https://doi.org/10.22319/rmcp.v15i3.6441 

Review

Genomic regions, genes, and single nucleotide polymorphisms in resistance to gastrointestinal nematodes in sheep. Review

 

Marcela Villegas-Castañeda a*

Vielka Jeanethe Castañeda-Bustos b

Juan Manuel Bello-López c

Clemente Cruz-Cruz 3

 

a Hospital Juárez de México. Escuela de enfermería del Hospital Juárez de México. Plaza San Pablo. No. 13. Col. Centro. Alc. Cuauhtémoc, 06090, CDMX, México.

b Universidad Autónoma de Baja California. Instituto de Ciencias Agrícolas. Mexicali, México.

c Hospital Juárez de México. División de investigación, CDMX, México.

 

*Corresponding author: marcela.villegas.casta@gmail.com 

 

Abstract:

Several factors can modify productivity in sheep flocks; one of them is gastrointestinal (GI) parasitosis by nematodes, which can cause weight loss, growth retardation, and, in extreme situations, death. Parasite infections involve the immune system for resistance or susceptibility; therefore, strategies are currently being sought that will currently be efficient in the long term to reduce this affectation. One of these strategies is precision livestock breeding, which consists in the identification and selection of genetically resistant animals, using molecular markers. The objective of this review is to gather novel information on quantitative traits (IQT) and genome-wide association studies (GWAS), which confirm the relevance of certain regions or genes in resistance to ovine gastrointestinal parasitosis. Likewise, the potential relevance of new regions was analyzed to perform finer mappings and find sets of polymorphisms that may allow a more efficient selection, while also considering the particular conditions of the sheep herds.

Keywords: Polymorphisms, Resistance, Gastrointestinal parasitosis, Sheep.

 

Received: 06/04/2023

Accepted: 04/03/2024

 

Introduction

One of the factors that can modify sheep productivity is gastrointestinal (GI) parasitosis; among its adverse effects are weight loss(1) growth retardation and, in extreme situations, death(1,2), aspects that directly affect the economy of sheep producers. Strategies are continually being developed to lessen the effects of infection, either by testing new drugs or by searching for animals that are genetically more resistant in order to reproduce them. Some authors mention that these strategies tend to be more efficient, in the long run, when they are based on multiple approaches(3,4).

Genetic variation between or within breeds allows the detection and genetic selection of individuals with a greater capacity to resist the consequences of helminth infections. Selection of sheep or goats for improved parasite resistance is considered a valuable option to complement other control measures(5). The term disease resistance is commonly used generically to refer to resistance to infection or resistance to the consequences of disease, i.e. disease tolerance. Strictly speaking, however, disease resistance describes the host's ability to interact with and control the parasite's life cycle. Within the context of GI parasitosis, this may include the probability of establishment of ingested larvae, the rate and degree of parasite development within the host, mortality and fecundity of the parasites, and thus the fecal egg count. On the other hand, disease tolerance is used to describe the ability of the host to resist the pathogenic effects of infection(1)

Genetic improvement for resistance is possible due to the existence of a wide genetic variation in animals. When looking for genetic associations, traits of resistance or susceptibility (fecal egg count (FEC), parasite load, worm size and fecundity): immune response (major histocompatibility complex-MHC, concentration of antibodies such as IgA, IgG and IgM); impact of infection (anemia, presence of pepsinogen, or fructosamine concentrations), or resilience (growth rate and frequency of treatment required) are regularly studied(3,6). There are a number of important scientific contributions linking genes of the ovine major histocompatibility complex (Ovar-MHC) to the ability of sheep to resist infection by gastrointestinal parasites(7–16); however, the effect of the MHC is reportedly small and accounts for approximately 11 % of the total phenotypic variation(7). Class 1 genes are among the most polymorphic genes; this diversity, together with the lack of clarity of genomic organization, makes it difficult to identify new alleles of interest in sheep, which results in an evident scarcity of information(17). The products of Class I and II genes are glycoproteins that present antigenic peptides to the T-cell receptor (TCR) of cytotoxic CD8+ and helper CD4+ lymphocytes, respectively(18,19). Class II DRB genes have been more extensively studied(18,20) and have shown consistent associations with the phenotype of resistance to GI nematodes(7). Current approaches may neglect this important molecule because statistical analyses to detect association between MHC alleles and disease depend partly on haplotype frequencies(21), whereas the ability to discriminate with causal point mutations depends on the degree of linkage disequilibrium (LD), as, when LD is high, alleles at different loci are often inherited together by the offspring, and the effects of the various loci cannot be easily disentangled(22). Due to the high polymorphic variation in MHC, it is necessary to construct haplotype combinations to associate it with resistance/susceptibility traits(21,23), since most MHC genes are inherited in bloc as a haplotype with rare recombination events(24). Use of MHC as a generalized marker requires deepening of knowledge to the point of sequencing already associated haplotypes and knowing in which race and for which parasite they can be validly utilized; the association of haplotypes may then become stronger than the allelic association with single nucleotide polymorphisms (SNPs).

New strategies such as the use of arrays or chips with thousands of polymorphisms for simultaneous genotyping may predict the genetic merit of an individual through clusters of single nucleotide polymorphisms(25). Genotyping arrays have been developed by such companies as Illumina and Affymetrix, together with the Sheep Genomics Consortium, at different densities and with different coverage within the genome; currently the most commonly used in genome-wide association studies or determination of genetic merit are high and medium density, which can detect approximately 606,000 and 50,000 (50 K) evenly spaced SNPs. It has been observed that the estimated level of development for markers separated by less than 1Mb in arrays of up to 12 K can be a suitable tool to identify genomic regions associated with traits related to resistance to GI parasites(26). In addition to this, there are intergenic regions, called "genetic deserts"(27), which are regions with non-coding sequences but also with unannotated regulatory elements that offer a promising potential for future research.

While resistance or susceptibility to gastrointestinal parasitosis may be controlled by multiple loci with small effects, epistasis relationships could be evaluated as part of the resistance architecture. In addition, epistatic relationships allow the regulation of the expression of neighboring genes, which in turn make the expression of other genes possible. To date, no studies have identified major genes as the only genes involved in resistance in gastrointestinal parasitic infections. Therefore, the objective of this review is to present genomic information that confirms the relevance of certain regions or genes, and to give new relevance to others in GI parasitosis by nematodes in sheep.

 

Recent findings of genes or genomic regions involved in resistance/susceptibility to gastrointestinal nematodes

Nematodes can be located in different regions of the gastrointestinal tract; for example, in the abomasum the most frequent are Haemonchus contortus, Trichostrongylus axei, Mecistocirrus digitatus, and Telodorsagia circumcincta, while Trichostrongylus colubriformis, Cooperia spp., and Nematodirus spp. are prevalent in the small intestine, and Oesophagostumum spp., Chabertia ovina, and Trichuris ovis(28–30) predominate in the large intestine. In general, it can be said that the nematode usually found worldwide, especially in tropical and subtropical regions or climates, is H. contortus(31), whereas Telodorsagia circumcincta is one of the most prevalent in cold regions(32). It has been hypothesized that the inherent GI resistance to parasites is given by several genes (polygenic), and that it is related to the immune system(33–35).

Four main mechanisms can be identified that determine the host’s response to GI parasite infection: 1) the mechanism of the innate immune response, 2) protection of gastric mucous membranes, 3) hemostasis pathways, and 4) acquired immunity(36). On the other hand, among the mechanisms that allow the expulsion of parasites are the following: hypermotility, gastric hypersecretion and goblet cell hyperplasia with subsequent increased mucus production. In vitro and in vivo studies have shown that the immediate expulsion of parasites is associated with the presence of histamine and leukotrienes in the mucus of the abomasum that inhibit parasite motility(37,38). High concentrations of histamine in the abomasal mucosa of hemoncosis-resistant sheep may allow parasite expulsion, promoting abomasal hypersecretion that decreases worm fecundity and motility(39,40).

With respect to the host immune response, it has been observed that there is a clear difference in the immune response of lambs that have been challenged once or twice in their lives, and adult ewes that have been challenged several times during their productive lives with different larval or worm stages(31,37,40). Lambs demonstrate competent immunity by the age of 2 to 3 mo(41), and if larval exposure challenge is constant, immunity develops with a significant protective response by the age of 10 to 12 mo(42,43). In adult sheep this immunity tends to remain, rendering them relatively resistant to infection, and low-level exposures make them retain immunity(44). In some studies, protection against GI parasitosis has been associated with the Th2 helper immune response(45,46), characterized by the production of interleukin (IL) 4 which is an important cytokine in the immune control of parasitic GI diseases(47,48), essential in the maturation of virgin CD4+ T cells through the STAT6 pathway(49,50); it also promotes the differentiation of high synthesis rate B cells (changing the heavy chain from IgM to IgE and IgA)(51–53), as well as the recruitment of eosinophils, basophils and mast cells to control infection and participate in the expulsion of helminths(54–56). IL-13 acts in concert with IL-4 to stimulate IgE class switching, promote healing by tissue fibrosis, and enhance larval expulsion by increasing mucosal permeability, mucus production and muscle contraction(57–59); IL-5 too stimulates eosinophil maturation. The positive upregulation of these two cytokines after infection with T. columbriformis(58,60), coincides with the increase in IgE and IgA production(61). The development of the Th1 cellular response prevents the expression of proinflammatory cytokines such as interferon gamma (IFN-γ), and, therefore, the Th2 response(55,62). There is a link between IFN-γ and susceptibility, as it negatively regulates IL-4 and, consequently, the differentiation towards the Th2 response(63,64)

During parasite infection, the concentration of IgA is more important in the abomasum than in serum, and a negative correlation has been observed between the amount of specific IgA in abomasal mucus and the parasite load in infections by H. contortus(65). This situation has also been evident with the nematode T. circumcinta found in sheep abomasum, where high levels of specific IgA in the abomasal mucus have decreased the fertility and length of the nematode(66). A typical feature of helminth infections is the production of specific IgE as a result of a Th2-type response. IgE is able to induce antibody-dependent cytotoxicity of eosinophils, mast cells and macrophages. Increased local IgE levels have been associated with resistance to GI parasitosis in sheep and goats(67–69).

There are reports of resistance/susceptibility in such breeds as Churra, Red Maasai, Merino(70), Dorper x Red Maasai crosses(71), Scottish Blackface(72), St. Agnes(26), Soay feral sheep, Djallonké(73), Border Leicester x Merino, Poll Dorset x Suffolk or white Dorper crosses, Kathadin(74), Tunisian Native sheep(75), Corriedale, Pampinta(76), Sardas(77), and Florida Native sheep among others(78). In addition, other studies in hair breeds such as the Red Maasai(79), Florida, Santa Cruz, Barbados Black Belly, and Navajo(80) have shown that they are more resistant to nematode infection and its consequences than European breeds. However, there is variation between hair breeds, as shown in a study comparing Pelibuey versus Kathadin lambs; Pelibuey lambs showed greater resistance to natural GI nematode infection compared to Kathadin lambs, sharing the same climatic and grazing conditions, associated with the phenotype of egg counts per gram of feces and peripheral eosinophil counts(81).

Traditionally the association is made with such traits as fecal egg count, because it is a direct manifestation of the host's inability to control parasite reproduction(37,82). Another is the FAMACHA index, which is an indirect measure of the presence of parasites in the abomasum and the severity of the anemia they can cause to the host, and is related to the reduction of the agglomerated cell volume (measured in percentage), sequelae of infection with parasites such as H. contortus, which highlights the host's inability to replenish red blood cell levels and, in an extreme situation, could lead to death; but if the individual tolerates the acute infection and does not die, maintaining its zootechnical activity may be a resilience trait(76). Animals infected with H. contortus show more severe anemias(82,83). In initial studies, these traits have been associated with regions on chromosome 20 (OAR20; OARn = Ovis aries chromosome number "n"), which contain MHC II alleles, and OAR3 on the gamma interferon gene (IFN-ɣ) or genes close to this region(84–86). Another systematic review, mentions that there is sufficient evidence regarding the association of the IFN-ɣ gene and resistance to T. circumcincta, and it has been suggested that this region and its neighboring genes are of interest in host resistance(36,84–88)

Other studies in which no association has been found with the abovementioned regions, point to this difference as being attributable to the characteristics of the study subjects, as associations have been found only in lambs, not in adult sheep(87,89).

Other data provided for Red Maasai x Dorper sheep suggest that variation in SNP markers located in immune cell signaling genes such as suppressor of cytokine signaling (SOCS2), ubiquitin E2-conjugating enzyme (UBE2N) and protein tyrosine kinase substrate 15 (EPS15), could favor Th2 cytokine production to enhance the biological function of eosinophilia, mastocytosis and humoral response (high IgE levels) at the site of infection. Mucus production by the action of genes such as MUC15 or GALANT4 and hemostasis pathways (ATP2B1) may be important mechanisms contributing to the phenotype or in the differences in parasite resistance in the Red Maasai x Dorper population(72), and describes two regions that had not been associated until then: OAR2 (162-163Mpb) and OAR3 (44Mpb). The data also show that the OAR6_81718546 polymorphism (close to the platelet-derived growth factor receptor-α PDGFRA) is associated with effects on the aggregate cell volume, as has been previously reported in sheep of the Brazilian Morada Nova, Spanish Churra (34), and Soay Feral breeds(78) and in the Red Maasai x Dorper cross. Markers affecting fecal egg count (OAR5_111342555, OAR15_35337227, OAR5_100699982.1, DU183841_402.1, OAR15_40719719.1, OAR15_40926306.1, OAR7_4206430, and OAR17_42673146) do not affect agglomerated cell volume or live weight according to this study(72)

On the other hand, twelve SNP's listed in Table 1 were analyzed in the Soay Feral breed, of which, RORC2 p.A404T (100,653,186 bp) is associated with IgA; in addition it was concluded that the IL23R p.V32M polymorphism (42,512,431 bp) is related to the IL-23 receptor, an inflammatory cytokine that exhibited association with body weight at 20 days in blackface lambs(78).

In 2016, associated genes were found in Spanish Churra breed for the fecal egg count trait in OAR6 (with peak at 88. 1 cM) as AFP, ALB, AMBN, AMTN, AREG, BTC, CXCL1, CXCL10, CXCL11, CXCL9, EREG, GC, IGJ, IL8, MUC7, PF4, PPBP, RASSF6, SCARB2, TMPRSS11D; in OAR8 (peak at 2 cM) for the same trait, as CD109, COL12A1, MYO6, and in OAR 22 (peak at 3.4 cM), for the IgA trait of gene PCDH15. Among the nematode species found most frequently in this study were Trichostrongylus spp. and Teladorsagia spp. In addition, other genes encoding for chemokines were found, including IL-8, CXCL1, CXCL10, CXCL11, CXCL9, PF4, PPBP ⸺molecules that are of great importance in the immune system, since they are involved from leukocyte recruitment to cell communication and activation during infection; particularly IL-8, CXCL8, and CXCL1 are involved in the recruitment and activation of neutrophils. Notably, this author found no clear correspondence with previously described classical regions related to IFN-ɣ or those involving MHC class II genes(34).

On the other hand, when evaluating Santa Inés sheep using a SNP chip with 12 785 single nucleotide polymorphic markers, an association was found between regions in OAR1, OAR2, OAR3, OAR5, OAR8, and OAR15(78) (Table 1). Several candidate chromosomal regions described by the authors are related to the development of the immune system, its activation, inflammatory response, lymphocyte regulation, and leukocyte proliferation (B2M, SFXN1, IL25, BMP4, TSHR, CCL28, PIK3R1, FGF10, IL15, TP-1, BPMG, BCL10, HSPD1, MALT1), highlighting genes such as CD109, which is a surface antigen expressed by CD34 or IL-25 cells; coincidentally some of these were reported by another study(34) as potential genes in resistance to GI nematodes in sheep.

As of 2018, GWAS studies with high or medium density chips have elucidated more candidate genes that could be relevant in resistance/susceptibility to nematodes and other parasites. This is indicated by the findings of several authors(75,77,90), one of them in Djallonké lambs from West Africa, where five genes (TRIB3, CDK4, CSNK2A1, MARK1, and SPATA5) are associated with resistance traits related to immunity and cell proliferation. It is also suggested that the MBL2 gene (as the basis of a QTL) in OAR22 is related to IgA levels(27,82). Also, it has been hypothesized that genes involved in lamb growth and size (such as the ADAMTS17 gene in OAR18) may be pleiotropic with certain genes that determine resistance traits to GI parasite infection; however, the association between these genes has not yet been clearly determined(34,73,91).

OAR2 is also highlighted in a study in Australian sheep of breeds such as Merino or Border Leicester x Merino, Poll Dorset/Suffolk/Suffolk/White Suffolk/White Dorper/Border Leicester crosses, where, in a first analysis, the authors provide an outline of three SNP's in OAR2 with a strong association to the trait of fecal egg count (rs421630816, rs424521894, rs413835864): the SNP rs421630816 (position in OAR2: 110.8 Mbp) in the PALLD gene, while rs424521894 and rs413835864 (position in OAR2:107.3 and 107.4 Mbp, respectively) in the GALNTL6 gene related to the synthesis of mucin-like glycans, which influence host-pathogen interaction. Likewise, these authors point out a region in OAR6 that includes six SNP's, where rs416517011 stands out for its level of significance in the association; they also found other associated genes in OAR18 and OAR24, hypothesizing that these genes share certain mechanisms with the immune system, suggesting potential interaction effects between genes(70). Another contribution found significant associations in OAR2, 3, 16, 23, and 24 in Kathadin sheep(74). A relevant finding is a locus located in OAR3, close to the C3 complement pathway receptor 1 gene (C3AR1). C3AR1 has been reported to be differentially expressed in susceptible versus resistant sheep(92) and has been associated with the Th1 response(93), also located in OAR16, 87 kb towards 5' of the ITGA2 gene (α-2 integrin) that mediates adhesion of platelets and other cell types to the extracellular matrix. One region that stands out in OAR2 ⸺the DIS3L2 gene (rs406850490 and rs422243920), an exoribonuclease involved in regulating the relative expression of Toll receptor type 4⸺ was significantly associated and suggests a potential role in mediating resistance. The DIS3L2-associated SNP had a minor allele frequency (MAF) overrepresented in resistant sheep (0.479) compared to susceptible sheep (0.094); this exoribonuclease may affect IL-10 expression by repression of let-7, a miRNA. Other findings of importance in the study refer to OAR3 ALK-tyrosine kinase receptor (rs437558829 and rs407346502) and C3AR1, OAR19 (rs406978752) GRM7-(metabotropic glutamate receptor 7), OAR23 (rs399876637) SLC14A2 (urea transporter 2) and OAR24 ZP3 glycoprotein (rs423186265); however, it has been suggest that these findings need to be validated(74).

In order to give prominence to the effects of the immune system on the response to parasitosis, another group studied indigenous Tunisian sheep under traditional grazing management and they highlighted RUFy4 and VIL1, two IL-8 receptors (CXCR1 and CXCR2) as candidate genes involved in the immune response in the GI tract, hypothesizing that they may be involved in repairing damaged tissue in the intestine and enhancing neutrophil recruitment and inflammation. They also found two cation transporter genes such as SLC22A4 (OCTN1) and SLC22A5 (OCTN2) involved in the transport of oxourea. The authors stress that the traditional management of these sheep allows them to develop multiple adaptive strategies that make them resistant to parasitosis, and the information gathered from this type of native livestock is very valuable in understanding the architecture of resistance(75). In Mexico there are herds with Creole characteristics and extensive management; therefore, it would be interesting to determine if the adaptive strategies of the immune system coincide with those of other herds, or other breeds, managed under similar conditions, and thus be able to identify coinciding mechanisms for use as markers for resistance/susceptibility to nematodes or other parasitosis.

Notably, a fine mapping carried out by Argentine researchers in Pampinta and Corriedale lambs under natural challenge found that certain regions that had been previously associated(36), in OAR3 and OAR6, and OAR20, contain genes involved in MHC-mediated antigen processing and lymphocyte signaling pathways. The OLA-DRA1_479 SNP was the only SNP that showed a significant association for the traits under study in Corriedale lambs; it also associated polymorphism of C-type lectin receptors that mediate functions such as cell signaling transduction processes, pathogen recognition, and innate immunity, although CLEC12A acts by inhibiting the production of IL-12, TLR4-dependent TLR4 and IL-12(94). It also marked three significant de novo SNP ́s, FOS_109, IL20RA_422 and TIMP3_716 ⸺the first, located in FBJ murine viral osteosarcoma homologous gene; the next, in IL-20 receptor gene, and the last, located in TIMP a metalloproteinase inhibitor in OAR 3, 7 and 8, respectively⸺; FOS_109 belongs to a group of proteins that regulate cell proliferation, differentiation and transformation. The duplicated expression of this gene in abomasal tissue was found to be associated with resistance in Merino sheep and it is hypothesized to be a relevant gene in primary infections by H. contortus. In some cases, FOS gene expression has also been associated with cell death by apoptosis. TIMP3_716 showed evidence that suggests association when using fecal egg count as a breeding value estimated as an association phenotype, and may be involved in the remodeling of damaged tissue in response to parasitic infections. The results obtained confirm genomic regions previously reported to be associated with nematode resistance in other sheep breeds, both for innate immunity (MASP, CLR, NLR, TLR, IL20R, FOS, TIMP) and adaptive immunity (CLR, IL2, OLA-DRA, TIMP) reinforcing the role of the host immune response against parasites(76).

In Sarda ewes and crosses of this line with Lacune, 10 regions with significant association to the trait of fecal egg count were mapped, pointing to 3,538 polymorphisms causing high-impact effects that can generate termination codons (nonsense mutations) in genes coding for 530 proteins. The authors of this study hypothesize that QTLs located in OAR 1, 12, 19, and 20 are strongly implicated in a complex mechanism of resistance in sheep to GI parasitosis; some of the polymorphisms they report can be seen in Table 1(77). In OAR12, the missense mutation c.103G>A in exon 2, position 39, 567,687 bp, in the TNFRSF1B gene (member of the TNF1B receptor superfamily), which is also close to the SELE gene (selectin E gene, four relevant nonsense mutations), encodes a protein in endothelial cells and is responsible for the accumulation of leukocytes at sites of inflammation mediated by vascular lining cells. Another authors(95) mentions that the SELE gene is negatively expressed in abomasal lymph nodes of lambs recently infected with T. circumcincta, suggesting it as a component of the resistance response to infection in GI parasitosis. In OAR19 the most significant association was in the MGR gene (metabotropic glutamate receptor, associated with nervous mechanisms in humans), in addition to 13 nonsense variants in the IL5RA gene (α-subunit of rIL-5). This protein has been found to be expressed in animals resistant to T. circumcincta (Scottish blackface lambs, churras ewes, and merino lambs)(95–97). In the OAR20 region, a large region encompassing MHC class II was found, although these are reportedly located at a distance of 4 to 6 Mb from the most significant location, highlighting that, due to the polymorphic nature of the gene, it is difficult to identify causal mutations or SNP's that are useful in resistance selection(98). Also reported were mutations in IL17A, IL17F, TRIM26, TRIM38, TNFRSF21, LOC1011118999, VEGFA and TNF. A significant SNP (rs404860664) was reported in the LOC101111058 gene (butyrophilin-like protein); however, butyrophilin-like proteins suggest that it plays a role in the regulation of local intestinal inflammation in other species(99), with nine mutations in TRIM 26; these proteins play roles in the regulation of pathogenesis in autoimmune diseases and pathogen defense in particular against viruses(100), and they also may be involved in the down-regulation of several immune response genes(77).

In a first study detecting repeat variants by GWAS in native sheep in Florida, 8124 copy number variations (CNV) were identified, although only 14 were significantly associated with the traits under study, such as fecal egg count and aggregate cell volume. The genes that stand out in this study in relation to the immune response are CCL1, CCL2, CCL8, CCL11, NOS2, TNF, CSF3, and STAT34, which may play an important role in the resistance to H. contortus. These genes could be used as potential markers of resistance in this breed; it is also possible that genes close to repeat regions such as LOC101110424, DOCK9, ITGBL1, BIVM, TNFSF13B, ING1, F7, F10, PCID2, and GAS6 may have important effects on the immune response against the parasite(90). For example, ITGBL1 gene expression is associated with immune cell infiltration(101), while genes F7 and F10 play a relevant role in the initiation of coagulation and defense against pathogens(102). The CCL1 gene is part of an eotaxin chemokine and promotes the migration of activated eosinophils(103), eosinophilia is a common event in sheep infected with H. contortus(104), and this gene is commonly used as a marker of resistance(92,105). In addition, three galectin genes (LOC101117947, LOC101118202 and LOC101102156) near a repeat region were associated with the egg count trait at day 28. Galectins are proteins involved in the immune response to parasitic infections of the gastrointestinal tract in sheep and are upregulated during infection with H. contortus(106). Some of these galectins, like No. 11, can regulate larval growth and development by binding to H. contortus larvae No. 4 and adults(107). The associated repeats for the cell package at day 0 and 28 (LOC101108321) are contained in genes related to multidrug resistance proteins (MRP), expressed at the same level in CD3+/CD4+ T cells according to a study performed in the peripheral blood of normal and refractory lymphoma patients(108); they can also regulate the inflammation of intestinal mucous epithelia(109). There is a possibility that all the repeated sequences found in this study may be segregated among the population, but as in other studies, they must be validated in other populations. These findings may contribute to the development of new strategies to improve parasite resistance in sheep and promote selective breeding through marker-assisted selection(90).



 

Conclusions

Parasitosis and parasitic resistance are an issue that affects sheep production systems, especially grazing sheep. Knowledge about the architecture of resistance/susceptibility in sheep contributes to genetic improvement at a faster rate, resulting in higher productivity in flocks that contribute to precision livestock breeding. Although effective drug treatments to fight parasitosis can be found in parallel, when new formulations become available, they can potentially be more expensive. There is also a growing interest in reducing the use of anthelmintics to contribute to the environment by reducing their excretion into the environment. The information from QTLs has been refined by GWAS analysis with high-density chips, creating the need for further fine mapping of candidate genes, so that the information may be utilized to screen sheep for resistance to GI parasites or to elucidate epistatic relationships between immune response genes in order to generate areas of research for functional or expression studies, providing greater clarity on the function of the immune system. Dissection of the architecture of resistance and susceptibility to gastrointestinal parasitosis, as well as the validation of associated loci in different herds, create the challenge of generating a marker test with the best possible combination of SNPs to allow characterizing individuals resistant to GI parasitosis among certain populations as a strategy to address parasitic resistance and implement more effective and direct selection programs. 




 

Table 1: Findings of genomic regions, SNP's and genes linked to association variables in resistance to gastrointestinal parasitosis in sheep

Author

Parasite

Association variables or traits

QTL

SNP

Genes

Benavides, 2015

Haemonchus contortus

AVCA*, LW**

OAR2 (15 Mbp), OAR11 (58 Mbp) OAR15 (54 Mbp).

New OAR2 (162-163Mpb) and OAR3 (44Mpb).  

OAR6_81718546, OAR5_111342555, OAR15_35337227, OAR5_100699982.1

DU183841_402.1, y OAR15_40719719.1 OAR15_40926306.1 

OAR7_4206430 y OAR17_42673146,

SOCS2, UBE2N y EPS15

ATP2B1 y LRP8

MUC15 y GALNT4

Atlija, 2016

Trichostrongylus spp y Teladorsagia spp

 

OAR6 (peak in 88.1 cM), OAR8 (peak in 2cM) y OAR22 (peak in o 3.4 cM)

 

AFP, ALB, AMBN, AMTN, AREG, BTC, CXCL1, CXCL10, CXCL11, CXCL9, EREG, GC, IGJ, IL8, MUC7, PF4, PPBP, RASSF6, SCARB2, TMPRSS11D, CD109, COL12A1, MYO6 PCDH15, IL8, CXCL1, CXCL10, CXCL11, CXCL9, PF4, PPBP, CxCL8 y CXCL1

Berton, 2017

Haemonchus contortus

FEC***, FAMACHA index, AVCA

OAR 2:91681809-9470993

2:140765269-143337545

OAR 3:195904655-195904655

OAR 1:56799547-56799547

 

OAR 16 :41876371-41876371

OAR 18:68738392-68738392


 
 

LPAR1; TXN; ALDOB; PLPPR1; CTSV; PTCH1; AGTPBP1; AQP3; ADRA1A; LOXL2; SFTPC; HR; LPL; LOC101123612; TGFBR1; GNA14; PCSK5; RORB; ALDH1A1; TYRP1; FREM1; PSIP1; CCDC171; BNC2; CNTLN; ADAMTSL1; RPS6; TP-1 XIRP2; LOC101109253; SCN7A; SCN9A; SCN1A; TTC21B; GALNT3; CSRNP3; LOC101110039; SCN2A; SCN3A, DHX57; GEMIN6; RSF7; GALM; HNRNPLL; LOC101119897; LOC101120157; ATL2; LOC101120655; LOC101120913; LOC101119706; RMDN2; CDC42EP3; TRNAC-GCA; TRNAS-GGA; QPCT; PRKD3; NDUFAF7; CEBPZ; SULT6B1; EIF2AK2; GPATCH11; HEATR5B; STRN; VIT; FEZ2; LOC101122183; LOC101122430; LOC101122685;  LOC101123283

GALNT2; TRNAE-UUC; PGBD5; LOC101103868; LOC101104120; LOC101104369; LOC101104630; LOC101104883; LOC101105131; LOC101105384; LOC101105628; LOC101105878; LOC101106137; LOC101106392; LOC101106652; LOC101106903; LOC101107159; LOC101107409; LOC101107663; LOC101107927; LOC101108188; LOC101108450; LOC101108625; LOC101108717; LOC101108881; LOC101109143; LOC101108983; LOC101109240; LOC101109508; LOC101109767

PDZD2; LOC101119673; C16H5orf22; DROSHA; CDH6

INF2; ADSSL1; SIVA1; AKT1; TMEM179; PLD4; LOC101104938; C18H14orf79; LOC101105444; GPR132; LOC101105953; BTBD6; BRF1; LOC101106466; LOC101106718; C18H14orf80; TMEM121; LOC101107475; LOC101107738; LOC101107998; LOC101108260; LOC101108522; LOC101108781

Wilkie, 2017

****NE

FEC, IgA, LW

 

RORC2 c*25T>C and RORC2 c.*109ª>g

E294Q y A404T) IL23R p.V324M y RORC2 p. A404T

TBX21, RORC2 e IL23R

Álvarez, 2019

NE

AVCA, FEClog, AVCA, FAMACHA

 

OAR1_55820164.1

OAR2_117867801.1

OAR8_16568165.1

OAR15_88875909.1

OAR18_43101149.1

OAR2_140684314.1

S16493.1 (OAR16)

S43307.1 (OAR7)

OAR8_8982479.1

OAR15_2525103.1

OAR17_3451123_X.1

S43852.1 (OAR19)

OAR2_64824262.1

OAR3_77774489.1

OAR3_161498140.1

OAR12_22189408.1

S32476.1

S09612.1 (OAR13)

OAR18_5508052_X.1

OAR22_6293170.1

OARX_107840506.1

TMOD1; TDRD7, MFSD6, INPPI, HIBCH, C2H2orf88, SV2C, IQGAP2, NUDT6

TRIB3, CDK4, CSNK2A1, MARK1 y SPATA5, MBL2, ATP6V1E2, TMEM247, EPAS1, ATP23, CTDSP2, AVIL, TSFM, METTL21B, METTL1, LOC101116039, MARCH9, CDK4, TSPAN31, MARK1

Kaladeh, 2019

H. contotus,       T. colubriformis, T. circumcincta

FEC

 

rs421630816, rs424521894, rs413835864,

rs421630816, rs424521894 y rs413835864, rs413835864, rs424521894 y rs421630816, rs416517011

PALLD, GALNTL6

Becker, 2020

Haemonchus contorutus

Estimated genetic values and FEC, and FAMACHA index

 

rs406850490 y rs422243920, rs437558829 y rs407346502, (rs406978752, rs399876637, rs423186265

C3AR1, DIS3L2

Ahbara, 2021

NS

 

QTL FECGEN

 

SLC22A4, SLC22A5, IL-4, IL-13, IL-4, VIL1, CXCR1, CXCR2, IL-4, IL-13, FECGEN, TFEC_1, HFEC, NFEC, LATRICH_2, IGA, OSAS, WORMCT, PEPSL y CEOSIN QTL, RUFy4 y VIL1, ITLN

* AVCA= average volume of the cell agglomerate; ** LW= live weight; *** FEC= fecal egg count; ****NS= not specified.



 

Acknowledgments and conflicts of interest

The authors would like to thank Dr. Mónica A. Cureño Díaz, MSc, Director of the Research and Teaching Division of the Hospital Juárez de México, for her kind words; Dr. Verónica Fernández, PhD, Head of the Research Division, and Tolina Alcántara, MSc, of the School of Nursing of HJM, for the facilities provided for this review.

The authors declare that they have no conflict of interest.

 

Literature cited:

  1. Bishop SC, Stear MJ. Modeling of host genetics and resistance to infectious diseases: understanding and controlling nematode infections. Vet Parasitol 2003;115(2):147–166.
  2. Jennings FW. The anaemias of parasitic infections. In: Soulsby EJL ed. Pathophysiology of parasitic infection. 1rst ed. Acaddemic Press, 1976:41-67 https://linkinghub.elsevier.com/retrieve/pii/B9780126553659500095. Accessed Sep 15, 2022..
  3. Bishop SC. Possibilities to breed for resistance to nematode parasite infections in small ruminants in tropical production systems. Animal 2012;6(5):741–747.
  4. Jackson F, Bartley D, Bartley Y, Kenyon F. Worm control in sheep in the future. Small Ruminant Res 2009;86(1–3):40–45.
  5. Bishop SC. Genetic resistance to infections in sheep. Vet Microbiol 2015;181(1–2):2–7. 
  6. Bishop SC, Morris CA. Genetics of disease resistance in sheep and goats. Small Ruminant Res 2007;70(1):48–59.
  7. Buitkamp J, Filmether P, Stear MJ, Epplen JT. Class I and class II major histocompatibility complex alleles are associated with faecal egg counts following natural, predominantly Ostertagia circumcincta infection. Parasitol Res 1996;82(8):693–696.
  8. Stear M. An ovine lymphocyte antigen is associated with reduced faecal egg counts in four-month-old lambs following natural, predominantly Ostertagia circumcincta infection. Int J Parasitol 1996;26(4):423–428.
  9. Charon KM, Moskwa B, Kury J, Gruszczynska J, Rutkowski R. Relationship between polymorphism in locus OMHC1 (MHC class I) and resistance to nematodes in Polish Heatherhead Sheep. Anim Sci Pap Rep 2001;19(4):285–292.
  10. Behnke JM, Iraqi F, Menge D, Baker RL, Gibson J, Wakelin D. Chasing the genes that control resistance to gastrointestinal nematodes. J Helminthol 2003;77(2):99–109.
  11. Stear MJ, Bishop SC, Henderson NG, Scott I. A key mechanism of pathogenesis in sheep infected with the nematode Teladorsagia circumcincta. Anim Health Res Rev 2003;4(1):45–52.
  12. Sayers G, Good B, Hanrahan JP, Ryan M, Angles JM, Sweeney T. Major Histocompatibility Complex DRB1 gene: its role in nematode resistance in Suffolk and Texel sheep breeds. Parasitology 2005;131(3):403–409.
  13. Gao J, Liu K, Liu H, Blair HT, Li G, Chen C, et al. A complete DNA sequence map of the ovine Major Histocompatibility Complex. BMC Genomics 2010;11(1):466.
  14. Hassan M, Good B, Hanrahan JP, Campion D, Sayers G, Mulcahy G, et al. The dynamic influence of the DRB1*1101 allele on the resistance of sheep to experimental Teladorsagia circumcincta infection. Vet Res 2011;42(1):46.
  15. Hickford JGH, Forrest RHJ, Zhou H, Fang Q, Frampton CM. Association between ariation in faecal egg count for a mixed field-challenge of nematode parasites and ovine MHC-DQA2 polymorphism. Vet Immunol Immunopathol 2011;144(3–4):312–320.
  16. Lee CY, Munyard KA, Gregg K, Wetherall JD, Stear MJ, Groth DM. The influence of MHC and immunoglobulins A and E on host resistance to gastrointestinal nematodes in Sheep J Parasitol Res 2011;2011:1–11.
  17. Buitkamp J. Uncovering novel MHC alleles from RNA-Seq data: expanding the spectrum of MHC class I alleles in sheep. BMC Genomic Data 2023;24(1):1.
  18. Dukkipati V, Blair H, Garrick D, Murray A. ‘Ovar-Mhc-Ovine major histocompatibility complex: Role in genetic resistance to diseases. N Z Vet J 2006;54(4):153–160.
  19. Rammensee HG, Friede T, Stevanović S. MHC ligands and peptide motifs: first listing. Immunogenetics 1995;41(4):178–228.
  20. Stear MJ, Belch A, Donskow-Schmelter K, Fitton LA, Innocent GT, Ishikane C, et al. Detection of genes with moderate effects on disease resistance using ovine mhc and resistance to nematodes as an example. Vet Immunol Immunopathol 2007;120(1–2):3–9.
  21. Stear MJ, Fitton L, Innocent GT, Murphy L, Rennie K, Matthews L. The dynamic influence of genetic variation on the susceptibility of sheep to gastrointestinal nematode infection. J R Soc Interface 2007;4(16):767–776.
  22. Ali AOA, Stear A, Fairlie-Clarke K, Brujeni GN, Isa NMM, Salisi MSB, et al. The genetic architecture of the MHC class II region in British Texel sheep. Immunogenetics 2017;69(3):157–163.
  23. Ali AOA, Stear A, Fairlie-Clarke K, Brujeni GN, Isa NMM, Salisi MSB, et al. The genetic architecture of the MHC class II region in British Texel sheep. Immunogenetics 2017;69(3):157–163.
  24. Begovich AB, McClure GR, Suraj VC, Helmuth RC, Fildes N, Bugawan TL, et al. Polymorphism, recombination, and linkage disequilibrium within the HLA class II region. J Immunol Baltim Md 1950. 1992;148(1):249–258.
  25. Meuwissen THE, Hayes BJ, Goddard ME. Prediction of total genetic value using genome-wide dense marker maps. Genetics 2001;157(4):1819–1829.
  26. Berton MP, de Oliveira Silva RM, Peripolli E, Stafuzza NB, Martin JF, Álvarez MS, et al. Genomic regions and pathways associated with gastrointestinal parasites resistance in Santa Inês breed adapted to tropical climate. J Anim Sci Biotechnol 2017;8(1):73.
  27. Bahbahani H, Salim B, Almathen F, Al Enezi F, Mwacharo JM, Hanotte O. Signatures of positive selection in African Butana and Kenana dairy zebu cattle. Tesfaye D, editor. PLOS One 2018;13(1):e0190446.
  28. Meana MA, Rojo VFA. Tricostrongiliosis y otras nematodosis. Parasitologia veterinaria.  Cordero CM, Rojo VFA et al. editores México: Mc Graw Hill Interamericana; 1999.
  29. Quiroz-Romero H. Parasitología y enfermedades parasitarias de animales domésticos. 1a ed. México, DF: Limusa; 2003.
  30. Soulsby EJL. Parasitología y enfermedades parasitarias en los animales domésticos. 7a México: Editorial Interamericana; 1988.
  31. Miller JE, Horohov DW. Immunological aspects of nematode parasite control in sheep. J Anim Sci 2006;84(suppl 13): E124–132.
  32. Venturina VM, Gossner AG, Hopkins J. The immunology and genetics of resistance of sheep to Teladorsagia circumcincta. Vet Res Commun 2013;37(2):171–181.
  33. Aguerre S, Jacquiet P, Brodier H, Bournazel JP, Grisez C, Prévot F, et al. Resistance to gastrointestinal nematodes in dairy sheep: Genetic variability and relevance of artificial infection of nucleus rams to select for resistant ewes on farms. Vet Parasitol 2018;256:16–23.
  34. Atlija M, Arranz JJ, Martínez-Valladares M, Gutiérrez-Gil B. Detection and replication of QTL underlying resistance to gastrointestinal nematodes in adult sheep using the ovine 50K SNP array. Genet Sel Evol 2016;48(1):4.
  35. Saddiqi HA, Jabbar A, Sarwar M, Iqbal Z, Muhammad G, Nisa M, et al. Small ruminant resistance against gastrointestinal nematodes: a case of Haemonchus contortus. Parasitol Res 2011;109(6):1483–1500.
  36. Benavides MV, Sonstegard TS, Van Tassell C. Genomic regions associated with sheep resistance to gastrointestinal nematodes. Trends Parasitol 2016;32(6):470–480.
  37. Alba-Hurtado F, Muñoz-Guzmán MA. Immune responses associated with resistance to Haemonchosis in sheep. BioMed Res Int 2013;2013:1–11.
  38. Karrow NA, Goliboski K, Stonos N, Schenkel F, Peregrine A. Review: Genetics of helminth resistance in sheep. Can J Anim Sci 2014;94(1):1–9.
  39. Balic A, Bowles VM, Meeusen ENT. Mechanisms of immunity to Haemonchus contortus infection in sheep. Parasite Immunol 2002;24(1):39–46.
  40. Miller HRP. Prospects for the immunological control of ruminant gastrointestinal nematodes: Natural immunity, can it be harnessed? Int J Parasitol 1996;26(8–9):801–811.
  41. Bishop SC, Bairden K, McKellar QA, Park M, Stear MJ. Genetic parameters for faecal egg count following mixed, natural, predominantly Ostertagia circumcincta infection and relationships with live weight in young lambs. Anim Sci 1996;63(3):423–428.
  42. Brunsdon RV. Seasonal changes in the level and composition of nematode worm burdens in young sheep. N Z J Agric Res 1970;13(1):126–148.
  43. Seaton DS, Jackson F, Smith WD, Angus KW. Development of immunity to incoming radiolabelled larvae in lambs continuously infected with Ostertagia circumcincta. Res Vet Sci 1989;46(2):241–246.
  44. McKenna PB. The diagnostic value and interpretation of faecal egg counts in sheep. N Z Vet J 1981;29(8):129–132.
  45. Chen F, Liu Z, Wu W, Rozo C, Bowdridge S, Millman A, et al. An essential role for TH2-type responses in limiting acute tissue damage during experimental helminth infection. Nat Med 2012;18(2):260–266.
  46. Moncada DM, Kammanadiminti SJ, Chadee K. Mucin and Toll-like receptors in host defense against intestinal parasites. Trends Parasitol 2003;19(7):305–311.
  47. Finkelman FD, Shea-Donohue T, Morris SC, Gildea L, Strait R, Madden KB, et al. Interleukin-4- and interleukin-13-mediated host protection against intestinal nematode parasites. Immunol Rev 2004;201(1):139–155.
  48. Reynolds LA, Filbey KJ, Maizels RM. Immunity to the model intestinal helminth parasite Heligmosomoides polygyrus. Semin Immunopathol 2012;34(6):829–846.
  49. Zheng WP, Flavell RA. Pillars Article: the transcription factor gata-3 is necessary and sufficient for Th2 cytokine gene expression in CD4 T cells. J Immunol Baltim Md 1950. 2016;196(11):4426–4435.
  50. Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell populations. Annu Rev Immunol 2011;28(1):445–489.
  51. Ansel KM, Djuretic I, Tanasa B, Rao A. Regulation of TH2 differentiation and Il4 locus accessibility. Annu Rev Immunol 2006;24(1):607–756.
  52. Finkelman FD, Holmes J, Katona IM, Urban JF, Beckmann MP, Park LS, et al. Lymphokine control of in vivo immunoglobulin isotype selection. Annu Rev Immunol 1990;8(1):303–333.
  53. Nelms K, Keegan AD, Zamorano J, Ryan JJ, Paul WE. The IL-4 Receptor: Signaling mechanisms and biologic functions. Annu Rev Immunol 1999;17(1):701–738.
  54. McRae KM, Stear MJ, Good B, Keane OM. The host immune response to gastrointestinal nematode infection in sheep. Parasite Immunol 2015;37(12):605–613.
  55. Begley CG, Nicola NA. Resolving conflicting signals: cross inhibition of cytokine signaling pathways. Blood 1999;93(5):1443–1447.
  56. Hussaarts L, Yazdanbakhsh M, Guigas B. Priming dendritic cells for th2 polarization: lessons learned from helminths and implications for metabolic disorders. Front Immunol http://journal.frontiersin.org/article/10.3389/fimmu.2014.00499/abstract. Accessed Aug 11, 2022.
  57. Madden KB, Whitman L, Sullivan C, Gause WC, Urban JF, Katona IM, et al. Role of STAT6 and Mast Cells in IL-4- and IL-13-induced alterations in Murine intestinal epithelial cell function. J Immunol 2002;169(8):4417–4422.
  58. Meeusen ENT, Balic A, Bowles V. Cells, cytokines and other molecules associated with rejection of gastrointestinal nematode parasites. Vet Immunol Immunopathol 2005;108(1–2):121–125.
  59. Wynn TA. IL-13 Effector functions. Annu Rev Immunol 2003;21(1):425–456.
  60. Lacroux C, Nguyen THC, Andreoletti O, Prevot F, Grisez C, Bergeaud JP, et al. Haemonchus contortus (Nematoda: Trichostrongylidae) infection in lambs elicits an unequivocal Th2 immune response. Vet Res 2006;37(4):607–622.
  61. Kooyman, Schallig, Van Leeuwen, Mackellar, Huntley, Cornelissen, et al. Protection in lambs vaccinated with Haemonchus contortus antigens is age related, and correlates with IgE rather than IgG1 antibody: Serum IgE in vaccinated sheep. Parasite Immunol 2000;22(1):13–20.
  62. Couper KN, Blount DG, Riley EM. IL-10: The master regulator of immunity to infection. J Immunol 2008;180(9):5771–5777.
  63. Bancroft AJ, Grencis RK. Th1 and Th2 cells and immunity to intestinal helminths. In: MacDonald TT, editor. Chemical immunology and allergy. Basel: KARGER; 1998.: https://www.karger.com/Article/FullText/58711. Accessed Aug 23, 2023
  64. Pulendran B. Modulating Th1/Th2 Responses with microbes, dendritic cells, and pathogen recognition receptors. Immunol Res 2004;29(1–3):187–196.
  65. Amarante AFT, Bricarello PA, Huntley JF, Mazzolin LP, Gomes JC. Relationship of abomasal histology and parasite-specific immunoglobulin A with the resistance to Haemonchus contortus infection in three breeds of sheep. Vet Parasitol 2005;128(1–2):99–107.
  66. Martínez-Valladares M, Vara-Del Rio MP, Cruz-Rojo MA, Rojo-Vazquez FA. Genetic resistance to Teladorsagia circumcincta: IgA and parameters at slaughter in Churra sheep. Parasite Immunol 2005;27(6):213–218.
  67. De la Chevrotière C, Bambou JC, Arquet R, Jacquiet P, Mandonnet N. Genetic analysis of the potential role of IgA and IgE responses against Haemonchus contortus in parasite resistance of Creole goats. Vet Parasitol 2012;186(3–4):337–343.
  68. Pernthaner A, Shaw RJ, McNeill MM, Morrison L, Hein WR. Total and nematode-specific IgE responses in intestinal lymph of genetically resistant and susceptible sheep during infection with Trichostrongylus colubriformis. Vet Immunol Immunopathol 2005;104(1–2):69–80.
  69. Pernthaner A, Cole SA, Morrison L, Green R, Shaw RJ, Hein WR. Cytokine and antibody subclass responses in the intestinal lymph of sheep during repeated experimental infections with the nematode parasite Trichostrongylus colubriformis. Vet Immunol Immunopathol 2006;114(1–2):135–148.
  70. Al Kalaldeh M, Gibson J, Lee SH, Gondro C, van der Werf JHJ. Detection of genomic regions underlying resistance to gastrointestinal parasites in Australian sheep. Genet Sel Evol 2019;51(1):37.
  71. Marshall K, Mugambi JM, Nagda S, Sonstegard TS, Van Tassell CP, Baker RL, et al. Quantitative trait loci for resistance to Haemonchus contortus artificial challenge in Red Maasai and Dorper sheep of East Africa. Anim Genet 2013;44(3):285–295.
  72. Benavides MV, Sonstegard TS, Kemp S, Mugambi JM, Gibson JP, Baker RL, et al. Identification of novel loci associated with gastrointestinal parasite resistance in a Red Maasai x Dorper Backcross population. PLoS ONE 2015;10(4):e0122797.
  73. Álvarez I, Fernández I, Soudré A, Traoré A, Pérez-Pardal L, Sanou M, et al. Identification of genomic regions and candidate genes of functional importance for gastrointestinal parasite resistance traits in Djallonké sheep of Burkina Faso. Arch Anim Breed 2019;62(1):313–323.
  74. Becker GM, Davenport KM, Burke JM, Lewis RM, Miller JE, Morgan JLM, et al. Genome‐wide association study to identify genetic loci associated with gastrointestinal nematode resistance in Katahdin sheep. Anim Genet 2020;51(2):330–335.
  75. Ahbara AM, Rouatbi M, Gharbi M, Rekik M, Haile A, Rischkowsky B, et al. Genome-wide insights on gastrointestinal nematode resistance in autochthonous Tunisian sheep. Sci Rep 2021;11(1):9250.
  76. Raschia MA, Donzelli MV, Medus PD, Cetrá BM, Maizon DO, Suarez VH, et al. Single nucleotide polymorphisms from candidate genes associated with nematode resistance and resilience in Corriedale and Pampinta sheep in Argentina. Gene 2021;770:145345.
  77. Casu S, Usai MG, Sechi T, Salaris SL, Miari S, Mulas G, et al. Association analysis and functional annotation of imputed sequence data within genomic regions influencing resistance to gastro-intestinal parasites detected by an LDLA approach in a nucleus flock of Sarda dairy sheep. Genet Sel Evol 2022;54(1):2.
  78. Wilkie H, Riggio V, Matika O, Nicol L, Watt KA, Sinclair R, et al. A candidate gene approach to study nematode resistance traits in naturally infected sheep. Vet Parasitol 2017;243:71–74.
  79. Preston JM, Allonby EW. The influence of breed on the susceptibility of sheep of Haemonchus contortus infection in Kenya. Res Vet Sci 1979;26(2):134–139.
  80. Courtney CH, Parker CF, McClure KE, Herd RP. Resistance of exotic and domestic lambs to experimental infection with Haemonchus contortus. Int J Parasitol 1985;15(1):101–109.
  81. Palomo-Couoh JG, Aguilar-Caballero AJ, Torres-Acosta JFJ, González-Garduño R. Comparing the phenotypic susceptibility of Pelibuey and Katahdin female lambs against natural gastrointestinal nematode infections under hot humid tropical conditions. Parasitol Res 2017;116(6):1627–1636.
  82. Besier RB, Kahn LP, Sargison ND, Van Wyk JA. Diagnosis, treatment and management of Haemonchus contortus in small ruminants. In: Grassr RB, Samson GB editors: Advances in parasitology. . Elsevier. 2016:181-238. https://linkinghub.elsevier.com/retrieve/pii/S0065308X16300240. Accessed Oct 17, 2022
  83. Van Wyk JA, Bath GF. The FAMACHA system for managing haemonchosisin sheep and goats by clinically identifying individual animals for treatment. Vet Res 2002;33(5):509–529.
  84. Coltman DW, Wilson K, Pilkington JG, Stear MJ, Pemberton JM. A microsatellite polymorphism in the gamma interferon gene is associated with resistance to gastrointestinal nematodes in a naturally-parasitized population of Soay sheep. Parasitology 2001;122(5):571–582.
  85. Davies G, Stear MJ, Benothman M, Abuagob O, Kerr A, Mitchell S, et al. Quantitative trait loci associated with parasitic infection in Scottish blackface sheep. Heredity 2006;96(3):252–258.
  86. Paterson KA, Mcewan JC, Dodds KG, Morris CA, Crawford AM. Fine mapping a locus affecting host resistance to internal parasites in sheep. http://rgdoi.net/10.13140/2.1.3789.2486. Accessed Oct 6, 2022
  87. Beraldi D, McRae AF, Gratten J, Pilkington JG, Slate J, Visscher PM, et al. Quantitative trait loci (QTL) mapping of resistance to strongyles and coccidia in the free-living Soay sheep (Ovis aries). Int J Parasitol 2007;37(1):121–129.
  88. Sayers G, Good B, Hanrahan JP, Ryan M, Sweeney T. Intron 1 of the interferon γ gene: Its role in nematode resistance in Suffolk and Texel sheep breeds. Res Vet Sci 2005;79(3):191–196.
  89. Gutiérrez-Gil B, Pérez J, Álvarez L, Martínez-Valladares M, De La Fuente LF, Bayón Y, et al. Quantitative trait loci for resistance to trichostrongylid infection in Spanish Churra sheep. Genet Sel Evol 2009;41(1):46.
  90. Estrada‐Reyes ZM, Ogunade IM, Pech‐Cervantes AA, Terrill TH. Copy number variant‐based genome wide association study reveals immune‐related genes associated with parasite resistance in a heritage sheep breed from the United States. Parasite Immunol; https://onlinelibrary.wiley.com/doi/10.1111/pim.12943. Accessed Sep 12, 2022.
  91. Silva MVB, Sonstegard TS, Hanotte O, Mugambi JM, Garcia JF, Nagda S, et al. Identification of quantitative trait loci affecting resistance to gastrointestinal parasites in a double backcross population of Red Maasai and Dorper sheep: Parasite indicator QTL of Red Maasai sheep. Anim Genet 2012;43(1):63–71.
  92. Ahmed AM, Sebastiano SR, Sweeney T, Hanrahan JP, Glynn A, Keane OM, et al. Breed differences in humoral and cellular responses of lambs to experimental infection with the gastrointestinal nematode Teladorsagia circumcincta. Vet Res 2015;46(1):8.
  93. Ghannam A, Fauquert JL, Thomas C, Kemper C, Drouet C. Human complement C3 deficiency: Th1 induction requires T cell-derived complement C3a and CD46 activation. Mol Immunol 2014;58(1):98–107.
  94. Geijtenbeek TBH, Gringhuis SI. Signalling through C-type lectin receptors: shaping immune responses. Nat Rev Immunol 2009;9(7):465–479.
  95. Gossner A, Wilkie H, Joshi A, Hopkins J. Exploring the abomasal lymph node transcriptome for genes associated with resistance to the sheep nematode Teladorsagia circumcincta. Vet Res 2013;44(1):68.
  96. Chitneedi PK, Suárez-Vega A, Martínez-Valladares M, Arranz JJ, Gutiérrez-Gil B. Exploring the mechanisms of resistance to Teladorsagia circumcincta infection in sheep through transcriptome analysis of abomasal mucosa and abomasal lymph nodes. Vet Res 2018;49(1):39.
  97. Zhang R, Liu F, Hunt P, Li C, Zhang L, Ingham A, et al. Transcriptome analysis unraveled potential mechanisms of resistance to Haemonchus contortus infection in Merino sheep populations bred for parasite resistance. Vet Res 2019;50(1):7.
  98. Sweeney T, Hanrahan JP, Ryan MT, Good B. Immunogenomics of gastrointestinal nematode infection in ruminants - breeding for resistance to produce food sustainably and safely. Parasite Immunol 2016;38(9):569–586.
  99. Yamazaki T, Goya I, Graf D, Craig S, Martin-Orozco N, Dong C. A butyrophilin family member critically inhibits T cell activation. J Immunol 2010;185(10):5907–5914.
  100. Yang W, Gu Z, Zhang H, Hu H. To TRIM the immunity: From innate to adaptive immunity. Front Immunol 2020;11:02157.
  101. Han TS, Hur K, Xu G, Choi B, Okugawa Y, Toiyama Y, et al. MicroRNA-29c mediates initiation of gastric carcinogenesis by directly targeting ITGB1. Gut. 2015;64(2):203–214.
  102. Iwanaga S, Lee BL. Recent advances in the innate immunity of invertebrate animals. BMB Rep 2005;38(2):128–150.
  103. Rosenberg HF, Dyer KD, Foster PS. Eosinophils: changing perspectives in health and disease. Nat Rev Immunol 2013;13(1):9–22.
  104. Balic A, Cunningham CP, Meeusen ENT. Eosinophil interactions with Haemonchus contortus larvae in the ovine gastrointestinal tract. Parasite Immunol 2006;28(3):107–115.
  105. Bisset SA, Morris CA, Squire DR, Hickey SM. Genetics of resilience to nematode parasites in young Romney sheep)-use of weight gain under challenge to assess individual anthelmintic treatment requirements. N Z J Agric Res 1996;39(3):313–323.
  106. Robinson N, Pleasance J, Piedrafita D, Meeusen EN. The kinetics of local cytokine and galectin expression after challenge infection with the gastrointestinal nematode, Haemonchus contortus. Int J Parasitol 2011;41(5):487–493.
  107. Preston SJM, Beddoe T, Walkden-Brown S, Meeusen E, Piedrafita D. Galectin-11: A novel host mediator targeting specific stages of the gastrointestinal nematode parasite, Haemonchus contortus. Int J Parasitol 2015;45(12):791–796.
  108. Legrand O, Perrot J, Tang R, Simonin G, Gurbuxani S, Zittoun R, et al. Expression of the multidrug resistance‐associated protein (MRP) mRNA and protein in normal peripheral blood and bone marrow haemopoietic cells. Br J Haematol 1996;94(1):23–33.
  109. Pazos M, Siccardi D, Mumy KL, Bien JD, Louie S, Shi HN, et al. Multidrug resistance-associated transporter 2 regulates mucosal inflammation by facilitating the synthesis of hepoxilin A3. J Immunol 2008;181(11):8044–8052.