Skip to main content

Candidate genes for mastitis resistance in dairy cattle: a data integration approach

Abstract

Background

Inflammation of the mammary tissue (mastitis) is one of the most detrimental health conditions in dairy ruminants and is considered the most economically important infectious disease of the dairy sector. Improving mastitis resistance is becoming an important goal in dairy ruminant breeding programmes. However, mastitis resistance is a complex trait and identification of mastitis-associated alleles in livestock is difficult. Currently, the only applicable approach to identify candidate loci for complex traits in large farm animals is to combine different information that supports the functionality of the identified genomic regions with respect to a complex trait.

Methods

To identify the most promising candidate loci for mastitis resistance we integrated heterogeneous data from multiple sources and compiled the information into a comprehensive database of mastitis-associated candidate loci. Mastitis-associated candidate genes reported in association, expression, and mouse model studies were collected by searching the relevant literature and databases. The collected data were integrated into a single database, screened for overlaps, and used for gene set enrichment analysis.

Results

The database contains candidate genes from association and expression studies and relevant transgenic mouse models. The 2448 collected candidate loci are evenly distributed across bovine chromosomes. Data integration and analysis revealed overlaps between different studies and/or with mastitis-associated QTL, revealing promising candidate genes for mastitis resistance.

Conclusion

Mastitis resistance is a complex trait influenced by numerous alleles. Based on the number of independent studies, we were able to prioritise candidate genes and propose a list of the 22 most promising. To our knowledge this is the most comprehensive database of mastitis associated candidate genes and could be helpful in selecting genes for functional validation studies.

Introduction

Inflammation of the mammary tissue is one of the most common harmful health issues in dairy ruminants and is considered the most economically important infectious disease of the dairy sector. The major consequences of mastitis are reduced milk production and veterinary costs [1], animal welfare concerns [2, 3], extensive use of antibiotics contributing to increasing antimicrobial resistance [4], and impact on the safety and quality of milk and dairy products [5, 6]. The improvement in milk yield has been tremendous in the last decades, doubling milk production in the last fifty years [7]. However, intensive selection focused on milk yield led to deterioration of animal fertility and mammary health traits [8, 9]. Selection for high milk flow, preferred due to machine milking, resulted in weakening of the mammary streak canal sphincter that represents a physical barrier for pathogen entry [10]. Characteristics of milk flow were clearly associated with mammary health status and correlated indicators (e.g., somatic cell score – SCC) [11,12,13]. Unfavourable correlations between milk production traits and mastitis can be offset to some extent by appropriate herd management [14], but mammary infections and fertility problems are generally more common in high producing herds [15,16,17]. The situation is somewhat different in the Nordic countries, where direct and indirect mastitis indicators (such as clinical mastitis and somatic cell count) are routinely measured and have been used in breeding programmes to select for mastitis resistance since the 1990s [18]. Despite the low heritability of the traits, such phenotype-based selection has been shown to be beneficial in improving mastitis resistance [19], but causal genetic mechanisms behind the traits remain unknown.

The conventional method of treating mastitis is intramammary administration of antibiotics, which is not always effective [20], and has other adverse effects (e.g., antimicrobial resistance, contamination of dairy products). In addition, the relative efficacy of intramammary therapies using different antimicrobials to treat mastitis is difficult to assess and remains inconclusive [21]. Improving disease resistance is therefore considered a priority goal in modern breeding programmes [22, 23]. There is an increasing trend towards monitoring the physiological status and welfare of farm animals in real-time by recording parameters directly on the animal using electronic devices (so called “indicator traits”) [24], which could also be used for early detection of mammary infections.

The discovery of candidate (causative) genes for complex traits in large livestock is difficult because of the high breeding costs of experimental herds, long generation intervals, inefficient genetic engineering techniques, and the heterogeneous genetic background of outbred ruminant populations that exhibit population-specific genetic interactions between loci. The situation is quite different in model organisms (e.g., mice) where inbred lines and well-established genetic manipulation techniques (targeted gene disruptions/alterations) are available that can directly reveal phenotypes associated loci. Genetically modified mouse models are therefore a valuable source of data on traits of interest in all mammalian species. The mouse model has been used extensively as a tool to identify gene functions. Despite some anatomical and physiological differences between mice and ruminants, mouse models provide a cost-effective alternative for studying intramammary infections [25]. On the other hand, in large farm animals, as suggested by Mackay [26], the only applicable approach for identifying and prioritising quantitative trait loci (QTL) and candidate genes is to integrate diverse information that all together demonstrate the involvement of identified candidate genomic regions in a complex phenotype. However, despite the efforts invested in candidate gene discovery in large farm animals over the past decades, the success of the classical QTL-to-candidate gene approach has been limited. Successful examples of quantitative trait nucleotide (QTN) identification in dairy cattle include single nucleotide polymorphisms (SNPs) associated with milk composition and yield in DGAT1 [27], GHR [28] and ABCG2 [29]. To our knowledge, no causative gene/QTN for mastitis resistance has yet been validated due to the complexity of the trait, specific host-pathogen interactions, and other factors that complicate identification and validation of candidate genes.

In this study, we performed an extensive literature data mining and collected heterogeneous mastitis-associated data from various sources, including different ruminant species (sheep, goat, and cattle) and mastitis associated mouse models. The relevant information has been integrated into a database of bovine candidate genes for mastitis resistance, which we hope will be useful to researchers investigating the genetic background of mastitis resistance. The database represents a collection of known candidate genes for mastitis resistance, which can be prioritised according to different criteria and validated in functional studies for possible detection of mastitis-associated QTN.

Materials and methods

Data collection

Mastitis-associated loci were collected by manual review of the relevant literature and extraction of data from various sources. Mastitis-associated candidate genes reported in association and expression studies were collected by searching for relevant publications in PubMed [30] (using combinations of keywords: “mastitis”, “somatic cells”, “mammary”, “infection”, “QTL”, “SNP”, “association”, “expression”, and “candidate genes”). For association studies, all relevant articles on ruminants were manually reviewed and candidate genes with reported associations to mastitis were included in the database. Regarding expression studies in ruminants, it is impossible to extract all relevant information from the literature because of the large amount of experimental data available. Therefore, we have attempted to provide a representative sample of the available transcriptomic data indicative of expression changes during mastitis with the most common mastitis-causing pathogens (e.g., Escherichia coli and Staphylococcus aureus). In the present database, we included mastitis associated expression data from a database published in 2009 [31] and updated the collection of candidate genes with more recent expression data obtained from studies combining heterogeneous transcriptomic information (meta-analyses) and studies selected as most relevant, based on our review and meeting specific criteria (published after 2009, reported differentially expressed in controlled infections, performed on ruminant in vitro or in vivo challenge systems, studying immune response against the most common mastitis pathogens – including E. coli or S. aureus or their virulence factors, and using microarrays or RNA sequencing methods) (e.g., [32,33,34,35,36,37,38,39]). The genes reported differentially expressed in the selected studies were included to the database.

Mouse knockout and transgene model data were retrieved from the Mouse Genome Informatics (MGI) database 6.13 [40] using the phenotype ontology term “mastitis”. The availability of well-annotated mouse and bovine genomes allows identification of orthologs between the species, enabling use of a comparative approach. Therefore, for candidate genes associated with mastitis phenotype in mouse models, cattle orthologs were included in the database.

QTL in cattle were examined at Animal QTL Database [41], specifically in the Cattle QTL database [42] using traits view “Cattle QTLdb trait hierarchies”, selecting “mastitis” in “health traits” category and considering all mastitis-associated traits; that is “somatic cell score” (SCS), “somatic cell count” (SCC), and “clinical mastitis” (CM).

Data analysis

Candidate genes reported by multiple independent studies, regardless of study approach, were considered the most promising. The most promising candidates were screened for positional overlaps with mastitis associated QTL (CM, SCS, and SCC). Physical positions of mastitis associated QTL were extracted from the Cattle QTLdb using “QTL/associations analysis tool” that enables data download and QTL locations compared with the genomic positions of candidate genes extracted from GenBank [43], using annotations from bovine genome assembly ARS-UCD1.2 [44]. In case candidate gene shared at least part of its sequence with QTL of interest we considered the QTL as overlapping.

Pathway enrichment analysis of all the collected candidate genes was performed using g:Profiler [45]. The standard analysis implemented in g:Profiler searched for pathways, biological processes (BP), molecular functions (MF), and cellular components (CC) in which genes from the collected candidate gene list were significantly enriched compared to all genes in the genome. For the most promising genes pathway redundancy was addressed with EnrichmentMap [46] and visualised in Cytoscape [47]. The list of the most promising candidate genes was analysed for protein-protein interactions using STRING database [48]. Lists of candidate genes obtained by different approaches in ruminants were compared using an interactive tool for comparing lists with Venn’s diagrams [49].

Results

We collected 157 candidate genes from association studies, 2300 candidate genes from expression studies, and six genes from mouse models yielding a total of 2448 candidate genes that are likely to represent a genetic background for mastitis resistance. In addition, there are currently 668 QTL directly associated with mastitis or correlated traits (i.e., clinical mastitis, somatic cell score, and somatic cell count) reported in Cattle QTLdb. The identified candidate genes and QTL show an even distribution across all bovine autosomes and chromosome X (Fig. 1).

Fig. 1
figure 1

Schematic overview of the chromosomal arrangement of the collected mastitis-associated candidate genes (left – physical map) and QTL (right – linkage map) from Cattle QTLdb. Candidate genes involved in the immune response to Escherichia coli infection are highlighted in green, those involved in Staphylococcus aureus infection are highlighted in orange, whereas red indicates candidate genes associated with other pathogens. Red colour in linkage maps represents significant QTL and blue represents suggestive

Transgenic and knockout models in mice

In the MGI database, we found one gene disruption (Mfge8) and five transgenic mouse models for Lao1, Enpp2, Lpar1, Lpar2, and Lpar3 that resulted in abnormal mammary gland physiology and were also associated with mastitis (Additional file 1: Table S1). Bovine orthologs of the Mfge8 [50] and Lpar3 [51] associated with mastitis phenotypes in knockout and transgenic mouse models were also reported differentially expressed during mammary infection in cattle.

Association studies

Associations between genotypes (mainly SNPs) and mastitis-associated phenotypes (mainly SCS) have been demonstrated for 157 candidate genes in ruminants (Additional file 1: Table S2), several of which have been reported in multiple independent association studies (e.g., CXCL8, CXCR1, TLR4). Polymorphisms in genes associated with mastitis resistance represent potential causative mutations (QTNs).

Transcriptomic studies

We found 2300 candidate genes differentially expressed during mastitis in transcriptomic studies (Additional file 1: Table S3). When considering the pathogen-specific response [52], there were 1825 genes reported differentially expressed during Escherichia coli (E.coli) infection, 480 during Staphylococcus aureus (S. aureus) infection, of which 231 were differentially regulated in response to both pathogens, while the rest are associated with other pathogens. Some of the genes differentially regulated during mastitis have been reported in multiple independent expression studies (e.g., CXCL8, CXCR1, LTF, TLR4).

QTL

The list of QTL associated with mastitis-related traits (CM, SCC, and SCS) currently includes 668 QTL in Cattle QTLdb. The number of mastitis associated QTL per different bovine chromosomes ranged from 4 (BTA17 and 24) to 49 (BTA9). The highest density of mastitis-associated QTL is located on chromosomes 9 (49 QTL), 14 (45), 5 (43), and 10 (42).

The most promising candidate genes

Candidate genes associated with mastitis in multiple independent studies represent candidate genes of particular interest (Table 1). Candidates found by a high number of independent studies and/or in studies using different research approaches (e.g., CXCL8, CXCR1) could be considered of the highest priority for validation studies.

Table 1 List of the most promising (associated with mastitis in multiple independent studies, regardless of study approach) candidate genes for mastitis resistance

Enrichment analysis

The g:Profiler search for the collected candidate genes, revealed the most enriched pathways from KEGG and REACTOME databases (Table 2). Functional enrichment analysis mapped our gene list to known sources of functional information and identified statistically significant enriched biological processes (BP), molecular functions (MF) and cellular components (CC), which clearly show the association of the candidate genes to immune response related activities (Table 3; Fig. 2).

Table 2 Significantly enriched pathways for the input list of the collected candidate genes obtained from KEGG and REACTOME
Table 3 Functional enrichment analysis for the input list of the collected candidate genes
Fig. 2
figure 2

Results of the candidate gene analysis. A Overlaps between the collected candidate genes found in different studies on ruminants compared to the list of candidate genes reported in multiple studies (considered the most promising candidate genes), showing eight genes in the cross section with reported sequence polymorphisms associated with mastitis resistance and differentially expressed during E. coli and S. aureus intramammary infections. B Pathway enrichment analysis using g:Profiler and Cytoscape showing major biological pathways within the analysed gene set of the most promising candidates (node cutoff: P value = 0.01; Q value = 0.01. C Protein-protein interactions of the 22 most promising candidate genes as suggested by STRING database of which 13 are connected in a network. Different edge colours represent different types of associations (turquoise: known interactions – from curated databases; magenta: known interactions – experimentally determined; green: predicted interactions – gene neighbourhood; red: predicted interactions – gene fusions; blue: predicted interactions – gene co-occurrence; lime: others – text mining; black: others – co-expression; lavender: others – protein homology)

Discussion

The database created provides a comprehensive overview of the current knowledge on the genetic background of mastitis resistance in dairy cattle and could be extrapolated to other dairy ruminant species. The database is a practical example of genetic dissection of a complex trait that could serve as an example for other quantitative phenotypes in different animal species. A similar database for milk- and mastitis-related traits has already been created [31]. This study focuses exclusively on mastitis-related traits and is updated with the newly acquired knowledge.

Over the past decades, numerous experiments (in vivo and in vitro) have been performed to search for mastitis resistance candidate genes at the DNA/RNA level using high-throughput technologies (e.g., RNA-Seq, microarrays). Whole genome/transcriptome studies have generated a tremendous amount of data. Consequently, many loci have been proposed as candidates for mastitis resistance. Large lists of candidate genes present a challenge for researchers in analysing and interpreting data from expression and association experiments. Integration of data and enrichment analyses are needed to gain better mechanistic insight into the genetic background of mastitis resistance. However, the problem with these studies is that they are difficult to integrate and combine because they use different methodological approaches, experimental designs, incubation periods, and pathogen/virulence factors for immune challenges that elicit pathogen-specific immune responses [105]. As a result, studies often show conflicting results, making it difficult to combine data into a meaningful meta-analysis to extract relevant information. We believe that standardisation of such experiments would represent an important advance in the field. In addition, it should be kept in mind that existing reports may lead new studies to focus on already identified candidate genes. Therefore, whole-genome/transcriptome-level studies should be used to collect relevant data and identify potential candidate genes more broadly, followed by targeted functional studies of the most promising candidates.

Genes influencing complex traits are assumed to be additive in their contribution to phenotype and to contribute only to a minor extent. The number (2448 candidate genes and 668 QTL) and distribution of candidate loci across all bovine chromosomes illustrate the complexity of the trait and support the theory of a large number of candidate loci with a small contribution of a single locus to the phenotype. Given the “infinitesimal model” paradigm [189], identification of causative genes for mastitis resistance would be difficult and probably not reasonable in the context of marker-assisted selection (MAS). However, if alleles with moderate to large influence on mastitis resistance were identified, it would be feasible to implement such alleles in breeding strategies via MAS. Combining MAS and genomic selection would likely be an optimal trait improvement strategy in such a case. In addition, there are suggestions that to better understand the genetic architecture of complex traits, the current additive effects-based quantitative genetic framework needs to be improved and refined to include the effects of a range of multi-allelic markers, epigenetic inheritance, and genetic interactions, or even that an entirely new paradigm in quantitative genetic analysis should be proposed [190]. Epigenetic processes have been shown to influence multiple traits, including disease resistance [191,192,193] and consideration of non-Mendelian inheritance may be a next step toward better understanding the genetic background of complex traits [194]. The effect of clinical mastitis on methylation patterns has been demonstrated in the case of DNA remethylation around the STAT5-binding enhancer in the CSN1S1 promoter leading to disruption of αS1-casein synthesis during intramammary infection [195]. In addition, microRNAs (miRNAs) [196] and large non-coding RNAs (lncRNAs) [197] play a role in regulating gene expression and may serve as biomarkers for various pathophysiological conditions [198], including mastitis [199].

Integration of the collected candidate genes from multiple sources into a single database, overlaid with known QTL, allows identification of regions densely populated with candidate loci, revealing potential genomic hotspots (positional candidates) for mastitis resistance. Five of the genes considered the most promising candidates (Table 1) (i.e., BoLA-DRB3, CXCL8, GC, NPFFR2, and STAT5A) overlap with mastitis-associated QTL (CM or SCS). Eight of the most promising candidates (C6, C9, CARD6, CD14, CXCL8, LTF, TLR2, TLR4) were associated with mastitis in multiple studies and with different study approaches (expression and association studies) and were differentially expressed during infections with E. coli and S. aureus, respectively (Fig. 2A). Pathway enrichment clearly shows the most promising candidate genes are involved in immune response activation (Fig. 2B). A protein-protein network as suggested by the STRING database shows C6, C9, CD14, CXCL8, LTF, TLR2, TLR4 (Fig. 2C) are connected in a dense network. This (over)simplified diagram illuminates signal transduction pattern for activation of mammary innate immunity beginning at recognition receptors (TLR2/TLR4) and co-receptors (CD14), which induce chemokines (CXCL8) through G protein-coupled receptors (CXCR1/2), activate complement system (MBL2, C6, C9) and induce expression of antimicrobial peptides (LTF).

An example of a top-priority candidate from our list is CXCL8 (IL8), a known proinflammatory cytokine with a described function in neutrophil chemotaxis [200]. CXCL8 has been shown to be upregulated in mammary infections and variability in its sequence has been associated with mastitis related traits in several independent studies (Table 1). In addition, the gene overlaps with QTL for clinical mastitis. CXCL8 could therefore be considered a positional and functional candidate for mastitis resistance.

In 2020 a comprehensive study using a two-step QTL-GWAS approach has been conducted on French dairy cattle breeds that validated a SNP variant (rs436532576) of the GC (vitamin D binding protein) as a probable causative variant for mastitis resistance in Holstein breed [201]. GC was also identified as one of the most promising genes in our study. Interestingly, it is located in the vicinity of CXCL8 (around 88.8 Mb on BTA6) and the region around GC and CXCL8 is densely populated with mastitis associated QTL (Cattle QTLdb). DCK and NPFFR2 suggested among the most promising candidates in our study are also located on BTA6, around 86.3 Mb and 87.3 Mb, respectively. Moreover, Cai et al. [202] prioritized mastitis resistance candidate genes using multiple data sources by combining genome-wide association statistics with expression data, and also suggested GC as a putative causative gene for clinical mastitis QTL on BTA6, while DCK and NPFFR2 were suggested to be among top-five markers showing significant signals in the QTL region. This identifies BTA6 region between 86 and 89 Mb especially interesting for locating QTN for mastitis resistance. However, in cases where multiple candidate genes are located in proximity to each other it is difficult to determine the causative mutation as the haplotypes may be linked. Despite the fragmentation of mastitis-associated data, the demonstrated consistency between the results of studies, which combine information from multiple sources, supports the validity of the integrative approach we used.

Novel gene editing techniques (e.g., CRISPR, TALEN, ZFN) have dramatically changed and simplified gene modifications, making it possible to generate candidate mutations, edit genomes, and even regulate gene expression of endogenous genes (e.g., transfection of transcription elements fused with CRISPR/dCas9). Combined with protocols for establishing primary ruminant cell cultures [203, 204] and the availability of immortalized ruminant cell lines (e.g., MAC-T [205], BME-UV [206]), a methodological platform is available for generating and validating the suggested candidate mutations in an in vitro setting, which may eventually be followed by (ethically more controversial) gene editing experiments in vivo.

Conclusion

The compiled database contains 2448 candidate genes associated with mastitis resistance in cattle, which are evenly distributed across bovine chromosomes, illustrating the complexity of the trait. The database provides a comprehensive source of mastitis associated candidate genes available to researchers interested in genetic background of mastitis resistance. The list of the most promising candidate genes represents a priority list for validation in functional studies, which may eventually lead to discovery of QTN and improved mastitis resistance in the future, employing selection or gene editing approaches.

Availability of data and materials

All data generated or analysed during this study are included in this published article and its supplementary information files.

Abbreviations

BP:

Biological process

CC:

Cellular component

CM:

Clinical mastitis

CRISPR:

Clustered regularly interspaced short palindromic repeats

DNA:

Deoxyribonucleic acid

GO:

Gene Ontology

KEGG:

Kyoto Encyclopaedia of Genes and Genomes

MAS:

Marker-assisted selection

MF:

Molecular function

MGI:

Mouse Genome Informatics

QTL:

Quantitative trait loci

QTN:

Quantitative trait nucleotide

RNA:

Ribonucleic acid

SCC:

Somatic cell count

SCS:

Somatic cell score

SNPs:

Single nucleotide polymorphisms

TALEN:

Transcription activator-like effector nucleases

ZFN:

Zinc-finger nuclease.

References

  1. Hogeveen H, Huijps K, Lam TJ. Economic aspects of mastitis: new developments. N Z Vet J. 2011;59(1):16–23.

    Article  CAS  Google Scholar 

  2. Leslie KE, Petersson-Wolfe CS. Assessment and management of pain in dairy cows with clinical mastitis. Vet Clin North Am Food Anim Pract. 2012;28(2):289–305.

    Article  Google Scholar 

  3. Medrano-Galarza C, Gibbons J, Wagner S, de Passillé AM, Rushen J. Behavioral changes in dairy cows with mastitis. J Dairy Sci. 2012;95(12):6994–7002.

    Article  CAS  Google Scholar 

  4. Krömker V, Leimbach S. Mastitis treatment-reduction in antibiotic usage in dairy cows. Reprod Domest Anim. 2017;52(Suppl 3):21–9.

    Article  Google Scholar 

  5. Auldist MJ, Coats S, Sutherland BJ, Mayes JJ, McDowell GH, Rogers GL. Effects of somatic cell count and stage of lactation on raw milk composition and the yield and quality of Cheddar cheese. J Dairy Res. 1996;63(2):269–80.

    Article  CAS  Google Scholar 

  6. Klei L, Yun J, Sapru A, Lynch J, Barbano D, Sears P, et al. Effects of milk somatic cell count on cottage cheese yield and quality. J Dairy Sci. 1998;81(5):1205–13.

    Article  CAS  Google Scholar 

  7. Gyles C. Industrial farm animal production. Can Vet J. 2010;51(2):125–8.

    Google Scholar 

  8. Roxström A, Strandberg E, Emanuelson U, Philipsson J. Genetic and environmental correlations among female fertility traits, and between the ability to show oestrus and milk production in dairy cattle. Acta Agr Scand Sect A-Anim Sc. 2001;51:192–9.

    Google Scholar 

  9. Lucy MC. Reproductive loss in high-producing dairy cattle: where will it end? J Dairy Sci. 2001;84(6):1277–93.

    Article  CAS  Google Scholar 

  10. Grindal RJ, Hillerton JE. Influence of milk flow rate on new intramammary infection in dairy cows. J Dairy Res. 1991;58(3):263–8.

    Article  CAS  Google Scholar 

  11. Tancin V, Ipema AH, Hogewerf P. Interaction of somatic cell count and quarter milk flow patterns. J Dairy Sci. 2007;90(5):2223–8.

    Article  CAS  Google Scholar 

  12. Sandrucci A, Tamburini A, Bava L, Zucali M. Factors affecting milk flow traits in dairy cows: results of a field study. J Dairy Sci. 2007;90(3):1159–67.

    Article  CAS  Google Scholar 

  13. Tamburini A, Bava L, Piccinini R, Zecconi A, Zucali M, Sandrucci A. Milk emission and udder health status in primiparous dairy cows during lactation. J Dairy Res. 2010;77(1):13–9.

    Article  CAS  Google Scholar 

  14. Windig JJ, Calus MP, Beerda B, Veerkamp RF. Genetic correlations between milk production and health and fertility depending on herd environment. J Dairy Sci. 2006;89(5):1765–75.

    Article  CAS  Google Scholar 

  15. Ingvartsen KL, Dewhurst RJ, Friggens NC. On the relationship between lactational performance and health: is it yield or metabolic imbalance that cause production diseases in dairy cattle? A position paper. Livest Prod Sci. 2003;83(2):277–308.

    Article  Google Scholar 

  16. Simianer H, Solbu H, Schaeffer LR. Estimated genetic correlations between disease and yield traits in dairy cattle. J Dairy Sci. 1991;74(12):4358–65.

    Article  CAS  Google Scholar 

  17. Berry DP, Wall E, Pryce JE. Genetics and genomics of reproductive performance in dairy and beef cattle. Animal. 2014;8(Suppl 1):105–21.

    Article  Google Scholar 

  18. Heringstad B, Klemetsdal G, Ruane J. Selection for mastitis resistance in dairy cattle: a review with focus on the situation in the nordic countries. Livest Prod Sci. 2000;64(2):95–106.

    Article  Google Scholar 

  19. Østerås O, Landin H, Kulkas L, Farre M, Reiersen J. The udder health in the nordic countries. The IDF Mastitis Conference; Copenhagen, Denmark: IDF; 2019. p. 52 – 6.

  20. Sugiyama M, Watanabe M, Sonobe T, Kibe R, Koyama S, Kataoka Y. Efficacy of antimicrobial therapy for bovine acute Klebsiella pneumoniae mastitis. J Vet Med Sci. 2022;84(7):1023–8.

    Article  Google Scholar 

  21. Winder CB, Sargeant JM, Hu D, Wang C, Kelton DF, Godkin MA, et al. Comparative efficacy of antimicrobials for treatment of clinical mastitis in lactating dairy cattle: a systematic review and network meta-analysis. Anim Health Res Rev. 2019;20(2):229–46.

    Article  CAS  Google Scholar 

  22. Oltenacu PA, Algers B. Selection for increased production and the welfare of dairy cows: are new breeding goals needed? Ambio. 2005;34(4–5):311–5.

    Article  Google Scholar 

  23. Berghof TVL, Poppe M, Mulder HA. Opportunities to improve resilience in animal breeding programs. Front Genet. 2018;9:692.

    Article  Google Scholar 

  24. Egger-Danner C, Cole JB, Pryce JE, Gengler N, Heringstad B, Bradley A, et al. Invited review: overview of new traits and phenotyping strategies in dairy cattle with a focus on functional traits. Animal. 2015;9(2):191–207.

    Article  CAS  Google Scholar 

  25. Notebaert S, Meyer E. Mouse models to study the pathogenesis and control of bovine mastitis. A review. Vet Q. 2006;28(1):2–13.

    Article  CAS  Google Scholar 

  26. Mackay TF. The genetic architecture of quantitative traits: lessons from Drosophila. Curr Opin Genet Dev. 2004;14(3):253–7.

    Article  CAS  Google Scholar 

  27. Grisart B, Coppieters W, Farnir F, Karim L, Ford C, Berzi P, et al. Positional candidate cloning of a QTL in dairy cattle: identification of a missense mutation in the bovine DGAT1 gene with major effect on milk yield and composition. Genome Res. 2002;12(2):222–31.

    Article  CAS  Google Scholar 

  28. Blott S, Kim JJ, Moisio S, Schmidt-Küntzel A, Cornet A, Berzi P, et al. Molecular dissection of a quantitative trait locus: a phenylalanine-to-tyrosine substitution in the transmembrane domain of the bovine growth hormone receptor is associated with a major effect on milk yield and composition. Genetics. 2003;163(1):253–66.

    Article  CAS  Google Scholar 

  29. Cohen-Zinder M, Seroussi E, Larkin DM, Loor JJ, Everts-van der Wind A, Lee JH, et al. Identification of a missense mutation in the bovine ABCG2 gene with a major effect on the QTL on chromosome 6 affecting milk yield and composition in Holstein cattle. Genome Res. 2005;15(7):936–44.

    Article  CAS  Google Scholar 

  30. PubMed. http://www.ncbi.nlm.nih.gov. Accessed on date: 10 Sept 2021.

  31. Ogorevc J, Kunej T, Razpet A, Dovc P. Database of cattle candidate genes and genetic markers for milk production and mastitis. Anim Genet. 2009;40(6):832–51.

    Article  CAS  Google Scholar 

  32. Sun Y, Li L, Li C, Wang G, Xing G. Gene microarray integrated with iTRAQ-based proteomics for the discovery of NLRP3 in LPS-induced inflammatory response of bovine mammary epithelial cells. J Dairy Res. 2019;86(4):416–24.

    Article  CAS  Google Scholar 

  33. Genini S, Badaoui B, Sclep G, Bishop SC, Waddington D, Pinard van der Laan MH, et al. Strengthening insights into host responses to mastitis infection in ruminants by combining heterogeneous microarray data sources. BMC Genom. 2011;12(1):225.

    Article  Google Scholar 

  34. Buitenhuis B, Røntved CM, Edwards SM, Ingvartsen KL, Sørensen P. In depth analysis of genes and pathways of the mammary gland involved in the pathogenesis of bovine Escherichia coli-mastitis. BMC Genom. 2011;12:130.

    Article  CAS  Google Scholar 

  35. Brand B, Hartmann A, Repsilber D, Griesbeck-Zilch B, Wellnitz O, Kühn C, et al. Comparative expression profiling of E. coli and S. aureus inoculated primary mammary gland cells sampled from cows with different genetic predispositions for somatic cell score. Genet Sel Evol. 2011;43:24.

    Article  Google Scholar 

  36. Jensen K, Günther J, Talbot R, Petzl W, Zerbe H, Schuberth H-J, et al. Escherichia coli- and Staphylococcus aureus-induced mastitis differentially modulate transcriptional responses in neighbouring uninfected bovine mammary gland quarters. BMC Genom. 2013;14(1):36.

    Article  CAS  Google Scholar 

  37. Younis S, Javed Q, Blumenberg M. Meta-analysis of transcriptional responses to mastitis-causing Escherichia coli. PLoS ONE. 2016;11(3):e0148562.

    Article  Google Scholar 

  38. Gilbert FB, Cunha P, Jensen K, Glass EJ, Foucras G, Robert-Granié C, et al. Differential response of bovine mammary epithelial cells to Staphylococcus aureus or Escherichia coli agonists of the innate immune system. Vet Res. 2013;44(1):40.

    Article  CAS  Google Scholar 

  39. Kirsanova E, Boysen P, Johansen GM, Heringstad B, Lewandowska-Sabat A, Olsaker I. Expression analysis of candidate genes for chronic subclinical mastitis in Norwegian Red cattle. J Dairy Sci. 2020;103(10):9142–9.

    Article  CAS  Google Scholar 

  40. Blake JA, Eppig JT, Kadin JA, Richardson JE, Smith CL, Bult CJ, et al. Mouse genome database (MGD)-2017: community knowledge resource for the laboratory mouse. Nucleic Acids Res. 2017;45(D1):D723–9.

  41. Hu ZL, Park CA, Reecy JM. Bringing the animal QTLdb and CorrDB into the future: meeting new challenges and providing updated services. Nucleic Acids Res. 2022;50(D1):D956–61.

  42. CattleQTLdb. https://www.animalgenome.org/QTLdb. Accessed on date: 15 Sept 2021.

  43. Benson DA, Cavanaugh M, Clark K, Karsch-Mizrachi I, Lipman DJ, Ostell J, et al. GenBank Nucleic Acids Res. 2012;41(D1):D36–42.

    Article  Google Scholar 

  44. Rosen BD, Bickhart DM, Schnabel RD, Koren S, Elsik CG, Tseng E, et al. De novo assembly of the cattle reference genome with single-molecule sequencing. GigaScience. 2020;9(3):giaa021.

    Article  CAS  Google Scholar 

  45. Raudvere U, Kolberg L, Kuzmin I, Arak T, Adler P, Peterson H, et al. G:profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 2019;47(W1):W191–8.

  46. Merico D, Isserlin R, Stueker O, Emili A, Bader GD. Enrichment map: a network-based method for gene-set enrichment visualization and interpretation. PLoS ONE. 2010;5(11):e13984.

    Article  Google Scholar 

  47. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.

    Article  CAS  Google Scholar 

  48. Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607–13.

  49. Oliveros JC. Venny. An interactive tool for comparing lists with Venn’s diagrams. 2007–2015. https://bioinfogp.cnb.csic.es/tools/venny/index.html.

  50. Atabai K, Fernandez R, Huang X, Ueki I, Kline A, Kline A, et al. Mfge8 is critical for mammary gland remodeling during involution. Mol Biology Cell. 2005;16(12):5528–37.

    Article  CAS  Google Scholar 

  51. Liu S, Umezu-Goto M, Murph M, Lu Y, Liu W, Zhang F, et al. Expression of autotaxin and lysophosphatidic acid receptors increases mammary tumorigenesis, invasion, and metastases. Cancer Cell. 2009;15(6):539–50.

    Article  CAS  Google Scholar 

  52. Petzl W, Zerbe H, Günther J, Seyfert H-M, Hussen J, Schuberth H-J. Pathogen-specific responses in the bovine udder. Models and immunoprophylactic concepts. Res Vet Sci. 2018;116:55–61.

    Article  CAS  Google Scholar 

  53. Kelm SC, Detilleux JC, Freeman AE, Kehrli ME, Dietz AB, Fox LK, et al. Genetic association between parameters of inmate immunity and measures of mastitis in periparturient holstein cattle. J Dairy Sci. 1997;80(8):1767–75.

    Article  CAS  Google Scholar 

  54. Yoshida T, Mukoyama H, Furuta H, Kondo Y, Takeshima SN, Aida Y, et al. Association of BoLA-DRB3 alleles identified by a sequence-based typing method with mastitis pathogens in japanese holstein cows. Anim Sci J. 2009;80(5):498–509.

    Article  CAS  Google Scholar 

  55. Yoshida T, Furuta H, Kondo Y, Mukoyama H. Association of BoLA-DRB3 alleles with mastitis resistance and susceptibility in japanese holstein cows. Anim Sci J. 2012;83(5):359–66.

    Article  CAS  Google Scholar 

  56. Baltian LR, Ripoli MV, Sanfilippo S, Takeshima SN, Aida Y, Giovambattista G. Association between BoLA-DRB3 and somatic cell count in Holstein cattle from Argentina. Mol Biol Rep. 2012;39(7):7215–20.

    Article  CAS  Google Scholar 

  57. Pokorska J, Kułaj D, Dusza M, Ochrem A, Makulska J. The influence of BoLA-DRB3 alleles on incidence of clinical mastitis, cystic ovary disease and milk traits in Holstein Friesian cattle. Mol Biol Rep. 2018;45(5):917–23.

    Article  CAS  Google Scholar 

  58. Rupp R, Hernandez A, Mallard BA. Association of bovine leukocyte antigen (BoLA) DRB3.2 with immune response, mastitis, and production and type traits in canadian holsteins. J Dairy Sci. 2007;90(2):1029–38.

    Article  CAS  Google Scholar 

  59. Kulberg S, Heringstad B, Guttersrud OA, Olsaker I. Study on the association of BoLA-DRB3.2 alleles with clinical mastitis in norwegian red cows. J Anim Breed Genet. 2007;124(4):201–7.

    Article  CAS  Google Scholar 

  60. Prajapati BM, Gupta JP, Pandey DP, Parmar GA, Chaudhari JD. Molecular markers for resistance against infectious diseases of economic importance. Vet World. 2017;10(1):112–20.

    Article  CAS  Google Scholar 

  61. Sharif S, Mallard BA, Wilkie BN, Sargeant JM, Scott HM, Dekkers JC, et al. Associations of the bovine major histocompatibility complex DRB3 (BoLA-DRB3) alleles with occurrence of disease and milk somatic cell score in canadian dairy cattle. Anim Genet. 1998;29(3):185–93.

    Article  CAS  Google Scholar 

  62. Sharif S, Mallard BA, Wilkie BN, Sargeant JM, Scott HM, Dekkers JC, et al. Associations of the bovine major histocompatibility complex DRB3 (BoLA-DRB3) with production traits in canadian dairy cattle. Anim Genet. 1999;30(2):157–60.

    Article  CAS  Google Scholar 

  63. Sharif S, Mallard BA, Sargeant JM. Presence of glutamine at position 74 of pocket 4 in the BoLA-DR antigen binding groove is associated with occurrence of clinical mastitis caused by Staphylococcus species. Vet Immunol Immunopathol. 2000;76(3–4):231–8.

    Article  CAS  Google Scholar 

  64. Derakhshani H, Plaizier JC, De Buck J, Barkema HW, Khafipour E. Association of bovine major histocompatibility complex (BoLA) gene polymorphism with colostrum and milk microbiota of dairy cows during the first week of lactation. Microbiome. 2018;6(1):203.

    Article  Google Scholar 

  65. Kirsanova E, Heringstad B, Lewandowska-Sabat A, Olsaker I. Identification of candidate genes affecting chronic subclinical mastitis in Norwegian Red cattle: combining genome-wide association study, topologically associated domains and pathway enrichment analysis. Animal Genet. 2020;51(1):22–31.

    Article  CAS  Google Scholar 

  66. Tiezzi F, Parker-Gaddis KL, Cole JB, Clay JS, Maltecca C. A genome-wide association study for clinical mastitis in first parity US Holstein cows using single-step approach and genomic matrix re-weighting procedure. PLoS ONE. 2015;10(2):e0114919.

    Article  Google Scholar 

  67. Meredith BK, Berry DP, Kearney F, Finlay EK, Fahey AG, Bradley DG, et al. A genome-wide association study for somatic cell score using the Illumina high-density bovine beadchip identifies several novel QTL potentially related to mastitis susceptibility. Front Genet. 2013;4:229.

    Article  Google Scholar 

  68. Banos G, Bramis G, Bush SJ, Clark EL, McCulloch MEB, Smith J, et al. The genomic architecture of mastitis resistance in dairy sheep. BMC Genom. 2017;18(1):624.

    Article  CAS  Google Scholar 

  69. Mitterhuemer S, Petzl W, Krebs S, Mehne D, Klanner A, Wolf E, et al. Escherichia coli infection induces distinct local and systemic transcriptome responses in the mammary gland. BMC Genom. 2010;11:138.

    Article  Google Scholar 

  70. Sahana G, Guldbrandtsen B, Thomsen B, Lund MS. Confirmation and fine-mapping of clinical mastitis and somatic cell score QTL in Nordic Holstein cattle. Anim Genet. 2013;44(6):620–6.

    Article  CAS  Google Scholar 

  71. Olsen HG, Knutsen TM, Lewandowska-Sabat AM, Grove H, Nome T, Svendsen M, et al. Fine mapping of a QTL on bovine chromosome 6 using imputed full sequence data suggests a key role for the group-specific component (GC) gene in clinical mastitis and milk production. Genet Sel Evol. 2016;48(1):79.

    Article  Google Scholar 

  72. Yuan ZR, Li J, Liu L, Zhang LP, Zhang LM, Chen C, et al. Single nucleotide polymorphism of CACNA2D1 gene and its association with milk somatic cell score in cattle. Mol Biol Rep. 2011;38(8):5179–83.

    Article  CAS  Google Scholar 

  73. Silva AA, Silva DA, Silva FF, Costa CN, Silva HT, Lopes PS, et al. GWAS and gene networks for milk-related traits from test-day multiple lactations in portuguese holstein cattle. J Appl Genet. 2020;61(3):465–76.

    Article  CAS  Google Scholar 

  74. Magotra A, Gupta ID, Verma A, Alex R, MR V, Ahmad T. Candidate SNP of CACNA2D1 gene associated with clinical mastitis and production traits in Sahiwal (Bos taurus indicus) and Karan fries (Bos taurus taurus × Bos taurus indicus). Anim Biotechnol. 2019;30(1):75–81.

  75. Deb R, Singh U, Kumar S, Kumar A, Singh R, Sengar G, et al. Genotypic to expression profiling of bovine calcium channel, voltage-dependent, alpha-2/delta subunit 1 gene, and their association with bovine mastitis among Frieswal (HFX Sahiwal) crossbred cattle of indian origin. Anim Biotechnol. 2014;25(2):128–38.

    Article  CAS  Google Scholar 

  76. Bagheri M, Zahmatkesh A. Dominance effects estimation of TLR4 and CACNA2D1 genes for health and production traits using logistic regression. J Genet. 2017;96(6):1027–31.

    Article  CAS  Google Scholar 

  77. Kadri NK, Guldbrandtsen B, Lund MS, Sahana G. Genetic dissection of milk yield traits and mastitis resistance quantitative trait loci on chromosome 20 in dairy cattle. J Dairy Sci. 2015;98(12):9015–25.

    Article  CAS  Google Scholar 

  78. Huang JM, Wang XG, Jiang Q, Sun Y, Yang CH, Ju ZH, et al. Identification of CD14 transcript in blood polymorphonuclear neutrophil leukocytes and functional variation in Holsteins. Genet Mol Res. 2016;15(2):gmr.15027932.

  79. Selvan AS, Gupta ID, Verma A, Chaudhari MV, Magotra A. Molecular characterization and combined genotype association study of bovine cluster of differentiation 14 gene with clinical mastitis in crossbred dairy cattle. Vet World. 2016;9(7):680–4.

    Article  CAS  Google Scholar 

  80. Wang Y, Zarlenga DS, Paape MJ, Dahl GE. Recombinant bovine soluble CD14 sensitizes the mammary gland to lipopolysaccharide. Vet Immunol Immunopathol. 2002;86(1–2):115–24.

    Article  CAS  Google Scholar 

  81. Li J, Wang Q, Chen F, Wang H, Chen J, Wang Z, et al. SNPs of CD14 change the mastitis morbidity of chinese holstein. Mol Med Rep. 2017;16(6):9102–10.

    Article  CAS  Google Scholar 

  82. Leyva-Baca I, Schenkel F, Sharma BS, Jansen GB, Karrow NA. Identification of single nucleotide polymorphisms in the bovine CCL2, IL8, CCR2 and IL8RA genes and their association with health and production in canadian holsteins. Anim Genet. 2007;38(3):198–202.

    Article  CAS  Google Scholar 

  83. Sodeland M, Kent MP, Olsen HG, Opsal MA, Svendsen M, Sehested E, et al. Quantitative trait loci for clinical mastitis on chromosomes 2, 6, 14 and 20 in norwegian red cattle. Anim Genet. 2011;42(5):457–65.

    Article  CAS  Google Scholar 

  84. Nani JP, Raschia MA, Carignano H, Poli MA, Calvinho LF, Amadio AF. Single nucleotide polymorphisms in candidate genes and their relation with somatic cell scores in argentinean dairy cattle. J Appl Genet. 2015;56(4):505–13.

    Article  CAS  Google Scholar 

  85. Sharifi S, Pakdel A, Ebrahimi M, Reecy JM, Fazeli Farsani S, Ebrahimie E. Integration of machine learning and meta-analysis identifies the transcriptomic bio-signature of mastitis disease in cattle. PLoS ONE. 2018;13(2):e0191227.

    Article  Google Scholar 

  86. Gorji AE, Roudbari Z, Sadeghi B, Javadmanesh A, Sadkowski T. Transcriptomic analysis on the promoter regions discover gene networks involving mastitis in cattle. Microb Pathog. 2019;137:103801.

    Article  CAS  Google Scholar 

  87. Dai H, Wei G, Wang Y, Ma N, Chang G, Shen X. Sodium butyrate promotes lipopolysaccharide-induced innate immune responses by enhancing mitogen-activated protein kinase activation and histone acetylation in bovine mammary epithelial cells. J Dairy Sci. 2020;103(12):11636–52.

    Article  CAS  Google Scholar 

  88. Rainard P, Cunha P, Bougarn S, Fromageau A, Rossignol C, Gilbert FB, et al. T helper 17-associated cytokines are produced during antigen-specific inflammation in the mammary gland. PLoS ONE. 2013;8(5):e63471.

    Article  CAS  Google Scholar 

  89. Russell CD, Widdison S, Leigh JA, Coffey TJ. Identification of single nucleotide polymorphisms in the bovine toll-like receptor 1 gene and association with health traits in cattle. Vet Res. 2012;43:17.

    Article  CAS  Google Scholar 

  90. Barber MR, Yang TJ. Chemotactic activities in nonmastitic and mastitic mammary secretions: presence of interleukin-8 in mastitic but not nonmastitic secretions. Clin Diag Lab Immunol. 1998;5(1):82–6.

    Article  CAS  Google Scholar 

  91. Nagahata H, Moriyama A, Sawada C, Asai Y, Kokubu C, Gondaira S, et al. Innate immune response of mammary gland induced by intramammary infusion of Bifidobacterium breve in lactating dairy cows. J Vet Med Sci. 2020;82(12):1742–9.

    Article  CAS  Google Scholar 

  92. Chen R, Yang Z, Ji D, Mao Y, Chen Y, Li Y, et al. Polymorphisms of the IL8 gene correlate with milking traits, SCS and mRNA level in chinese holstein. Mol Biol Rep. 2011;38(6):4083–8.

    Article  CAS  Google Scholar 

  93. Chen R, Wang Z, Yang Z, Zhu X, Ji D, Mao Y. Association of IL8 -105G/A with mastitis somatic cell score in chinese holstein dairy cows. Anim Biotechnol. 2015;26(2):143–7.

    Article  CAS  Google Scholar 

  94. Purba FY, Suzuki N, Isobe N. Association of endometritis and ovarian follicular cyst with mastitis in dairy cows. J Vet Med Sci. 2021;83(2):338–43.

    Article  CAS  Google Scholar 

  95. Hughes K, Watson CJ. The mammary microenvironment in mastitis in humans, dairy ruminants, rabbits and rodents: a one health focus. J Mammary Gland Biol Neoplasia. 2018;23(1–2):27–41.

    Article  Google Scholar 

  96. Stojkovic B, Mullen MP, Donofrio G, McLoughlin RM, Meade KG. Interleukin 8 haplotypes drive divergent responses in uterine endometrial cells and are associated with somatic cell score in Holstein-Friesian cattle. Vet Immunol Immunopathol. 2017;184:18–28.

    Article  CAS  Google Scholar 

  97. Bohl LP, Isaac P, Breser ML, Orellano MS, Correa SG, Tolosa de Talamoni NG, et al. Interaction between bovine mammary epithelial cells and planktonic or biofilm Staphylococcus aureus: the bacterial lifestyle determines its internalization ability and the pathogen recognition. Microb Pathog. 2021;152:104604.

    Article  CAS  Google Scholar 

  98. El-Deeb W, Fayez M, Alhumam N, Elsohaby I, Quadri SA, Mkrtchyan H. The effect of staphylococcal mastitis including resistant strains on serum procalcitonin, neopterin, acute phase response and stress biomarkers in Holstein dairy cows. PeerJ. 2021;9:e11511-e.

    Article  Google Scholar 

  99. Mount JA, Karrow NA, Caswell JL, Boermans HJ, Leslie KE. Assessment of bovine mammary chemokine gene expression in response to lipopolysaccharide, lipotechoic acid + peptidoglycan, and CpG oligodeoxynucleotide 2135. Can J Vet Res. 2009;73(1):49–57.

    CAS  Google Scholar 

  100. Fonseca I, Silva PV, Lange CC, Guimarães MFM, Weller MMDCA, Sousa KRS, et al. Expression profile of genes associated with mastitis in dairy cattle. Genet Mol Biol. 2009;32(4):776–81.

    Article  CAS  Google Scholar 

  101. Tao W, Mallard B. Differentially expressed genes associated with Staphylococcus aureus mastitis of canadian holstein cows. Vet Immunol Immunopathol. 2007;120(3–4):201–11.

    Article  CAS  Google Scholar 

  102. Moyes KM, Drackley JK, Morin DE, Rodriguez-Zas SL, Everts RE, Lewin HA, et al. Mammary gene expression profiles during an intramammary challenge reveal potential mechanisms linking negative energy balance with impaired immune response. Physiol Genomics. 2010;41(2):161–70.

    Article  CAS  Google Scholar 

  103. Niedziela DA, Murphy MP, Grant J, Keane OM, Leonard FC. Clinical presentation and immune characteristics in first-lactation holstein-friesian cows following intramammary infection with genotypically distinct Staphylococcus aureus strains. J Dairy Sci. 2020;103(9):8453–66.

    Article  CAS  Google Scholar 

  104. Stevens MG, Peelman LJ, De Spiegeleer B, Pezeshki A, Van De Walle GR, Duchateau L, et al. Differential gene expression of the toll-like receptor-4 cascade and neutrophil function in early- and mid-lactating dairy cows. J Dairy Sci. 2011;94(3):1277–88.

    Article  CAS  Google Scholar 

  105. Fu Y, Zhou E, Liu Z, Li F, Liang D, Liu B, et al. Staphylococcus aureus and Escherichia coli elicit different innate immune responses from bovine mammary epithelial cells. Vet Immunol Immunopathol. 2013;155(4):245–52.

    Article  CAS  Google Scholar 

  106. Tsuchida S, Yamad Y, Fukui E, Kawada T, Omi T, Tsuchida A, et al. Distribution of single nucleotide polymorphisms in the CXCR1 gene and association with calf diseases in japanese black cattle. J Vet Med Sci. 2010;72(12):1609–14.

    Article  CAS  Google Scholar 

  107. Galvão KN, Pighetti GM, Cheong SH, Nydam DV, Gilbert RO. Association between interleukin-8 receptor-α (CXCR1) polymorphism and disease incidence, production, reproduction, and survival in Holstein cows. J Dairy Sci. 2011;94(4):2083–91.

    Article  Google Scholar 

  108. Verbeke J, Piepers S, Peelman L, Van Poucke M, De Vliegher S. Pathogen-group specific association between CXCR1 polymorphisms and subclinical mastitis in dairy heifers. J Dairy Res. 2012;79(3):341–51.

    Article  CAS  Google Scholar 

  109. Verbeke J, Van Poucke M, Peelman L, Piepers S, De Vliegher S. Associations between CXCR1 polymorphisms and pathogen-specific incidence rate of clinical mastitis, test-day somatic cell count, and test-day milk yield. J Dairy Sci. 2014;97(12):7927–39.

    Article  CAS  Google Scholar 

  110. Verbeke J, Van Poucke M, Peelman L, De Vliegher S. Differential expression of CXCR1 and commonly used reference genes in bovine milk somatic cells following experimental intramammary challenge. BMC Genet. 2015;16:40.

    Article  Google Scholar 

  111. Verbeke J, Boulougouris X, Rogiers C, Burvenich C, Peelman L, De Spiegeleer B, et al. Reactive oxygen species generation by bovine blood neutrophils with different CXCR1 (IL8RA) genotype following Interleukin-8 incubation. BMC Vet Res. 2015;11:104.

    Article  Google Scholar 

  112. Verbeke J, Piccart K, Piepers S, Van Poucke M, Peelman L, De Visscher A, et al. Somatic cell count and milk neutrophil viability of dairy heifers with specific CXCR1 genotypes following experimental intramammary infection with Staphylococcus chromogenes originating from milk. Vet J. 2015;204(3):322–6.

    Article  CAS  Google Scholar 

  113. Zhou L, Wang HM, Ju ZH, Zhang Y, Huang JM, Qi C, et al. Association of novel single nucleotide polymorphisms of the CXCR1 gene with the milk performance traits of chinese native cattle. Genet Mol Res. 2013;12(3):2725–39.

    Article  CAS  Google Scholar 

  114. Bagheri M, Moradi-Sharhrbabak M, Miraie-Ashtiani R, Safdari-Shahroudi M, Abdollahi-Arpanahi R. Case-control approach application for finding a relationship between candidate genes and clinical mastitis in Holstein dairy cattle. J Appl Genet. 2016;57(1):107–12.

    Article  CAS  Google Scholar 

  115. Pokorska J, Dusza M, Kułaj D, Żukowski K, Makulska J. Single nucleotide polymorphisms in the CXCR1 gene and its association with clinical mastitis incidence in polish holstein-friesian cows. Genet Mol Res. 2016;15(2):gmr.15027247.

  116. Pawlik A, Sender G, Kapera M, Korwin-Kossakowska A. Association between interleukin 8 receptor α gene (CXCR1) and mastitis in dairy cattle. Cent-Europ J Immunol. 2015;40(2):153–8.

    Article  Google Scholar 

  117. Rambeaud M, Pighetti GM. Differential calcium signaling in dairy cows with specific CXCR1 genotypes potentially related to interleukin-8 receptor functionality. Immunogenetics. 2007;59(1):53–8.

    Article  CAS  Google Scholar 

  118. Moretti R, Soglia D, Chessa S, Sartore S, Finocchiaro R, Rasero R, et al. Identification of SNPs associated with somatic cell score in candidate genes in italian Holstein Friesian bulls. Animals (Basel). 2021;11(2):366.

    Article  Google Scholar 

  119. Fontanesi L, Calò DG, Galimberti G, Negrini R, Marino R, Nardone A, et al. A candidate gene association study for nine economically important traits in italian holstein cattle. Anim Genet. 2014;45(4):576–80.

    Article  CAS  Google Scholar 

  120. Beecher C, Daly M, Childs S, Berry DP, Magee DA, McCarthy TV, et al. Polymorphisms in bovine immune genes and their associations with somatic cell count and milk production in dairy cattle. BMC Genet. 2010;11:99.

    Article  Google Scholar 

  121. Chen R, Yang Z, Ji D, Mao Y, Chen Y, Zhang Y, et al. SNPs of CXCR1 gene and its associations with somatic cell score in chinese holstein cattle. Anim Biotechnol. 2011;22(3):133–42.

    Article  Google Scholar 

  122. Leyva-Baca I, Schenkel F, Martin J, Karrow NA. Polymorphisms in the 5’ upstream region of the CXCR1 chemokine receptor gene, and their association with somatic cell score in Holstein cattle in Canada. J Dairy Sci. 2008;91(1):407–17.

    Article  CAS  Google Scholar 

  123. Siebert L, Headrick S, Lewis M, Gillespie B, Young C, Wojakiewicz L, et al. Genetic variation in CXCR1 haplotypes linked to severity of Streptococcus uberis infection in an experimental challenge model. Vet Immunol Immunopathol. 2017;190:45–52.

    Article  CAS  Google Scholar 

  124. Mao YJ, Zhu XR, Li R, Chen D, Xin SY, Zhu YH, et al. Methylation analysis of CXCR1 in mammary gland tissue of cows with mastitis induced by Staphylococcus aureus. Genet Mol Res. 2015;14(4):12606–15.

    Article  CAS  Google Scholar 

  125. Alhussien M, Manjari P, Mohammed S, Sheikh AA, Reddi S, Dixit S, et al. Incidence of mastitis and activity of milk neutrophils in Tharparkar cows reared under semi-arid conditions. Trop Anim Health Prod. 2016;48(6):1291–5.

    Article  Google Scholar 

  126. Alhussien MN, Dang AK. Pathogen-dependent modulation of milk neutrophils competence, plasma inflammatory cytokines and milk quality during intramammary infection of Sahiwal (Bos indicus) cows. Microb Pathog. 2018;121:131–8.

    Article  CAS  Google Scholar 

  127. Youngerman SM, Saxton AM, Oliver SP, Pighetti GM. Association of CXCR2 polymorphisms with subclinical and clinical mastitis in dairy cattle. J Dairy Sci. 2004;87(8):2442–8.

    Article  CAS  Google Scholar 

  128. Rambeaud M, Clift R, Pighetti GM. Association of a bovine CXCR2 gene polymorphism with neutrophil survival and killing ability. Vet Immunol Immunopathol. 2006;111(3–4):231–8.

    Article  CAS  Google Scholar 

  129. El Nahas SM, El Kasas AH, Abou Mossallem AA, Abdelhamid MI, Warda M. A study on IL8RB gene polymorphism as a potential immuno-compromised adherent in exaggeration of parenteral and mammo-crine oxidative stress during mastitis in buffalo. J Adv Res. 2017;8(6):617–25.

    Article  Google Scholar 

  130. Wu X, Lund MS, Sahana G, Guldbrandtsen B, Sun D, Zhang Q, et al. Association analysis for udder health based on SNP-panel and sequence data in danish holsteins. Genet Sel Evol. 2015;47:50.

    Article  Google Scholar 

  131. Abdel-Shafy H, Bortfeldt RH, Reissmann M, Brockmann GA. Validating genome-wide associated signals for clinical mastitis in german holstein cattle. Anim Genet. 2018;49(1):82–5.

    Article  CAS  Google Scholar 

  132. Nayeri S, Schenkel F, Fleming A, Kroezen V, Sargolzaei M, Baes C, et al. Genome-wide association analysis for β-hydroxybutyrate concentration in milk in Holstein dairy cattle. BMC Genet. 2019;20(1):58.

    Article  CAS  Google Scholar 

  133. Khan MZ, Wang D, Liu L, Usman T, Wen H, Zhang R, et al. Significant genetic effects of JAK2 and DGAT1 mutations on milk fat content and mastitis resistance in Holsteins. J Dairy Res. 2019;86(4):388–93.

    Article  CAS  Google Scholar 

  134. Cai Z, Dusza M, Guldbrandtsen B, Lund MS, Sahana G. Distinguishing pleiotropy from linked QTL between milk production traits and mastitis resistance in Nordic Holstein cattle. Genet Sel Evol. 2020;52(1):19.

    Article  Google Scholar 

  135. Li L, Tang W, Zhao M, Gong B, Cao M, Li J. Study on the regulation mechanism of lipopolysaccharide on oxidative stress and lipid metabolism of bovine mammary epithelial cells. Physiol Res. 2021;70(5):777–85.

    Article  CAS  Google Scholar 

  136. Sahana G, Guldbrandtsen B, Thomsen B, Holm LE, Panitz F, Brøndum RF, et al. Genome-wide association study using high-density single nucleotide polymorphism arrays and whole-genome sequences for clinical mastitis traits in dairy cattle. J Dairy Sci. 2014;97(11):7258–75.

    Article  CAS  Google Scholar 

  137. Cai ZA-O, Guldbrandtsen B, Lund MS, Sahana G. Prioritizing candidate genes post-GWAS using multiple sources of data for mastitis resistance in dairy cattle. BMC Genom. 2018;19:656.

    Article  Google Scholar 

  138. Freebern E, Santos DJA, Fang L, Jiang J, Parker Gaddis KL, Liu GE, et al. GWAS and fine-mapping of livability and six disease traits in Holstein cattle. BMC Genom. 2020;21(1):41.

    Article  CAS  Google Scholar 

  139. Huang J, Wang H, Wang C, Li J, Li Q, Hou M, et al. Single nucleotide polymorphisms, haplotypes and combined genotypes of lactoferrin gene and their associations with mastitis in chinese holstein cattle. Mol Biol Rep. 2010;37(1):477–83.

    Article  CAS  Google Scholar 

  140. Carvajal AM, Huircan P, Lepori A. Single nucleotide polymorphisms in immunity-related genes and their association with mastitis in chilean dairy cattle. Genet Mol Res. 2013;12(3):2702–11.

    Article  CAS  Google Scholar 

  141. Wojdak-Maksymiec K, Szyda J, Strabel T. Parity-dependent association between TNF-α and LTF gene polymorphisms and clinical mastitis in dairy cattle. BMC Vet Res. 2013;9:114.

    Article  Google Scholar 

  142. Chopra A, Gupta ID, Verma A, Chakravarty AK, Vohra V. Lactoferrin gene promoter variants and their association with clinical and subclinical mastitis in indigenous and crossbred cattle. Pol J Vet Sci. 2015;18(3):465–71.

    Article  CAS  Google Scholar 

  143. Dinesh K, Verma A, Das Gupta I, Thakur YP, Verma N, Arya A. Identification of polymorphism in exons 7 and 12 of lactoferrin gene and its association with incidence of clinical mastitis in Murrah buffalo. Trop Anim Health Prod. 2015;47(4):643–7.

    Article  Google Scholar 

  144. Soyeurt H, Bastin C, Colinet FG, Arnould VM, Berry DP, Wall E, et al. Mid-infrared prediction of lactoferrin content in bovine milk: potential indicator of mastitis. Animal. 2012;6(11):1830–8.

    Article  CAS  Google Scholar 

  145. Cheng Z, Buggiotti L, Salavati M, Marchitelli C, Palma-Vera S, Wylie A, et al. Global transcriptomic profiles of circulating leucocytes in early lactation cows with clinical or subclinical mastitis. Mol Biol Rep. 2021;48(5):4611–23.

    Article  CAS  Google Scholar 

  146. Huang JM, Wang ZY, Ju ZH, Wang CF, Li QL, Sun T, et al. Two splice variants of the bovine lactoferrin gene identified in Staphylococcus aureus isolated from mastitis in dairy cattle. Genet Mol Res. 2011;10(4):3199–203.

    Article  CAS  Google Scholar 

  147. Tanamati F, Stafuzza NB, Gimenez DFJ, Stella AAS, Santos DJA, Ferro MIT, et al. Differential expression of immune response genes associated with subclinical mastitis in dairy buffaloes. Animal. 2019;13(8):1651–7.

    Article  CAS  Google Scholar 

  148. Yang G, Yue Y, Li D, Duan C, Qiu X, Zou Y, et al. Antibacterial and immunomodulatory effects of Pheromonicin-NM on Escherichia coli-challenged bovine mammary epithelial cells. Int Immunopharmacol. 2020;84:106569.

    Article  CAS  Google Scholar 

  149. Wu J, Li L, Sun Y, Huang S, Tang J, Yu P, et al. Altered molecular expression of the TLR4/NF-κB signaling pathway in mammary tissue of chinese holstein cattle with mastitis. PLoS ONE. 2015;10(2):e0118458-e.

    Article  Google Scholar 

  150. Liu J, Ju Z, Li Q, Huang J, Li R, Li J, et al. Mannose-binding lectin 1 haplotypes influence serum MBL-A concentration, complement activity, and milk production traits in chinese holstein cattle. Immunogenetics. 2011;63(11):727–42.

    Article  CAS  Google Scholar 

  151. Wang C, Liu M, Li Q, Ju Z, Huang J, Li J, et al. Three novel single-nucleotide polymorphisms of MBL1 gene in chinese native cattle and their associations with milk performance traits. Vet Immunol Immunopathol. 2011;139(2–4):229–36.

    Article  CAS  Google Scholar 

  152. Yuan Z, Li J, Gao X, Xu S. SNPs identification and its correlation analysis with milk somatic cell score in bovine MBL1 gene. Mol Biol Rep. 2013;40(1):7–12.

    Article  CAS  Google Scholar 

  153. Fraser RS, Lumsden JS, Lillie BN. Identification of polymorphisms in the bovine collagenous lectins and their association with infectious diseases in cattle. Immunogenetics. 2018;70(8):533–46.

    Article  CAS  Google Scholar 

  154. Kamaldeep K, Magotra A, Pander BL, Dalal DS, Malik BS, Garg AR, et al. Evaluation of candidate genotype of immune gene MBL1 associated with udder health and performance traits in dairy cattle and buffalo of India. Trop Anim Health Prod. 2021;53(4):429.

    Article  CAS  Google Scholar 

  155. Wang X, Ju Z, Huang J, Hou M, Zhou L, Qi C, et al. The relationship between the variants of the bovine MBL2 gene and milk production traits, mastitis, serum MBL-C levels and complement activity. Vet Immunol Immunopathol. 2012;148(3–4):311–9.

    Article  CAS  Google Scholar 

  156. Suravajhala P, Benso A. Prioritizing single-nucleotide polymorphisms and variants associated with clinical mastitis. Adv Appl Bioinform Chem. 2017;10:57–64.

    Google Scholar 

  157. Oget C, Allain C, Portes D, Foucras G, Stella A, Astruc JM, et al. A validation study of loci associated with mastitis resistance in two french dairy sheep breeds. Genet Sel Evol. 2019;51(1):5.

    Article  Google Scholar 

  158. Rupp R, Senin P, Sarry J, Allain C, Tasca C, Ligat L, et al. A point mutation in suppressor of cytokine signalling 2 (Socs2) increases the susceptibility to inflammation of the mammary gland while associated with higher body weight and size and higher milk production in a sheep model. PLoS Genet. 2015;11(12):e1005629-e.

    Article  Google Scholar 

  159. Bakhtiarizadeh MR, Mirzaei S, Norouzi M, Sheybani N, Vafaei Sadi MS. Identification of gene modules and hub genes involved in mastitis development using a systems biology approach. Front Genet. 2020;11:722.

    Article  CAS  Google Scholar 

  160. Arun SJ, Thomson PC, Sheehy PA, Khatkar MS, Raadsma HW, Williamson P. Targeted analysis reveals an important role of JAK-STAT-SOCS genes for milk production traits in australian dairy cattle. Front Genet. 2015;6:342.

    Article  Google Scholar 

  161. Piechotta M, Holzhausen L, Araujo MG, Heppelmann M, Sipka A, Pfarrer C, et al. Antepartal insulin-like growth factor concentrations indicating differences in the metabolic adaptive capacity of dairy cows. J Vet Sci. 2014;15(3):343–52.

    Article  Google Scholar 

  162. Riley LG, Gardiner-Garden M, Thomson PC, Wynn PC, Williamson P, Raadsma HW, et al. The influence of extracellular matrix and prolactin on global gene expression profiles of primary bovine mammary epithelial cells in vitro. Anim Genet. 2010;41(1):55–63.

    Article  CAS  Google Scholar 

  163. Usman T, Yu Y, Liu C, Wang X, Zhang Q, Wang Y. Genetic effects of single nucleotide polymorphisms in JAK2 and STAT5A genes on susceptibility of chinese holsteins to mastitis. Mol Biol Rep. 2014;41(12):8293–301.

    Article  CAS  Google Scholar 

  164. Martin P, Palhière I, Maroteau C, Clément V, David I, Klopp GT, et al. Genome-wide association mapping for type and mammary health traits in french dairy goats identifies a pleiotropic region on chromosome 19 in the Saanen breed. J Dairy Sci. 2018;101(6):5214–26.

    Article  CAS  Google Scholar 

  165. Kiyici JM, Akyüz B, Kaliber M, Arslan K, Aksel EG, Cinar MU. Association of GH, STAT5A, MYF5 gene polymorphisms with milk somatic cell count, EC and pH levels of Holstein dairy cattle. Anim Biotechnol. 2022;33(3):401–7.

    Article  CAS  Google Scholar 

  166. Goldammer T, Zerbe H, Molenaar A, Schuberth HJ, Brunner RM, Kata SR, et al. Mastitis increases mammary mRNA abundance of beta-defensin 5, toll-like-receptor 2 (TLR2), and TLR4 but not TLR9 in cattle. Clin Diagn Lab Immunol. 2004;11(1):174–85.

    CAS  Google Scholar 

  167. Ruiz-Rodriguez CT, Brandt JR, Oliverio R, Ishida Y, Guedj N, Garrett EF, et al. Polymorphisms of the toll-like receptor 2 of goats (Capra hircus) may be associated with somatic cell count in milk. Anim Biotechnol. 2017;28(2):112–9.

    Article  CAS  Google Scholar 

  168. Elmaghraby MM, El-Nahas AF, Fathala MM, Sahwan FM, Tag El-Dien MA. Association of toll-like receptors 2 and 6 polymorphism with clinical mastitis and production traits in Holstein cattle. Iran J Vet Res. 2018;19(3):202–7.

    CAS  Google Scholar 

  169. Bai J, Lin J, Li W, Liu M. Association of toll-like receptor 2 polymorphisms with somatic cell score in Xinjiang Brown cattle. Anim Sci J. 2012;83(1):23–30.

    Article  CAS  Google Scholar 

  170. An R, Gao M, Meng Y, Tong X, Chen J, Wang J. Infective mastitis due to bovine-associated Streptococcus dysgalactiae contributes to clinical persistent presentation in a murine mastitis model. Vet Med Sci. 2021;7(5):1600–10.

    Article  CAS  Google Scholar 

  171. Zhang LP, Gan QF, Ma TH, Li HD, Wang XP, Li JY, et al. Toll-like receptor 2 gene polymorphism and its relationship with SCS in dairy cattle. Anim Biotechnol. 2009;20(3):87–95.

    Article  CAS  Google Scholar 

  172. Farhat K, Sauter KS, Brcic M, Frey J, Ulmer AJ, Jungi TW. The response of HEK293 cells transfected with bovine TLR2 to established pathogen-associated molecular patterns and to bacteria causing mastitis in cattle. Vet Immunol Immunopathol. 2008;125(3–4):326–36.

    Article  CAS  Google Scholar 

  173. Ma JL, Zhu YH, Zhang L, Zhuge ZY, Liu PQ, Yan XD, et al. Serum concentration and mRNA expression in milk somatic cells of toll-like receptor 2, toll-like receptor 4, and cytokines in dairy cows following intramammary inoculation with Escherichia coli. J Dairy Sci. 2011;94(12):5903–12.

    Article  CAS  Google Scholar 

  174. Ogorevc J, Simčič M, Zorc M, Škrjanc M, Dovč P. Polymorphism (rs650082970) is associated with somatic cell count in goat milk. PeerJ. 2019;7:e7340.

    Article  Google Scholar 

  175. Swain DK, Kushwah MS, Kaur M, Patbandha TK, Mohanty AK, Dang AK. Formation of NET, phagocytic activity, surface architecture, apoptosis and expression of toll like receptors 2 and 4 (TLR2 and TLR4) in neutrophils of mastitic cows. Vet Res Commun. 2014;38(3):209–19.

    Article  Google Scholar 

  176. Sharma BS, Leyva I, Schenkel F, Karrow NA. Association of toll-like receptor 4 polymorphisms with somatic cell score and lactation persistency in Holstein bulls. J Dairy Sci. 2006;89(9):3626–35.

    Article  CAS  Google Scholar 

  177. de Mesquita AQ, Rezende CSM, de Mesquita AJ, da Veiga Jardim EAJ, Kipnis APJ. Association of TLR4 polymorphisms with subclinical mastitis in brazilian holsteins. Braz J Microbiol. 2012;43(2):692–7.

  178. Panigrahi M, Kumar H, Nayak SS, Rajawat D, Parida S, Bhushan B, et al. Molecular characterization of CRBR2 fragment of TLR4 gene in association with mastitis in Vrindavani cattle. Microb Pathog. 2022;165:105483.

    Article  CAS  Google Scholar 

  179. Catalani E, Amadori M, Vitali A, Lacetera N. Short communication: lymphoproliferative response to lipopolysaccharide and incidence of infections in periparturient dairy cows. J Dairy Sci. 2013;96(11):7077–81.

    Article  CAS  Google Scholar 

  180. Chen H, Liu C, Xiang M, Yu J, Xia Y, Hu X, et al. Contribution of the mutation rs8193069 in TLR4 to mastitis resistance and performance in Holstein cows in southern China. Vet Med Sci. 2022;8(1):357–66.

    Article  CAS  Google Scholar 

  181. Wang XP, Luoreng ZM, Gao SX, Guo DS, Li JY, Gao X, et al. Haplotype analysis of TLR4 gene and its effects on milk somatic cell score in chinese commercial cattle. Mol Biol Rep. 2014;41(4):2345–51.

    Article  CAS  Google Scholar 

  182. Blum SE, Heller ED, Jacoby S, Krifucks O, Leitner G. Comparison of the immune responses associated with experimental bovine mastitis caused by different strains of Escherichia coli. J Dairy Res. 2017;84(2):190–7.

    Article  CAS  Google Scholar 

  183. Kandasamy S, Kerr DE. Genomic analysis of between-cow variation in dermal fibroblast response to lipopolysaccharide. J Dairy Sci. 2012;95(7):3852–64.

    Article  CAS  Google Scholar 

  184. Akhtar M, Guo S, Guo Y-f, Zahoor A, Shaukat A, Chen Y, et al. Upregulated-gene expression of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) via TLRs following NF-κB and MAPKs in bovine mastitis. Acta Trop. 2020;207:105458.

    Article  CAS  Google Scholar 

  185. Panigrahi M, Sharma A, Bhushan B. Molecular characterization and expression profile of partial TLR4 gene in association to mastitis in crossbred cattle. Anim Biotechnol. 2014;25(3):188–99.

    Article  CAS  Google Scholar 

  186. Ibeagha-Awemu EM, Lee JW, Ibeagha AE, Bannerman DD, Paape MJ, Zhao X. Bacterial lipopolysaccharide induces increased expression of toll-like receptor (TLR) 4 and downstream TLR signaling molecules in bovine mammary epithelial cells. Vet Res. 2008;39(2):11.

    Article  Google Scholar 

  187. Korkmaz FT, Elsasser TH, Kerr DE. Variation in fibroblast expression of toll-like receptor 4 and lipopolysaccharide-induced cytokine production between animals predicts control of bacterial growth but not severity of Escherichia coli mastitis. J Dairy Sci. 2018;101(11):10098–115.

    Article  CAS  Google Scholar 

  188. Sorg D, Danowski K, Korenkova V, Rusnakova V, Küffner R, Zimmer R, et al. Microfluidic high-throughput RT-qPCR measurements of the immune response of primary bovine mammary epithelial cells cultured from milk to mastitis pathogens. Animal. 2013;7(5):799–805.

    Article  CAS  Google Scholar 

  189. Fisher RAS. The genetical theory of natural selection. Oxford: Clarendon Press; 1930.

    Book  Google Scholar 

  190. Nelson RM, Pettersson ME, Carlborg Ö. A century after Fisher: time for a new paradigm in quantitative genetics. Trends Genet. 2013;29(12):669–76.

    Article  CAS  Google Scholar 

  191. Wang XS, Yu Y. Regulation mechanisms of epigenetics on inflammation and its perspective on breeding for mastitis resistance in dairy cattle. Yi Chuan (Hereditas). 2010;32(7):663–9.

    Article  CAS  Google Scholar 

  192. Singh K, Molenaar AJ, Swanson KM, Gudex B, Arias JA, Erdman RA, et al. Epigenetics: a possible role in acute and transgenerational regulation of dairy cow milk production. Animal. 2012;6(3):375–81.

    Article  CAS  Google Scholar 

  193. Wang N, Zhou C, Basang W, Zhu Y, Wang X, Li C, et al. Mechanisms by which mastitis affects reproduction in dairy cow: a review. Reprod Domest Anim. 2021;56(9):1165–75.

    Article  Google Scholar 

  194. Ibeagha-Awemu EM, Yu Y. Consequence of epigenetic processes on animal health and productivity: is additional level of regulation of relevance? Anim Front. 2021;11(6):7–18.

    Article  Google Scholar 

  195. Vanselow J, Yang W, Herrmann J, Zerbe H, Schuberth HJ, Petzl W, et al. DNA-remethylation around a STAT5-binding enhancer in the alphaS1-casein promoter is associated with abrupt shutdown of alphaS1-casein synthesis during acute mastitis. J Mol Endocrinol. 2006;37(3):463–77.

    Article  CAS  Google Scholar 

  196. Xiao C, Rajewsky K. MicroRNA control in the immune system: basic principles. Cell. 2009;136(1):26–36.

    Article  CAS  Google Scholar 

  197. Wang H, Wang X, Li X, Wang Q, Qing S, Zhang Y, et al. A novel long non-coding RNA regulates the immune response in MAC-T cells and contributes to bovine mastitis. FEBS J. 2019;286(9):1780–95.

    Article  CAS  Google Scholar 

  198. Weber JA, Baxter DH, Zhang S, Huang DY, Huang KH, Lee MJ, et al. The microRNA spectrum in 12 body fluids. Clin Chem. 2010;56(11):1733–41.

    Article  CAS  Google Scholar 

  199. Sun J, Aswath K, Schroeder SG, Lippolis JD, Reinhardt TA, Sonstegard TS. MicroRNA expression profiles of bovine milk exosomes in response to Staphylococcus aureus infection. BMC Genom. 2015;16:806.

    Article  CAS  Google Scholar 

  200. Caswell JL, Middleton DM, Gordon JR. Production and functional characterization of recombinant bovine interleukin-8 as a specific neutrophil activator and chemoattractant. Vet Immunol Immunopathol. 1999;67(4):327–40.

    Article  CAS  Google Scholar 

  201. Tribout T, Croiseau P, Lefebvre R, Barbat A, Boussaha M, Fritz S, et al. Confirmed effects of candidate variants for milk production, udder health, and udder morphology in dairy cattle. Genet Sel Evol. 2020;52(1):55.

    Article  CAS  Google Scholar 

  202. Cai Z, Guldbrandtsen B, Lund MS, Sahana G. Prioritizing candidate genes post-GWAS using multiple sources of data for mastitis resistance in dairy cattle. BMC Genom. 2018;19(1):656.

    Article  Google Scholar 

  203. Tsugami Y, Suzuki N, Kawahara M, Suzuki T, Nishimura T, Kobayashi K. Establishment of an in vitro culture model to study milk production and the blood-milk barrier with bovine mammary epithelial cells. Anim Sci J. 2020;91(1):e13355.

    Article  CAS  Google Scholar 

  204. Hillreiner M, Müller NI, Koch HM, Schmautz C, Küster B, Pfaffl MW, et al. Establishment of a 3D cell culture model of primary bovine mammary epithelial cells extracted from fresh milk. In Vitro Cell Dev Biol Anim. 2017;53(8):706–20.

    Article  CAS  Google Scholar 

  205. Huynh HT, Robitaille G, Turner JD. Establishment of bovine mammary epithelial cells (MAC-T): an in vitro model for bovine lactation. Exp Cell Res. 1991;197(2):191–9.

    Article  CAS  Google Scholar 

  206. Zavizion B, van Duffelen M, Schaeffer W, Politis I. Establishment and characterization of a bovine mammary epithelial cell line with unique properties. In Vitro Cell Dev Biol Anim. 1996;32(3):138–48.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

We thank the Slovenian Research Agency for funding through the programme grant no. P40220, and postgraduate “young researchers” funding to ZB. The funder had no role in this study and any other activities in relation to writing or publishing it.

Author information

Authors and Affiliations

Authors

Contributions

JO designed and conceptualized this study, ZB and JO collected, analysed and interpreted the data. JO and ZB co-wrote this manuscript. ZB wrote the initial draft of the manuscript. JO revised and edited the manuscript. Both authors read and approved the last modified submitted version and have agreed to be personally accountable for the author’s own contributions.

Corresponding author

Correspondence to Jernej Ogorevc.

Ethics declarations

Ethics approval and consent to participate

Not applicable. No animal experiments were conducted in this study.

Consent for publication

Not applicable. No human subjects were involved in this study.

Competing interests

The authors declare that they have no competing interests.

Supplementary Information

Additional file 1: Table S1.

Genetically modified mouse models associated with mastitis (MGI). Table S2. Associations between genotypes and mastitis related traits (mostly SCS) in dairy ruminants. Table S3. Candidate genes differentially expressed during mastitis.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Brajnik, Z., Ogorevc, J. Candidate genes for mastitis resistance in dairy cattle: a data integration approach. J Animal Sci Biotechnol 14, 10 (2023). https://doi.org/10.1186/s40104-022-00821-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40104-022-00821-0

Keywords