Indian Journal of Animal Research

  • Chief EditorK.M.L. Pathak

  • Print ISSN 0367-6722

  • Online ISSN 0976-0555

  • NAAS Rating 6.50

  • SJR 0.263

  • Impact Factor 0.4 (2024)

Frequency :
Monthly (January, February, March, April, May, June, July, August, September, October, November and December)
Indexing Services :
Science Citation Index Expanded, BIOSIS Preview, ISI Citation Index, Biological Abstracts, Scopus, AGRICOLA, Google Scholar, CrossRef, CAB Abstracting Journals, Chemical Abstracts, Indian Science Abstracts, EBSCO Indexing Services, Index Copernicus

Mycobacterium avium Subspecies paratuberculosis Infection and Immunopathological Changes in Animals: A Review

M. Kumar1,4, K.K. Chaubey2,5,*, D. Dayal1, S.V. Singh1, S. Sharma3, U. Goyal1, S. Tyagi1, A. Pramanik2
1Department of Biotechnology, Institute of Applied Sciences and Humanities, GLA University, Mathura-281 406, Uttar Pradesh, India.
2Division of Research and Innovation, School of Applied and Life Sciences, Uttaranchal University, Arcadia Grant, Dehradun-248 007, Uttarakhand, India.
3Department of Veterinary Physiology and Biochemistry, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar-125 004, Haryana, India.
4Department of Veterinary Clinical Complex, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar-125004, Haryana, India.
5ICAR-Central Institute for Research on Goats, Makhdoom, Farah, Mathura-281 122, Uttar Pradesh, India.
6School of Basic and Applied Sciences, Sanskriti University, Mathura-281 401, Uttar Pradesh, India.
Mycobacterium avium subspecies paratuberculosis (MAP) is a chronic granulomatous intestinal disease and causative agent of Johne’s disease (JD) in domestic and wild ruminants. This study explores the intricate dynamics of the host immune response in MAP infection and its implications for disease progression and control. Our investigation revealed the pivotal role of interleukin-10 (IL-10) in mediating the communication between the host’s innate and adaptive immunological responses. IL-10 inhibits interferon-gamma (IFN-γ) secretion, altering the balance between Th1 and Th2 responses. In the early stages, a robust Th1 response marked by increased IFN-γ production is observed, but as the disease progresses, a shift towards a humoral Th2 response occurs. Furthermore, we found that IL-10 exerts suppressive effects on macrophage (Mφ) anti-mycobacterial action, leading to increased intracellular survival of mycobacteria. IL-10 also hampers Mφ antimicrobial activity by suppressing the production of essential cytokines such as IL-12 and tumor necrosis factor alpha (TNF-α), crucial for activating natural killer (NK) cells and inducing differentiation of CD4+ T lymphocytes into Th1 effector cells. Importantly, our study sheds light on the intricate interplay between past infections or concurrent active co-infections and their influence on the immune response to unrelated pathogens in the context of paratuberculosis. This review underscores the multifaceted role of IL-10 in the immunopathological manifestation of MAP infection and host immunomodulation. These findings provide valuable insights for both animal and human research in the fight against MAP infection, emphasizing the importance of balancing immune responses and considering the influence of concurrent infections in disease progression. 
Mycobacterium avium subspecies paratuberculosis (MAP) is the causative agent of Johne’s disease (JD), a chronic granulomatous intestinal illness that affects cattle, primates and small/wild ruminants (Garvey, 2018). The disease poses a significant economic burden due to its negative impact on livestock farming and animal welfare (Chaubey et al., 2017). MAP infection is known to cause paratuberculosis (pTB) in a wide range of animals (Chaubey et al., 2016). With its slow and insidious spread, pTB exhibits a long incubation period, with initial asymptomatic signs typically manifesting in young calves after approximately a year (Karcher et al., 2008). The global prevalence of pTB has been a cause of concern, as it is documented in specific regions and continues to spread rapidly within the livestock industry, leading to substantial financial losses (Roller et al., 2020). Diagnosis of MAP infection is very tough and challenging because infection producing non-specific symptoms and it comes under observation after long incubation period of the exposure, very long or variable sub-clinical stage of disease in individual infected animals and intermittent or continuous shedding of the MAP bacilli in their milk and faeces (Sweeney et al., 2011; Chaubey et al., 2016; 2017). Chaubey et al., (2019) compared different screening test where they reported histopathological analysis and immunomodulation in host due to MAP infection and also showed pathological test having higher sensitivity than other screening tests. Sharma et al., (2022) explained that changes in microbiome profile of the host may modulate the host immune responses. The complex interplay between MAP and the host’s immune responses has been a subject of extensive research, shedding light on the immunopathological changes that occur during the course of the infection (Kravitz et al., 2021).

In this comprehensive review, we aim to investigate into the diverse aspects of MAP infection and its association with immunopathological alterations in various animal species. By findings from multiple studies, we seek to provide a deeper understanding of the mechanisms underlying the establishment and progression of pTB. Additionally, we will explore the implications of MAP infection on animal health, agriculture and the economy, with a focus on the global impact of the disease. Through this exploration, we hope to contribute to the growing body of knowledge surrounding MAP infection and its consequences, which could potentially pave the way for improved diagnostics, prevention strategies and therapeutic interventions. As the understanding of pTB continues to evolve, addressing this significant health concern will be crucial for safeguarding animal populations and promoting sustainable livestock practices.
 
Phases of pTB infection
 
The disease progresses through three clinical stages. In stage I (subclinical with undetectable excretion), there is appreciable shedding of bacteria. There are no clinical indications during the silent infection period, however animals periodically shed MAP in their faeces (Cocito et al., 1994). This is followed by a subclinical excretory phase (stage II), during which the concentration of mycobacteria in the intestinal mucosa and lumen progressively increases. The terminal phase (stage III, clinical and excretory) is characterized by an intractable chronic diarrhoea and the symptoms of a generalized infectious process (Whitlock and Buergelt, 1996). The majority of the animals die in the clinical phase caused by severe dehydration and emaciation due to disease.
 
Host-pathogen responses in pTB
 
The immune response to pTB starts with a strong T-helper1 (Th1) response that is marked by increased interferon-gamma (IFN-γ) production (Stabel and Whitlock, 2001). With the advancement of the disease, the immunological response shifts from a cell-mediated Th1-type response to a body’s immune Th2-type response (Leite et al., 2015). Strong Th1 immunological responses targeted against the causal pathogen, MAP is thought to be required to keep bacterial reproduction under control (Begg et al., 2018). Macrophages (Mφ) then take up MAP and engage with gut-associated lymphoid tissue (GALT), causing a variety of local alterations. Therefore, the result of MAP infection is believed to be determined by the local immune responses induced by lymphocytes in various layers of the gut (Munjal et al., 2005). Th1 response, which is defined by the generation of IFN-γ controls the early host response to MAP infections (Park et al., 2018).

Furthermore, in pTB bovine model, as shown in Fig 1 about the overview of immune responses, the innate immune response in natural and challenged MAP infected animals declining with progression of the disease (Begg et al., 2011). Pathogenic mycobacteria communicate through toll like receptor 2 (TLR2) to avoid Mj antigen presentation and antimicrobial responses, according to several investigations (Banaiee et al., 2006). By interacting with TLR2 on bovine mononuclear phagocytes, MAP activates mitogen-activated protein kinase-p38 (MAPK-p38), resulting in enormous levels of interleukins-10 (IL-10) synthesis and a small level of IL-12 synthesis. For the elimination of microbial infection, successful phago-lysosomal maturation is a critical innate immunological response. During mycobacterial infection, MAPK-p38 plays a key role in signalling pathways (Khalifeh and Stabel, 2004) and it’s linked to blocking phagosome-lysosome fusion and maturation. IL-10 can be a powerful anti-inflammatory cytokine that prevents phagosome formation and apoptosis. We also discuss techniques for targeting IL-10, MAPK and signal transducer and activators of transcription 3 (STAT3)  in various intracellular pathogen contamination (Hussain et al., 2016). MAP infection alters miRNA expression, which is important for fine-tuning the host antibody response and could be used as biomarkers and diagnostic tools for MAP infection (Malvisi et al., 2016). MAP infection has a higher susceptibility rate than other diseases because pTB can spread horizontally and vertically in the animals (Nielsen et al., 2013). MAP infection has been also reported as chronic inflammatory bowel disease in human and present in the blood, intestine and breast milk (Naser et al., 2004). At the time of infection of subclinical phases, Mφ form granulomas in the gut lymphoid tissue due to the insistence of MAP. Faecal shedding is known to take place with the down regulation of cell-mediated immunological response and over expression of the humoral immune response however, in a few cases sick animals pointed enhanced faecal shedding along when cellular immunity is activated (Ganusov et al., 2015).
 

Fig 1: Overview of immune response in MAP infection.



Pathophysiological connection between Mφ and other immune cells in MAP infection
 
In certain situations, the production of IL-1 can reach excessive levels, raising the chances that few clinical JD symptoms because of IL-1 toxicity and that increased IL-1 production at infection sites coincides with increased expression of tumor necrosis factor receptor-associated protein 1  (raf-1) in invading Mφ. The pathophysiology of MAP infection is thus consistent with increased IL-1 expression, especially because lesions associated with JD often feature a large number of Mφ (Aho et al., 2003). IFN-γ and IL-6 transcript expression is similarly increased in intestinal tissues when MAP infection is active (Coussens et al., 2004). Cluster of differentiation 40 (CD40) signalling is impaired on Th1 cells, which is a key Mφ target for CD40 ligand (CD40L) to keep a Th1 immunological response. Mononuclear phagocytic cells (MPC) provide an intracellular environment for MAP survival and proliferation. Pathogen recognition receptors (PRRs) on Mj are important in the absorption of mycobacteria. One of the immunological evasion strategies: MAP can impede the formation and acidification of the phagocytic vacuole within Mφ, preventing bacteria from being exposed to bactericidal lysosomal enzymes and oxygen-obtaining radicals (Sohal et al., 2008). As a result, MAP can proliferate within Mj phagosomes while utilising resources of host cell. MAP is thought to cause apoptosis and infect nearby Mφ, drift into the adjacent lymphatic organ, allowing bacteria to spread to lymph nodes, including the mesenteric lymph nodes (Allen et al., 2001). MAP can block Mφ apoptosis, lowering the immunological response and permitting intracellular reproduction, based at the level of strain’s infection virulence (Kabara and Coussens, 2012).

Infected mice’s spleens had an increased number of cytokines and MAP-reactive T cells and their serum had modestly raised levels of IFN-γ (Abdissa et al., 2020). The pathological alterations found in groups leporine and bovine were clearly demarcated, non-encapsulated granuloma comprising epithelioid cells, Mφ and the infrequent giant cell. Although no acid-fast bacteria (AFB) was seen in some granulomata, a varied no. of AFB had been detected in particular granulomata’s cells. But lesions have been found in the jejunum of an organism (B3), with granuloma in both the jejunal Peyer’s patch (JPP) and the overlaying lamina propria, as well as a limited variety of AFB in the JPP granulomata (Beard et al., 2001).

The use of macroscopic alterations in the mesenteric lymph nodes and small intestine as a prescription in the post-mortem diagnosis of JD illness in goats have been observed. Macroscopical alterations were frequently absent or non-specific. Caseous and/or calcified foci in infected animals’ mesenteric lymph nodes were frequently detected, whereas reported intestinal alterations were surprisingly rare. However, substantial epithelioid cell infiltrations and numerous acid-fast bacilli were frequent in portions of macroscopically unaltered gut. Infected animals had productive inflammation with tubercle development in lymph nodes on a random basis (Fodstad and Gunnarsson, 1979).
 
Immunological biomarkers and their role in MAP infection
 
Antibodies (Ab)
 
In spite of the fact that the significance of the production of antibodies in immunity of host to intracellular mycobacterial infections is unknown, certain Abs detected in the serum, can play a role in preservative immunity (Achkar and Prados-Rosales, 2018). Although less widely used in Bovine tuberculosis (bTB), ELISA conjugated with serum and milk antibody is common diagnostics for pTB. Commercially available pTB test methods have the maximum diagnostic performance in the later phases of the disease, when animals are infected, but lower sensitivity in the initial stages of infection (Nielsen and Toft, 2008). In an experiment in sheep, animals designated as disease resistant has a greater response of antibody than those in which the infection developed (De Silva et al., 2018). This opens up few latest possibilities for serological testing to be utilized to detect hardy animals during the initial phases of sickness. In a study, a indigenously developed ELISA was employed to screen serum samples from cattle, evaluating the antibody response to MAP infection and the results indicated a 27.22% prevalence of MAP infection in the cattle population in the Madhya Pradesh region (Audarya et al., 2022).

Multiplex bead-based assays exhibit a dependable capability for detecting MAP infection, even in its initial stages, when conventional commercial ELISA tests often fail to detect antibody responses in animals. In terms of improving early diagnosis in both pTB and bTB, a variety of antigens have been investigated (Li et al., 2017). The most common subtype employed in mycobacterial antibody ELISA is immunoglobulin G (IgG); although, targeting alternative isoforms may be more revealing. IgA, the most common subtype seen in mucosal secretion, has also holds promise in identifying resilience, since it has been linked to protective responses in mycobacterial infection in mice (Tjarnlund et al., 2006). During the course of pTB disease, MAP-specific faecal IgA immunoglobulin can be detected; however this is temporary and seems to be related to ambient MAP infection (Begg et al., 2015).
 
Cytokines, IFN-γ, chemokines and ILs in MAP
 
Assessing the IFN-γ response directly at the infection site could offer a more pertinent parameter for gaining a deeper understanding of the cytokine’s involvement in fecal shedding. Unfortunately, implementing such a diagnostic test is not feasible (De Silva et al., 2018). Sheep has a larger initial IFN-γ response when they are immature, this cytokine has the potential to be used as a biomarker of resistance. Other cytokines and chemokines that are differentially regulated between infected and uninfected individuals and between active and hidden infection, have been reported and these are likely relevant to other mycobacterial diseases. In research on human mycobacterial infections, activated T cells and associated cytokines, such as IL-2, IL-3, IL-6, IL-7, IL-8, IL-9 and IL-10 were found to vary considerably even between infected and normal groups (Anbarasu et al., 2013). Hence, further investigation is imperative, especially concerning pathogen-specific reactions, to ascertain whether cytokine levels can serve as a reliable means of determining and differentiating in clinical contexts (Table 1).

Table 1: Cytokine, chemokine and interleukin responses were observed in MAP infection.



As the clinical condition worsens in both sheep and goats, the immune response is reduced, most likely due to overexpression of immune-suppressive cytokines (Lybeck et al., 2009). The release of IFN-γ from Th-1 in goats is induced by neutralisation of IL-10 in vitro with monoclonal antibodies supporting IL-10’s inhibitory impact. IFN-γ assays results in the context of pTB can offer valuable insights into the extent of exposure within a herd, serving as a robust indicator of herd-level exposure. Existing diagnostic tests in cattle use IFN-γ as a marker of MAP exposure; however, due to its low specificity, it is not a suitable marker of infection (Plain et al., 2012). An elevation in IL-10 was detected in sheep, four months following experimental infection with MAP, implying that infected sheep as alternative develop Th1 immune response over Th2 immunological response in initial phase of the disease. As a result, the significance of IL-10 in the response to MAP is debatable and the basis for the disparity in reaction between the two types has unexplained. Overall total leucocyte counts and lymphocytes have been shown to be significantly diminished in natural cases of MAP infection in buffaloes (Sharma et al., 2020).

MAP targets the host’s lymphoid tissues linked with the mucosa. The micro fold cells (M cells) of neonatal calves, goat kids and mice’s ileal Peyer’s patches are one way that MAP attacks the organism (Bermudez et al., 2010). Fig 2 depicts the infection of MAP in calves and its subsequent host interactions. M cells play a pivotal role as antigen-presenting cells and have been demonstrated to internalize and transport various pathogenic microorganisms, such as Mycobacterium species, without undergoing degradation. Bacteria can remain and delivered whole to the fundamentally immune-competent, sub-epithelial lymphoid organ due to lacking of lysosomes and hydrolytic enzymes by M-cells (Sigurardottir et al., 2004). On its cell wall, MAP produces fibronectin attachment protein (FAP), allowing it to be opsonized by fibronectin and afterwards uptaken by M cells, that have numerous β1 integrin receptors at its luminal side. During MAP absorption, M cells activate defensins, which are AMP to guard the host. A transitory rise in β-defensin is observed in the first four hours after exploratory MAP infection of cattle. Enterocytes in lambs and mice have also been proven to occupy MAP (Bermudez et al., 2010). IL-12 plays a pivotal role in fostering the development of Th1 cells that produce IFN-γ when naive CD4+ T cells are activated. Furthermore, in the context of infection caused by intracellular parasites, IL-12 produced by macrophages stimulates NK cells to generate IFN-g, thereby contributing to the protective response during the acute phase of infection.

Fig 2: Infection of MAP in calf and the mode of entrance into intestine.


 
Clusters of differentiation (CD) markers, Mj and Th cells
 
MAP is relocates the mucosal epithelium after initial invasion by M cells, then penetrates and continue in sub-epithelial Mj, triggering a cellular immunological response centred at the CD4+ Th1 cell mileu. Complement receptors, immunoglobulin receptors and scavenger receptors all play a role in Mj mediated MAP uptake (Whittington et al., 2012). MAP is delivered to CD4+ Th-1 cells by Mj at the time of primary infection. IFN-g is secreted by CD4+ T-cells, which inhibits bacterial proliferation. Throughout the subclinical stage of JD, the amount of IFN-g rises in the ileal and cecal lymph nodes of cattle. MAP is thought to infiltrate and inactivate Mj by interfering with their potential to respond to normal T-cell signalling, according to some studies. T-cells have compelled to acknowledge to extracellular indicator via the CD154-CD40 pathway during MAP infection, favouring unsuitable Th2-like pursuit, as well as IL-10 production and unable to start phagosome acidification, which should fight infection (Sommer et al., 2009). The progression of JD from subclinical to clinical forms has linked to a movement  in immunological response from Th1 to Th2, which results in the development of a significant cellular response as well as a simultaneous antibody and IFN-γ response (Begg et al., 2011). Lambs infected with MAP’s C strain developed a combined cellular and humoral response, but animals affected with S strain only developed a weak innate immunity (Fernandez et al., 2014). Sheep’s immunological response to MAP infection has influenced by the strain, which influences the clinical condition they suffer. Antigen mediated humoral immune response of Th1 cells and CD4+/CD8+ ratio >1 has been seen during paucibacillary enteritis (Reddacliff et al., 2004). Sheep with the paucibacillary degenerative disease have elevated amounts of IFN-g, whereas those with the multibacillary structure have a strong humoral response to MAP and a weak cell-mediated immunological response to MAP, as well as a lower ratio of CD4+ and CD8+ cells. IFN-γ production is lower in this stage of the disease (Smeed et al., 2007).
 
TH1- Mφ interaction
 
A cytotoxic and proinflammatory Th1-like response is required to sway MAP infections. Maintaining Th1 response and Mj activation requires interaction between CD40 receptors and CD154 (CD40L) on Mφ and activated T cells respectively (Sommer et al., 2009). Although MAP is mostly restricted to Mj, infection control requires a proinflammatory and cytotoxic Th1 response marked by proinflammatory cytokines like IL-12 and IFN-γ, as well as the generation of nitric oxide (NO). An adequate Th1 response emerges soon after MAP exposure (Coussens, 2001). The proinflammatory Th1-like response is shifted during the subclinical stage of MAP infection and a Th2-like response, marked by antibody formation, takes over (Stabel, 2000).
 
Th1/TH17axis
 
In the initial stages of mycobacterial infection, Th17-derived cytokines play a critical role.  IL-26 and IL-17F are linked to host defence against intracellular microorganisms (Coulter et al., 2017). IL-26 causes immune cells to be primed and infections to be killed directly by forming membrane pores. Furthermore, through modulating the Th1 response and neutrophil recruitment, IL-17 provides protection against intracellular infections (Umemura et al., 2007). IL-17, IL-23 and IL-26 are Th17-derived cytokines that improve granuloma integrity by modulating neutrophil recruitment via CXCR3 signalling. In sick animals, peripheral and intestinal CMI responses had been lower than in subclinical animals. The destruction of CD4+ T cells and leads to a steady decline in T cells appeared to be linked to disease progression. Deregulation of Mj function mediated by MAP is the underlying problem that could be linked to CMI depression (Tooker et al., 2002). Activation of gene and signalling along genes encoding TNF-α, IL-1, 12 and g, can result in disruption in the immunological response that controls disease. Mj that active, containing MAP travel to lymph nodes and available an antigen, which stimulates natural T cells to produce IFN-g and cytokines like IL-6, IL-1 and IL-2 inducing the Th1 response (Coussens, 2001). Activated Th1 cells cause the immunological response that cell-mediated by producing IL-2, TNF and IFN-γ during the subclinical phages. Tim3 galectin 9 interaction have also been shown to regulate antiviral (Machala et al., 2019; Sharma et al., 2011) and anti-bacterial (Sada-Ovalle et al., 2012) immune responses.
 
Toll like receptors (TLRs)
 
TLRs recognise PAMPS originating from diverse microorganisms and emulate in innate immunological system. TLRs trigger the activation of the transcription factors NF-κB and IFN-regulatory factors (IRFs), which control the consequence of innate immunological responses, by attracting certain adaptor molecules (Kawasaki and Kawai, 2014). Innate immune cells including dendritic cells (DCs) and Mj, as well as non-immune cells like fibroblast cells and epithelial cells, express TLRs. TLR2 identifies a broad range of pathogen-associated molecular patterns (PAMPs), as well as lipoproteins, lipotechoic acids, peptidoglycans, zymosan, mannan and tGPI-mucin, in combination with TLR1 or TLR6 (Kawai and Akira, 2010). For the early identification of microorganisms, the innate immunological system uses germline-encoded pattern recognition receptors (PRRs). PAMPs and own generated molecules formed from injured cells, known as damage-associated molecular patterns (DAMPs), are recognized by PRRs. By generating inflammatory cytokines, such as type I IFN and other mediators, PRRs activate downstream signalling pathways which contribute in the development of innate immunological responses. Antigen-specific adaptive immunological responses are primed and orchestrated by these pathways, which not only generate immediate host defensive responses like inflammation. These reactions are required for both the clearance of invading microorganisms and the subsequent training of antigen-specific adaptive immune responses (Kawasaki and Kawai, 2014).

Changes in TLR gene expression in sheep at the time of experimental infection with MAP have been observed in both in vitro and in vivo experiments (Plain et al., 2010). TLR2 regulates the specific immune system in the initial phases of JD and influences development of disease in selected tissues and in sheep PBMCs (Fiorentina et al., 2021). TLR2 mRNA expression rises in the multibacillary form of JD, owing to a stronger inflammatory response in regions at which the majority of microbes are located (Nalubamba et al., 2008). TLR4 expression is higher in animals suffering from the multibacillary type of the disease. In both paucibacillary and multibacillary stages of disease, expression of TLR1 and TLR 6, particularly TLR6 in sheep, increases in target organs. TLR6 and TLR2 expression levels rise, allowing the TLR2/TLR6 heterodimer to form in response to MAP (Plain et al., 2010). TLR9 expression is likewise up in the PBMC of calves who have been experimentally infected with MAP (Arsenault et al., 2013). On the other hand, no alterations have been observed in TLR9 over expression in PBMCs of sheep and cattle spontaneously diseased with MAP (Thirunavuk karasu et al., 2013). TLR activation in Mj and dendritic cells, especially in response to mycobacterial infection, stimulates intracellular signalling pathways, which result in the generation of proinflammatory cytokines (TNF-α, IL-6, IL-1) via NF-κB pathway (Fig 3). MAP infection in sheep and goats has been studied using these expression markers (Purdie et al., 2012).

Fig 3: TLR response in Infection of MAP.


 
Micro-RNA (miRNA)
 
The role of miRNAs and their influence on Mj activation during MAP pathogenesis is still unknown. MiRNAs have been identified as a significant arbitrator of the host immunological response to infection in recent research, primarily by modulating proteins that are intricate in both innate and adaptive immunological pathways (Drury et al., 2017). According to certain studies, both in vivo and in vitro, Micro-RNA-27a-3p (miRNA-27a-3p) modification is down regulated during MAP infection. miRNAs are short non-coding endogenous fragments of RNA with a length of 21-25 nucleotides that control gene expression by binding to the 32 -UTR of target mRNA genes (Hoentjen et al., 2005). Furthermore, miRNAs play a role in Mj polarization as well as T and B cell differentiation. The miRNA-27a and miRNA-27b have antimicrobial activity against murine CMV infection in distinct mouse cell lines, according to early research (Buck et al., 2010). Farrell unveiled several newly discovered circulating miRNAs present in bovine serum and pinpointed miRNAs whose levels are associated with bovine development, which could be regarded a new technique for pTB control. These miRNAs retain their stability after being stored for a long time and are used as a diagnostic marker (Farrell et al., 2015). The role of miRNAs in the regulation of innate immunity in pTB remains unknown (Meng et al., 2014).
In pTB, the significance of IL-10 is widely recognized as it plays a pivotal role in mediating the communication between the host’s innate and adaptive immunological responses. Studies have demonstrated that IL-10 inhibits the secretion of IFN-γ in the peripheral blood of calves and goats infected with MAP. Notably, antigen-presenting cells (APCs) generated from ovine and bovine monocytes secrete IL-10 in response to MAP. While IL-10 serves as a vital immunoregulator, it also exerts suppressive effects on Mφ anti-mycobacterial action, leading to increased intracellular survival of mycobacteria. Moreover, IL-10 inhibits Mj antimicrobial activity by suppressing the production of other crucial cytokines such as IL-12 and TNF-α. IL-12, an essential pro-inflammatory cytokine, is produced by APCs like monocytes, Mφ and dendritic cells. IL-12 primarily activates natural killer cells (NK cells) and induces differentiation of CD4+ T lymphocytes into Th1 effector cells, which produce IFN-γ. In the context of pTB, IL-10 plays a multifaceted role in the disease process, including guiding Th2-type cell growth in the lungs. Animal models and human investigations have revealed that past infections or concurrent active co-infections can significantly influence the immune response to unrelated pathogens. This study highlights the immunopathological manifestation caused due to MAP infection and host immunomodulation due to MAP infections and also provides valuable insights in animal and human research against MAP infection. Paucibacillary and multibacillary pathologies are associated with type 1 and type 2 Th responses, respectively. Nevertheless, numerous other cytokines exhibit distinct expression patterns, underscoring their involvement in a intricate network of interactions that contribute to the development of these final pathologies. The notable rise in IL10 expression concurrent with IFN-γ expression following M. bovis infection could suggest that the host is attempting to mitigate the pathological damage linked to robust pro-inflammatory immune responses.
We are thankful to DBT project [grant number BT/PR32758/AAQ/1/760/2019 dt. 29.01.2020], DBT, New Delhi for providing funds for this work.
All authors declare no conflict of interest.

  1. Abdissa, K., Ruangkiattikul, N., Ahrend, W., Nerlich, A., Beineke, A., Laarmann, K., Janze, N., Lobermeyer, U., Suwandi, A., Falk, C., Schleicher, U., Weiss, S., Bogdan, C., Goethe, R. (2020). Relevance of inducible nitric oxide synthase for immune control of Mycobacterium avium subspecies paratuberculosis infection in mice. Virulence. 11(1): 465- 481. 

  2. Achkar, J.M., Prados-Rosales, R. (2018). Updates on antibody functions in Mycobacterium tuberculosis infection and their relevance for developing a vaccine against tuberculosis. Current Opinion in Immunology. 53: 30-37.

  3. Aho, A.D., McNulty, A.M., Coussens, P.M. (2003). Enhanced expression of interleukin-1á and tumor necrosis factor receptor- associated protein 1 in ileal tissues of cattle infected with Mycobacterium avium subsp. paratuberculosis. Infection and Immunity. 71(11): 6479-6486.

  4. Allen, S., Sotos, J., Sylte, M.J., Czuprynski, C.J. (2001). Use of Hoechst 33342 staining to detect apoptotic changes in bovine mononuclear phagocytes infected with Mycobacterium avium subsp. paratuberculosis. Clinical Diagnostic Laboratory Immunology. 8(2): 460-464.

  5. Anbarasu, D., Raja, C.P. and Raja, A. (2013). Multiplex analysis of cytokines/chemokines as biomarkers that differentiate healthy contacts from tuberculosis patients in high endemic settings. Cytokine. 61(3): 747-754.

  6. Aranday-Cortes, E., Hogarth, P.J., Kaveh, D.A., Whelan, A.O., Villarreal-Ramos, B. (2012). Transcriptional profiling of disease-induced host responses in bovine tuberculosis and the identification of potential diagnostic biomarkers. Plos One. 7(2): 30626. DOI: 10.1371/journal.pone.0030626. 

  7. Arsenault, R.J., Li, Y., Maattanen, P., Scruten, E., Doig, K., Potter, A., Griebel, P., Kusalik, A., Napper, S. (2013). Altered Toll-like receptor 9 signaling in Mycobacterium avium subsp. paratuberculosis-infected bovine monocytes reveals potential therapeutic targets. Infection and Immunity. 81(1): 226-237. 

  8. Audarya, S.D., Singh, M., Chaubey, K.K., Gupta, S., Sikrodia, R. (2022). Antibodies against Mycobacterium avium subspecies paratuberculosis in Cattle of Indore District in Madhya Pradesh. Indian Journal of Animal Research. 1: 6. doi: 10.18805/IJAR.B-4773.

  9. Banaiee, N., Kincaid, E.Z., Buchwald, U., Jacobs, W.R., Ernst, J.D. (2006). Potent inhibition of macrophage responses to IFN-gamma by live virulent Mycobacterium tuberculosis is independent of mature mycobacterial lipoproteins but dependent on TLR2. Journal of Immunology (Baltimore, Md./ : 1950). 176(5): 3019-3027. 

  10. Beard, P.M., Stevenson, K., Pirie, A., Rudge, K., Buxton, D., Rhind, S.M., Sinclair, M.C., Wildblood, L.A., Jones, D.G., Sharp, J.M. (2001). Experimental Paratuberculosis in calves following inoculation with a rabbit isolate of Mycobacterium avium subsp. paratuberculosis. Journal of Clinical Microbiology. 39(9): 3080-4. https://doi.org/10.1128/ JCM.39.9.3080-3084.2001.

  11. Begg, D.J., de Silva, K., Carter, N., Plain, K.M., Purdie, A., Whittington, R.J. (2011). Does a Th1 over Th2 dominancy really exist in the early stages of Mycobacterium avium subspecies paratuberculosis infections? Immunobiology. 216(7): 840-846. 

  12. Begg, D.J., de Silva, K., Plain, K.M., Purdie, A.C., Dhand, N., Whittington, R.J. (2015). Specific faecal antibody responses in sheep infected with Mycobacterium avium subspecies paratuberculosis. Veterinary Immunology and Immunopathology. 166(3-4): 125-131. 

  13. Begg, D.J., Plain, K.M., de Silva, K., Gurung, R., Gunn, A., Purdie, A.C., Whittington, R.J. (2018). Immunopathological changes and apparent recovery from infection revealed in cattle in an experimental model of Johne’s disease using a lyophilised culture of Mycobacterium avium subspecies paratuberculosis. Veterinary Microbiology. 219: 53-62. 

  14. Bermudez, L.E., Petrofsky, M., Sommer, S., Barletta, R.G. (2010). Peyer’s patch-deficient mice demonstrate that Mycobacterium avium subsp. paratuberculosis translocates across the mucosal barrier via both M cells and enterocytes but has inefficient dissemination. Infection and Immunity. 78(8): 3570-3577. 

  15. Berry, A., Wu, C.W., Venturino, A.J., Talaat, A.M. (2018). Biomarkers for early stages of johne’s disease infection and immunization in goats. Frontiers in Microbiology. 9: 2284. doi: 10.3389/ fmicb.2018.02284.  

  16. Buck, A.H., Perot, J., Chisholm, M.A., Kumar, D.S., Tuddenham, L., Cognat, V., Marcinowski, L., Dölken, L., Pfeffer, S. (2010). Post-transcriptional regulation of miR-27 in murine cytomegalovirus infection. RNA (New York, N.Y.). 16(2): 307-315. 

  17. Chaubey, K.K., Gupta, R.D., Gupta, S., Singh, S.V., Bhatia, A.K., Jayaraman, S., Kumar, N., Goel, A., Rathore, A.S., Sahzad and Sohal, J.S. (2016). Trends and advances in the diagnosis and control of paratuberculosis in domestic livestock. Veterinary Quarterly. 36(4): 203-227.

  18. Chaubey, K.K., Singh, S.V., Gupta, S., Singh, M., Sohal, J.S., Kumar, N., Singh, M.K., Bhatia, A.K. and Dhama, K. (2017). Mycobacterium avium subspecies paratuberculosis- an important food borne pathogen of high public health significance with special reference to India: An update. Veterinary Quarterly. 37(1): 282-299.

  19. Chaubey, K.K., Gangwar, N., Pawaiya, R.S., Jatav, G.P., Sohal, J.S., Singh, S.V., Singh, M., Gupta, S., Kumaresan, G., Kumar, N. and Jayaraman, S. (2019). Evaluation of newly developed ‘six recombinant secretary proteins based ‘cocktail ELISA’and ‘whole cell lysate’based ‘indigenous ELISA’and tissue microscopy’with ‘Gold standard’ histo- pathology for the diagnosis of Johne’s disease in slaughtered goats and buffaloes. Comparative Immunology, Microbiology and Infectious Diseases. 66: 101338. 10.1016/j.cimid. 2019.101338.

  20. Cocito, C., Gilot, P., Coene, M., De Kesel, M., Poupart, P. and amp; Vannuffel, P. (1994). Paratuberculosis. Clinical Microbiology Reviews. 7(3): 328-345.

  21. Coulter, F., Parrish, A., Manning, D., Kampmann, B., Mendy, J., Garand, M., Lewinsohn, D.M., Riley, E.M., Sutherland, J.S. (2017). IL-17 Production from T Helper 17, Mucosal- Associated Invariant T and gd Cells in Tuberculosis Infection and Disease. Frontiers in Immunology. 8(OCT). Article 1252. https://doi.org/10.3389/fimmu.2017.01252.

  22. Coussens, P.M. (2001). Mycobacterium paratuberculosis and the bovine immune system. Animal Health Research Reviews. 2(2): 141-162. 

  23. Coussens, P.M., Verman, N., Coussens, M.A., Elftman, M.D., McNulty,  A.M. (2004). Cytokine gene expression in peripheral blood mononuclear cells and tissues of cattle infected with Mycobacterium avium subsp. paratuberculosis: Evidence for an inherent proinflammatory gene expression pattern. Infection and Immunity. 72(3): 1409-1422. 

  24. De Silva, K., Plain, K., Purdie, A., Begg, D., Whittington, R. (2018). Defining resilience to mycobacterial disease: Characteristics of survivors of ovine paratuberculosis. Veterinary Immunology and Immunopathology. 195: 56-64.

  25. Drury, R.E., O’Connor, D., Pollard, A.J. (2017). The clinical application of MicroRNAs in infectious disease. Frontiers in Immunology. 8: 1182. doi: 10.3389/fimmu.2017.01182.

  26. Dudemaine, P.L., Fecteau, G., Lessard, M., Labrecque, O., Roy, J.P., Bissonnette, N. (2014). Increased blood-circulating interferon-g, interleukin-17 and osteopontin levels in bovine paratuberculosis. Journal of Dairy Science. 97(6): 3382-3393. 

  27. Farrell, D., Shaughnessy, R.G., Britton, L., MacHugh, D.E., Markey, B., Gordon, S.V. (2015). The identification of circulating MiRNA in bovine serum and their potential as novel biomarkers of early Mycobacterium avium subsp paratuberculosis Infection. PloS One. 10(7).  DOI:10.1371/ journal.pone.0134310.

  28. Fernández, M., Benavides, J., Sevilla, I. A., Fuertes, M., Castaño, P., Delgado, L., Marín, J.F.G., Garrido, J.M., Ferreras, M.C., Pérez, V. (2014). Experimental infection of lambs with C and S-type strains of Mycobacterium avium subspecies paratuberculosis: Immunological and pathological findings. Veterinary Research. 45(1): 1-15. 

  29. Fiorentina, P., Martino, C., Mancini, Y., De Iorio, M.G., Williams, J.L., Minozzi, G. (2021). Using omics approaches in the discovery of biomarkers for early diagnosis of johne’s disease in sheep and goats. Animals/ : An Open Access Journal from MDPI. 11(7): 1912. https://doi.org/10.3390/ ani11071912 

  30. Fodstad, F. H., Gunnarsson, E. (1979). Post-mortem examination in the diagnosis of johne’s disease in goats. Acta Veterinaria Scandinavica. 20(2): 157-167. 

  31. Ganusov, V.V., Klinkenberg, D., Bakker, D., Koets, A.P. (2015). Evaluating contribution of the cellular and humoral immune responses to the control of shedding of Mycobacterium avium spp. paratuberculosis in cattle. Veterinary Research. 46(1). DOI: 10.1186/s13567-015-0204-1.

  32. Garvey, M. (2018). Mycobacterium avium subspecies paratuberculosis: A possible causative agent in human morbidity and risk to public health safety. Open Veterinary Journal. 8(2): 172-181.

  33. Hodgkinson, J.W., Ge, J.Q., Grayfer, L., Stafford, J., Belosevic, M. (2012). Analysis of the immune response in infections of the goldfish (Carassius auratus L.) with Mycobacterium marinum. Developmental and Comparative Immunology. 38(3): 456-465. 

  34. Hoentjen, F., Sartor, R.B., Ozaki, M., Jobin, C. (2005). STAT3 regulates NF-kappaB recruitment to the IL-12p40 promoter in dendritic cells. Blood. 105(2): 689-696. 

  35. Hussain, T., Shah, S.Z.A., Zhao, D., Sreevatsan, S., Zhou, X. (2016). The role of IL-10 in Mycobacterium avium subsp. paratuberculosis infection. Cell Communication and Signaling. 14(1): 1-14. 

  36. Kabara, E., Coussens, P.M. (2012). Infection of primary bovine macrophages with Mycobacterium avium subspecies paratuberculosis suppresses host cell apoptosis. Frontiers in Microbiology.  20(3): 215. doi: 10.3389/fmicb.2012.00215.

  37. Karcher, E.L., Beitz, D.C., Stabel, J.R. (2008). Modulation of cytokine gene expression and secretion during the periparturient period in dairy cows naturally infected with Mycobacterium avium subsp. paratuberculosis. Veterinary Immunology and Immunopathology. 123: 277-288. 

  38. Kawai, T., Akira, S. (2010). The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors. Nature Immunology. 11(5): 373-384. 

  39. Kawasaki, T., Kawai, T. (2014). Toll-like receptor signaling pathways. Frontiers in Immunology. 5: 461. doi: 10.3389/fimmu. 2014.00461. 

  40. Khalifeh, M.S., Stabel, J.R. (2004). Effects of gamma interferon, interleukin-10 and transforming growth factor beta on the survival of Mycobacterium avium subsp. paratuberculosis in monocyte-derived macrophages from naturally infected cattle. Infection and Immunity. 72: 1974-1982. 

  41. Kravitz, A., Pelzer, K., Sriranganathan, N. (2021). The Paratuberculosis paradigm examined: A review of host genetic resistance and innate immune fitness in Mycobacterium avium subsp. paratuberculosis infection. Frontiers in Veterinary Science. 8: 721706. doi: 10.3389/fvets.2021.721706. 

  42. Leite, F.L., Eslabão, L.B., Pesch, B., Bannantine, J.P., Reinhardt, T.A., Stabel, J.R. (2015). ZAP-70, CTLA-4 and proximal T cell receptor signaling in cows infected with Mycobacterium avium subsp. paratuberculosis. Veterinary Immunology and Immunopathology. 167: 15-21. 

  43. Li, L., Wagner, B., Freer, H., Schilling, M., Bannantine, J.P., Campo, J.J., Katani, R., Grohn, Y.T., Radzio-Basu, J., Kapur, V. (2017). Early detection of Mycobacterium avium subsp. paratuberculosis infection in cattle with multiplex-bead based immunoassays. PloS One. 12(12): e0189783. https:/ /doi.org/10.1371/journal.pone.0189783. 

  44. Lybeck, K.R., Storset, A.K., Olsen, I. (2009). Neutralization of interleukin-10 from CD14(+) monocytes enhances gamma interferon production in peripheral blood mononuclear cells from Mycobacterium avium subsp. paratuberculosis- infected goats. Clinical and Vaccine Immunology: CVI. 16: 1003-1011. 

  45. Machala, E.A., McSharry, B.P., Rouse, B.T., Abendroth, A., Slobedman, B. (2019). Gal power: the diverse roles of galectins in regulating viral infections. The Journal of General Virology. 100: 333-349. 

  46. Malvisi, M., Palazzo, F., Morandi, N., Lazzari, B., Williams, J.L., Pagnacco, G., Minozzi, G. (2016). Responses of bovine innate immunity to Mycobacterium avium subsp. paratuberculosis infection revealed by changes in gene expression and levels of MicroRNA. Plos One. 11: e0164461.  doi:10.1371/ journal.pone.0164461.

  47. Meng, Q.L., Liu, F., Yang, X.Y., Liu, X.M., Zhang, X., Zhang, C., Zhang, Z.D. (2014). Identification of latent tuberculosis infection-related microRNAs in human U937 macrophages  expressing Mycobacterium tuberculosis Hsp16.3. BMC Microbiology. 12(14): 37. doi: 10.1186/1471-2180-14-37. 

  48. Munjal, S.K., Tripathi, B.N., Paliwal, O.P. (2005). Progressive immunopathological changes during early stages of experimental infection of goats with Mycobacterium avium subspecies paratuberculosis. Veterinary Pathology. 42: 427-436. 

  49. Nalubamba, K., Smeed, J., Gossner, A., Watkins, C., Dalziel, R., Hopkins, J. (2008). Differential expression of pattern recognition receptors in the three pathological forms of sheep paratuberculosis. Microbes and Infection. 10: 598-604. 

  50. Naser, S. A., Ghobrial, G., Romero, C., Valentine, J. F. (2004). Culture of Mycobacterium avium subspecies paratuberculosis from the blood of patients with Crohn’s disease. Lancet. 364: 1039-1044. 

  51. Nielsen, S.S., Toft, N. (2008). Ante mortem diagnosis of paratuberculosis: A review of accuracies of ELISA, interferon-g assay and faecal culture techniques. Veterinary Microbiology.  129: 217-235.

  52. Nielsen, S.S., Toft, N., Okura, H. (2013). Dynamics of Specific Anti- Mycobacterium avium Subsp. paratuberculosis Antibody Response through Age. Plos One. 8: e63009. doi: 10.1371/ journal.pone.0063009.

  53. Palmer, M.V., Thacker, T.C., Waters, W.R. (2016a). Differential Cytokine gene expression in granulomas from lungs and lymph nodes of cattle experimentally infected with aerosolized Mycobacterium bovis. Plos One. 11(11): e0167471. doi: 10.1371/journal.pone.0167471.

  54. Palmer, M.V., Thacker, T.C., Waters, W.R. (2016b). Multinucleated giant cell cytokine expression in pulmonary granulomas of cattle experimentally infected with Mycobacterium bovis. Veterinary Immunology and Immunopathology. 180: 34-39. 

  55. Park, H.E., Park, H.T., Jung, Y.H., Yoo, H.S. (2018). Gene expression profiles of immune-regulatory genes in whole blood of cattle with a subclinical infection of Mycobacterium avium subsp. paratuberculosis. Plos One. 13(4): e0196502.  doi: 10.1371/journal.pone.0196502. 

  56. Plain, K.M., Begg, D.J., de Silva, K., Purdie, A.C., Whittington, R.J. (2012). Enhancement of the interferon gamma assay to detect paratuberculosis using interleukin-7 and interleukin- 12 potentiation. Veterinary Immunology and Immunopathology. 149: 28-37. 

  57. Plain, K.M., Purdie, A.C., Begg, D.J., de Silva, K., Whittington, R.J. (2010). Toll-like receptor (TLR)6 and TLR1 differentiation in gene expression studies of Johne’s disease. Veterinary Immunology and Immunopathology. 137: 142-148. 

  58. Purdie, A.C., Plain, K.M., Begg, D.J., de Silva, K., Whittington, R.J. (2012). Expression of genes associated with the antigen presentation and processing pathway are consistently regulated in early Mycobacterium avium subsp. paratuberculosis infection. Comparative Immunology, Microbiology and Infectious Diseases. 35: 151-162. 

  59. Reddacliff, L.A., McClure, S.J., Whittington, R.J. (2004). Immunoperoxidase studies of cell mediated immune effector cell populations in early Mycobacterium avium subsp. paratuberculosis infection in sheep. Veterinary Immunology and Immunopathology. 97: 149-162. 

  60. Roller, M., Hansen, S., Knauf-Witzens, T., Oelemann, W.M.R., Czerny, C.P., Abd El Wahed, A., Goethe, R. (2020). Mycobacterium avium Subspecies paratuberculosis infection in zoo animals: A review of susceptibility and disease process. Frontiers in Veterinary Science. 7. DOI: https://doi.org/10.3389/fvets.2020.572724.

  61. Sada-Ovalle, I., Chávez-Galán, L., Torre-Bouscoulet, L., Nava- Gamiño, L., Barrera, L., Jayaraman, P., Torres-Rojas, M., Salazar-Lezama, M.A., Behar, S.M. (2012). The Tim3- galectin 9 pathway induces antibacterial activity in human macrophages infected with Mycobacterium tuberculosis. Journal of Immunology (Baltimore, Md./ : 1950). 189: 5896-5902. 

  62. Sharma, S., Sundararajan, A., Suryawanshi, A., Kumar, N., Veiga- Parga, T., Kuchroo, V.K. and Rouse, B.T. (2011). T cell immunoglobulin and mucin protein-3 (Tim-3)/Galectin-9 interaction regulates influenza A virus-specific humoral and CD8 T-cell responses. Proceedings of the National Academy of Sciences. 108(47): 19001-19006.

  63. Sharma, S., Gautam, A., Singh, S.V., Chaubey, K.K., Mehta, R., Gupta, S., Sharma, M., Rose, M.K., Jain, V.K. (2020). Prevalence of Mycobacterium avium subspecies paratuberculosis (MAP) infection in suspected diarrhoeic buffaloes and cattle reporting at Veterinary University in India. Comparative Immunology, Microbiology and Infectious  Diseases. 73: 101533.  doi: 10.1016/j.cimid.2020.101533.

  64. Sharma, S., Chaubey, K.K., Singh, S.V., Gupta, S. (2022). Symbiotic microbiota: A class of potent immunomodulators. Science Asia. 48(6): 855-865.

  65. Sigurardóttir, Ó.G., Valheim, M., Press, C.M.L. (2004). Establishment of Mycobacterium avium subsp. paratuberculosis infection in the intestine of ruminants. Advanced Drug Delivery Reviews. 56: 819-834. 

  66. Smeed, J.A., Watkins, C.A., Rhind, S.M., Hopkins, J. (2007). Differential cytokine gene expression profiles in the three pathological forms of sheep paratuberculosis. BMC Veterinary Research. 3: 1-11. 

  67. Sohal, J.S., Singh, S.V., Tyagi, P., Subhodh, S., Singh, P.K., Singh, A.V., Narayanasamy, K., Sheoran, N., Singh, S.K. (2008). Immunology of mycobacterial infections: With special reference to Mycobacterium avium subspecies paratuberculosis. Immunobiology. 213: 585-598. 

  68. Sommer, S., Pudrith, C.B., Colvin, C.J. and Coussens, P.M. (2009) Mycobacterium avium subspecies paratuberculosis suppresses expression of IL-12p40 and iNOS genes induced by signalling through CD40 in bovine monocyte- derived macrophages. Veterinary immunology and immunopathology. 128(1-3): 44-52.

  69. Stabel, J.R. (2000). Transitions in immune responses to Mycobacterium paratuberculosis. Veterinary Microbiology. 77: 465-473. 

  70. Stabel, J.R. and Whitlock, R.H. (2001). An evaluation of a modified interferon-gamma assay for the detection of paratuberculosis in dairy herds. Veterinary Immunology and Immunopathology. 79: 69-81. 

  71. Sweeney, R.W. (2011). Pathogenesis of paratuberculosis. Veterinary Clinics: Food Animal Practice. 27(3): 537-546.

  72. Thacker, T.C., Palmer, M.V., Waters, W.R. (2007). Associations between cytokine gene expression and pathology in Mycobacterium bovis infected cattle. Veterinary Immunology  and Immunopathology. 119: 204-213.

  73. Thirunavukkarasu, S., de Silva, K., Whittington, R.J. and Plain, K.M. (2013). In vivo and in vitro expression pattern of Toll-like receptors in Mycobacterium avium subspecies paratuberculosis infection. Veterinary immunology and immunopathology. 156(1-2): 20-31.

  74. Tjärnlund, A., Rodríguez, A., Cardona, P.J., Guirado, E., Ivanyi, J., Singh, M., Fernández, C. (2006). Polymeric IgR knockout mice are more susceptible to mycobacterial infections in the respiratory tract than wild-type mice. International Immunology. 18(5): 807-816.

  75. Tooker, B.C., Burton, J.L., Coussens, P.M. (2002). Survival tactics of M. paratuberculosis in bovine macrophage cells. Veterinary Immunology and Immunopathology. 87: 429- 437. 

  76. Umemura, M., Yahagi, A., Hamada, S., Begum, M.D., Watanabe, H., Kawakami, K., Suda, T., Sudo, K., Nakae, S., Iwakura, Y., Matsuzaki, G. (2007). IL-17-mediated regulation of innate and acquired immune response against pulmonary Mycobacterium bovis bacille Calmette-Guerin infection. Journal of Immunology (Baltimore, Md./ : 1950). 178: 3786-3796. 

  77. Whitlock, R.H., Buergelt, C. (1996). Preclinical and clinical manifestations of paratuberculosis (including pathology). The Veterinary Clinics of North America. Food Animal Practice. 12: 345- 356. 

  78. Whittington, R.J., Begg, D.J., de Silva, K., Plain, K.M., Purdie, A.C. (2012). Comparative immunological and microbiological aspects of paratuberculosis as a model mycobacterial infection. Veterinary Immunology and Immunopathology. 148: 29-47. 

  79. Widdison, S., Schreuder, L.J., Villarreal-Ramos, B., Howard, C.J., Watson, M., Coffey, T.J. (2006). Cytokine expression profiles of bovine lymph nodes: effects of Mycobacterium bovis infection and bacille Calmette-Guérin vaccination. Clinical and Experimental Immunology. 144: 281-289. 

  80. Witchell, J., Maddipatla, S.V.P.K., Wangoo, A., Vordermeier, M., Goyal, M. (2010). Time dependent expression of cytokines in Mycobacterium bovis infected cattle lymph nodes. Veterinary Immunology and Immunopathology. 138: 79-84.

Editorial Board

View all (0)