Next Article in Journal
Cytogenetic and Molecular Effects of Kaolin’s Foliar Application in Grapevine (Vitis vinifera L.) under Summer’s Stressful Growing Conditions
Previous Article in Journal
Human Endogenous Retroviruses in Neurodegenerative Diseases
Previous Article in Special Issue
The Effect of Short- and Long-Term Cryopreservation on Chicken Primordial Germ Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Unraveling the Genetic Foundations of Broiler Meat Quality: Advancements in Research and Their Impact

Jiangxi Provincial Key Laboratory of Poultry Genetic Improvement, Nanchang Normal University, Nanchang 330032, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work and shared first authorship.
Genes 2024, 15(6), 746; https://doi.org/10.3390/genes15060746
Submission received: 25 April 2024 / Revised: 1 June 2024 / Accepted: 3 June 2024 / Published: 6 June 2024
(This article belongs to the Special Issue Poultry Breeding: Genetics and Genomics)

Abstract

:
As societal progress elevates living standards, the focus on meat consumption has shifted from quantity to quality. In broiler production, optimizing meat quality has become paramount, prompting efforts to refine various meat attributes. Recent advancements in sequencing technologies have revealed the genome’s complexity, surpassing previous conceptions. Through experimentation, numerous genetic elements have been linked to crucial meat quality traits in broiler chickens. This review synthesizes the current understanding of genetic determinants associated with meat quality attributes in broilers. Researchers have unveiled the pivotal insights detailed herein by employing diverse genomic methodologies such as QTL-based investigations, candidate gene studies, single-nucleotide polymorphism screening, genome-wide association studies, and RNA sequencing. These studies have identified numerous genes involved in broiler meat quality traits, including meat lightness (COL1A2 and ACAA2), meat yellowness (BCMO1 and GDPD5), fiber diameter (myostatin and LncIRS1), meat pH (PRDX4), tenderness (CAPN1), and intramuscular fat content (miR-24-3p and ANXA6). Consequently, a comprehensive exploration of these genetic elements is imperative to devise novel molecular markers and potential targets, promising to revolutionize strategies for enhancing broiler meat quality.

1. Introduction

Chicken muscle is one of the foremost sources of meat globally. It is characterized by its high protein content, nutritional richness, and versatile applications, making it a staple in many diets. Broiler meat quality refers to the characteristics obtained from broiler chickens specifically bred and raised for meat production. Chicken meat’s quality is a pivotal determinant of its palatability and sensory attributes. As consumer preferences increasingly veer towards high-quality food products, investigating chicken meat (broiler meat) quality traits garners escalating attention [1,2,3,4,5].
Critical chicken meat quality traits include meat color, water-holding capacity, pH levels, tenderness, and nutrient composition. These attributes influence the meat’s appearance, taste, shelf life, and nutritional profile. Consequently, a comprehensive understanding and exploration of chicken meat qualities are imperative to elevating poultry standards, meeting consumer expectations, and propelling advancements in the poultry industry.
In recent years, significant strides have been made in studying chicken meat quality traits [6,7,8,9]. Researchers have embraced various methodologies to assess, compare, and analyze chicken characteristics encompassing color, texture, pH, fat content, and nutritional composition. These endeavors have unveiled the underlying factors impacting these traits and elucidated strategies and methodologies for enhancing chicken meat quality [10,11].
However, knowledge gaps and challenges persist despite the progress in studying chicken meat quality traits. Notably, the full extent of different chicken hybrids and strains’ influence on meat quality traits remains elusive. At the same time, the effects of feed composition adjustments and breeding management strategies warrant further exploration. Therefore, this paper reviews and synthesizes the latest research advancements in chicken quality traits. Specifically, we will delve into meat color, water retention, pH levels, tenderness, and intramuscular fat. We will scrutinize the factors influencing these traits and elucidate policies and methodologies to enhance chicken meat quality. By providing comprehensive references and scientific insights, we aim to underpin the continual evolution of the chicken industry, offering guidance and inspiration for future research endeavors.

2. Genetic Factors Influencing Meat Quality Traits in Chicken

Genetic determinants play a pivotal role in shaping the qualitative attributes of chicken meat, encompassing characteristics such as flesh color, water retention, pH value, and tenderness. These genetic factors collectively contribute to chicken meat’s overall quality and sensory experience (Table 1).

2.1. Meat Color

Meat color is a vital visual indicator of quality for consumers, with brighter hues often associated with superior nutritional value and desirability [34]. Recent research has delved into the genetic underpinnings of meat color formation, unveiling candidate genes that influence flesh coloration. Studies have identified genes associated with glycolysis, fatty acid metabolism, protein metabolism, and heme concentration as potential regulators of meat color [7,12,14]. Notably, genes such as ACAA2, ACSS3, APOH, ATP5L, CAV3, COL1A2, COX7C, GDPD5, MMP27, RBP4A, SLC2A6, TBXAS1, and UQCR10 have emerged as key players in modulating chicken meat color [6,7,8,12,13,15]. Understanding the genetic basis of meat color variation enables targeted breeding strategies and management practices to enhance meat quality and meet consumer preferences.

2.2. Water-Holding Capacity

Water-holding capacity (WHC) is a critical attribute affecting meat quality, as excessive water loss results in dry and unpalatable meat. Genetic studies have identified candidate genes in muscle cell membrane permeability, aquaporin protein regulation, and hydro-regulation pathways influencing WHC [35,36,37]. Notably, genes such as ABCA1, COL6A1, GSTT1L, and the IIb sodium phosphate cotransporter have regulated WHC and intramuscular fat deposition [35,36]. The IIb sodium phosphate cotransporter reduces the WHC of thigh meat [35].
The promoters of ABCA1, COL6A1, and GSTT1L were found to be hypermethylated, leading to transcriptional downregulation in hens during the later laying period. This downregulation may account for the significant difference in meat quality observed between juvenile and later-laying-period hens [36]. Woody breast muscle disease has also been associated with genetic mutations affecting WHC and overall meat quality [37]. Strategies aimed at modulating gene expression and optimizing muscle hydration levels can enhance WHC and improve the sensory attributes of chicken meat.

2.3. pH Value

The pH value of chicken meat plays a pivotal role in determining its flavor, texture, and overall quality. Genetic studies have identified genes involved in muscle pH regulation, including ACOT9, APOO, CAV3, CUEO, E1BTD2, EIF2S3, KLH15, LOC107052650, MAPKAPK3, PCYT1B, PHKA1, PITX2, PPP1R3A, PRDX4, PRKAG2, RGCC, RHOC, SIX1, SLC25A30, SLC2A1, SLC37A4, and VTI1B, which influence postmortem pH decline and ultimate meat pH [10,11,13,16,17]. Variations in these genes can impact muscle acidification rates and subsequent protein denaturation, affecting WHC and meat texture. Moreover, the temporal expression of genes such as PITX2 and SIX1 has been correlated with meat pH and sensory attributes, underscoring their role in meat quality determination [13].

2.4. Tenderness

Meat tenderness is a crucial factor influencing consumer perception and satisfaction. Genetic studies have identified candidate genes, including CAPN1 and CAST, which regulate muscle protein degradation and influence tenderness [18,19]. Specific genetic variations within these genes, such as the SNP CAPN1 G3535A and G37868A variant of the CAST gene, have been linked to variations in shear force values and intramuscular fat content [18,19]. Understanding the genetic mechanisms underlying meat tenderness enables targeted breeding programs and tenderization strategies to improve overall meat quality.

2.5. Muscle Fiber Diameter

Muscle fiber diameter is a crucial metric in assessing muscle structure and development, directly impacting the chewiness and texture of chicken meat [29,38]. Birds that grow rapidly have more muscle fibers than those that grow more slowly. Fast-growing chickens display larger muscle fiber diameters and a more significant proportion of glycolytic fibers (type II or quick-twitch, glycolytic fibers) than slow-growing chickens [39,40,41]. The fiber type distribution in different muscles influences their dominant metabolic capabilities, subsequently affecting the meat quality of slaughtered animals [42,43].
Understanding the genetic factors influencing muscle fiber diameter is paramount, prompting extensive research into associated genes. Identified candidate genes such as myostatin, IGF-1, and PDK4 are implicated in various stages of muscle growth, fiber formation, and composition [28]. During chicken embryogenesis, myostatin downregulates myf5, Pax3, and MyoD, leading to a deficiency in limb muscle size [31]. Zhu et al. [28] employed qRT-PCR to measure the expression of these genes and utilized scanning electron microscopy for fiber diameter analysis. Their findings suggest that myostatin, IGF-1, and PDK4 may significantly contribute to observed variations in chicken muscle characteristics.
Further investigations by Li et al. [27] highlighted the regulatory role of the hub gene DYNLL2 and its target miR-148-3p in chicken myogenesis. MyoD and MyoG, members of the myogenic regulator family, activate transcription for myogenic agents by binding to a conserved E-box. They are crucial in muscle cell differentiation and development [30]. Interestingly, the long noncoding RNA LncIRS1 has increased muscle fiber diameter and muscle mass in vivo in chickens [29]. Additionally, genetic strategies to modulate muscle hydration levels in broiler meat quality could include targeting myomaker, as elucidated by Luo et al. [30], which is essential for chicken myoblast fusion and subsequently impacts muscle development and possibly muscle hydration.
Moreover, the IGFBP3 gene, integral to mammalian embryonic and postnatal development, inhibits chicken primary myoblast proliferation while promoting differentiation, thereby influencing muscle fiber diameter [8]. Boonlaos et al. [26] found that Toll-like receptor gene TLR2 and TLR4 mRNA expression correlate with the muscle fiber diameter of white striping chicken. Furthermore, the IGF1 gene encoding Insulin-like Growth Factor 1 is crucial in muscle development [29,44]. Studies have revealed a correlation between IGF1 gene polymorphism and chicken muscle fiber diameter, impacting meat texture and taste [44].

2.6. Crude Protein Content

Protein is a fundamental component of the muscles and tissues in chicken meat, directly influencing its nutritional value and overall quality. However, variations in protein composition among different chicken meat products lead to discrepancies in production and consumption. Consequently, researchers have explored genes associated with crude protein content in chicken meat to enhance its protein levels. Leveraging advancements in genomics and bioinformatics, researchers have identified candidate genes linked to crude protein synthesis and metabolism in chicken meat [32,33]. Bordini et al. [32] suggested that breast muscle width and total crude protein concentration can indicate enhanced breast yield and reduced aberrant composition in contemporary broilers. Hub genes within interconnected modules, such as THRAP3, PRPF40A, and BIRC2, play roles in oxidative stress adaptation and apoptosis inhibition, potentially influencing protein levels.
Moreover, genes encoding collagen variants (COL5A2 and COL6A3) and SPARC and MMP2 correlate strongly with crude protein content, indicating their significance in chicken meat production [32]. The ATGL gene, associated with chicken growth and fat traits, is strongly correlated with body weight and pectoral muscle crude protein content [33]. Similarly, studies suggest a relationship between FABP gene polymorphism and chicken meat crude protein content, potentially mediated through fatty acid–protein interactions [45]. Additionally, research indicates a correlation between IGF1 gene polymorphism and chicken meat’s crude protein content, emphasizing its role in protein regulation [46].
Investigating the interplay between candidate genes and chicken meat’s crude protein content offers novel pathways for regulating protein synthesis and metabolism. However, current research on candidate genes in chicken meat remains nascent and requires further experimentation. Future studies, particularly utilizing gene editing techniques, can elucidate these genes’ functionality and regulatory mechanisms, contributing to enhanced chicken meat quality and increased protein content to meet the demand for high-quality meat [47].

2.7. Shear Force

The shear force of chicken meat, which represents the resistance encountered when cutting through the meat, is a crucial indicator for assessing the quality and tenderness of poultry meat. It is a significant parameter in determining meat quality, reflecting the strength and chewiness of muscle fibers. However, this force varies among different breeds and individuals, influenced by genetic factors. Hence, researchers have initiated studies on candidate genes associated with chicken meat’s shear force to unravel its genetic regulatory mechanisms [48].
Understanding the shear force of chicken meat can enhance food texture and cater to diverse consumer preferences. Piórkowska et al. [48] emphasized its importance in determining tenderness, a critical quality criterion. They identified genes like ASB2, associated with meat conversion by promoting protein breakdown, and genes involved in lipogenesis and collagen synthesis, potentially impacting shear forces. For instance, in regulating shear force, the CAST gene exhibited differential expression in lean and fat muscle [48]. Zhang et al. [49] highlighted its potential role in muscle fiber development and chicken shear force. Similarly, the MSTN gene encoding Mystatin, a muscle growth suppressor, showed associations with muscle growth and satellite cell proliferation, potentially influencing shear force [50].
Moreover, comprehensive gene expression analyses revealed essential genes like PRKAG3, ATP2A2, and PPARGC1A implicated in muscle fiber composition and transition [51]. PPARGC1A, in particular, emerged as crucial in muscle fiber dynamics in chickens. However, understanding these candidate genes’ functions and regulatory mechanisms is still in its infancy, necessitating further experimental validation. Future research employing gene editing and transgenic technology could unravel the mechanisms of these genes, optimizing the shear force and texture of chicken meat. Large-scale genome association studies and candidate gene screening hold promise in identifying additional genes related to chicken meat’s shear force, paving the way for enhancing meat quality and meeting consumer demands [47].

2.8. Intramuscular Fat Content

Intramuscular fat (IMF), nestled between muscle fibers, enhances the tenderness and flavor of chicken meat [52]. Research suggests that varying fat content indirectly influences meat quality characteristics such as tenderness, water content, and juiciness while significantly impacting flavor [53]. Optimal IMF levels maintain muscle wettability, ensuring meat softness and tenderness during cooking [53]. A higher fat content enhances juiciness and flavor richness [52]. Liu et al. [54] demonstrated that genetic selection for increased IMF strengthens fatty acid synthesis and transportation, improving chicken quality. Additionally, Lin et al. [22] revealed that miR-24-3p inhibits adipocyte differentiation, affecting intramuscular adipocyte deposition via ANXA6 and, consequently, meat quality. ANXA6 represses intramuscular preadipocyte proliferation while promoting differentiation [22]. Sun et al. [23] identified miR-18b-3p’s influence on IMF formation by targeting the ACOT13 gene, potentially altering chicken meat quality characteristics. miR-18b-3p inhibits intramuscular preadipocyte differentiation, whereas ACOT13 promotes differentiation. IMF deposition in breast muscle is associated with pyruvate and citric acid metabolism through GAPDH, LDHA, GPX1, and GBE1 [21]. In Beijing-You and Cobb chickens, ACADL, ACAD9, HADHA, and HADHB were identified as candidate biomarkers for IMF deposition [24]. Cui et al. [55] explored the biochemical mechanisms underlying IMF deposition in chicken strains with increased fat content. Recently, a co-expression network analysis identified miR-29c-3p-PIK3R1, miR-363-3p-PTEN, miR-6701-3p-PTEN, miR-449c/d-5p-TRAF6, miR-6701-3p-BMPR1B, and miR-1563-WWP1 as key miRNA and mRNA pairs controlling IMF accumulation [20] (Table 1).
The IMF content significantly influences chicken meat quality, with optimal levels enhancing tenderness, though excessive fat content can lead to greasiness. Therefore, controlling intramuscular fat content during breeding is crucial to ensuring favorable meat quality traits in chickens [55].
In conclusion, genetic factors are pivotal in determining meat quality traits in chickens, influencing characteristics such as flesh color, water-holding capacity, pH value, tenderness, muscle fiber diameter, crude protein content, shear force, and IMF content. By unraveling the genetic basis of these traits, researchers can develop targeted breeding strategies and management practices to enhance meat quality, meet consumer preferences, and drive advancements in the poultry industry.

3. Future Perspectives

Research into chicken meat quality traits has advanced significantly in recent decades, yet noteworthy challenges and opportunities warranting further exploration remain. Future investigations can delve into the foundational aspects of chicken meat quality traits, particularly by understanding their genetic underpinnings. By elucidating the genetic mechanisms governing these traits, breeders can enhance chicken meat’s quality and nutritional value through targeted breeding programs. Additionally, emphasis can be placed on refining feeding techniques and nutritional management practices, including optimizing feed composition, utilizing growth promoters, and enhancing feeding environments to maximize chicken meat quality.
The progression of science and technology opens up new avenues for studying chicken meat quality traits, with emerging technologies offering fresh opportunities. Modern biotechnological tools such as gene editing technology and metabolomics can be harnessed to analyze chicken meat quality traits, ultimately contributing to quality improvement. For instance, Tizard et al. [56] demonstrated the potential of gene editing and transgenic technology in controlling disease-related traits and removing allergens, showcasing the transformative impact of modern technologies in various production scenarios. Moreover, studies like that of Xu et al. [57] utilizing gene editing technology to elucidate the role of specific genes in chicken meat development further underscore the potential of these technologies.
However, integrating various analytical approaches can provide deeper insights into chicken meat quality development [20,58]. Research on chicken meat traits should encompass genetic exploration, muscle structure, physiological processes, feeding management, and advancements in nutritional technology. Such holistic investigations will be instrumental in elevating chicken meat quality to meet evolving consumer demands.

4. Summary

This article provides a comprehensive overview of the advancements in understanding the factors influencing chicken meat quality. The studies discussed have identified numerous genes associated with crucial broiler meat quality traits, such as meat lightness (COL1A2 and ACAA2), meat yellowness (BCMO1 and GDPD5), fiber diameter (myostatin and LncIRS1), meat pH (PRDX4), tenderness (CAPN1), and intramuscular fat content (miR-24-3p and ANXA6). These findings underscore the importance of investigating candidate genes to enhance chicken meat quality further and establish a solid foundation for future research.

Author Contributions

Conceptualization, X.X.; preparation of the original draft, T.L. and B.A.A.G.; proofreading and editing, X.X.; project supervision and administration, X.X. and J.X.; financing acquisition, X.X. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Natural Science Foundation of China, grant number 32060740.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

All authors thank colleagues for their help during the research process.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ABCA1—ATP-binding cassette sub-family A member 1, ACAA2—Acetyl-CoA acyltransferase 2, ANXA6—Annexin A6, APOH—Apolipoprotein H, ATP5L—ATP synthase membrane subunit L, BCMO1—Beta-carotene 15,15′-monooxygenase 1, BIRC2—Baculoviral IAP repeat-containing protein 2, CAPN1—Calpain-1 catalytic subunit, CAV3—Caveolin-3, CAST—Calpastatin, COL1A2—Collagen type I alpha 2 chain, COL5A2—Collagen type V alpha 2 chain, COL6A—Collagen type VI, COL6A3—Collagen type VI alpha 3 chain, COX7C—Cytochrome c oxidase subunit 7C, mitochondrial, CUEO—Copper-resistance protein CUEO, DEG—Differentially expressed in gastric cancer, DYNLL2—Dynein light chain LC8-type 2, FABP—Fatty acid-binding protein, GDPD5—Glycerophosphodiester phosphodiesterase domain-containing protein 5, GSTT1L—Glutathione S-transferase theta 1-like, IGF-1—Insulin-like growth factor 1, IGFBP3—Insulin-like growth factor-binding protein 3, MMP2—Matrix metalloproteinase-2, MMP27—Matrix metalloproteinase-27, MyoD—Myoblast determination protein 1, PDK4—Pyruvate dehydrogenase kinase 4, PITX1—Pituitary homeobox 1, PITX2—Pituitary homeobox 2, PPP1R3A—Protein phosphatase 1 regulatory subunit 3A, PRDX4—Peroxiredoxin-4, PRKAG2—Protein kinase AMP-activated non-catalytic subunit gamma 2, PRPF40A—Pre-mRNA-processing factor 40 homolog A, PPKAG—Protein kinase AMP-activated non-catalytic subunit gamma, RBP4A1—Retinol-binding protein 4, SLC2A6—Solute carrier family 2, facilitated glucose transporter member 6, SIX1—Homeobox protein SIX1, SPARC—Secreted protein acidic and rich in cysteine, TBXAS1—Thromboxane A synthase 1, THRAP3—Thyroid hormone receptor-associated protein 3, TLR2—Toll-like receptor 2, TLR4—Toll-like receptor 4, TNNT3—Troponin T3, fast skeletal type, UQCP10—Ubiquinol-cytochrome c reductase complex protein 10, WGCNA—Weighted gene co-expression network analysis.

References

  1. Berri, C.; Wacrenier, N.; Millet, N.; Le Bihan-Duval, E. Effect of Selection for Improved Body Composition on Muscle and Meat Characteristics of Broilers from Experimental and Commercial Lines. Poult. Sci. 2001, 80, 833–838. [Google Scholar] [CrossRef] [PubMed]
  2. Jaturasitha, S.; Srikanchai, T.; Kreuzer, M.; Wicke, M. Differences in Carcass and Meat Characteristics between Chicken Indigenous to Northern Thailand (Black-Boned and Thai Native) and Imported Extensive Breeds (Bresse and Rhode Island Red). Poult. Sci. 2008, 87, 160–169. [Google Scholar] [CrossRef]
  3. da Silva, D.C.F.; de Arruda, A.M.V.; Gonçalves, A.A. Quality Characteristics of Broiler Chicken Meat from Free-Range and Industrial Poultry System for the Consumers. J. Food Sci. Technol. 2017, 54, 1818–1826. [Google Scholar] [CrossRef]
  4. Abdalla, B.A.; Chen, J.; Nie, Q.; Zhang, X. Genomic Insights Into the Multiple Factors Controlling Abdominal Fat Deposition in a Chicken Model. Front. Genet. 2018, 9, 262. [Google Scholar] [CrossRef] [PubMed]
  5. Marchewka, J.; Sztandarski, P.; Solka, M.; Louton, H.; Rath, K.; Vogt, L.; Rauch, E.; Ruijter, D.; de Jong, I.C.; Horbańczuk, J.O. Linking Key Husbandry Factors to the Intrinsic Quality of Broiler Meat. Poult. Sci. 2023, 102, 102384. [Google Scholar] [CrossRef] [PubMed]
  6. Xiong, X.; Liu, X.; Zhu, X.; Tan, Y.; Wang, Z.; Xu, J.; Tu, X.; Rao, Y.; Duan, J.; Zhao, W.; et al. A Mutation in PHKG1 Causes High Drip Loss and Low Meat Quality in Chinese Ningdu Yellow Chickens. Poult. Sci. 2022, 101, 101556. [Google Scholar] [CrossRef]
  7. Sun, J.; Tan, X.; Yang, X.; Bai, L.; Kong, F.; Zhao, G.; Wen, J.; Liu, R. Identification of Candidate Genes for Meat Color of Chicken by Combing Selection Signature Analyses and Differentially Expressed Genes. Genes 2022, 13, 307. [Google Scholar] [CrossRef]
  8. Guo, X.; Zhang, H.; Wang, H.; He, X.-X.; Wang, J.-X.; Wei, W.; Liu, M.; Xu, J.-M.; Liu, Y.-N.; Jiang, R.-S. Identification of Key Modules and Hub Genes Involved in Regulating the Color of Chicken Breast Meat Using WGCNA. Animals 2023, 13, 2356. [Google Scholar] [CrossRef]
  9. Xiao, L.; Qi, L.; Fu, R.; Nie, Q.; Zhang, X.; Luo, W. A Large-Scale Comparison of the Meat Quality Characteristics of Different Chicken Breeds in South China. Poult. Sci. 2024, 103, 103740. [Google Scholar] [CrossRef]
  10. Nadaf, J.; Berri, C.; Dunn, I.; Godet, E.; Le Bihan-Duval, E.; De Koning, D.J. An Expression QTL of Closely Linked Candidate Genes Affects pH of Meat in Chickens. Genetics 2014, 196, 867–874. [Google Scholar] [CrossRef]
  11. Beauclercq, S.; Hennequet-Antier, C.; Praud, C.; Godet, E.; Collin, A.; Tesseraud, S.; Métayer-Coustard, S.; Bourin, M.; Moroldo, M.; Martins, F.; et al. Muscle Transcriptome Analysis Reveals Molecular Pathways and Biomarkers Involved in Extreme Ultimate pH and Meat Defect Occurrence in Chicken. Sci. Rep. 2017, 7, 6447. [Google Scholar] [CrossRef] [PubMed]
  12. Jiang, H.; Yang, D.; Ma, Z. Genome-Wide Association Study of Meat Color Traits in Danzhou Chickens. Chin. J. Anim. Sci. 2022, 58, 117–122. [Google Scholar]
  13. Cao, H.; Zhou, W.; Tan, Y.; Xu, X.; Mao, H.; Dong, X.; Xu, N.; Yin, Z. Chronological Expression of PITX2 and SIX1 Genes and the Association between Their Polymorphisms and Chicken Meat Quality Traits. Animals 2021, 11, 445. [Google Scholar] [CrossRef] [PubMed]
  14. Sun, Y.; Zhao, G.; Liu, R.; Zheng, M.; Hu, Y.; Wu, D.; Zhang, L.; Li, P.; Wen, J. The Identification of 14 New Genes for Meat Quality Traits in Chicken Using a Genome-Wide Association Study. BMC Genom. 2013, 14, 458. [Google Scholar] [CrossRef] [PubMed]
  15. Le Bihan-Duval, E.; Nadaf, J.; Berri, C.; Pitel, F.; Graulet, B.; Godet, E.; Leroux, S.Y.; Demeure, O.; Lagarrigue, S.; Duby, C.; et al. Detection of a Cis [Corrected] eQTL Controlling BCMO1 Gene Expression Leads to the Identification of a QTG for Chicken Breast Meat Color. PLoS ONE 2011, 6, e14825. [Google Scholar] [CrossRef] [PubMed]
  16. Bihan-Duval, E.L.; Hennequet-Antier, C.; Berri, C.; Beauclercq, S.A.; Bourin, M.C.; Boulay, M.; Demeure, O.; Boitard, S. Identification of Genomic Regions and Candidate Genes for Chicken Meat Ultimate pH by Combined Detection of Selection Signatures and QTL. BMC Genom. 2018, 19, 294. [Google Scholar] [CrossRef]
  17. Li, X.; Liu, X.; Nadaf, J.; Le Bihan-Duval, E.; Berri, C.; Dunn, I.; Talbot, R.; De Koning, D.-J. Using Targeted Resequencing for Identification of Candidate Genes and SNPs for a QTL Affecting the pH Value of Chicken Meat. G3 Genes Genomes Genet. 2015, 5, 2085–2089. [Google Scholar] [CrossRef] [PubMed]
  18. Zhou, Y.-G.; Xiong, Y.; Yang, C.-W.; Jiang, X.-S.; Ran, J.-S.; Jin, J.; Wang, Y.; Lan, D.; Ren, P.; Hu, Y.-D.; et al. Experimental Verification of CAPN1 and CAST Gene Polymorphisms in Different Generations of Da-Heng Broilers. Biomed. Res. Int. 2017, 2017, 7968450. [Google Scholar] [CrossRef]
  19. Shu, J.T.; Zhang, M.; Shan, Y.J.; Xu, W.J.; Chen, K.W.; Li, H.F. Analysis of the Genetic Effects of CAPN1 Gene Polymorphisms on Chicken Meat Tenderness. Genet. Mol. Res. 2015, 14, 1393–1403. [Google Scholar] [CrossRef]
  20. Gai, K.; Ge, Y.; Liu, D.; Zhang, H.; Cong, B.; Guo, S.; Liu, Y.; Xing, K.; Qi, X.; Wang, X.; et al. Identification of Key Genes Related to Intramuscular Fat Deposition in Beijing-You Chicken by mRNA and miRNA Transcriptome Analysis. Poult. Sci. 2023, 102, 103035. [Google Scholar] [CrossRef]
  21. Luo, N.; Shu, J.; Yuan, X.; Jin, Y.; Cui, H.; Zhao, G.; Wen, J. Differential Regulation of Intramuscular Fat and Abdominal Fat Deposition in Chickens. BMC Genom. 2022, 23, 308. [Google Scholar] [CrossRef]
  22. Lin, Z.; Tang, Y.; Li, Z.; Li, J.; Yu, C.; Yang, C.; Liu, L.; Wang, Y.; Liu, Y. miR-24-3p Dominates the Proliferation and Differentiation of Chicken Intramuscular Preadipocytes by Blocking ANXA6 Expression. Genes 2022, 13, 635. [Google Scholar] [CrossRef]
  23. Sun, G.; Li, F.; Ma, X.; Sun, J.; Jiang, R.; Tian, Y.; Han, R.; Li, G.; Wang, Y.; Li, Z.; et al. Gga-miRNA-18b-3p Inhibits Intramuscular Adipocytes Differentiation in Chicken by Targeting the ACOT13 Gene. Cells 2019, 8, 556. [Google Scholar] [CrossRef]
  24. Liu, R.; Wang, H.; Liu, J.; Wang, J.; Zheng, M.; Tan, X.; Xing, S.; Cui, H.; Li, Q.; Zhao, G.; et al. Uncovering the Embryonic Development-Related Proteome and Metabolome Signatures in Breast Muscle and Intramuscular Fat of Fast-and Slow-Growing Chickens. BMC Genom. 2017, 18, 816. [Google Scholar] [CrossRef] [PubMed]
  25. Yang, Y.; Xiong, D.; Yao, L.; Zhao, C. An SNP in Exon 11 of Chicken 5′-AMP-Activated Protein Kinase Gamma 3 Subunit Gene Was Associated with Meat Water Holding Capacity. Anim. Biotechnol. 2016, 27, 13–16. [Google Scholar] [CrossRef]
  26. Boonlaos, A.; Uddin, M.J.; Temyord, K.; Jattawa, D.; Kayan, A. Muscle Fiber Characteristics and Expression Level of Troponin T3, Toll-like Receptor 2, and Toll-like Receptor 4 Genes in Chicken Meat with White Striping. Vet. World 2023, 16, 1415–1420. [Google Scholar] [CrossRef] [PubMed]
  27. Li, Y.; Yuan, P.; Fan, S.; Zhai, B.; Jin, W.; Li, D.; Li, H.; Sun, G.; Han, R.; Liu, X.; et al. Weighted Gene Co-Expression Network Indicates That the DYNLL2 Is an Important Regulator of Chicken Breast Muscle Development and Is Regulated by miR-148a-3p. BMC Genom. 2022, 23, 258. [Google Scholar] [CrossRef]
  28. Zhu, M.; Wang, M.; Shao, Y.; Nan, Y.; Blair, H.T.; Morris, S.T.; Zhao, Z.; Zhang, H. Characterization of Muscle Development and Gene Expression in Early Embryos of Chicken, Quail, and Their Hybrids. Gene 2021, 768, 145319. [Google Scholar] [CrossRef]
  29. Li, Z.; Cai, B.; Abdalla, B.A.; Zhu, X.; Zheng, M.; Han, P.; Nie, Q.; Zhang, X. LncIRS1 Controls Muscle Atrophy via Sponging miR-15 Family to Activate IGF1-PI3K/AKT Pathway. J. Cachexia Sarcopenia Muscle 2019, 10, 391–410. [Google Scholar] [CrossRef]
  30. Luo, W.; Li, E.; Nie, Q.; Zhang, X. Myomaker, Regulated by MYOD, MYOG and miR-140-3p, Promotes Chicken Myoblast Fusion. Int. J. Mol. Sci. 2015, 16, 26186–26201. [Google Scholar] [CrossRef]
  31. Amthor, H.; Huang, R.; McKinnell, I.; Christ, B.; Kambadur, R.; Sharma, M.; Patel, K. The Regulation and Action of Myostatin as a Negative Regulator of Muscle Development during Avian Embryogenesis. Dev. Biol. 2002, 251, 241–257. [Google Scholar] [CrossRef]
  32. Bordini, M.; Soglia, F.; Davoli, R.; Zappaterra, M.; Petracci, M.; Meluzzi, A. Molecular Pathways and Key Genes Associated With Breast Width and Protein Content in White Striping and Wooden Breast Chicken Pectoral Muscle. Front. Physiol. 2022, 13, 936768. [Google Scholar] [CrossRef]
  33. Nie, Q.-H.; Fang, M.-X.; Xie, L.; Shen, X.; Liu, J.; Luo, Z.-P.; Shi, J.-J.; Zhang, X.-Q. Associations of ATGL Gene Polymorphisms with Chicken Growth and Fat Traits. J. Appl. Genet. 2010, 51, 185–191. [Google Scholar] [CrossRef] [PubMed]
  34. Harford, I.D.; Pavlidis, H.O.; Anthony, N.B. Divergent Selection for Muscle Color in Broilers. Poult. Sci. 2014, 93, 1059–1066. [Google Scholar] [CrossRef] [PubMed]
  35. Han, J.C.; Yang, X.D.; Zhang, T.; Li, H.; Li, W.L.; Zhang, Z.Y.; Yao, J.H. Effects of 1alpha-Hydroxycholecalciferol on Growth Performance, Parameters of Tibia and Plasma, Meat Quality, and Type IIb Sodium Phosphate Cotransporter Gene Expression of One- to Twenty-One-Day-Old Broilers. Poult. Sci. 2009, 88, 323–329. [Google Scholar] [CrossRef] [PubMed]
  36. Zhang, M.; Yan, F.-B.; Li, F.; Jiang, K.-R.; Li, D.-H.; Han, R.-L.; Li, Z.-J.; Jiang, R.-R.; Liu, X.-J.; Kang, X.-T.; et al. Genome-Wide DNA Methylation Profiles Reveal Novel Candidate Genes Associated with Meat Quality at Different Age Stages in Hens. Sci. Rep. 2017, 7, 45564. [Google Scholar] [CrossRef]
  37. Zhang, Y.; Wang, P.; Xu, X.; Xia, T.; Li, Z.; Zhao, T. Effect of Wooden Breast Myopathy on Water-Holding Capacity and Rheological and Gelling Properties of Chicken Broiler Breast Batters. Poult. Sci. 2020, 99, 3742–3751. [Google Scholar] [CrossRef]
  38. Devatkal, S.K.; Naveena, B.M.; Kotaiah, T. Quality, Composition, and Consumer Evaluation of Meat from Slow-Growing Broilers Relative to Commercial Broilers. Poult. Sci. 2019, 98, 6177–6186. [Google Scholar] [CrossRef] [PubMed]
  39. Dransfield, E.; Sosnicki, A.A. Relationship between Muscle Growth and Poultry Meat Quality. Poult. Sci. 1999, 78, 743–746. [Google Scholar] [CrossRef]
  40. Verdiglione, R.; Cassandro, M. Characterization of Muscle Fiber Type in the Pectoralis Major Muscle of Slow-Growing Local and Commercial Chicken Strains. Poult. Sci. 2013, 92, 2433–2437. [Google Scholar] [CrossRef]
  41. Du, Y.F.; Ding, Q.L.; Li, Y.M.; Fang, W.R. Identification of Differentially Expressed Genes and Pathways for Myofiber Characteristics in Soleus Muscles between Chicken Breeds Differing in Meat Quality. Anim. Biotechnol. 2017, 28, 83–93. [Google Scholar] [CrossRef] [PubMed]
  42. Choi, Y.M.; Ryu, Y.C.; Kim, B.C. Influence of Myosin Heavy- and Light Chain Isoforms on Early Postmortem Glycolytic Rate and Pork Quality. Meat Sci. 2007, 76, 281–288. [Google Scholar] [CrossRef] [PubMed]
  43. Joo, S.T.; Kim, G.D.; Hwang, Y.H.; Ryu, Y.C. Control of Fresh Meat Quality through Manipulation of Muscle Fiber Characteristics. Meat Sci. 2013, 95, 828–836. [Google Scholar] [CrossRef] [PubMed]
  44. Yu, M.; Wang, H.; Xu, Y.; Yu, D.; Li, D.; Liu, X.; Du, W. Insulin-like Growth Factor-1 (IGF-1) Promotes Myoblast Proliferation and Skeletal Muscle Growth of Embryonic Chickens via the PI3K/Akt Signalling Pathway. Cell Biol. Int. 2015, 39, 910–922. [Google Scholar] [CrossRef] [PubMed]
  45. Khosinklang, W.; Kubota, S.; Riou, C.; Kaewsatuan, P.; Molee, A.; Molee, W. Omega-3 Meat Enrichment and L-FABP, PPARA, and LPL Genes Expression Are Modified by the Level and Period of Tuna Oil Supplementation in Slow-Growing Chickens. J. Anim. Sci. 2023, 101, skad267. [Google Scholar] [CrossRef] [PubMed]
  46. Zhou, H.; Mitchell, A.D.; McMurtry, J.P.; Ashwell, C.M.; Lamont, S.J. Insulin-like Growth Factor-I Gene Polymorphism Associations with Growth, Body Composition, Skeleton Integrity, and Metabolic Traits in Chickens. Poult. Sci. 2005, 84, 212–219. [Google Scholar] [CrossRef] [PubMed]
  47. Petracci, M.; Cavani, C. Muscle Growth and Poultry Meat Quality Issues. Nutrients 2012, 4, 1. [Google Scholar] [CrossRef] [PubMed]
  48. Piórkowska, K.; Żukowski, K.; Nowak, J.; Połtowicz, K.; Ropka-Molik, K.; Gurgul, A. Genome-Wide RNA-Seq Analysis of Breast Muscles of Two Broiler Chicken Groups Differing in Shear Force. Anim. Genet. 2016, 47, 68–80. [Google Scholar] [CrossRef]
  49. Zhang, Z.-R.; Jiang, X.-S.; Du, H.-R.; Zhu, Q.; Li, X.-C.; Yang, C.-W.; Liu, Y.-P. Characterization of the Expression Profiles of Calpastatin (CAST) Gene in Chicken. Mol. Biol. Rep. 2012, 39, 1839–1843. [Google Scholar] [CrossRef]
  50. Xu, T.S.; Gu, L.H.; Zhang, X.H.; Ye, B.G.; Liu, X.L.; Hou, S.S. Characterization of Myostatin Gene (MSTN) of Pekin Duck and the Association of Its Polymorphism with Breast Muscle Traits. Genet. Mol. Res. 2013, 12, 3166–3177. [Google Scholar] [CrossRef]
  51. Jingting, S.; Qin, X.; Yanju, S.; Ming, Z.; Yunjie, T.; Gaige, J.; Zhongwei, S.; Jianmin, Z. Oxidative and Glycolytic Skeletal Muscles Show Marked Differences in Gene Expression Profile in Chinese Qingyuan Partridge Chickens. PLoS ONE 2017, 12, e0183118. [Google Scholar] [CrossRef]
  52. Hocquette, J.F.; Gondret, F.; Baéza, E.; Médale, F.; Jurie, C.; Pethick, D.W. Intramuscular Fat Content in Meat-Producing Animals: Development, Genetic and Nutritional Control, and Identification of Putative Markers. Animal 2010, 4, 303–319. [Google Scholar] [CrossRef] [PubMed]
  53. Peña-Saldarriaga, L.M.; Fernández-López, J.; Pérez-Alvarez, J.A. Quality of Chicken Fat By-Products: Lipid Profile and Colour Properties. Foods 2020, 9, 1046. [Google Scholar] [CrossRef]
  54. Liu, L.; Cui, H.; Xing, S.; Zhao, G.; Wen, J. Effect of Divergent Selection for Intramuscular Fat Content on Muscle Lipid Metabolism in Chickens. Animals 2019, 10, 4. [Google Scholar] [CrossRef]
  55. Cui, H.; Liu, L.; Liu, X.; Wang, Y.; Luo, N.; Tan, X.; Zhu, Y.; Liu, R.; Zhao, G.; Wen, J. A Selected Population Study Reveals the Biochemical Mechanism of Intramuscular Fat Deposition in Chicken Meat. J. Anim. Sci. Biotechnol. 2022, 13, 54. [Google Scholar] [CrossRef]
  56. Tizard, M.L.; Jenkins, K.A.; Cooper, C.A.; Woodcock, M.E.; Challagulla, A.; Doran, T.J. Potential Benefits of Gene Editing for the Future of Poultry Farming. Transgenic Res. 2019, 28, 87–92. [Google Scholar] [CrossRef]
  57. Xu, K.; Han, C.X.; Zhou, H.; Ding, J.M.; Xu, Z.; Yang, L.Y.; He, C.; Akinyemi, F.; Zheng, Y.M.; Qin, C.; et al. Effective MSTN Gene Knockout by AdV-Delivered CRISPR/Cas9 in Postnatal Chick Leg Muscle. Int. J. Mol. Sci. 2020, 21, 2584. [Google Scholar] [CrossRef] [PubMed]
  58. Li, J.; Zhang, D.; Yin, L.; Li, Z.; Yu, C.; Du, H.; Jiang, X.; Yang, C.; Liu, Y. Integration Analysis of Metabolome and Transcriptome Profiles Revealed the Age-Dependent Dynamic Change in Chicken Meat. Food Res. Int. 2022, 156, 111171. [Google Scholar] [CrossRef]
Table 1. Genes associated with meat quality attributes in chickens.
Table 1. Genes associated with meat quality attributes in chickens.
AttributesGene SymbolFunctionStudy Model or Breeds Technology Used for
Identification
Refs.
Meat colorATP5L, UQCR10, COX7CPositively affects breast meat lightness and yellownessYellow-feather chickens (Huainan and Wannan chickens)RNA-Seq/WGCNA[8]
CAV3, RBP4A and APOHNegatively affects breast meat lightness and yellowness
TBXAS1Regulates breast muscle rednessWhite-feathered chickens (Arbor Acres and Line B) and yellow-feathered chickens (Beijing-You and Jingxing Yellow) exhibit notable variations in meat colormRNA sequencing/Selection signature analysis/qPCR[7]
SLC2A6, MMP27 and COL1A2Regulates breast
muscle lightness
GDPD5Positively affects breast
muscle yellowness
PHKG1Associated with breast muscle lightnessNingdu yellow chickensAssociation analysis of SNPs using DNA resequencing[6]
ACAA2Regulates breast muscle lightnessDanzhou chickensGWAS[12]
ACSS3Regulates breast muscle redness
PITX2CT genotype associated with highest breast muscle lightnessWuliang Mountain Black-bone chickensAssociation analysis of SNPs by DNA direct sequencing[13]
COL1A2Associated with breast muscle lightnessF2 cross population between Beijing-You chickens and Cobb-VantressGWAS[14]
BCMO1Associated with breast
muscle yellowness
F2 cross population between high-growth and low-growth chicken linesExpression QTL analysis[15]
pH valueSIX1GG genotype associated with the highest breast muscle pHWuliang Mountain Black-bone chickensAssociation analysis of SNPs by DNA direct sequencing[13]
PITX2CT genotype associated with highest breast muscle pH.
PPP1R3A and SLC37A4Associated with muscle pH, potentially reduces pHTwo broiler lines with muscle high pH or low pH valuesQTL and selection signature[16]
MAPKAPK3, SLC25A30, RGCCRegulates breast muscle pHTwo broiler lines with muscle high pH (~6.34) or low pH (~5.55) valuesMicroarray[11]
PRDX4, EIF2S3, PCYT1B, E1BTD2Associated with muscle pH, potentially reduces pHF2 cross population between high-growth and low-growth chicken linesTarget enrichment analyses for a QTL and next-generation sequencing[17]
PRDX4Associated with muscle pH and its expression downregulatedF2 cross population between high-growth and low-growth chicken linesExpression QTL[10]
ACOT9
KLH15, APOOAssociated with muscle pH and its expression upregulated
TendernessCAPN1Associated with breast meat tendernessDa-Heng BroilerAssociation analysis of SNPs by DNA direct sequencing[18]
CAPN1Associated with breast meat tendernessQingyuan partridge chicken and Recessive White chickenWarner–Bratzler shear force/SNP by DNA direct sequencing[19]
Intramuscular fat contentmiR-29c-3p-PIK3R1, miR-363-3p-PTEN, miR-6701-3p-PTEN, miR-449c/d-5p-TRAF6, miR-6701-3p-BMPR1B, and miR-1563-WWP1Regulates breast muscle IMF depositionBeijing-You chickenmRNA-Seq/small RNA-seq/WGCNA[20]
LDHA, GPX1, GBE1Upregulated in high IMFHigh and low IMF chickens from breast muscleRNA-seq/network analysis[21]
miR-24-3pPromotes intramuscular preadipocyte proliferation while inhibiting its differentiation by targeting ANXA6In vitroOverexpression and knockdown[22]
ANXA6Inhibits intramuscular preadipocyte proliferation and promotes its differentiationIn vitroOverexpression and knockdown[22]
miR-18b-3pInhibits intramuscular preadipocyte differentiationIn vitromiRNA-Seq/miR-qPCR [23]
ACOT13Enhances intramuscular preadipocyte differentiationIn vitromiRNA-Seq/Luciferase assay/qPCR[23]
ACADL, ACAD9, HADHA and HADHBRegulates IMF deposition. Upregulated before day 1 and downregulated from day 1 to day 14 after hatchBeijing-You (slow-growing) and Cobb (fast-growing) chickenMass spectrometry-based approaches[24]
Water Holding CapacityPHKG1Causes high drip lossNingdu yellow chickensAssociation analysis of SNP by DNA direct sequencing[6]
PRKAG3Associated with water-holding capacityWhite Plymouth RockAssociation analysis of SNP by DNA direct sequencing[25]
Muscle fiber diameterTLR2 and TLR4mRNA expression correlates with muscle fiber diameterCommercial poultry processing plantMuscle cross-sectional area/qPCR[26]
miR-148a-3pmiR-148a-3p upregulates MYHC expression by targeting DYNLL2, promotes myoblast differentiationIn vitromRNA and miRNA analysis/overexpression/inhibition[27]
DYNLL2Reduces MYHC expression, inhibits myoblast differentiationIn vitromRNA and miRNA analysis/overexpression/inhibition[27]
Myostatin, IGF-1, and PDK4Expression correlates with muscle fiber diameter during embryogenesisIn vitroElectron microscopy/qPCR[28]
LncIRS1Increases muscle mass and mean muscle fiberIn vivoBioinformatic analysis/overexpression/shRNA/fiber cross-sectional area[29]
miR-140-3pInhibits myoblast fusion by targeting myomakerIn vitromiRNA mimic and inhibitor/luciferase assay[30]
MyomakerPromotes myoblast fusionIn vitroOverexpression/siRNA[30]
MyostatinMyostatin downregulates Myf5, Pax3, and MyoD, resulting in a deficiency in limb muscle sizeIn vivoOverexpression/wholemount in situ hybridization[31]
Crude protein contentCOL5A2, COL6A3, SPARC and MMP2mRNA levels covaried with crude protein content in normal and White Striping/Wooden BreastsRoss 708 broilerMicroarray data collection/Co-expression network analysis; cytoHubba[32]
ATGLAssociated with crude protein content of breast muscleF2 cross population between White Recessive Rock and Xinghua chickensAssociation analysis of SNP by DNA direct sequencing[33]
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

Lu, T.; Abdalla Gibril, B.A.; Xu, J.; Xiong, X. Unraveling the Genetic Foundations of Broiler Meat Quality: Advancements in Research and Their Impact. Genes 2024, 15, 746. https://doi.org/10.3390/genes15060746

AMA Style

Lu T, Abdalla Gibril BA, Xu J, Xiong X. Unraveling the Genetic Foundations of Broiler Meat Quality: Advancements in Research and Their Impact. Genes. 2024; 15(6):746. https://doi.org/10.3390/genes15060746

Chicago/Turabian Style

Lu, Tian, Bahareldin Ali Abdalla Gibril, Jiguo Xu, and Xinwei Xiong. 2024. "Unraveling the Genetic Foundations of Broiler Meat Quality: Advancements in Research and Their Impact" Genes 15, no. 6: 746. https://doi.org/10.3390/genes15060746

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