxygen transport.[155]

3.4. Reproductive Biology and Hatchery Management

Effective breeding and hatchery management are critical for the sustainability of aquaculture production. Key components include broodstock management, reproductive control, and genetic enhancement [156]. Advances in hatchery technology have significantly improved production through innovations such as formulated broodfish diets, GnRH-induced spawning, and enhanced micro-diets for larvae [157]. Hormonal manipulations are often required to induce oocyte maturation and spermiation in captive fish [158]. Breeding programs play a vital role in enhancing productivity and sustainability. Hatcheries are indispensable for providing high-quality juvenile fish, which are essential for sustainable fish production [159]. Proteomics has emerged as a valuable tool for investigating the molecular mechanisms underlying reproduction and development in fish. Research in fish proteomics has primarily focused on gametogenesis and the development of novel biomedical applications, significantly influencing aquaculture and fisheries. High-quality seeds lead to healthier individuals, and understanding the molecular basis of reproduction supports advances in aquaculture. Egg quality is a critical determinant of reproductive fitness in aquatic species [160]. Proteomic studies have identified putative protein markers associated with oocyte quality, such as vitellogenin, ApoA1, and MBL proteins, in the ovarian fluid of rainbow trout, using shotgun proteomics [161]. In captive hapuku (Polyprion oxygeneios), iTRAQ-based analyses identified HSP-70 and IDS proteins as potential biomarkers for assessing egg quality [162]. Proteomic studies of sperm and seminal plasma have also gained importance due to their role in sperm quality and fertilization success [163]. Cryopreservation protein markers are essential for the long-term ex situ conservation of germplasm. Proteomic analyses using 1DE and 2DE have investigated alterations in the semen proteome of rainbow trout induced by cryopreservation, aiding in identifying biomarkers for cryoinjury and high-quality germplasm [164]. Fish roe and caviar, derived from fish eggs, are highly nutritious sources of proteins and lipids. Caviar, particularly from sturgeon eggs, is the most expensive, while roe is also harvested from trout, salmon, carp, and other species [165]. Molecular-level studies of fish gametes have provided insights into reproductive biology and help identify quality markers for these food products. The proteome profiling of female sturgeon ova using 2DE and MALDI-TOF-TOF has provided a foundation for identifying molecular markers related to caviar quality [166]. Findings from proteomic studies have contributed to a deeper understanding of reproductive success in fish and help preserve the quality and integrity of these valuable food sources.

4. Challenges and Limitations in the Use of Proteomics in Livestock and Aquaculture Production

Although proteomics has become invaluable in livestock and aquaculture research, offering critical insights into growth, reproduction, disease resistance, and environmental adaptation, its application still faces significant challenges and limitations that hinder its full potential in these fields.

4.1. Management and Maintenance of Proteomics Data

A major challenge in proteomics research is managing and maintaining vast datasets. Meticulous data processing is essential for preventing errors and biases during analysis. For instance, proteomics has been applied to study milk protein composition in sheep (Ovis aries) and dairy cattle (Bos taurus), requiring stringent normalization and scaling techniques to account for individual variability in milk yield and quality [167,168,169]. Similarly, in aquaculture species like Nile tilapia (Oreochromis niloticus), proteomics has been used to analyze the liver and muscle proteins under various feeding regimes [170,171]. However, the absence of robust, species-specific proteomics databases restricts the scope of such studies. While databases like ASlive have been developed to catalog alternative splicing events in livestock, equivalent databases for aquaculture species remain underdeveloped, limiting the identification of novel proteins and biomarkers [12].

4.2. Lack of Comprehensive Phenomics Data

The lack of detailed phenomics data—comprehensive organism-wide phenotypic information—hinders the accurate interpretation of proteomics data. Key traits such as adaptability, fitness, disease resistance, and production performance should be systematically recorded in livestock and aquaculture species [172]. For example, in Gallus gallus domesticus, proteomics could be used to study stress biomarkers during heatwaves [173]. However, inconsistent phenomic data, including body temperature and feed intake, limit comprehensive analysis. Similarly, species such as Pacific white shrimp (Penaeus vannamei) and Atlantic salmon (Salmo salar) could benefit from detailed growth and disease resistance data under varying environmental conditions [174].

4.3. Sample Preparation and Variability

Sample preparation introduces significant variability in proteomics studies. Protein extraction, purification, and fractionation vary across protocols, equipment, and environmental conditions, affecting reproducibility. While gel- and MS-based techniques have been successfully applied in many organisms, quantifying aquatic protein hydrolases remains challenging due to their inherent complexities. In zebrafish (Danio rerio), proteomics studies of the reproductive system often require multiple validation techniques to ensure reliable results [175,176]. In livestock, biological sample heterogeneity—such as blood plasma from different breeds of cattle or pigs—complicates protein biomarkers [177,178]. Additionally, the clinical stage of a disease influences protein expression patterns, as observed in the urinary proteomics of cattle infected with bovine spongiform encephalopathy (BSE), underscoring the need for careful sample selection and study design [179].

4.4. Species-Specific and Tissue-Specific Challenges

Proteomic analyses must address species- and tissue-specific challenges. For instance, aquatic protein hydrolases are often contaminated with nonprotein substances such as polysaccharides, complicating protein extraction and analysis [180]. Similarly, the protein composition of blood plasma in cattle differs significantly from that of muscle tissue, necessitating customized preparation methods for each sample type [177,181].

4.5. Cost and Accessibility

The high costs of proteomics research and technical variability in methodologies hinder its widespread adoption, particularly in developing regions where livestock and aquaculture are economically important industries. For example, mass spectrometry for protein analysis remains prohibitively expensive for many institutions engaged in livestock and aquaculture research [182,183].

5. Future Directions

Proteomics is an increasingly valuable tool in aquaculture and livestock research, offering promising future directions that can advance our understanding of biological processes, improve production efficiency, and enhance health management. Integrating proteomics with other omics technologies and leveraging emerging innovations can enhance the benefit of groundbreaking discoveries and practical applications in the fields of aquaculture and livestock (Figure 3).

5.1. Multi-Omics Integration and Systems Biology Approaches

Integrating proteomics with genomics, transcriptomics, and metabolomics provides a comprehensive understanding of the physiological and molecular mechanisms underlying key traits such as growth, reproduction, and disease resistance in livestock and aquaculture. Multi-omics approaches in livestock have revealed how the milk microbiome modulates host physiology, improves genetic potential, and aids in disease prevention [184,185]. Similarly, combining proteomics with other omics tools in aquaculture enables researchers to track protein expression changes in fish exposed to environmental stressors such as high salinity or extreme temperatures, aiding in the identification of biomarkers for health monitoring and adaption [186,187]. Multi-omics studies in livestock have also assessed the effects of low-dose antibiotics (LDA) on hepatocellular functions, lipid metabolism, and immunity, revealing epigenetic and transcriptional changes critical to growth promotion [188]. Such approaches can be adopted in aquaculture to investigate the molecular effects of nutritional supplements, antibiotics, or environmental toxins, leading to targeted interventions to improve fish welfare and productivity.

5.2. Single-Cell and Spatial Proteomics

Single-cell and spatial proteomics are emerging as transformative technologies in aquaculture and livestock, enabling the investigation of cellular heterogeneity and spatially resolved protein expression patterns in unprecedented detail [177,189]. In livestock, single-cell proteomics has been applied to study immune responses, such as protein-level changes in neutrophils during uterine diseases in cattle, which can guide therapeutic developments [190]. Spatial proteomics, on the other hand, allows for the mapping of protein expression in tissues such as the liver or mammary glands, enhancing our understanding of metabolic functions and disease mechanisms [191]. In aquaculture, these approaches can be used to examine the localized immune responses in pathogen-infected fish tissues or to investigate protein changes in specific organs under environmental stress. Single-cell techniques could also aid in identifying novel therapeutic targets for fish diseases [10].

5.3. Proteogenomics and Proteotranscriptomics

Proteogenomics and proteotranscriptomics, which integrate proteomics with genomic and transcriptomic datasets, enable a more precise, functional annotation of genes and proteins [192,193]. These approaches have been employed in livestock to identify novel antimicrobial resistance genes and proteins associated with meat quality traits [194,195]. In aquaculture, proteogenomics can help decipher the complexity of fish biology by linking protein expression to phenotypic traits such as disease resistance and stress tolerance, enabling more effective breeding programs and health management strategies [196,197]. These insights can also aid in vaccine development, targeted drug delivery systems, and the creation of diagnostic tools for aquaculture species.

5.4. Artificial Intelligence in Proteomics

Artificial intelligence (AI) and machine learning (ML) are revolutionizing proteomics data analysis in both aquaculture and livestock. AI can process complex datasets, identify protein expression patterns, and build predictive models for disease progression or production traits [198,199]. In livestock, AI-based tools such as DIABLO have successfully integrated multi-omics datasets to identify biomarkers related to milk production efficiency and disease resistance [200]. Similarly, in aquaculture, AI could assist in predicting fish health outcomes, optimizing feed formulations, and identifying stress biomarkers for environmental monitoring [201]. These technologies also hold promise for real-time applications, including biosensors or microfluidic devices for rapid field diagnostics in both aquaculture and livestock production systems.

5.5. Standardized Protocols and Data Sharing

The advances in proteomics in aquaculture and livestock requires the development of standardized protocols for sample preparation, data acquisition, and analysis [202,203]. In livestock research, repositories such as mzTab, PRIDE, and PeptideAtlas have facilitated data sharing and cross-study comparisons, which are essential for validating findings and expanding the field [204]. However, a major challenge remains in the lack of universal protocols and species-specific reference databases for aquaculture. Developing such resources for fish species will facilitate cross-species comparisons and accelerate discoveries in fish biology and aquaculture management.

6. Conclusions

Advanced technologies such as proteomics, genomics, and real-time monitoring are revolutionizing precision livestock and aquaculture farming by enabling personalized management strategies, enhanced disease control, and sustainable practices. These innovations address consumer demand for ethical production and align with sustainability goals. However, challenges related to cost and scalability require further research and innovation. Ultimately, these advances have the potential to transform livestock and aquaculture into more sustainable, resilient, and ethically responsible industries.

Author Contributions

Conceptualization, J.J. and N.V.L.; resources, A.T., J.-C.L., S.W., S.C. and L.T.P.K.; writing—original draft preparation, J.J., A.T., J.-C.L., S.W., S.C., C.-J.L., L.T.P.K., N.D.-H., N.V.L. and P.S.; writing—review and editing, J.J., N.V.L., C.-J.L., P.-C.T. and T.M. All authors have read and agreed to the published version of the manuscript.

Funding

This study was partially supported by Chiang Mai University.

Institutional Review Board Statement

Only the literature mentioned was studied in the current article, and no experiments on animals were carried out.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Farmakis, D.; Papingiotis, G.; Parissis, J.; Filippatos, G. Ups and downs in heart failure: The case of proteomics. Eur. J. Heart Fail. 2018, 20, 63–66. [Google Scholar] [CrossRef]
  2. Alsagaby, S.A. Understanding the fundamentals of proteomics. Curr. Top. Pept. Protein. Res. 2019, 20, 25–33. [Google Scholar] [CrossRef]
  3. Sahayarayan, J.J.; Enogochitra, A.; Chandrasekaran, M. Basic Concepts in Proteomics and Applications. Comput. Bioinform. Multidiscip. Appl. 2021, 279–293. [Google Scholar]
  4. Dolomatov, S.; Kazakova, V.P.; Zukow, W. Proteomics. J. Educ. Health Sport 2021, 11, 158–189. [Google Scholar] [CrossRef]
  5. Yokota, H. Applications of proteomics in pharmaceutical research and development. Biochim. Biophys. Acta (BBA)-Proteins Proteom. 2019, 1867, 17–21. [Google Scholar] [CrossRef] [PubMed]
  6. Manes, N.P.; Nita-Lazar, A. Application of targeted mass spectrometry in bottom-up proteomics for systems biology research. J. Proteom. 2018, 189, 75–90. [Google Scholar] [CrossRef]
  7. Channaveerappa, D.; Ngounou Wetie, A.G.; Darie, C.C. Bottlenecks in proteomics: An update. Adv. Mass Spectrom. Biomed. Res. 2019, 1140, 753–769. [Google Scholar]
  8. Mathpal, D. An analysis of proteomics and its applications. Asian J. Multidimens. Res. 2021, 10, 633–640. [Google Scholar] [CrossRef]
  9. Jaiswal, S.; Rasal, K.D.; Chandra, T.; Prabha, R.; Iquebal, M.A.; Rai, A.; Kumar, D. Proteomics in fish health and aquaculture productivity management: Status and future perspectives. Aquaculture 2023, 566, 739159. [Google Scholar] [CrossRef]
  10. Moreira, M.; Schrama, D.; Farinha, A.P.; Cerqueira, M.; Raposo de Magalhaes, C.; Carrilho, R.; Rodrigues, P. Fish pathology research and diagnosis in aquaculture of farmed fish; a proteomics perspective. Animals 2021, 11, 125. [Google Scholar] [CrossRef]
  11. Kumar, S.; Mohanty, A.; Kauhsik, J.; Mukesh, M. Proteomics-based advancements in research toward sustainable production from dairy livestock. In Advances in Animal Experimentation and Modeling; Elsevier: Amsterdam, The Netherlands, 2022; pp. 353–358. [Google Scholar]
  12. Chakraborty, D.; Sharma, N.; Kour, S.; Sodhi, S.S.; Gupta, M.K.; Lee, S.J.; Son, Y.O. Applications of omics technology for livestock selection and improvement. Front. Genet. 2022, 13, 774113. [Google Scholar] [CrossRef]
  13. YANG, Y.-l.; Rong, Z.; Kui, L. Future livestock breeding: Precision breeding based on multi-omics information and population personalization. J. Integr. Agric. 2017, 16, 2784–2791. [Google Scholar] [CrossRef]
  14. Al-Sharif, M.; Radwan, H.; Hendam, B.; Ateya, A. DNA polymorphisms of FGFBP1, leptin, κ-casein, and αs1-casein genes and their association with reproductive performance in dromedary she-camels. Theriogenology 2022, 178, 18–29. [Google Scholar] [CrossRef]
  15. Da Silva Diniz, W.J.; Ward, A.K. 282 Multi-omics approaches to improve animal production. J. Anim. Sci. 2021, 99, 20–21. [Google Scholar] [CrossRef]
  16. Mullen, A.; Stapleton, P.; Corcoran, D.; Hamill, R.; White, A. Understanding meat quality through the application of genomic and proteomic approaches. Meat Sci. 2006, 74, 3–16. [Google Scholar] [CrossRef] [PubMed]
  17. Rexroad, C.; Vallet, J.; Matukumalli, L.K.; Reecy, J.; Bickhart, D.; Blackburn, H.; Boggess, M.; Cheng, H.; Clutter, A.; Cockett, N. Genome to phenome: Improving animal health, production, and well-being—A new USDA blueprint for animal genome research 2018–2027. Front. Genet. 2019, 10, 327. [Google Scholar] [CrossRef]
  18. Peddinti, D.; Nanduri, B.; Kaya, A.; Feugang, J.M.; Burgess, S.C.; Memili, E. Comprehensive proteomic analysis of bovine spermatozoa of varying fertility rates and identification of biomarkers associated with fertility. BMC Syst. Biol. 2008, 2, 1–13. [Google Scholar] [CrossRef]
  19. Itze-Mayrhofer, C.; Brem, G. Quantitative proteomic strategies to study reproduction in farm animals: Female reproductive fluids. J. Proteom. 2020, 225, 103884. [Google Scholar] [CrossRef]
  20. Kosteria, I.; Anagnostopoulos, A.K.; Kanaka-Gantenbein, C.; Chrousos, G.P.; Tsangaris, G.T. The use of proteomics in assisted reproduction. In Vivo 2017, 31, 267–283. [Google Scholar] [CrossRef]
  21. Braundmeier, A.; Miller, D. Invited review: The search is on: Finding accurate molecular markers of male fertility. J. Dairy Sci. 2001, 84, 1915–1925. [Google Scholar] [CrossRef]
  22. Arnold, G.J.; Frohlich, T. Dynamic proteome signatures in gametes, embryos and their maternal environment. Reprod. Fertil. Dev. 2010, 23, 81–93. [Google Scholar] [CrossRef]
  23. Lamy, J.; Labas, V.; Harichaux, G.; Tsikis, G.; Mermillod, P.; Saint-Dizier, M. Regulation of the bovine oviductal fluid proteome. Reproduction 2016, 152, 629–644. [Google Scholar] [CrossRef]
  24. Muñoz, M.; Corrales, F.J.; Caamaño, J.N.; Díez, C.; Trigal, B.; Mora, M.I.; Martín, D.; Carrocera, S.; Gómez, E. Proteome of the early embryo–maternal dialogue in the cattle uterus. J. Proteome Res. 2012, 11, 751–766. [Google Scholar] [CrossRef]
  25. Zachut, M.; Sood, P.; Levin, Y.; Moallem, U. Proteomic analysis of preovulatory follicular fluid reveals differentially abundant proteins in less fertile dairy cows. J. Proteom. 2016, 139, 122–129. [Google Scholar] [CrossRef]
  26. McIlveen, H.; Buchanan, J. The impact of sensory factors on beef purchase and consumption. Nutr. Food Sci. 2001, 31, 286–292. [Google Scholar] [CrossRef]
  27. Gagaoua, M.; Bonnet, M.; Picard, B. Protein array-based approach to evaluate biomarkers of beef tenderness and marbling in cows: Understanding of the underlying mechanisms and prediction. Foods 2020, 9, 1180. [Google Scholar] [CrossRef]
  28. Gagaoua, M.; Hughes, J.; Terlouw, E.C.; Warner, R.D.; Purslow, P.P.; Lorenzo, J.M.; Picard, B. Proteomic biomarkers of beef colour. Trends Food Sci. Technol. 2020, 101, 234–252. [Google Scholar] [CrossRef]
  29. Picard, B.; Gagaoua, M. Meta-proteomics for the discovery of protein biomarkers of beef tenderness: An overview of integrated studies. Food Res. Int. 2020, 127, 108739. [Google Scholar] [CrossRef]
  30. Gagaoua, M.; Monteils, V.; Couvreur, S.; Picard, B. Identification of biomarkers associated with the rearing practices, carcass characteristics, and beef quality: An integrative approach. J. Agric. Food Chem. 2017, 65, 8264–8278. [Google Scholar] [CrossRef]
  31. Abd El-Salam, M.H. Application of proteomics to the areas of milk production, processing and quality control—A review. Int. J. Dairy Technol. 2014, 67, 153–166. [Google Scholar] [CrossRef]
  32. Manso, M.; Léonil, J.; Jan, G.; Gagnaire, V. Application of proteomics to the characterisation of milk and dairy products. Int. Dairy J. 2005, 15, 845–855. [Google Scholar] [CrossRef]
  33. Agregán, R.; Echegaray, N.; López-Pedrouso, M.; Kharabsheh, R.; Franco, D.; Lorenzo, J.M. Proteomic advances in milk and dairy products. Molecules 2021, 26, 3832. [Google Scholar] [CrossRef]
  34. Le, T.T.; Deeth, H.C.; Larsen, L.B. Proteomics of major bovine milk proteins: Novel insights. Int. Dairy J. 2017, 67, 2–15. [Google Scholar] [CrossRef]
  35. Elolimy, A.A.; Zeineldin, M.; Abdelmegeid, M.; Abdelatty, A.M.; Alharthi, A.S.; Bakr, M.H.; Elghandour, M.M.; Salem, A.Z.; Loor, J.J. Metabolomics and Proteomics Signatures in Feed-Efficient Beef and Dairy Cattle. Sustain. Agric. Rev. 54 Anim. Biotechnol. Livest. Prod. 1 2021, 54, 153–165. [Google Scholar]
  36. Pires, B.V.; Reolon, H.G.; Abduch, N.G.; Souza, L.L.; Sakamoto, L.S.; Mercadante, M.E.Z.; Silva, R.M.; Fragomeni, B.O.; Baldi, F.; Paz, C.C. Effects of feeding and drinking behavior on performance and carcass traits in beef cattle. Animals 2022, 12, 3196. [Google Scholar] [CrossRef]
  37. Taiwo, G.A.; Idowu, M.; Denvir, J.; Cervantes, A.P.; Ogunade, I.M. Identification of key pathways associated with residual feed intake of beef cattle based on whole blood transcriptome data analyzed using gene set enrichment analysis. Front. Vet. Sci. 2022, 9, 848027. [Google Scholar] [CrossRef]
  38. Mullins, Y.; Keogh, K.; Blackshields, G.; Kenny, D.A.; Kelly, A.K.; Waters, S.M. Transcriptome assisted label free proteomics of hepatic tissue in response to both dietary restriction and compensatory growth in cattle. J. Proteom. 2021, 232, 104048. [Google Scholar] [CrossRef]
  39. Zhang, J.; Shi, H.; Li, S.; Cao, Z.; Yang, H.; Wang, Y. Integrative hepatic metabolomics and proteomics reveal insights into the mechanism of different feed efficiency with high or low dietary forage levels in Holstein heifers. J. Proteom. 2019, 194, 1–13. [Google Scholar] [CrossRef]
  40. Chagas, L.; Bass, J.; Blache, D.; Burke, C.; Kay, J.; Lindsay, D.; Lucy, M.; Martin, G.; Meier, S.; Rhodes, F. Invited review: New perspectives on the roles of nutrition and metabolic priorities in the subfertility of high-producing dairy cows. J. Dairy Sci. 2007, 90, 4022–4032. [Google Scholar] [CrossRef]
  41. Mukiibi, R.; Vinsky, M.; Keogh, K.A.; Fitzsimmons, C.; Stothard, P.; Waters, S.M.; Li, C. Transcriptome analyses reveal reduced hepatic lipid synthesis and accumulation in more feed efficient beef cattle. Sci. Rep. 2018, 8, 7303. [Google Scholar] [CrossRef]
  42. Serna-García, M.; Fonseca, L.F.S.; Panadero Romero, J.J.; Carretero Asuncion, J.; dos Santos Silva, D.B.; Salatta, B.M.; Frezarim, G.B.; Mercadante, M.E.Z.; Bonilha, S.F.M.; Ferro, J.A. Transcriptome Profiling of the Liver in Nellore Cattle Phenotypically Divergent for RFI in Two Genetic Groups. Animals 2023, 13, 359. [Google Scholar] [CrossRef]
  43. Hiller, B. Recent developments in lipid metabolism in ruminants–the role of fat in maintaining animal health and performance. Anim. Prod. Sci. 2014, 54, 1549–1560. [Google Scholar] [CrossRef]
  44. Wang, Q.; Zhao, X.; Zhang, Z.; Zhao, H.; Huang, D.; Cheng, G.; Yang, Y. Proteomic analysis of physiological function response to hot summer in liver from lactating dairy cows. J. Therm. Biol. 2017, 65, 82–87. [Google Scholar] [CrossRef]
  45. Da Poian, A.T.; Da Poian, A.T.; Castanho, M.A.; Castanho, M.A. Integrative Human Biochemistry: A Textbook for Medical Biochemistry; Springer: Berlin/Heidelberg, Germany, 2015. [Google Scholar]
  46. Moyes, K.; Bendixen, E.; Codrea, M.C.; Ingvartsen, K.L. Identification of hepatic biomarkers for physiological imbalance of dairy cows in early and mid lactation using proteomic technology. J. Dairy Sci. 2013, 96, 3599–3610. [Google Scholar] [CrossRef]
  47. Schäff, C.; Börner, S.; Hacke, S.; Kautzsch, U.; Albrecht, D.; Hammon, H.M.; Röntgen, M.; Kuhla, B.R. Increased anaplerosis, TCA cycling, and oxidative phosphorylation in the liver of dairy cows with intensive body fat mobilization during early lactation. J. Proteome Res. 2012, 11, 5503–5514. [Google Scholar]
  48. Alexandre, P.A.; Naval-Sanchez, M.; Porto-Neto, L.R.; Ferraz, J.B.S.; Reverter, A.; Fukumasu, H. Systems biology reveals NR2F6 and TGFB1 as key regulators of feed efficiency in beef cattle. Front. Genet. 2019, 10, 230. [Google Scholar] [CrossRef]
  49. Taussat, S.; Boussaha, M.; Ramayo-Caldas, Y.; Martin, P.; Venot, E.; Cantalapiedra-Hijar, G.; Hozé, C.; Fritz, S.; Renand, G. Gene networks for three feed efficiency criteria reveal shared and specific biological processes. Genet. Sel. Evol. 2020, 52, 1–14. [Google Scholar] [CrossRef]
  50. Xi, Y.; Yang, Z.; Wu, F.; Han, Z.; Wang, G. Gene expression profiling of hormonal regulation related to the residual feed intake of Holstein cattle. Biochem. Biophys. Res. Commun. 2015, 465, 19–25. [Google Scholar] [CrossRef]
  51. Lamy, E.; Mau, M. Saliva proteomics as an emerging, non-invasive tool to study livestock physiology, nutrition and diseases. J. Proteom. 2012, 75, 4251–4258. [Google Scholar] [CrossRef]
  52. Castillo-Lopez, E.; Pacífico, C.; Sener-Aydemir, A.; Hummel, K.; Nöbauer, K.; Ricci, S.; Rivera-Chacon, R.; Reisinger, N.; Razzazi-Fazeli, E.; Zebeli, Q. Diet and phytogenic supplementation substantially modulate the salivary proteome in dairy cows. J. Proteom. 2023, 273, 104795. [Google Scholar] [CrossRef]
  53. Schulz, B.L.; Cooper-White, J.; Punyadeera, C.K. Saliva proteome research: Current status and future outlook. Crit. Rev. Biotechnol. 2013, 33, 246–259. [Google Scholar] [CrossRef]
  54. Wang, J.; Liang, Y.; Wang, Y.; Cui, J.; Liu, M.; Du, W.; Xu, Y. Computational prediction of human salivary proteins from blood circulation and application to diagnostic biomarker identification. PLoS ONE 2013, 8, e80211. [Google Scholar] [CrossRef]
  55. Marco-Ramell, A.; Pato, R.; Peña, R.; Saco, Y.; Manteca, X.; De La Torre, J.R.; Bassols, A. Identification of serum stress biomarkers in pigs housed at different stocking densities. Vet. J. 2011, 190, e66–e71. [Google Scholar] [CrossRef]
  56. Hazard, D.; Fernandez, X.; Pinguet, J.; Chambon, C.; Letisse, F.; Portais, J.-C.; Wadih-Moussa, Z.; Rémignon, H.; Molette, C. Functional genomics of the muscle response to restraint and transport in chickens. J. Anim. Sci. 2011, 89, 2717–2730. [Google Scholar] [CrossRef]
  57. Cruzen, S.; Pearce, S.; Baumgard, L.; Gabler, N.; Huff-Lonergan, E.; Lonergan, S. Proteomic changes to the sarcoplasmic fraction of predominantly red or white muscle following acute heat stress. J. Proteom. 2015, 128, 141–153. [Google Scholar] [CrossRef]
  58. Zeng, T.; Jiang, X.; Li, J.; Wang, D.; Li, G.; Lu, L.; Wang, G. Comparative proteomic analysis of the hepatic response to heat stress in Muscovy and Pekin ducks: Insight into thermal tolerance related to energy metabolism. PLoS ONE 2013, 8, e76917. [Google Scholar] [CrossRef]
  59. Rakib, M.; Messina, V.; Gargiulo, J.; Lyons, N.; Garcia, S. Potential use of HSP70 as an indicator of heat stress in dairy cows—A review. J. Dairy Sci. 2024, 107, 11597–11610. [Google Scholar] [CrossRef]
  60. Baek, Y.-C.; Kim, M.; Jeong, J.-Y.; Oh, Y.-K.; Lee, S.-D.; Lee, Y.-K.; Ji, S.-Y.; Choi, H. Effects of short-term acute heat stress on physiological responses and heat shock proteins of Hanwoo steer (Korean cattle). J. Anim. Reprod. Biotechnol. 2019, 34, 173–182. [Google Scholar] [CrossRef]
  61. Chen, X.; Shu, H.; Sun, F.; Yao, J.; Gu, X. Impact of heat stress on blood, production, and physiological indicators in heat-tolerant and heat-sensitive dairy cows. Animals 2023, 13, 2562. [Google Scholar] [CrossRef]
  62. Humer, E.; Khol-Parisini, A.; Metzler-Zebeli, B.U.; Gruber, L.; Zebeli, Q. Alterations of the lipid metabolome in dairy cows experiencing excessive lipolysis early postpartum. PLoS ONE 2016, 11, e0158633. [Google Scholar] [CrossRef]
  63. Cairoli, F.; Battocchio, M.; Veronesi, M.C.; Brambilla, D.; Conserva, F.; Eberini, I.; Wait, R.; Gianazza, E. Serum protein pattern during cow pregnancy: Acute-phase proteins increase in the peripartum period. Electrophoresis 2006, 27, 1617–1625. [Google Scholar] [CrossRef]
  64. Yang, Y.; Wang, J.; Bu, D.; Li, S.; Yuan, T.; Zhou, L.; Yang, J.; Sun, P. Comparative proteomics analysis of plasma proteins during the transition period in dairy cows with or without subclinical mastitis after calving. Czech J. Anim. Sci. 2012, 57, 481–489. [Google Scholar] [CrossRef]
  65. Lu, J.; Antunes Fernandes, E.; Páez Cano, A.E.; Vinitwatanakhun, J.; Boeren, S.; van Hooijdonk, T.; van Knegsel, A.; Vervoort, J.; Hettinga, K.A. Changes in milk proteome and metabolome associated with dry period length, energy balance, and lactation stage in postparturient dairy cows. J. Proteome Res. 2013, 12, 3288–3296. [Google Scholar] [CrossRef]
  66. Xu, W.; Van Knegsel, A.; Saccenti, E.; Van Hoeij, R.; Kemp, B.; Vervoort, J. Metabolomics of milk reflects a negative energy balance in cows. J. Proteome Res. 2020, 19, 2942–2949. [Google Scholar] [CrossRef]
  67. Di Luca, A.; Ianni, A.; Henry, M.; Martino, C.; Meleady, P.; Martino, G. Label-free quantitative proteomics and stress responses in pigs—The case of short or long road transportation. PLoS ONE 2022, 17, e0277950. [Google Scholar] [CrossRef]
  68. Fuente-García, C.; Sentandreu, M.A.; Aldai, N.; Oliván, M.; Sentandreu, E. Proteomic pipeline for biomarker hunting of defective bovine meat assisted by liquid chromatography-mass spectrometry analysis and chemometrics. J. Proteom. 2021, 238, 104153. [Google Scholar] [CrossRef]
  69. Herzberg, D.; Strobel, P.; Müller, H.; Meneses, C.; Werner, M.; Bustamante, H. Proteomic profiling of proteins in the dorsal horn of the spinal cord in dairy cows with chronic lameness. PLoS ONE 2020, 15, e0228134. [Google Scholar] [CrossRef]
  70. Yong, Y.; Li, J.; Yu, T.; Fang, B.; Liu, X.; Yu, Z.; Ma, X.; Gooneratne, R.; Abd El-Aty, A.; Ju, X. Overexpression of heat shock protein 70 induces apoptosis of intestinal epithelial cells in heat-stressed pigs: A proteomics approach. J. Therm. Biol. 2022, 108, 103289. [Google Scholar] [CrossRef]
  71. Arroyo, L.; Valent, D.; Carreras, R.; Pato, R.; Sabrià, J.; Velarde, A.; Bassols, A. Neurobiology of environmental enrichment in pigs: Hanges in monoaminergic neurotransmitters in several brain areas and in the hippocampal proteome. J. Proteom. 2020, 229, 103943. [Google Scholar] [CrossRef]
  72. Govindaiah, P.M.; Maheswarappa, N.B.; Banerjee, R.; Muthukumar, M.; Manohar, B.B.; Mishra, B.P.; Sen, A.R.; Biswas, A.K. Decoding halal and jhatka slaughter: Novel insights into welfare and protein biomarkers in slow-growing broiler chicken. J. Sci. Food Agric. 2024, 104, 9160–9168. [Google Scholar] [CrossRef]
  73. Kiran, M.; Maheswarappa, N.B.; Banerjee, R.; Ch, V.; Rapole, S. Impact of stunning before slaughter on expression of skeletal muscles proteome in sheep. Anim. Biotechnol. 2023, 34, 495–502. [Google Scholar] [CrossRef]
  74. McCabe, C.J.; Aryal, U.K.; Casey, T.; Boerman, J. Impact of exposure to chronic light–dark phase shifting circadian rhythm disruption on muscle proteome in periparturient dairy cows. Proteomes 2021, 9, 35. [Google Scholar] [CrossRef] [PubMed]
  75. Sammad, A.; Hu, L.; Luo, H.; Abbas, Z.; Umer, S.; Zhao, S.; Xu, Q.; Khan, A.; Wang, Y.; Zhu, H. Investigation of metabolome underlying the biological mechanisms of acute heat stressed granulosa cells. Int. J. Mol. Sci. 2022, 23, 2146. [Google Scholar] [CrossRef] [PubMed]
  76. Ceciliani, F.; Eckersall, D.; Burchmore, R.; Lecchi, C. Proteomics in veterinary medicine: Applications and trends in disease pathogenesis and diagnostics. Vet Pathol 2014, 51, 351–362. [Google Scholar] [CrossRef]
  77. Bilić, P.; Kuleš, J.; Galan, A.; Gomes de Pontes, L.; Guillemin, N.; Horvatić, A.; Festa Sabes, A.; Mrljak, V.; Eckersall, P.D. Proteomics in veterinary medicine and animal science: Neglected scientific opportunities with immediate impact. Proteomics 2018, 18, 1800047. [Google Scholar] [CrossRef]
  78. O’Reilly, E.L.; Viora, L.; Malcata, F.; Pepler, P.T.; Zadoks, R.; Brady, N.; Hanh, H.Q.; McLaughlin, M.; Horvatic, A.; Gelemanovic, A. Biomarker and proteome analysis of milk from dairy cows with clinical mastitis: Determining the effect of different bacterial pathogens on the response to infection. Res. Vet. Sci. 2024, 172, 105240. [Google Scholar] [CrossRef] [PubMed]
  79. Turk, R.; Piras, C.; Kovačić, M.; Samardžija, M.; Ahmed, H.; De Canio, M.; Urbani, A.; Meštrić, Z.F.; Soggiu, A.; Bonizzi, L. Proteomics of inflammatory and oxidative stress response in cows with subclinical and clinical mastitis. J. Proteom. 2012, 75, 4412–4428. [Google Scholar] [CrossRef]
  80. Ibeagha-Awemu, E.M.; Ibeagha, A.E.; Messier, S.; Zhao, X. Proteomics, genomics, and pathway analyses of Escherichia coli and Staphylococcus aureus infected milk whey reveal molecular pathways and networks involved in mastitis. J. Proteome Res. 2010, 9, 4604–4619. [Google Scholar] [CrossRef]
  81. Kim, Y.; Atalla, H.; Mallard, B.; Robert, C.; Karrow, N. Changes in Holstein cow milk and serum proteins during intramammary infection with three different strains of Staphylococcus aureus. BMC Vet. Res. 2011, 7, 1–13. [Google Scholar] [CrossRef]
  82. Lim, B.; Kim, S.; Lim, K.-S.; Jeong, C.-G.; Kim, S.-C.; Lee, S.-M.; Park, C.-K.; Te Pas, M.F.; Gho, H.; Kim, T.-H. Integrated time-serial transcriptome networks reveal common innate and tissue-specific adaptive immune responses to PRRSV infection. Vet. Res. 2020, 51, 1–18. [Google Scholar] [CrossRef]
  83. Liu, Y.; Zhang, K.; Zheng, H.; Shang, Y.; Guo, J.; Tian, H.; Lu, G.; Jin, Y.; He, J.; Cai, X. Proteomics analysis of porcine serum proteins by LC-MS/MS after foot-and-mouth disease virus (FMDV) infection. J. Vet. Med. Sci. 2011, 73, 1569–1572. [Google Scholar] [CrossRef]
  84. Sun, J.-f.; Shi, Z.-x.; Guo, H.-c.; Li, S.; Tu, C.-c. Proteomic analysis of swine serum following highly virulent classical swine fever virus infection. Virol. J. 2011, 8, 107. [Google Scholar] [CrossRef] [PubMed]
  85. Thongboonkerd, V.; Chiangjong, W.; Mares, J.; Moravec, J.; Tuma, Z.; Karvunidis, T.; Sinchaikul, S.; Chen, S.-T.; Opatrný Jr, K.; Matejovic, M. Altered plasma proteome during an early phase of peritonitis-induced sepsis. Clin. Sci. 2009, 116, 721–730. [Google Scholar] [CrossRef] [PubMed]
  86. Sutton, K.; Nash, T.; Sives, S.; Borowska, D.; Mitchell, J.; Vohra, P.; Stevens, M.P.; Vervelde, L. Disentangling the innate immune responses of intestinal epithelial cells and lamina propria cells to Salmonella Typhimurium infection in chickens. Front. Microbiol. 2023, 14, 1258796. [Google Scholar] [CrossRef] [PubMed]
  87. Ducatelle, R.; Goossens, E.; De Meyer, F.; Eeckhaut, V.; Antonissen, G.; Haesebrouck, F.; Van Immerseel, F. Biomarkers for monitoring intestinal health in poultry: Present status and future perspectives. Vet. Res. 2018, 49, 43. [Google Scholar] [CrossRef]
  88. Baxter, M.F.; Latorre, J.D.; Dridi, S.; Merino-Guzman, R.; Hernandez-Velasco, X.; Hargis, B.M.; Tellez-Isaias, G. Identification of serum biomarkers for intestinal integrity in a broiler chicken malabsorption model. Front. Vet. Sci. 2019, 6, 144. [Google Scholar] [CrossRef]
  89. De Meyer, F.; Eeckhaut, V.; Ducatelle, R.; Dhaenens, M.; Daled, S.; Dedeurwaerder, A.; De Gussem, M.; Haesebrouck, F.; Deforce, D.; Van Immerseel, F. Host intestinal biomarker identification in a gut leakage model in broilers. Vet. Res. 2019, 50, 1–14. [Google Scholar] [CrossRef]
  90. Kaewsatuan, P.; Poompramun, C.; Kubota, S.; Yongsawatdigul, J.; Molee, W.; Uimari, P.; Molee, A. Comparative proteomics revealed duodenal metabolic function associated with feed efficiency in slow-growing chicken. Poult. Sci. 2022, 101, 101824. [Google Scholar] [CrossRef]
  91. Fonseca, L.D.; Eler, J.P.; Pereira, M.A.; Rosa, A.F.; Alexandre, P.A.; Moncau, C.T.; Salvato, F.; Rosa-Fernandes, L.; Palmisano, G.; Ferraz, J.B. Liver proteomics unravel the metabolic pathways related to feed efficiency in beef cattle. Sci. Rep. 2019, 9, 5364. [Google Scholar] [CrossRef]
  92. Regidor-Cerrillo, J.; Xia, D.; Jiménez-Pelayo, L.; García-Sánchez, M.; Collantes-Fernández, E.; Randle, N.; Wastling, J.; Ortega-Mora, L.-M.; Horcajo, P. Proteomic characterization of host-pathogen interactions during bovine trophoblast cell line infection by Neospora caninum. Pathogens 2020, 9, 749. [Google Scholar] [CrossRef]
  93. Adetunji, A.; Casey, T.; Franco, J.; Shah, D.; Fasina, Y. Proteomic analysis of the effect of Salmonella challenge on broiler chicken. Molecules 2022, 27, 7277. [Google Scholar] [CrossRef]
  94. Zubair, M.; Muhamed, S.A.; Khan, F.A.; Zhao, G.; Menghwar, H.; Faisal, M.; Zhang, H.; Zhu, X.; Rasheed, M.A.; Chen, Y. Identification of 60 secreted proteins for Mycoplasma bovis with secretome assay. Microb. Pathog. 2020, 143, 104135. [Google Scholar] [CrossRef] [PubMed]
  95. Korbonits, L.; Kleinwort, K.J.; Amann, B.; Didier, A.; Märtlbauer, E.; Hauck, S.M.; Deeg, C.A. Mycobacterium avium subsp. paratuberculosis infected cows reveal divergent immune response in bovine peripheral blood derived lymphocyte proteome. Metabolites 2022, 12, 924. [Google Scholar] [CrossRef]
  96. Wu, Z.-L.; Chen, S.-Y.; Hu, S.; Jia, X.; Wang, J.; Lai, S.-J. Metabolomic and proteomic profiles associated with ketosis in dairy cows. Front. Genet. 2020, 11, 551587. [Google Scholar] [CrossRef] [PubMed]
  97. He, J.-J.; Ma, J.; Wang, J.-L.; Zhang, F.-K.; Li, J.-X.; Zhai, B.-T.; Elsheikha, H.M.; Zhu, X.-Q. iTRAQ-based quantitative proteomics analysis identifies host pathways modulated during Toxoplasma gondii infection in swine. Microorganisms 2020, 8, 518. [Google Scholar] [CrossRef] [PubMed]
  98. Liang, M.; Lu, M.; Aleem, M.T.; Zhang, Y.; Wang, M.; Wen, Z.; Song, X.; Xu, L.; Li, X.; Yan, R. Identification of excretory and secretory proteins from Haemonchus contortus inducing a Th9 immune response in goats. Vet. Res. 2022, 53, 36. [Google Scholar] [CrossRef]
  99. Du, X.; Zhou, D.; Zhou, J.; Xue, J.; Cheng, Z. Marek’s disease virus and Reticuloendotheliosis virus coinfection enhances viral replication and alters cellular protein profiles. Front. Vet. Sci. 2022, 9, 854007. [Google Scholar] [CrossRef]
  100. Zhou, J.; Huang, S.; Fan, B.; Niu, B.; Guo, R.; Gu, J.; Gao, S.; Li, B. iTRAQ-based proteome analysis of porcine group A rotavirus-infected porcine IPEC-J2 intestinal epithelial cells. J. Proteom. 2021, 248, 104354. [Google Scholar] [CrossRef]
  101. Zhou, X.; Zhou, L.; Ge, X.; Guo, X.; Han, J.; Zhang, Y.; Yang, H. Quantitative proteomic analysis of porcine intestinal epithelial cells infected with porcine deltacoronavirus using iTRAQ-coupled LC-MS/MS. J. Proteome Res. 2020, 19, 4470–4485. [Google Scholar] [CrossRef]
  102. Alnakip, M.E.; Rhouma, N.R.; Abd-Elfatah, E.N.; Quintela-Baluja, M.; Böhme, K.; Fernández-No, I.; Bayoumi, M.A.; Abdelhafez, M.M.; Taboada-Rodriguez, A.; Calo-Mata, P. Discrimination of major and minor streptococci incriminated in bovine mastitis by MALDI-TOF MS fingerprinting and 16S rRNA gene sequencing. Res. Vet. Sci. 2020, 132, 426–438. [Google Scholar] [CrossRef]
  103. Rodrigues, P.M.; Martin, S.A.; Silva, T.S.; Boonanuntanasarn, S.; Schrama, D.; Moreira, M.; Raposo, C. Proteomics in fish and aquaculture research. Proteom. Domest. Anim. Farm Syst. Biol. 2018, 311–338. [Google Scholar]
  104. Carrera, M.; Piñeiro, C.; Martinez, I. Proteomic strategies to evaluate the impact of farming conditions on food quality and safety in aquaculture products. In Mass Spectrometry in Food Analysis; CRC Press: Boca Raton, FL, USA, 2022; pp. 43–59. [Google Scholar]
  105. Nissa, M.U.; Pinto, N.; Parkar, H.; Goswami, M.; Srivastava, S. Proteomics in fisheries and aquaculture: An approach for food security. Food Control 2021, 127, 108125. [Google Scholar] [CrossRef]
  106. Rajan, B.; Fernandes, J.M.; Caipang, C.M.; Kiron, V.; Rombout, J.H.; Brinchmann, M.F. Proteome reference map of the skin mucus of Atlantic cod (Gadus morhua) revealing immune competent molecules. Fish Shellfish Immunol. 2011, 31, 224–231. [Google Scholar] [CrossRef]
  107. Dietrich, M.A.; Arnold, G.J.; Nynca, J.; Fröhlich, T.; Otte, K.; Ciereszko, A. Characterization of carp seminal plasma proteome in relation to blood plasma. J. Proteom. 2014, 98, 218–232. [Google Scholar] [CrossRef] [PubMed]
  108. Carrera, M.; Cañas, B.; Gallardo, J.M. The sarcoplasmic fish proteome: Pathways, metabolic networks and potential bioactive peptides for nutritional inferences. J. Proteom. 2013, 78, 211–220. [Google Scholar] [CrossRef]
  109. Li, T.; Li, J.; Hu, W. Changes in microbiological, physicochemical and muscle proteins of post mortem large yellow croaker (Pseudosciaena crocea). Food Control 2013, 34, 514–520. [Google Scholar] [CrossRef]
  110. Patel, D.M.; Brinchmann, M.F. Skin mucus proteins of lumpsucker (Cyclopterus lumpus). Biochem. Biophys. Rep. 2017, 9, 217–225. [Google Scholar] [CrossRef]
  111. Jurado, J.; Fuentes-Almagro, C.A.; Guardiola, F.A.; Cuesta, A.; Esteban, M.Á.; Prieto-Álamo, M.-J. Proteomic profile of the skin mucus of farmed gilthead seabream (Sparus aurata). J. Proteom. 2015, 120, 21–34. [Google Scholar] [CrossRef]
  112. Sanahuja, I.; Ibarz, A. Skin mucus proteome of gilthead sea bream: A non-invasive method to screen for welfare indicators. Fish Shellfish Immunol. 2015, 46, 426–435. [Google Scholar] [CrossRef]
  113. Borges, M.H.; Andrich, F.; Lemos, P.H.; Soares, T.G.; Menezes, T.N.; Campos, F.V.; Neves, L.X.; Castro-Borges, W.; Figueiredo, S.G. Combined proteomic and functional analysis reveals rich sources of protein diversity in skin mucus and venom from the Scorpaena plumieri fish. J. Proteom. 2018, 187, 200–211. [Google Scholar] [CrossRef]
  114. Li, B.; Gou, M.; Han, J.; Yuan, X.; Li, Y.; Li, T.; Jiang, Q.; Xiao, R.; Li, Q. Proteomic analysis of buccal gland secretion from fasting and feeding lampreys (Lampetra morii). Proteome Sci. 2018, 16, 9. [Google Scholar] [CrossRef]
  115. Nynca, J.; Arnold, G.J.; Fröhlich, T.; Otte, K.; Flenkenthaler, F.; Ciereszko, A. Proteomic identification of rainbow trout seminal plasma proteins. Proteomics 2014, 14, 133–140. [Google Scholar] [CrossRef]
  116. Matos, E.; Silva, T.S.; Wulff, T.; Valente, L.M.; Sousa, V.; Sampaio, E.; Gonçalves, A.; Silva, J.M.; de Pinho, P.G.; Dinis, M.T. Influence of supplemental maslinic acid (olive-derived triterpene) on the post-mortem muscle properties and quality traits of gilthead seabream. Aquaculture 2013, 396, 146–155. [Google Scholar] [CrossRef]
  117. Addis, M.F.; Cappuccinelli, R.; Tedde, V.; Pagnozzi, D.; Porcu, M.C.; Bonaglini, E.; Roggio, T.; Uzzau, S. Proteomic analysis of muscle tissue from gilthead sea bream (Sparus aurata L.) farmed in offshore floating cages. Aquaculture 2010, 309, 245–252. [Google Scholar] [CrossRef]
  118. Li, J.; Levitan, B.; Gomez-Jimenez, S.; Kültz, D. Development of a gill assay library for ecological proteomics of threespine sticklebacks (Gasterosteus aculeatus). Mol. Cell. Proteom. 2018, 17, 2146–2163. [Google Scholar] [CrossRef] [PubMed]
  119. Kumar, G.; Hummel, K.; Razzazi-Fazeli, E.; El-Matbouli, M. Proteome profiles of head kidney and spleen of rainbow trout (Oncorhynchus mykiss). Proteomics 2018, 18, 1800101. [Google Scholar] [CrossRef]
  120. Rajan, B.; Patel, D.M.; Kitani, Y.; Viswanath, K.; Brinchmann, M.F. Novel mannose binding natterin-like protein in the skin mucus of Atlantic cod (Gadus morhua). Fish Shellfish Immunol. 2017, 68, 452–457. [Google Scholar] [CrossRef]
  121. Jennings, S.; Stentiford, G.D.; Leocadio, A.M.; Jeffery, K.R.; Metcalfe, J.D.; Katsiadaki, I.; Auchterlonie, N.A.; Mangi, S.C.; Pinnegar, J.K.; Ellis, T. Aquatic food security: Insights into challenges and solutions from an analysis of interactions between fisheries, aquaculture, food safety, human health, fish and human welfare, economy and environment. Fish Fish. 2016, 17, 893–938. [Google Scholar] [CrossRef]
  122. Saleh, M.; Kumar, G.; Abdel-Baki, A.-A.; Dkhil, M.A.; El-Matbouli, M.; Al-Quraishy, S. Quantitative shotgun proteomics distinguishes wound-healing biomarker signatures in common carp skin mucus in response to Ichthyophthirius multifiliis. Vet. Res. 2018, 49, 1–12. [Google Scholar] [CrossRef]
  123. Wu, N.; Song, Y.-L.; Wang, B.; Zhang, X.-Y.; Zhang, X.-J.; Wang, Y.-L.; Cheng, Y.-Y.; Chen, D.-D.; Xia, X.-Q.; Lu, Y.-S. Fish gut-liver immunity during homeostasis or inflammation revealed by integrative transcriptome and proteome studies. Sci. Rep. 2016, 6, 36048. [Google Scholar] [CrossRef]
  124. Causey, D.R.; Kim, J.-H.; Stead, D.A.; Martin, S.A.; Devlin, R.H.; Macqueen, D.J. Proteomic comparison of selective breeding and growth hormone transgenesis in fish: Unique pathways to enhanced growth. J. Proteom. 2019, 192, 114–124. [Google Scholar] [CrossRef]
  125. Liu, P.-f.; Du, Y.; Meng, L.; Li, X.; Yang, D.; Liu, Y. Phosphoproteomic analyses of kidneys of Atlantic salmon infected with Aeromonas salmonicida. Sci. Rep. 2019, 9, 2101. [Google Scholar] [CrossRef]
  126. Babaheydari, S.B.; Keyvanshokooh, S.; Dorafshan, S.; Johari, S.A. Proteomic analysis of skeletal deformity in diploid and triploid rainbow trout (Oncorhynchus mykiss) larvae. Comp. Biochem. Physiol. Part D Genom. Proteom. 2016, 19, 1–7. [Google Scholar] [CrossRef] [PubMed]
  127. Luo, Z.; Fu, J.P.; Li, N.; Liu, Z.-X.; Qin, T.; Zhang, X.; Nie, P. Immunogenic proteins and their vaccine development potential evaluation in outer membrane proteins (OMPs) of Flavobacterium columnare. Aquac. Fish. 2016, 1, 1–8. [Google Scholar] [CrossRef]
  128. Wang, N.; Yang, Z.; Zang, M.; Liu, Y.; Lu, C. Identification of Omp38 by immunoproteomic analysis and evaluation as a potential vaccine antigen against Aeromonas hydrophila in Chinese breams. Fish Shellfish Immunol. 2013, 34, 74–81. [Google Scholar] [CrossRef]
  129. Pang, H.Y.; Li, Y.; Wu, Z.H.; Jian, J.C.; Lu, Y.S.; Cai, S.H. Immunoproteomic analysis and identification of novel immunogenic proteins from Vibrio harveyi. J. Appl. Microbiol. 2010, 109, 1800–1809. [Google Scholar] [CrossRef] [PubMed]
  130. Shahin, K.; Thompson, K.D.; Inglis, N.F.; Mclean, K.; Ramirez-Paredes, J.G.; Monaghan, S.J.; Hoare, R.; Fontaine, M.; Metselaar, M.; Adams, A. Characterization of the outer membrane proteome of Francisella noatunensis subsp. orientalis. J. Appl. Microbiol. 2018, 125, 686–699. [Google Scholar] [CrossRef]
  131. Sneddon, L.U.; Wolfenden, D.C.; Thomson, J.S. Stress management and welfare. In Fish physiology; Elsevier: Amsterdam, The Netherlands, 2016; Volume 35, pp. 463–539. [Google Scholar]
  132. Barreto, M.O.; Rey Planellas, S.; Yang, Y.; Phillips, C.; Descovich, K. Emerging indicators of fish welfare in aquaculture. Rev. Aquac. 2022, 14, 343–361. [Google Scholar] [CrossRef]
  133. Rise, M.L.; Martyniuk, C.J.; Chen, M. Comparative physiology and aquaculture: Toward Omics-enabled improvement of aquatic animal health and sustainable production. Comp. Biochem. Physiol. Part D Genom. Proteom. 2019, 31, 100603. [Google Scholar] [CrossRef]
  134. Ye, H.; Lin, Q.; Luo, H. Applications of transcriptomics and proteomics in understanding fish immunity. Fish Shellfish Immunol. 2018, 77, 319–327. [Google Scholar] [CrossRef]
  135. Karim, M.; Puiseux-Dao, S.; Edery, M. Toxins and stress in fish: Proteomic analyses and response network. Toxicon 2011, 57, 959–969. [Google Scholar] [CrossRef]
  136. Alves, R.N.; Cordeiro, O.; Silva, T.S.; Richard, N.; de Vareilles, M.; Marino, G.; Di Marco, P.; Rodrigues, P.M.; Conceição, L.E. Metabolic molecular indicators of chronic stress in gilthead seabream (Sparus aurata) using comparative proteomics. Aquaculture 2010, 299, 57–66. [Google Scholar] [CrossRef]
  137. Cordero, H.; Morcillo, P.; Cuesta, A.; Brinchmann, M.F.; Esteban, M.A. Differential proteome profile of skin mucus of gilthead seabream (Sparus aurata) after probiotic intake and/or overcrowding stress. J. Proteom. 2016, 132, 41–50. [Google Scholar] [CrossRef] [PubMed]
  138. Veiseth-Kent, E.; Grove, H.; Færgestad, E.M.; Fjæra, S.O. Changes in muscle and blood plasma proteomes of Atlantic salmon (Salmo salar) induced by crowding. Aquaculture 2010, 309, 272–279. [Google Scholar] [CrossRef]
  139. Smith, R.W.; Moccia, R.D.; Seymour, C.B.; Mothersill, C.E. Irradiation of rainbow trout at early life stages results in a proteomic legacy in adult gills. Part A; proteomic responses in the irradiated fish and in non-irradiated bystander fish. Environ. Res. 2018, 163, 297–306. [Google Scholar] [CrossRef] [PubMed]
  140. Dietrich, M.A.; Ciereszko, A. Proteomic characterization of fresh spermatozoa and supernatant after cryopreservation in relation to freezability of carp (Cyprinus carpio L.) semen. PLoS ONE 2018, 13, e0192972. [Google Scholar] [CrossRef]
  141. Dietrich, M.A.; Irnazarow, I.; Ciereszko, A. Proteomic identification of seminal plasma proteins related to the freezability of carp semen. J. Proteom. 2017, 162, 52–61. [Google Scholar] [CrossRef]
  142. Nuez-Ortín, W.G.; Carter, C.G.; Nichols, P.D.; Cooke, I.R.; Wilson, R. Liver proteome response of pre-harvest Atlantic salmon following exposure to elevated temperature. BMC Genom. 2018, 19, 1–13. [Google Scholar] [CrossRef]
  143. Wen, X.; Zhang, X.; Hu, Y.; Xu, J.; Wang, T.; Yin, S. iTRAQ-based quantitative proteomic analysis of Takifugu fasciatus liver in response to low-temperature stress. J. Proteom. 2019, 201, 27–36. [Google Scholar] [CrossRef]
  144. Schauer, K.L.; Reddam, A.; Xu, E.G.; Wolfe, L.M.; Grosell, M. Interrogation of the Gulf toadfish intestinal proteome response to hypersalinity exposure provides insights into osmoregulatory mechanisms and regulation of carbonate mineral precipitation. Comp. Biochem. Physiol. Part D: Genom. Proteom. 2018, 27, 66–76. [Google Scholar] [CrossRef]
  145. Bresolin de Souza, K.; Jutfelt, F.; Kling, P.; Förlin, L.; Sturve, J. Effects of increased CO2 on fish gill and plasma proteome. PLoS ONE 2014, 9, e102901. [Google Scholar] [CrossRef]
  146. Eyckmans, M.; Benoot, D.; Van Raemdonck, G.A.; Zegels, G.; Van Ostade, X.W.; Witters, E.; Blust, R.; De Boeck, G. Comparative proteomics of copper exposure and toxicity in rainbow trout, common carp and gibel carp. Comp. Biochem. Physiol. Part D Genom. Proteom. 2012, 7, 220–232. [Google Scholar] [CrossRef] [PubMed]
  147. Banerjee, S.; Mahanty, A.; Mohanty, S.; Mazumder, D.G.; Cash, P.; Mohanty, B.P. Identification of potential biomarkers of hepatotoxicity by plasma proteome analysis of arsenic-exposed carp Labeo rohita. J. Hazard. Mater. 2017, 336, 71–80. [Google Scholar] [CrossRef] [PubMed]
  148. Liang, X.; Martyniuk, C.J.; Zha, J.; Wang, Z. Brain quantitative proteomic responses reveal new insight of benzotriazole neurotoxicity in female Chinese rare minnow (Gobiocypris rarus). Aquat. Toxicol. 2016, 181, 67–75. [Google Scholar] [CrossRef] [PubMed]
  149. Liang, X.; Zha, J.; Martyniuk, C.J.; Wang, Z.; Zhao, J. Histopathological and proteomic responses in male Chinese rare minnow (Gobiocypris rarus) indicate hepatotoxicity following benzotriazole exposure. Environ. Pollut. 2017, 229, 459–469. [Google Scholar] [CrossRef]
  150. Enerstvedt, K.S.; Sydnes, M.O.; Pampanin, D.M. Study of the plasma proteome of Atlantic cod (Gadus morhua): Effect of exposure to two PAHs and their corresponding diols. Chemosphere 2017, 183, 294–304. [Google Scholar] [CrossRef]
  151. Berg, K.; Puntervoll, P.; Klungsøyr, J.; Goksøyr, A. Brain proteome alterations of Atlantic cod (Gadus morhua) exposed to PCB 153. Aquat. Toxicol. 2011, 105, 206–217. [Google Scholar] [CrossRef]
  152. Wu, X.; Lu, C.; Dong, X.; Zhang, Z.; Yang, M.; Xu, H. Proteomics analysis of zebrafish brain following chronically exposed to bisphenol A. Toxicol. Environ. Chem. 2017, 99, 469–481. [Google Scholar] [CrossRef]
  153. Biales, A.D.; Bencic, D.C.; Flick, R.L.; Blocksom, K.A.; Lazorchak, J.M.; Lattier, D.L. Proteomic analysis of a model fish species exposed to individual pesticides and a binary mixture. Aquat. Toxicol. 2011, 101, 196–206. [Google Scholar] [CrossRef]
  154. Simmons, D.B.; Cowie, A.M.; Koh, J.; Sherry, J.P.; Martyniuk, C.J. Label-free and iTRAQ proteomics analysis in the liver of zebrafish (Danio rerio) following dietary exposure to the organochlorine pesticide dieldrin. J. Proteom. 2019, 202, 103362. [Google Scholar] [CrossRef]
  155. Lin, H.D.; Hsu, L.S.; Chien, C.C.; Chen, S.C. Proteomic analysis of ametryn toxicity in zebrafish embryos. Environ. Toxicol. 2018, 33, 579–586. [Google Scholar] [CrossRef]
  156. Sonesson, A.K.; Hallerman, E.; Humphries, F.; Hilsdorf, A.; Leskien, D.; Rosendal, K.; Bartley, D.; Hu, X.; Garcia Gomez, R.; Mair, G. Sustainable management and improvement of genetic resources for aquaculture. J. World Aquac. Soc. 2023, 54, 364–396. [Google Scholar] [CrossRef]
  157. Engdaw, F.; Geremew, A. Broodstock nutrition in Nile tilapia and its implications on reproductive efficiency. Front. Aquac. 2024, 3, 1281640. [Google Scholar] [CrossRef]
  158. Mylonas, C.C.; Fostier, A.; Zanuy, S. Broodstock management and hormonal manipulations of fish reproduction. Gen. Comp. Endocrinol. 2010, 165, 516–534. [Google Scholar] [CrossRef]
  159. Uddin, M.N.; Das, A.K.; Sarker, M.A.; Roy, D.; Mithun, M.N.A.S.; Rahman, S.; Uddin, M.S. Problems and Its Related Factors Affecting the Hatchery Owners in Producing Fish Seeds in Rural Bangladesh. Agric. Res. 2024, 1–12. [Google Scholar] [CrossRef]
  160. Yilmaz, O.; Patinote, A.; Nguyen, T.V.; Com, E.; Lavigne, R.; Pineau, C.; Sullivan, C.V.; Bobe, J. Scrambled eggs: Proteomic portraits and novel biomarkers of egg quality in zebrafish (Danio rerio). PLoS ONE 2017, 12, e0188084. [Google Scholar] [CrossRef]
  161. Nynca, J.; Arnold, G.J.; Fröhlich, T.; Ciereszko, A. Shotgun proteomics of rainbow trout ovarian fluid. Reprod. Fertil. Dev. 2015, 27, 504–512. [Google Scholar] [CrossRef] [PubMed]
  162. Kohn, Y.Y.; Symonds, J.E.; Kleffmann, T.; Nakagawa, S.; Lagisz, M.; Lokman, P.M. Proteomic analysis of early-stage embryos: Implications for egg quality in hapuku (Polyprion oxygeneios). Fish Physiol. Biochem. 2015, 41, 1403–1417. [Google Scholar] [CrossRef]
  163. Gombar, R.; Pitcher, T.E.; Lewis, J.A.; Auld, J.; Vacratsis, P.O. Proteomic characterization of seminal plasma from alternative reproductive tactics of Chinook salmon (Oncorhynchus tswatchysha). J. Proteom. 2017, 157, 1–9. [Google Scholar] [CrossRef]
  164. Nynca, J.; Arnold, G.J.; Fröhlich, T.; Ciereszko, A. Cryopreservation-induced alterations in protein composition of rainbow trout semen. Proteomics 2015, 15, 2643–2654. [Google Scholar] [CrossRef]
  165. Vilgis, T.A. The physics of the mouthfeel of caviar and other fish roe. Int. J. Gastron. Food Sci. 2020, 19, 100192. [Google Scholar] [CrossRef]
  166. Keyvanshokooh, S.; Vaziri, B. Proteome analysis of Persian sturgeon (Acipenser persicus) ova. Anim. Reprod. Sci. 2008, 109, 287–297. [Google Scholar] [CrossRef] [PubMed]
  167. Littlejohn, M.D.; Tiplady, K.; Lopdell, T.; Law, T.A.; Scott, A.; Harland, C.; Sherlock, R.; Henty, K.; Obolonkin, V.; Lehnert, K. Expression variants of the lipogenic AGPAT6 gene affect diverse milk composition phenotypes in Bos taurus. PLoS ONE 2014, 9, e85757. [Google Scholar] [CrossRef]
  168. Janjanam, J.; Singh, S.; Jena, M.K.; Varshney, N.; Kola, S.; Kumar, S.; Kaushik, J.K.; Grover, S.; Dang, A.K.; Mukesh, M. Comparative 2D-DIGE proteomic analysis of bovine mammary epithelial cells during lactation reveals protein signatures for lactation persistency and milk yield. PLoS ONE 2014, 9, e102515. [Google Scholar] [CrossRef] [PubMed]
  169. Ha, M.; Sabherwal, M.; Duncan, E.; Stevens, S.; Stockwell, P.; McConnell, M.; Bekhit, A.E.-D.; Carne, A. In-depth characterization of sheep (Ovis aries) milk whey proteome and comparison with cow (Bos taurus). PLoS ONE 2015, 10, e0139774. [Google Scholar] [CrossRef]
  170. Ali, A.; Shaalan, W.M.; Al-Tobasei, R.; Salem, M. Coding and noncoding genes involved in atrophy and compensatory muscle growth in Nile Tilapia. Cells 2022, 11, 2504. [Google Scholar] [CrossRef]
  171. Oliveira, J.; Oliva-Teles, A.; Couto, A. Tracking Biomarkers for the Health and Welfare of Aquaculture Fish. Fishes 2024, 9, 289. [Google Scholar] [CrossRef]
  172. Hartman, S.O. Genomic Studies of Novel Behavior Traits in Lactating Sows and their Relationship to Heat Stress Resilience and Maternal Performance. Master’s Thesis, Purdue University, West Lafayette, IN, USA, 2024. [Google Scholar]
  173. da Silva, J.; Andrade, L.; Rodrigues, P.; Cordeiro, L.; Lima, G.; Lopes, J.; Castillo, E.; Martins, R.; Assunção, A.; Vieira, J. Plasma Proteome Alterations of Laying Hens Subjected to Heat Stress and Fed a Diet Supplemented with Pequi Oil (Caryocar brasiliense Camb.): New Insights in the Identification of Heat Stress Biomarkers. Biomolecules 2024, 14, 1424. [Google Scholar] [CrossRef]
  174. Houston, R.D.; Kriaridou, C.; Robledo, D. Animal board invited review: Widespread adoption of genetic technologies is key to sustainable expansion of global aquaculture. Animal 2022, 16, 100642. [Google Scholar] [CrossRef]
  175. Purushothaman, K.; Das, P.P.; Presslauer, C.; Lim, T.K.; Johansen, S.D.; Lin, Q.; Babiak, I. Proteomics analysis of early developmental stages of zebrafish embryos. Int. J. Mol. Sci. 2019, 20, 6359. [Google Scholar] [CrossRef]
  176. Yan, J.; Ding, Y.; Peng, Z.; Qin, L.; Gu, J.; Wan, C. Systematic Proteomics Study on the Embryonic Development of Danio rerio. J. Proteome Res. 2023, 22, 2814–2826. [Google Scholar] [CrossRef]
  177. Adnane, M.; de Almeida, A.M.; Chapwanya, A. Unveiling the power of proteomics in advancing tropical animal health and production. Trop. Anim. Health Prod. 2024, 56, 182. [Google Scholar] [CrossRef]
  178. Eckersall, P.D. Proteins, proteomics, and the dysproteinemias. Clin. Biochem. Domest. Anim. 2008, 6, 117–155. [Google Scholar]
  179. Plews, M.; Lamoureux, L.; Simon, S.L.; Graham, C.; Ruddat, V.; Czub, S.; Knox, J.D. Factors affecting the accuracy of urine-based biomarkers of BSE. Proteome Sci. 2011, 9, 1–11. [Google Scholar] [CrossRef]
  180. Gajahin Gamage, N.T.; Miyashita, R.; Takahashi, K.; Asakawa, S.; Senevirathna, J.D.M. Proteomic applications in aquatic environment studies. Proteomes 2022, 10, 32. [Google Scholar] [CrossRef] [PubMed]
  181. D’Ambrosio, C.; Arena, S.; Talamo, F.; Ledda, L.; Renzone, G.; Ferrara, L.; Scaloni, A. Comparative proteomic analysis of mammalian animal tissues and body fluids: Bovine proteome database. J. Chromatogr. B 2005, 815, 157–168. [Google Scholar] [CrossRef]
  182. Rodrigues, P.M.; Silva, T.S.; Dias, J.; Jessen, F. Proteomics in aquaculture: Applications and trends. J. Proteom. 2012, 75, 4325–4345. [Google Scholar] [CrossRef] [PubMed]
  183. Soares, R.; Franco, C.; Pires, E.; Ventosa, M.; Palhinhas, R.; Koci, K.; de Almeida, A.M.; Coelho, A.V. Mass spectrometry and animal science: Protein identification strategies and particularities of farm animal species. J. Proteom. 2012, 75, 4190–4206. [Google Scholar] [CrossRef] [PubMed]
  184. Couvillion, S.P.; Mostoller, K.E.; Williams, J.E.; Pace, R.M.; Stohel, I.L.; Peterson, H.K.; Nicora, C.D.; Nakayasu, E.S.; Webb-Robertson, B.-J.M.; McGuire, M.A. Interrogating the role of the milk microbiome in mastitis in the multi-omics era. Front. Microbiol. 2023, 14, 1105675. [Google Scholar] [CrossRef]
  185. Kusumawati, A.; Mustopa, A.; Wibawan, I.; Setiyono, A.; Sudarwanto, M. Metagenomic analysis of pathogen mastitis in cow’s milk from Cicurug, Sukabumi, West Java, Indonesia. IOP Conf. Ser. Earth Environ. Sci. 2021, 762, 012064. [Google Scholar] [CrossRef]
  186. Luo, M.; Zhu, W.; Liang, Z.; Feng, B.; Xie, X.; Li, Y.; Liu, Y.; Shi, X.; Fu, J.; Miao, L. High-temperature stress response: Insights into the molecular regulation of American shad (Alosa sapidissima) using a multi-omics approach. Sci. Total Environ. 2024, 916, 170329. [Google Scholar] [CrossRef] [PubMed]
  187. Ma, Q.; Liu, X.; Feng, W.; Liu, S.; Zhuang, Z. Analyses of the molecular mechanisms associated with salinity adaption of Trachidermus fasciatus through combined iTRAQ-based proteomics and RNA sequencing-based transcriptomics. Prog. Biophys. Mol. Biol. 2018, 136, 40–53. [Google Scholar] [CrossRef]
  188. Hu, Y.; Zhang, Y.; Liu, C.; Qin, R.; Gong, D.; Wang, R.; Zhang, D.; Che, L.; Chen, D.; Xin, G. Multi-omics profiling highlights lipid metabolism alterations in pigs fed low-dose antibiotics. BMC Genet. 2020, 21, 1–12. [Google Scholar] [CrossRef] [PubMed]
  189. Bagger, F.O.; Probst, V. Single cell sequencing in cancer diagnostics. Single-Cell Seq. Methylation: Methods Clin. Appl. 2020, 1255, 175–193. [Google Scholar]
  190. Kania, S.; Reed, S.; Thomford, J.; BonDurant, R.; Hirata, K.; Corbeil, R.; North, M.; Corbeil, L. Degradation of bovine complement C3 by trichomonad extracellular proteinase. Vet. Immunol. Immunopathol. 2001, 78, 83–96. [Google Scholar] [CrossRef]
  191. Codeluppi, S.; Borm, L.E.; Zeisel, A.; La Manno, G.; van Lunteren, J.A.; Svensson, C.I.; Linnarsson, S. Spatial organization of the somatosensory cortex revealed by osmFISH. Nat. Methods 2018, 15, 932–935. [Google Scholar] [CrossRef] [PubMed]
  192. Fancello, L.; Burger, T. An analysis of proteogenomics and how and when transcriptome-informed reduction of protein databases can enhance eukaryotic proteomics. Genome Biol. 2022, 23, 132. [Google Scholar] [CrossRef]
  193. Menschaert, G.; Fenyö, D. Proteogenomics from a bioinformatics angle: A growing field. Mass Spectrom. Rev. 2017, 36, 584–599. [Google Scholar] [CrossRef]
  194. Dhindwal, P.; Thompson, C.; Kos, D.; Planedin, K.; Jain, R.; Jelinski, M.; Ruzzini, A. A neglected and emerging antimicrobial resistance gene encodes for a serine-dependent macrolide esterase. Proc. Natl. Acad. Sci. USA 2023, 120, e2219827120. [Google Scholar] [CrossRef]
  195. Wang, Y.-H.; Byrne, K.A.; Reverter, A.; Harper, G.S.; Taniguchi, M.; McWilliam, S.M.; Mannen, H.; Oyama, K.; Lehnert, S.A. Transcriptional profiling of skeletal muscle tissue from two breeds of cattle. Mamm. Genome 2005, 16, 201–210. [Google Scholar] [CrossRef]
  196. Mohanty, B.P.; Mohanty, S.; Mitra, T.; Mahanty, A.; Ganguly, S.; Singh, S. Omics technology in fisheries and aquaculture. Adv. Fish Res. 2019, 7, 1–30. [Google Scholar]
  197. Vitorino, R. Transforming clinical research: The power of high-throughput omics integration. Proteomes 2024, 12, 25. [Google Scholar] [CrossRef]
  198. Quazi, S. Artificial intelligence and machine learning in precision and genomic medicine. Med. Oncol. 2022, 39, 120. [Google Scholar] [CrossRef] [PubMed]
  199. Mann, M.; Kumar, C.; Zeng, W.-F.; Strauss, M.T. Artificial intelligence for proteomics and biomarker discovery. Cell Syst. 2021, 12, 759–770. [Google Scholar] [CrossRef]
  200. Singh, A.; Shannon, C.P.; Gautier, B.; Rohart, F.; Vacher, M.; Tebbutt, S.J.; Lê Cao, K.-A. DIABLO: An integrative approach for identifying key molecular drivers from multi-omics assays. Bioinformatics 2019, 35, 3055–3062. [Google Scholar] [CrossRef]
  201. Zhang, L.; Guo, W.; Lv, C.; Guo, M.; Yang, M.; Fu, Q.; Liu, X. Advancements in artificial intelligence technology for improving animal welfare: Current applications and research progress. Anim. Res. One Health 2024, 2, 93–109. [Google Scholar] [CrossRef]
  202. Hughes, C.S.; Sorensen, P.H.; Morin, G.B. A standardized and reproducible proteomics protocol for bottom-up quantitative analysis of protein samples using SP3 and mass spectrometry. Proteom. Biomark. Discov. Methods Protoc. 2019, 1959, 65–87. [Google Scholar]
  203. Gawor, A.; Bulska, E. A Standardized Protocol for Assuring the Validity of Proteomics Results from Liquid Chromatography–High-Resolution Mass Spectrometry. Int. J. Mol. Sci. 2023, 24, 6129. [Google Scholar] [CrossRef]
  204. Griss, J.; Jones, A.R.; Sachsenberg, T.; Walzer, M.; Gatto, L.; Hartler, J.; Thallinger, G.G.; Salek, R.M.; Steinbeck, C.; Neuhauser, N. The mzTab data exchange format: Communicating mass-spectrometry-based proteomics and metabolomics experimental results to a wider audience. Mol. Cell. Proteom. 2014, 13, 2765–2775. [Google Scholar] [CrossRef]
Figure 1. Overview of the proteomics workflow in livestock sciences: from data generation and storage to applications. The upper panel illustrates the process of generating and storing proteomics data, including sampling, sample preparation, data acquisition, and storage. The middle panel highlights the analyses with proteomics technologies, including protein extraction, separation, mass spectrometry, data processing, and bioinformatic analysis. The lower panel demonstrates various applications in livestock, such as genetic improvement and breeding programs, feed efficiency and nutritional optimization, stress and welfare management, and disease diagnosis and health management.
Figure 1. Overview of the proteomics workflow in livestock sciences: from data generation and storage to applications. The upper panel illustrates the process of generating and storing proteomics data, including sampling, sample preparation, data acquisition, and storage. The middle panel highlights the analyses with proteomics technologies, including protein extraction, separation, mass spectrometry, data processing, and bioinformatic analysis. The lower panel demonstrates various applications in livestock, such as genetic improvement and breeding programs, feed efficiency and nutritional optimization, stress and welfare management, and disease diagnosis and health management.
Animals 15 01946 g001
Figure 2. Proteomics workflow in aquaculture: from sample collection to applications. The upper panel outlines sample collection and preparation, involving the collection of aquaculture species samples and subsequent preparation steps. The middle panel describes proteomics technologies, including purification, detection, characterization, sequence analysis, quantification, and structural analysis using techniques like chromatography, ELISA, mass spectrometry, and spectroscopy. The lower panel illustrates the broad applications in aquaculture, such as nutrition and diet optimization, disease management, stress response analysis, and hatchery management.
Figure 2. Proteomics workflow in aquaculture: from sample collection to applications. The upper panel outlines sample collection and preparation, involving the collection of aquaculture species samples and subsequent preparation steps. The middle panel describes proteomics technologies, including purification, detection, characterization, sequence analysis, quantification, and structural analysis using techniques like chromatography, ELISA, mass spectrometry, and spectroscopy. The lower panel illustrates the broad applications in aquaculture, such as nutrition and diet optimization, disease management, stress response analysis, and hatchery management.
Animals 15 01946 g002
Figure 3. Emerging approaches in livestock proteomics for sustainable production and animal welfare. The figure highlights key methodologies, including multi-omics integration, single-cell and spatial proteomics, proteogenomics and proteotranscriptomics, artificial intelligence, and machine learning, as well as standardized protocols and data sharing, all contributing to improvements in sustainable production and animal welfare.
Figure 3. Emerging approaches in livestock proteomics for sustainable production and animal welfare. The figure highlights key methodologies, including multi-omics integration, single-cell and spatial proteomics, proteogenomics and proteotranscriptomics, artificial intelligence, and machine learning, as well as standardized protocols and data sharing, all contributing to improvements in sustainable production and animal welfare.
Animals 15 01946 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jitjumnong, J.; Taweechaipaisankul, A.; Lin, J.-C.; Wongchanla, S.; Chuwatthanakhajorn, S.; Lin, C.-J.; Khang, L.T.P.; Linh, N.V.; Sangsawad, P.; Dinh-Hung, N.; et al. An Overview of Advancements in Proteomic Approaches to Enhance Livestock Production and Aquaculture. Animals 2025, 15, 1946. https://doi.org/10.3390/ani15131946

AMA Style

Jitjumnong J, Taweechaipaisankul A, Lin J-C, Wongchanla S, Chuwatthanakhajorn S, Lin C-J, Khang LTP, Linh NV, Sangsawad P, Dinh-Hung N, et al. An Overview of Advancements in Proteomic Approaches to Enhance Livestock Production and Aquaculture. Animals. 2025; 15(13):1946. https://doi.org/10.3390/ani15131946

Chicago/Turabian Style

Jitjumnong, Jakree, Anukul Taweechaipaisankul, Jou-Ching Lin, Supatirada Wongchanla, Schwann Chuwatthanakhajorn, Chih-Jen Lin, Luu Tang Phuc Khang, Nguyen Vu Linh, Papungkorn Sangsawad, Nguyen Dinh-Hung, and et al. 2025. "An Overview of Advancements in Proteomic Approaches to Enhance Livestock Production and Aquaculture" Animals 15, no. 13: 1946. https://doi.org/10.3390/ani15131946

APA Style

Jitjumnong, J., Taweechaipaisankul, A., Lin, J.-C., Wongchanla, S., Chuwatthanakhajorn, S., Lin, C.-J., Khang, L. T. P., Linh, N. V., Sangsawad, P., Dinh-Hung, N., Tang, P.-C., & Moonmanee, T. (2025). An Overview of Advancements in Proteomic Approaches to Enhance Livestock Production and Aquaculture. Animals, 15(13), 1946. https://doi.org/10.3390/ani15131946

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop