Skip to main content

The role of interleukin-10 receptor alpha (IL10Rα) in Mycobacterium avium subsp. paratuberculosis infection of a mammary epithelial cell line

Abstract

Background

Johne’s disease is a chronic wasting disease caused by the bacterium Mycobacterium avium subspecies paratuberculosis (MAP). Johne’s disease is highly contagious and MAP infection in dairy cattle can eventually lead to death. With no available treatment for Johne’s disease, genetic selection and improvements in management practices could help reduce its prevalence. In a previous study, the gene coding interleukin-10 receptor subunit alpha (IL10Rα) was associated with Johne’s disease in dairy cattle. Our objective was to determine how IL10Rα affects the pathogenesis of MAP by examining the effect of a live MAP challenge on a mammary epithelial cell line (MAC-T) that had IL10Rα knocked out using CRISPR/cas9. The wild type and the IL10Rα knockout MAC-T cell lines were exposed to live MAP bacteria for 72 h. Thereafter, mRNA was extracted from infected and uninfected cells. Differentially expressed genes were compared between the wild type and the IL10Rα knockout cell lines. Gene ontology was performed based on the differentially expressed genes to determine which biological pathways were involved.

Results

Immune system processes pathways were targeted to determine the effect of IL10Rα on the response to MAP infection. There was a difference in immune response between the wild type and IL10Rα knockout MAC-T cell lines, and less difference in immune response between infected and not infected IL10Rα knockout MAC-T cells, indicating IL10Rα plays an important role in the progression of MAP infection. Additionally, these comparisons allowed us to identify other genes involved in inflammation-mediated chemokine and cytokine signalling, interleukin signalling and toll-like receptor pathways.

Conclusions

Identifying differentially expressed genes in wild type and ILR10α knockout MAC-T cells infected with live MAP bacteria provided further evidence that IL10Rα contributes to mounting an immune response to MAP infection and allowed us to identify additional potential candidate genes involved in this process. We found there was a complex immune response during MAP infection that is controlled by many genes.

Peer Review reports

Background

Johne’s disease is a highly contagious chronic wasting condition caused by Mycobacterium avium subspecies paratuberculosis (MAP). Infection with MAP causes thickening of the intestines, which leads to decreased nutrient absorption, diarrhea and weight loss [1]. There is no cure, and infections eventually lead to death. A long incubation period means there is the potential for subclinical signs to be missed, such as reduced milk, protein and fat yield [2]. The disease is clinical when there is diarrhea, reduced milk production and absorption of nutrients, emaciation and death [3]. MAP is transmitted feco-orally and the most common route of transmission is contaminated colostrum and milk given to a neonatal calf; there is evidence that MAP can even survive extended pasteurization [4]. MAP can survive in the barn for up to 16 months, and both calves and cows are at risk when feces builds up from infected animals [5]. Management practices are crucial to preventing spread of MAP because they are currently the only means of control.

Dairy cattle herd-level prevalence of Johne’s disease in Ontario, Canada is an estimated 26.2–27.2%, and within-herd prevalence is an estimated 2.3–4.2% [6]. Diagnosis of Johne’s disease for dairy cattle is most commonly done with a milk ELISA test in Canada. This non-invasive test is low-cost, has a quick turn-around time, and is practical to use on-farm to get real-time results. However, the low sensitivity (30.4%) of this test leads to many false negative results [7]. These false negative results, combined with a long incubation period, create the potential for subclinical signs to be missed. Vaccines are being developed, or are in development [8,9,10], however, none are currently available to Canadian producers. Breeding for increased resistance to Johne’s disease could contribute to reducing its effect on individual animals and the industry.

Though low (6%), the heritability estimate for resistance to Johne’s disease indicates that selection against the disease could be an effective long-term strategy [11]. Breeding for increased resistance to Johne’s disease could contribute to the control of this disease [12], however, due to an unreliable phenotype, low sensitivity of tests and the long incubation period, it is challenging to incorporate Johne’s disease resistance into breeding programs. Despite these challenges, studies show there is genetic variation in disease susceptibility, as well as single nucleotide polymorphisms (SNPs) associated with a positive Johne’s disease test [12,13,14,15,16,17,18,19,20,21,22,23,24,25]. By knowing which genes and genetic variants are involved in resistance, they can be better represented in SNP arrays used for genotyping animals for genomic selection. By including causal variants in genomic selection, the most informative variants are used in a routine evaluation [26].

Interleukin-10 (IL10) is a cytokine that can regulate the antimicrobial activity of macrophages, resulting in an anti-inflammatory effect and tissue repair [27]. During an infection of murine macrophages with MAP, the IL10 receptor (IL10R) has been shown to be upregulated in response [28]. Interleukin-10 receptor alpha (IL10Rα) is one dimer making up the IL10R that binds IL10. When exposed to MAP, IL10Rα is upregulated, which contributes to decreasing inflammation and is thought to aid survival of MAP [29]. Four tightly linked SNPs that were discovered in IL10Rα were found to have an additive and dominant effect on MAP infection status [30], thus, IL10Rα plays a role in the progression of Johne’s disease. Precise mechanisms of this association, however, are currently not yet fully understood.

One way to learn more about the role of IL10Rα in the pathogenesis of Johne’s disease and MAP infection is by knocking out IL10Rα in a cell line and carrying out in vitro functional studies. An IL10Rα knockout cell line was created using bovine mammary epithelial (MAC-T) cells [15], and a functional study was performed by stimulating cells with MAP lysate. When comparing wild-type and knock-out MAC-T cells stimulated with MAP lysate, researchers observed increased expression of pro-inflammatory cytokines IL1α, IL1β, IL6 and TNFα in knock-out cells providing evidence that IL10Rα is involved in response to MAP infection [15]. This supports that genetic variation within this gene could facilitate breeding for an increased resistance to Johne’s disease. However, by infecting IL10Rα knockout MAC-T cells with live MAP, a response that better represents what happens in vivo will likely be observed. Therefore, the objective of this study was to examine the effect of live MAP on IL10Rα knockout MAC-T cells by examining differentially expressed genes between wild type MAC-T cells without exposure to MAP (WT), wild type MAC-T cells with exposure to MAP (WT-MAP), IL10Rα knockout MAC-T cells without exposure to MAP (KO), and IL10Rα knockout MAC-T cells with exposure to MAP (KO-MAP).

Results

Differentially expressed (DE) genes

In total, there were 1388 DE genes between WT and WT-MAP (Additional file 1: Table S1), 1738 DE genes between WT and KO (Additional file 1: Table S2), 1613 DE genes between WT-MAP and KO-MAP (Additional file 1: Table S3), and 561 DE genes between KO and KO-MAP (Additional file 1: Table S4). Number of shared and exclusive DE genes from the four comparisons is available in the additional materials (Additional file 2: Figure S1). We identified the 20 DE genes with the highest absolute fold change among each of the comparison groups (Additional file 3: Tables S5 to S8). None of the top 20 DE genes were identified by the PANTHER classification system as being immune system processes. The top three biological processes within all of the comparison groups were biological regulation, cellular processes, and metabolic processes (Fig. 1).

Fig. 1
figure 1

PANTHER biological processes for differentially expressed genes from the four contrasts: wildtype MAC-T cells (WT) versus the wildtype MAC-T cells infected with Mycobacterium avium subsp. Paratuberculosis (WT-MAP), WT versus the IL10Rα-knockout MAC-T cells (KO), WT-MAP versus the IL10Rα-knockout MAC-T cells infected with Mycobacterium avium subsp. Paratuberculosis (KO-MAP), and KO versus KO-MAP

Metabolic processes and immune response

Metabolic processes was one of the most significant gene ontology (GO) biological processes enriched by genes among all four comparisons identified in PANTHER enrichment analysis [31, 32]. The protein-protein interaction enrichment analyses were statistically significant (p-value < 1.0e− 16) for all comparison groups. A total of 46 Kyoto encyclopedia of genes and genomes (KEGG) pathways related to metabolic processes were enriched for DE genes identified through the WT versus WT-MAP contrast, 32 KEGG pathways through the WT versus KO contrast, 23 KEGG pathways through the WT-MAP versus KO-MAP contrast, and 24 KEGG pathways through the KO versus KO-MAP contrast (Additional file 4: Tables S9 to S12).

Many genes involved in immune system processes were also differentially expressed within all comparison groups even though they did not fall within the top 20 DE genes. There were six identified KEGG pathways from DE genes for the WT versus WT-MAP contrast, 11 KEGG pathways for the WT versus KO contrast, and 13 KEGG pathways for the WT-MAP versus KO-MAP contrast (Additional file 5: Tables S13 to S15). While there were 21 total DE genes relating to immune system processes for KO versus KO-MAP (Additional file 5: Table S16), no KEGG pathways were identified, possibly due to the small number of known interactions between each of the genes.

There were 213 KEGG pathways related to inflammation mediated by chemokine and cytokine signalling pathways in the four contrasts. Of these, 13 KEGG pathways were identified from the WT versus WT-MAP contrast, 98 KEGG pathways were identified from the WT versus KO contrast, and 38 were identified from the WT-MAP versus KO-MAP, and 64 KEGG pathways were identified from the KO versus KO-MAP contrast (Additional file 6: S17 to S20).

There were 132 KEGG pathways involved in interleukin signalling and toll-like receptor signalling in the four contrasts identified. Of these, 18 KEGG pathways related to interleukin signalling were identified from the WT versus WT-MAP contrast, 42 KEGG pathways from the WT versus KO contrast, 52 KEGG pathways from the WT-MAP versus KO-MAP contrast, and three KEGG pathways from the KO versus KO-MAP contrast (Additional file 7: S21 to S24). Additionally, 17 KEGG pathways related to toll-like receptor signalling were identified from the WT versus WT-MAP contrast (Additional file 8: 25). No other contrasts identified the toll-like receptor signalling KEGG pathway.

Overall, there was little difference in immune response in the KO versus KO-MAP contrast. There were 21 DE genes related to immune system processes (Additional file 5: Table S16), 9 DE genes related to inflammation (Additional file 9: Table S26), and 2 DE genes related to the interleukin signalling pathway (Additional file 9: Table S27). Protein-Protein interaction analyses was performed in STRING [33] which revealed significant enrichment for the genes related to immune system processes (Additional file 10). However, there was no enrichment for DE genes involved in toll-like receptor and interleukin signalling pathways. Figure 2 shows all DE genes from the KO versus KO-MAP contrast involved in inflammation mediated by chemokine and cytokine signalling, interleukin signalling, toll-like receptor signalling, B cell activation, and T cell activation pathways. The majority of these genes were downregulated in the KO-MAP treatment, and most are exclusive to inflammation and no other immune system function.

Fig. 2
figure 2

PANTHER immune system processes for differentially expressed genes from the contrast of the IL10Rα-knockout MAC-T cells (KO) versus the IL10Rα-knockout MAC-T cells infected with Mycobacterium avium subsp. Paratuberculosis (KO-MAP)

Discussion

In this knockout study, we investigated the MAC-T cell response to MAP infection in the absence of IL10Rα. Our results highlighted the roles of metabolic and immune responses to MAP infection, specifically identifying genes involved in inflammation mediated chemokine and cytokine signalling, interleukin signalling and toll-like receptor pathways. This study has provided further evidence of the role of IL10Rα during MAP infection and has identified many other involved genes. The top three biological processes for all four comparisons were biological regulation, cellular processes, and metabolic processes (Fig. 1). These results were consistent with the literature because differences in the regulation of biological processes and cellular processes are typically observed and metabolic processes are affected in knockout gene studies [20, 34, 35], and infection with MAP bacteria has been shown to alter the metabolism of cells [36,37,38].

Metabolic response

Several studies have shown lipid metabolism and cholesterol pathways were affected by MAP infection, which was likely in order to facilitate MAP survival [36,37,38], however, we did not specifically assess MAP viability. For macrophages, alteration of these pathways is believed to help establish infection and its persistence within intracellular compartments [36, 38]. In our study, enrichments were found in DE genes in non-infected and infected cell line comparisons, which included the KEGG pathways: cholesterol metabolism; retinol metabolism; fat digestion and absorption; and peroxisome proliferator-activated receptors (PPAR) signalling. In this study we have used MAC-T cells and not macrophages, so DE genes related to metabolism may not be due to MAP infection, but could be simply because a gene (IL10Rα) has been knocked-out. Additionally, enrichment analyses can be biased towards well studied genes and pathways, so our results may not convey the entirety of what is occurring.

When further examining metabolic processes among all comparisons, common genes were found in WT versus WT-MAP, WT versus KO, and WT-MAP versus KO-MAP contrasts. This shows that gene editing had an effect on MAC-T cell metabolism, because these genes were only absent in the comparison where both cell lines had IL10Rα knocked out, the KO versus KO-MAP comparison. Many pathways relating to adipose cells were found, such as fat digestion and absorption, cholesterol metabolism, and the adipocytokine signalling pathway (Additional file 4). Cells in the current study were mammary epithelial cells and because the mammary gland is composed of fatty tissue, and mobilization of fat is needed in the mammary gland to produce milk, the presence of pathways relating to adipose tissue is expected.

Immune response

Immune system processes related to both innate and acquired immunity were also examined to gain a greater understanding of how IL10Rα affects the pathogenesis of MAP bacteria (Additional file 5). DE genes involved in the interleukin signalling pathway and the toll-like receptor signalling pathway were inspected as they play a crucial role in pathogen recognition, cell signalling and immune system activation (Additional files 7 and 8). As IL10Rα is a dimer making up the IL10 receptor for an anti-inflammatory cytokine IL10, the inflammation pathway regulated by chemokine and cytokine signalling was also examined as this pathway was expected to be significantly enriched (Additional file 6). Collectively examining DE genes involved in these pathways, as well as the B cell activation and T cell activation pathways, showed that there was an immune response due to MAP infection (Additional file 11).

In the comparison of WT versus WT-MAP, we identified DE genes involved in the immune system, including transcription factor Jun (Jun), which was downregulated in WT-MAP. The Jun pathway has been implicated in the activation of transcription of IL10 in T helper 2 cells, which are a major source of IL10 [39], and MAC-T cells are also capable of producing IL10 [40]. Another identified DE gene was interleukin 6 (IL6), which was upregulated in WT-MAP. Amongst IL6’s pleiotropic properties, IL6 can act as a pro-inflammatory cytokine [41,42,43], and has been found in previous studies to be associated with IL10 and Johne’s disease [19], chronic human inflammatory lung disease [44], and bovine respiratory disease [45].

In the WT versus KO contrast, we identified DE genes involved in the immune system, such as interleukin 36 alpha (IL36α). IL36α was downregulated in the KO and encodes for a pro-inflammatory cytokine, and is a member of the interleukin 1 (IL1) family of cytokines that plays a major role in initiating inflammation in response to infection, or tissue injury [46,47,48,49,50]. In this comparison, mitogen activated protein kinases (MAPK) 12 (MAPK12) and MAPK13 were downregulated in the KO. These are stress-activated protein kinases that play an important role in the innate immune system signalling and inflammation [51]. Toll-like receptor 6 (TLR6) was upregulated in the KO, and can also activate the nuclear factor kappa beta (NF-κβ) signalling pathway to generate a pro-inflammatory response [52]. In contrast, the bovine major histocompatibility complex class II DR alpha (BoLA-DRα) was downregulated in the KO and DRα is required for antigen presentation to initiate adaptive immunity [27]. IL6 was upregulated in the WT versus KO contrast, and all other contrasts (i.e. WT versus WT-MAP, WT-MAP versus KO-MAP, and KO versus KO-MAP).

In the comparison of WT-MAP versus KO-MAP, IL10Rα was downregulated in the KO-MAP, as expected, due to gene editing. Interleukin 20 receptor alpha (IL20Rα) was also downregulated, and it regulates the JAK-STAT signalling pathway [53, 54]. IL20Rα is expressed in the epidermis, which could explain its presence in MAC-T cells, however, the JAK-STAT signalling pathway is also important for immune cell development. Related to this signalling pathway, Janus kinase 3 (JAK3) was upregulated in the KO-MAP, and signal transducer and activator of transcription 3 (STAT3) was downregulated; JAK3 is involved in cytokine receptor-mediated intracellular signal transduction, and STAT3 acts as a transcription factor in response to cytokines and growth factors [54].

Response to infection

Of the four comparisons, there were fewer DE genes identified that are associated with immune response between KO versus KO-MAP, providing evidence that IL10Rα is a major gene of interest when examining MAP infection. When looking at DE genes relating to inflammation, only IL6 and chemokine (C-C motif) ligand 20 (CCL20) were upregulated in the KO-MAP, whereas the other seven inflammation-related genes were downregulated. CCL20 has been shown to have increased expression in human inflammatory bowel disease [55], and is involved in the trafficking of various immune cell types [27]. Two DE genes relating to the interleukin signalling pathway were present, and these were interleukin 12 receptor beta 2 (IL12Rβ2), which was downregulated in the KO-MAP, and IL6, which was upregulated. The IL12R is involved in the activation of T cells and natural killer (NK) cells [56]. Collectively, these findings indicate that upon MAP infection, IL10Rα knockout cells do still respond to MAP infection, however, the response is far less than what it would be in unedited cells.

Polygenic nature of immune response to MAP bacteria and application of results

Our results support that immune response to MAP bacteria is highly polygenic, which is expected, as genetic variation of disease-related traits is usually explained by a large number of genetic variants with small effects. An example of the highly polygenic nature of disease-related traits is chronic human inflammatory bowel disease (IBD), which has been associated with MAP infection [57, 58], where researchers have implicated more than 130 candidate genes [59,60,61]. Studies have shown that many genes are thought to contribute to MAP infection status [13], and incorporating resistance to Johne’s disease into genomic evaluations will involve taking into account variants in multiple genes and not just IL10Rα. Further research into causal variants is needed because this information could be useful in pharmaceutical development to treat Johne’s disease, and including causal variants in genotyping arrays may improve accuracy of genomic selection [26] for selecting animals more resistant to MAP infection.

Conclusions

Johne’s disease is devastating for the dairy cattle industry, with serious animal welfare implications and substantial economic loss. Prior research identified IL10Rα to be involved in the immune response in MAC-T cells to MAP cell lysate, however, this may not necessarily reflect the response to a live pathogen challenge. Targeting genes involved in immune system processes allowed us to identify specific pathways that were related to DE genes between wild type and IL10Rα knockout MAC-T cells, such as inflammation mediated by the chemokine and cytokine signalling pathway, the interleukin signalling pathway, and the toll-like receptor signalling pathway. This study provided further information on IL10Rα’s role in MAP infection and pathogenesis, offering further support that IL10Rα is a good candidate gene, and presenting results that could help with development of therapeutics. The immune response to Johne’s disease and progression of MAP infection is complex, and this in vitro study gives further insight into other genes and pathways involved that require further study.

Methods

Knockout cell line development, infection, and extraction of mRNA

The MAC-T cell cultures (Nexia Biotechnologies, Quebec, Canada) were developed by Huynh et al. [62], while IL10Rα knockout and confirmation of knockout were performed using the same methods as Mallikarjunappa et al. [15]. MAP infections were performed using the method of Lamont et al. [63] using MAP (Madonna strain) gifted from Dr. Lucy Mutharia (University of Guelph). All the experiments were carried out in quadruplicate at four independent times. Bacteria was cultured using the methods described by Shandilya et al. [40], and the bacterial load was added to each infected treatment group to form a 10:1 multiplicity of infection [40, 64]. There were four treatment groups, described in Table 1, with unedited and IL10Rα knockout cells infected with MAP bacteria or not. As described in Shandilya et al. [40], MAC-T cells had MAP bacteria added (for 72 h), or for uninfected cells, MAP carrier solution media was added. Extraction of mRNA was performed using the same method as Shandilya et al. [65] using the RNeasy Mini Kit (Quiagen, Germany). Before outsourcing (Genewiz, Azenta Life Sciences, US) the samples for RNA-Sequencing, the purity of the RNA samples was determined using the Cytation 5 Spectrophotometer (Biotek, USA) at A260/280 nm ratio. The integrity of the RNA samples was assessed by bioanalyzer, and the RNA concentration was determined using the Qubit® 2.0 Fluorometer. Paired-end (2 × 150 bp) reads were generated using the Illumina HiSeq 2500 sequencer.

Table 1 Experimental design: MAC-T cells and IL10Rα knockout MAC-T cells were infected with Mycobacterium avium subspecies paratuberculosis (MAP) for 72 h, after which mRNA was extracted for sequencing

The data in this study have been deposited in NCBI’s Gene Expression Omnibus [66] and are accessible through GEO series accession number GSE247921 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?&acc=GSE247921).

RNA-Sequencing analysis

To analyze the RNA-Sequencing data, the CLC Genomics Workbench software 20.0.4 (QIAGEN, Aarhus, Denmark) was used. FastQ files were imported into the software, and quality control was done following the parameters described in [67]. A Phred score measures the probability that a nucleobase was identified correctly during the sequencing process, with low scores indicating incorrect identification. A score of at least 20 is a standard quality control cut-off as that indicates a 99% accuracy [67]. In this study all Phred scores were above 35, and the accuracy was 99.97%. The data was then trimmed based on quality scores which included a maximum error probability of 0.05 and a maximum number of ambiguous nucleotides of 2.

For RNA-Sequencing analysis, the CLC Genomics Workbench 20.0.4 (QIAGEN, Aarhus, Denmark) environment, was used to align pair-end sequence reads of each sample against the bovine reference genome, ARS-UCD1.2 imported from Ensembl [68]. The parameters used for global alignments were 0.6 for length fraction and 0.6 for similarity fraction, which represents the minimum percentage of total alignment that must match the reference sequence with a minimum percentage identity between the aligned region of the read and the reference. In addition, two mismatches, three insertions and three deletions per read were allowed. To facilitate group comparisons, the gene levels were quantified in reads per kilobase per million mapped reads (RPKM) and transformed to log 2 [69].

Differentially expressed (DE) genes analyses for WT versus WT-MAP, WT versus KO, WT-MAP versus KO-MAP, and KO versus KO-MAP contrasts were performed using the CLC Genomics Workbench 20.0.4 (QIAGEN, Aarhus, Denmark). The two-group comparisons were carried out using samples obtained after 72 h of infection from the four groups, WT (n=3), WT-MAP (n=4), KO (n=4) and KO-MAP (n=4). The WT group only included three samples because one sample was removed due to failing the quality control. Empirical analysis was done using the DGE tool in CLC workbench and performed for each contrast, using the following parameter to keep data: total count ≥ 5.0. Transcript levels were quantified in reads per kilobase per million mapped reads (RPKM). DE genes between each contrast were defined by a p-value ≤0.01, FDR ≤0.05, and |fold change|≥2.

Gene ontology

After DE genes were identified, GO enrichment analysis was performed for each analysed contrast, using PANTHER [31, 32]. Genes specifically related to metabolic processes, as well as the immune system processes of inflammation mediated by chemokine and cytokine signalling pathways, the Toll-like receptor signalling pathway, the interleukin signalling pathway, the B cell activation pathway, and the T cell activation pathway were targeted. Venn diagrams were created to determine overlap between each of the comparison groups [70]. Knowledge about specific targeted genes of interest was gained from GeneCards [27], and protein-protein interaction network analysis was performed using STRING [33].

Data availability

The data in this study are accessible through NCBI GEO series accession number GSE247921 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?&acc=GSE247921).

References

  1. Hendrick SH, Kelton DF, Leslie KE, Lissemore KD, Archambault M, Duffield TF. Effect of paratuberculosis on culling, milk production, and milk quality in dairy herds. J Am Vet Med Assoc. 2005;227:1302–8.

    Article  PubMed  Google Scholar 

  2. Pritchard TC, Coffey MP, Bond KS, Hutchings MR, Wall E. Phenotypic effects of subclinical paratuberculosis (Johne’s disease) in dairy cattle. J Dairy Sci. 2017;100:679–90.

    Article  CAS  PubMed  Google Scholar 

  3. Coussens PM. Mycobacterium paratuberculosis and the bovine immune system. Anim Health Res Rev. 2001;2:141–62.

    Article  CAS  PubMed  Google Scholar 

  4. Grant IR, Rowe MT, Dundee L, Hitchings E. Mycobacterium avium ssp. paratuberculosis: its incidence, heat resistance and detection in milk and dairy products. Int J Dairy Technol. 2001;54:2–13.

    Article  CAS  Google Scholar 

  5. Slana I, Pribylova R, Kralova A, Pavlik I. Persistence of Mycobacterium avium subsp. paratuberculosis at a farm-scale Biogas Plant supplied with manure from paratuberculosis-affected dairy cattle. Appl Environ Microbiol. 2011;77:3115–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Pieper L, Sorge US, DeVries T, Godkin A, Lissemore K, Kelton D. Comparing ELISA test-positive prevalence, risk factors and management recommendations for Johne’s disease prevention between organic and conventional dairy farms in Ontario, Canada. Prev Vet Med. 2015;122:83–91.

    Article  PubMed  Google Scholar 

  7. Norton S, Johnson WO, Jones G, Heuer C. Evaluation of diagnostic tests for Johne’s Disease (Mycobacterium Avium subspecies paratuberculosis) in New Zealand dairy cows. J Vet Diagn Invest. 2010;22:341–51.

    Article  PubMed  Google Scholar 

  8. Stabel JR, Bannantine JP. Reduced tissue colonization of Mycobacterium avium subsp. paratuberculosis in neonatal calves vaccinated with a cocktail of recombinant proteins. Vaccine. 2021;39:3131–40.

    Article  CAS  PubMed  Google Scholar 

  9. Park KT, Allen AJ, Bannantine JP, Seo KS, Hamilton MJ, Abdellrazeq GS, et al. Evaluation of two mutants of Mycobacterium avium subsp. paratuberculosis as candidates for a live attenuated vaccine for Johne’s disease. Vaccine. 2011;29:4709–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Phanse Y, Wu C-W, Venturino AJ, Hansen C, Nelson K, Broderick SR, et al. A protective vaccine against Johne’s disease in cattle. Microorganisms. 2020;8:1427.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Brito LF, Mallikarjunappa S, Sargolzaei M, Koeck A, Chesnais J, Schenkel FS, et al. The genetic architecture of milk ELISA scores as an indicator of Johne’s disease (paratuberculosis) in dairy cattle. J Dairy Sci. 2018;101:10062–75.

    Article  CAS  PubMed  Google Scholar 

  12. van Hulzen KJE, Koets AP, Nielen M, Heuven HCM, van Arendonk JAM, Klinkenberg D. The effect of genetic selection for Johne’s disease resistance in dairy cattle: results of a genetic-epidemiological model. J Dairy Sci. 2014;97:1762–73.

    Article  PubMed  Google Scholar 

  13. Alpay F, Zare Y, Kamalludin MH, Huang X, Shi X, Shook GE, et al. Genome-Wide Association Study of Susceptibility to Infection by Mycobacterium avium subspecies paratuberculosis in Holstein cattle. PLoS ONE. 2014;9:e111704.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Pant SD, Verschoor CP, Skelding AM, Schenkel FS, You Q, Biggar GA, et al. Bovine IFNGR2, IL12RB1, IL12RB2, and IL23R polymorphisms and MAP infection status. Mamm Genome. 2011;22:583–8.

    Article  CAS  PubMed  Google Scholar 

  15. Mallikarjunappa S, Shandilya UK, Sharma A, Lamers K, Bissonnette N, Karrow NA, et al. Functional analysis of bovine interleukin-10 receptor alpha in response to Mycobacterium avium subsp. paratuberculosis lysate using CRISPR/Cas9. BMC Genet. 2020;21:121.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Shook GE, Chaffer M, Wu X-L, Ezra E. Genetic parameters for paratuberculosis infection and effect of infection on production traits in Israeli holsteins. Anim Genet. 2012;43:56–64.

    Article  PubMed  Google Scholar 

  17. Gonda MG, Chang YM, Shook GE, Collins MT, Kirkpatrick BW. Genetic variation of Mycobacterium avium ssp. paratuberculosis infection in US Holsteins. J Dairy Sci. 2006;89:1804–12.

    Article  CAS  PubMed  Google Scholar 

  18. MacHugh DE, Taraktsoglou M, Killick KE, Nalpas NC, Browne JA, DE Park S, et al. Pan-genomic analysis of bovine monocyte-derived macrophage gene expression in response to in vitro infection with Mycobacterium avium subspecies paratuberculosis. Vet Res. 2012;43:25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Mallikarjunappa S, Schenkel FS, Brito LF, Bissonnette N, Miglior F, Chesnais J, et al. Association of genetic polymorphisms related to Johne’s disease with estimated breeding values of Holstein sires for milk ELISA test scores. BMC Vet Res. 2020;16:165.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ariel O, Brouard J-S, Marete A, Miglior F, Ibeagha-Awemu E, Bissonnette N. Genome-wide association analysis identified both RNA-seq and DNA variants associated to paratuberculosis in Canadian Holstein cattle ‘in vitro’ experimentally infected macrophages. BMC Genomics. 2021;22:162.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Canive M, Badia-Bringué G, Vázquez P, González-Recio O, Fernández A, Garrido JM, et al. Identification of loci associated with pathological outcomes in Holstein cattle infected with Mycobacterium avium subsp. paratuberculosis using whole-genome sequence data. Sci Rep. 2021;11:20177.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Canive M, González-Recio O, Fernández A, Vázquez P, Badia-Bringué G, Lavín JL, et al. Identification of loci associated with susceptibility to Mycobacterium avium subsp. paratuberculosis infection in Holstein cattle using combinations of diagnostic tests and imputed whole-genome sequence data. PLoS ONE. 2021;16:e0256091.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sanchez M-P, Tribout T, Fritz S, Guatteo R, Fourichon C, Schibler L, et al. New insights into the genetic resistance to paratuberculosis in Holstein cattle via single-step genomic evaluation. Genet Sel Evol. 2022;54:67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Sanchez M-P, Guatteo R, Davergne A, Saout J, Grohs C, Deloche M-C, et al. Identification of the ABCC4, IER3, and CBFA2T2 candidate genes for resistance to paratuberculosis from sequence-based GWAS in Holstein and Normande dairy cattle. Genet Sel Evol. 2020;52:14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Badia-Bringué G, Canive M, Alonso-Hearn M. Control of Mycobacterium avium subsp. paratuberculosis load within infected bovine monocyte-derived macrophages is associated with host genetics. Front Immunol. 2023;14:1042638.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Boichard D, Ducrocq V, Fritz S. Sustainable dairy cattle selection in the genomic era. J Anim Breed Genet. 2015;132:135–43.

    Article  CAS  PubMed  Google Scholar 

  27. Stelzer G, Rosen N, Plaschkes I, Zimmerman S, Twik M, Fishilevich S et al. The GeneCards suite: from Gene Data Mining to Disease Genome sequence analyses. Curr Protoc Bioinforma. 2016;54.

  28. Hussain T, Zhao D, Shah SZA, Wang J, Yue R, Liao Y, et al. MicroRNA 27a-3p regulates antimicrobial responses of murine macrophages infected by Mycobacterium avium subspecies paratuberculosis by Targeting Interleukin-10 and TGF-β-Activated protein kinase 1 binding protein 2. Front Immunol. 2018;8:1915.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Casey ME, Meade KG, Nalpas NC, Taraktsoglou M, Browne JA, Killick KE et al. Analysis of the bovine monocyte-derived macrophage response to Mycobacterium avium subspecies paratuberculosis infection using RNA-seq. Front Immunol. 2015;6.

  30. Verschoor CP, Pant SD, You Q, Schenkel FS, Kelton DF, Karrow NA. Mycobacterium avium ssp. Paratuberculosis. 2010;:9.

  31. Mi H, Dong Q, Muruganujan A, Gaudet P, Lewis S, Thomas PD. PANTHER version 7: improved phylogenetic trees, orthologs and collaboration with the Gene Ontology Consortium. Nucleic Acids Res. 2010;38 suppl1:D204–10.

    Article  Google Scholar 

  32. Thomas PD, Campbell MJ, Kejariwal A, Mi H, Karlak B, Daverman R, et al. PANTHER: a Library of protein families and subfamilies indexed by function. Genome Res. 2003;13:2129–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Jensen LJ, Kuhn M, Stark M, Chaffron S, Creevey C, Muller J, et al. STRING 8–a global view on proteins and their functional interactions in 630 organisms. Nucleic Acids Res. 2009;37(Database):D412–6.

    Article  CAS  PubMed  Google Scholar 

  34. Asselstine V, Miglior F, Suárez-Vega A, Fonseca PAS, Mallard B, Karrow N, et al. Genetic mechanisms regulating the host response during mastitis. J Dairy Sci. 2019;102:9043–59.

    Article  CAS  PubMed  Google Scholar 

  35. Marete A, Ariel O, Ibeagha-Awemu E, Bissonnette N. Identification of long non-coding RNA isolated from naturally infected macrophages and Associated with bovine Johne’s Disease in Canadian Holstein using a combination of neural networks and logistic regression. Front Vet Sci. 2021;8:639053.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Johansen MD, de Silva K, Plain KM, Whittington RJ, Purdie AC. Mycobacterium avium subspecies paratuberculosis is able to manipulate host lipid metabolism and accumulate cholesterol within macrophages. Microb Pathog. 2019;130:44–53.

    Article  CAS  PubMed  Google Scholar 

  37. Alonso-Hearn M, Canive M, Blanco-Vazquez C, Torremocha R, Balseiro A, Amado J, et al. RNA-Seq analysis of ileocecal valve and peripheral blood from Holstein cattle infected with Mycobacterium avium subsp. paratuberculosis revealed dysregulation of the CXCL8/IL8 signaling pathway. Sci Rep. 2019;9:14845.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Ariel O, Gendron D, Dudemaine P-L, Gévry N, Ibeagha-Awemu EM, Bissonnette N. Transcriptome profiling of bovine macrophages infected by Mycobacterium avium spp. paratuberculosis depicts Foam Cell and Innate Immune Tolerance Phenotypes. Front Immunol. 2020;10:2874.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Wang Z-Y, Sato H, Kusam S, Sehra S, Toney LM, Dent AL. Regulation of IL-10 gene expression in Th2 cells by Jun Proteins. J Immunol. 2005;174:2098–105.

    Article  CAS  PubMed  Google Scholar 

  40. Shandilya UK, Wu X, McAllister C, Mutharia L, Karrow NA. Impact of Mycobacterium avium subsp. paratuberculosis infection on bovine IL10RA knockout mammary epithelial (MAC-T) cells. Vitro Cell Dev Biol - Anim. 2023. https://doi.org/10.1007/s11626-023-00758-2.

    Article  Google Scholar 

  41. Atsushi Muraguchi T, Hirano B, Tang T, Matsuda Y, Horii K, Nakajima, et al. The essential role of B cell stimulatory factor 2 (BSF-2/IL-6) for the terminal differentiation of B cells. J Exp Med. 1988;167:332–44.

    Article  Google Scholar 

  42. Tackey E, Lipsky PE, Illei GG. Rationale for interleukin-6 blockade in systemic lupus erythematosus. Lupus. 2004;:339–43.

  43. Sun Y, Biscarini F, Bovenhuis H, Parmentier HK, van der Poel JJ. Genetic parameters and across-line SNP associations differ for natural antibody isotypes IgM and IgG in laying hens. Anim Genet. 2012;44:413–24.

    Article  PubMed  Google Scholar 

  44. Rincon M, Irvin CG. Role of IL-6 in Asthma and other Inflammatory Pulmonary diseases. Int J Biol Sci. 2012;8:1281–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Neupane M, Kiser JN, the Bovine Respiratory Disease Complex Coordinated Agricultural Project Research Team, Neibergs HL. Gene set enrichment analysis of SNP data in dairy and beef cattle with bovine respiratory disease. Anim Genet. 2018;49:527–38.

    Article  CAS  PubMed  Google Scholar 

  46. Fields JK, Günther S, Sundberg EJ. Structural basis of IL-1 family Cytokine Signaling. Front Immunol. 2019;10:1412.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Henry CM, Sullivan GP, Clancy DM, Afonina IS, Kulms D, Martin SJ. Neutrophil-derived proteases escalate inflammation through activation of IL-36 Family cytokines. Cell Rep. 2016;14:708–22.

    Article  CAS  PubMed  Google Scholar 

  48. Kono H, Rock KL. How dying cells alert the immune system to danger. Nat Rev Immunol. 2008;8:279–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Sims JE, Smith DE. The IL-1 family: regulators of immunity. Nat Rev Immunol. 2010;10:89–102.

    Article  CAS  PubMed  Google Scholar 

  50. Afonina IS, Tynan GA, Logue SE, Cullen SP, Bots M, Lüthi AU, et al. Granzyme B-Dependent Proteolysis acts as a switch to enhance the proinflammatory activity of IL-1α. Mol Cell. 2011;44:265–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Escós A, Risco A, Alsina-Beauchamp D, Cuenda A. p38γ and p38δ mitogen activated protein kinases (MAPKs), New stars in the MAPK Galaxy. Front Cell Dev Biol. 2016;4.

  52. Kawai T, Akira S. Signaling to NF-κB by toll-like receptors. Trends Mol Med. 2007;13:460–9.

    Article  CAS  PubMed  Google Scholar 

  53. Lamichhane S, Mo J-S, Sharma G, Choi T-Y, Chae S-C. MicroRNA 452 regulates IL20RA-mediated JAK1/STAT3 pathway in inflammatory colitis and colorectal cancer. Inflamm Res. 2021;70:903–14.

    Article  CAS  PubMed  Google Scholar 

  54. Rawlings JS, Rosler KM, Harrison DA. The JAK/STAT signaling pathway. J Cell Sci. 2004;117:1281–3.

    Article  CAS  PubMed  Google Scholar 

  55. Kaser A, Ludwiczek O, Holzmann S, Moschen AR, Weiss G, Enrich B, et al. Increased expression of CCL20 in human inflammatory bowel disease. J Clin Immunol. 2004;24:74–85.

    Article  CAS  PubMed  Google Scholar 

  56. Ohyama H, Kato-Kogoe N, Nishimura F, Takeuchi-Hatanaka K, Matsushita S, Yamanegi K, et al. Differential effects of polymorphisms in the 5′ Flanking Region of IL12RB2 on NK- and T-Cell activity. J Interferon Cytokine Res. 2008;28:563–70.

    Article  CAS  PubMed  Google Scholar 

  57. Proietti E, Fuhler GM, Peppelenbosch MP. Mycobacterium Avium subspecies paratuberculosis infection and Biological Treatment of IBD: cause or Consequence? J Crohns Colitis. 2021;15:1247–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Van Der Sloot KWJ, Voskuil MD, Blokzijl T, Dinkla A, Ravesloot L, Visschedijk MC, et al. Isotype-specific antibody responses to Mycobacterium avium paratuberculosis antigens are Associated with the Use of Biologic Therapy in Inflammatory Bowel Disease. J Crohns Colitis. 2021;15:1253–63.

    Article  PubMed  Google Scholar 

  59. de Lange KM, Moutsianas L, Lee JC, Lamb CA, Luo Y, Kennedy NA, et al. Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat Genet. 2017;49:256–61.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Liu JZ, van Sommeren S, Huang H, Ng SC, Alberts R, Takahashi A, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 2015;47:979–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. the NIDDK IBD Genetics Consortium, the Belgian-French IBD Consortium, the Wellcome Trust Case Control Consortium, Barrett JC, Hansoul S, Nicolae DL, et al. Genome-wide association defines more than 30 distinct susceptibility loci for Crohn’s disease. Nat Genet. 2008;40:955–62.

    Article  Google Scholar 

  62. 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;197:191–9.

    Article  CAS  PubMed  Google Scholar 

  63. Lamont EA, Talaat AM, Coussens PM, Bannantine JP, Grohn YT, Katani R et al. Screening of Mycobacterium avium subsp. paratuberculosis mutants for attenuation in a bovine monocyte-derived macrophage model. Front Cell Infect Microbiol 2014;4.

  64. Shandilya UK, Wu X, McAllister C, Mutharia L, Karrow NA. Role of toll-like receptor 4 in Mycobacterium avium subsp. paratuberculosis infection of bovine mammary epithelial (MAC-T) cells in Vitro. Microbiol Spectr. 2023. https://doi.org/10.1128/spectrum.04393-22.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Shandilya UK, Sharma A, Mallikarjunappa S, Guo J, Mao Y, Meade KG, et al. CRISPR-Cas9–mediated knockout of TLR4 modulates Mycobacterium avium ssp. paratuberculosis cell lysate–induced inflammation in bovine mammary epithelial cells. J Dairy Sci. 2021;104:11135–46.

    Article  CAS  PubMed  Google Scholar 

  66. Edgar R, Domrachev M, Lash AE. Gene expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002;30:207–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Zhang S, Wang B, Wan L, Li LM. Estimating Phred scores of Illumina base calls by logistic regression and sparse modeling. BMC Bioinformatics. 2017;18:335.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Howe KL, Achuthan P, Allen J, Allen J, Alvarez-Jarreta J, Amode MR, et al. Ensembl 2021. Nucleic Acids Res. 2021;49:D884–91.

    Article  CAS  PubMed  Google Scholar 

  69. Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B. Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods. 2008;5:621–8.

    Article  CAS  PubMed  Google Scholar 

  70. van de Peer Y. Venn Diagrams.

Download references

Acknowledgements

This study was part of the Resilient Dairy Genome Project (RDGP). We gratefully acknowledge the funding and support from the Resilient Dairy Genome Project including Genome Canada (Canada), Genome Alberta (Canada), Ontario Ministry of Economic Development, Job Creation and Trade (Canada), Genome Quebec (Canada), Genome British Columbia (Canada), Lactanet (Canada), Dairy Farmers of Canada (Canada), Aarhus University (Denmark), The Council on Dairy Cattle Breeding (United States), Qualitas AG (Switzerland), USDA – Agricultural Research Service (United States), Allflex Livestock Intelligence (United States), Afimilk Ltd (Israel), Nedap NV (The Netherlands), Illumina (Canada), Ontario Genomics Institute (Canada), University of Guelph (Canada), University of Alberta (Canada), Université Laval (Canada), University of British Columbia (Canada), McGill University (Canada), Agriculture Research Department of Jobs, Precincts and Regions, La Trobe University (Australia), Instituto Nacional de Investigacion y Technologia Agraria y Alimentaria (INIA, Spain), Confederación de Asociaciones de Frisona Española (CONAFE, Spain), Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE, France), Vereinigte Informationssysteme Tierhaltung (VIT, Germany), University of Wisconsin-Madison (United States), University of Florida (United States), Purdue University (United States), University of Georgia (United States), Iowa State University (United States), Michigan State University (United States), University of Calgary (Canada), University of Prince Edward Island (Canada), Alberta Agriculture & Forestry (Canada), Agriculture and Agri-Food Canada (Canada), Freie Universität Berlin (Germany), São Paulo State University (Brazil), GenTORE Project (France), Semex (Canada), Foundation for Food and Agriculture Research (FFAR, US), Ministry of Agriculture Food & Rural Affairs (Canada), Ontario Ministry of Research and Innovation (Canada), ETH Zurich, Animal Nutrition Group (Switzerland), Agroscope Posieux (Switzerland), University of Kiel (Germany) As per the research agreement, researchers maintain independence in conducting their studies, own their data, and report the outcomes regardless of the results. The decision to publish the findings rests solely with the researchers. CFB acknowledges NSERC, NAK and UKS acknowledge NSERC and Semex, and CMR acknowledges Mitacs Elevate and Lactanet Canada for their support.

Funding

This research was part of the Genome Canada funded Resilient Dairy Genome Project, and was also funded by NSERC and Semex, and Mitacs Elevate and Lactanet Canada.

Author information

Authors and Affiliations

Authors

Contributions

CFB, FSS, and NAK designed the study. UKS performed laboratory work. AF, AS, CMR, MMMM, NAK performed data analyses. AF, CMR, CFB, FSS, and NAK interpreted results. AF and CMR wrote the manuscript, and all authors reviewed it.

Corresponding authors

Correspondence to Christina M. Rochus or Christine F. Baes.

Ethics declarations

Ethics approval

Ethical approval was not required for this study.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

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

Fong, A., Rochus, C.M., Shandilya, U.K. et al. The role of interleukin-10 receptor alpha (IL10Rα) in Mycobacterium avium subsp. paratuberculosis infection of a mammary epithelial cell line. BMC Genom Data 25, 58 (2024). https://doi.org/10.1186/s12863-024-01234-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12863-024-01234-w

Keywords