Open access peer-reviewed chapter

Common Strategies, Different Mechanisms to Infect the Host: Anaplasma and Mycobacterium

Written By

Vladimir López, Pilar Alberdi and José de la Fuente

Submitted: 19 June 2017 Reviewed: 06 October 2017 Published: 26 September 2018

DOI: 10.5772/intechopen.71535

From the Edited Volume

Tuberculosis

Edited by Jean-Marie Ntumba Kayembe

Chapter metrics overview

1,055 Chapter Downloads

View Full Metrics

Abstract

Intracellular bacteria such as Anaplasma spp. and Mycobacterium spp. pose a risk to human and animal populations worldwide. The main function of immune response cells is to eliminate invading pathogens. However, pathogens can deregulate host cell function and turn defense cells into suitable hosts. Intracellular bacterial have a smaller genome, compared to the host cell, thus requiring efficient mechanisms for survival and persistence within the host by inducing sustained changes in cell function and immune response. Bacterial epigenetic regulation of host cell gene transcription appears to be a general mechanism that enhances pathogen survival while altering host cell function and facilitating infection. Anaplasma phagocytophilum leads to modified host cell gene transcription and phenotype by epigenetically altering host chromatin. Mycobacterial infection of human cells also results in host gene silencing using a mechanism that involves HDAC complex formation and histone deacetylation. Membrane proteins are essential for cell invasion in both pathogens, and can regulate and protect the pathogen against the host response. Understanding the mechanisms employed by these bacteria to infect the host could contribute to develop effective interventions for the control of tuberculosis and anaplasmosis. This review focuses on the common strategies employed by two zoonotic pathogens, Anaplasma and Mycobacterium spp., highlighting also the different mechanisms used to infect host cells.

Keywords

  • Anaplasma
  • Mycobacteria
  • immunology
  • infection
  • tuberculosis

1. Introduction

Ticks and tick-borne diseases represent a growing problem for human and animal health worldwide whereas tuberculosis continues to be a global burden in both human and animal populations [1, 2]. Pathogenic organisms have evolved host mimicking properties and manipulate host responses for their own survival and propagation. To successfully establish and maintain a bacterial infection, the pathogens subvert the host cells defense response to survive, proliferate, and persist within the infected cell. To evade host defense systems, bacterial pathogens produce a variety of virulence factors that stimulate bacterial adherence and invasion and subvert host cell signaling cascades that regulate intracellular microbial survival and trafficking. Some of these mechanisms are mediated by factors released by the bacteria, whereas others rely on hijacking host components to prevent the production of an effective immune response thus promoting their survival within the host cell [2, 3]. Intracellular bacteria from Anaplasma and Mycobacterium genera produce similar genes expression patterns in infected ruminants [4]. Pathogen and host-specific differences could contribute to disease diagnosis and treatment of tuberculosis and anaplasmosis in ruminants.

In this review, we provide an overview of some of the mechanisms employed by Anaplasma and Mycobacterium to infect the host cell and the impact on their pathogenesis.

Advertisement

2. Anaplasma phagocytophilum, an intracellular bacterium with unusual tropism

The emergence of tick-borne pathogens has been promoted by the exploitation of environmental resources and the increase in human outdoor activities, allowing the contact with tick vectors normally present in the field [5]. Anaplasma phagocytophilum is an obligate intracellular rickettsial pathogen transmitted mainly by Ixodes spp. ticks causing human granulocytic anaplasmosis (HGA), equine, and canine granulocytic anaplasmosis, and tick-borne fever (TBF) in ruminants [6]. In the vertebrate host, A. phagocytophilum infects neutrophils where the pathogen multiplies within a parasitophorous vacuole or morula in the cytoplasm of tick and vertebrate host cells [7, 8]. These gram-negative bacteria are grouped within the family Anaplasmataceae [3]. Complications and fatality are rare but more common in the elderly, the immunocompromised, or if proper diagnosis and/or antibiotic therapy are delayed. Fatalities are usually not directly attributed to the infection itself; pathological findings suggest defects in host defense and the presence of secondary infections [9]. However, the severity of illness and fatality rates could also be due to underlying immunosuppression.

Anaplasma is a highly antigenically variant bacterial pathogen that displays a diversity of mechanisms to create the structural and antigenic variation necessary to escape the immune response and allows long-term persistence in the host thus being able to act as a reservoir for transmission. A. phagocytophilum strategies to infect vertebrate host cells include, among others, remodeling of the cytoskeleton, inhibition of cell apoptosis, manipulation of the immune response and modification of cell epigenetics and metabolism [10]. Hosts respond to infection by activating alternative pathways to regulate cell apoptosis, immunity, metabolism and stress response mediated by heat-shock proteins (Hsps) [1]. Unlike other bacteria, A. phagocytophilum is aflagellated and does not have a type III secretion system (T3SS) [11, 12].

Pathogens subvert cellular immune response to favor infection and multiplication. Host cell transcriptome and proteome studies have demonstrated an effect of A. phagocytophilum infection on the inhibition of cell innate immunity [13, 14, 15]. They employ a variety of mechanisms to create the structural and antigenic variation needed to subvert the host immune system and long-term persistence [3]. A. phagocytophilum also employs a type IV secretion system (T4SS) to deliver proteins or DNA into eukaryotic cells [16]. It also inhibits host cell apoptosis to allow the bacteria sufficient time to develop morulae [17].

Adaptation to a life in eukaryotic cells and transmission between hosts has been assisted by the deletion of many genes that are present in the genomes of free-living bacteria, including genes required for the biosynthesis of lipopolysaccharide and peptidoglycan that are involved in the activation of host leukocytes [18].

P44 (also known as MSP2) is a highly variable immunodominant surface protein that facilitates adherence to granulocytes [19]. The genome of Anaplasma consists of more than 100 msp2(p44) paralogs [20]. Antibodies specific to P44 inhibit A. phagocytophilum infection in mice and HL-60 cells, which suggests that antigenic variation of P44 proteins may help A. phagocytophilum to escape host immune surveillance [3]. Some Anaplasma strains are naturally persistent in lambs and can be used to analyze the mechanisms of persistence in the vertebrate host. Variation of the outer membrane protein MSP2(P44) is believed to play a key role in persistence of the organism [21].

A. phagocytophilum can avoid killing by innate immunity but it also induces some innate immune responses, such as the production of IFN-γ, that contribute to tissue injury and disease [22]. Signal transducer and activator of transcription 1 (Stat1) is important in host innate and adaptive immune responses to intracellular pathogens, including intracellular bacteria [23]. It mediates most of the biological functions of both type I interferon (IFNα/β) and type II IFN (IFNγ). A. phagocytophilum infection-induced IFNγ signaling leads to phosphorylation of Stat1 in mice and is critical for the generation of protection [24]. Experimental infections with mice have demonstrated that the absence of Stat1 converts the subclinical infection to a severe one [22] suggesting that Stat1 plays an important role in controlling the response to bacterial infections. Stat1 also participates in the IFN-γ signaling of mycobacterial immunity. IFN-γ signaling provides positive feedback to both macrophages and CD4+ T-cells, which amplifies the Th1 response [25]. Suppressor of cytokine signaling (SOCS) expression has been implicated in intracellular survival of A. phagocytophilum in neutrophils where expression of IFN-γ receptor alpha-chain CD119 is diminished leading to reduced Stat1 dimerization and signaling [26].

In neutrophils, the genes most downregulated in response to A. phagocytophilum infection include those coding for proteins involved in bacterial killing such as myeloperoxidase, transferrin, bactericidal/permeability-increasing protein and cell protection (mucin 12). Immune-system-related genes encoding interferons, cytokines, chemokines, and their receptors are upregulated in response to infection [13, 14, 15]. This suggests that pathogens have developed mechanisms to subvert the innate immune protective mechanisms in vertebrate hosts. However, some species can activate innate immune protective mechanisms to control infection and appear to play a minor role as reservoir hosts for the pathogen [27]. For instance, pigs naturally and experimentally infected with A. phagocytophilum control bacterial infection through activation of innate immune responses, phagocytosis, and autophagy [28] resulting in low infection levels or infection clearance.

A. phagocytophilum, a pathogen lacking the T3SS and flagellin, activates the NLRC4 inflammasome (a component of the innate immune system) and secretion of IL-1β [29]. IL-18 release mediated by the NLRC4 inflammasome regulates IFN-γ production by CD4+ T cells upon A. phagocytophilum infection [30]. The receptor-interacting serine/threonine-protein kinase 2 (RIPK2) appears to be a major regulator of the immune response against A. phagocytophilum. Ripk2−/− immune cells exhibit a defect in activation for the nuclear factor (NF)-κB and the NLRC4 inflammasome pathways [29]. Furthermore, experimental mice lacking COX2 (cyclooxygenase 2) are more susceptible to A. phagocytophilum, they do not secrete IL-18 and exhibit splenomegaly and damage to the splenic architecture [29].

A. phagocytophilum transiently infects bone-marrow derived macrophages (BMDMs) [31] and clinical features in animal models and infected patients suggest classical macrophage activation [32]. Deep sequencing analysis of experimentally infected macrophages indicated that the transcription of genes that encode for phospholipase A2 (pla2g12a, pla2g5 and pla2g2e), COX2 and PGE synthase (ptges) was increased upon A. phagocytophilum infection [29].

A. phagocytophilum use heat shock proteins (Hsps) for infection of vertebrate host cells [33, 34]. Host cells can also activate Hsps in response to infection [35, 36, 37]. The mammalian immune response against pathogen Hsps to control infection may trigger a detrimental autoimmune response to host Hsps [35, 36, 38]. However, recent evidence suggests that hosts may benefit from induction of Hsps in response to pathogen infection [1]. A mutant strain of Mycobacterium tuberculosis, that constitutively over produced Hsp70 proteins, was fully virulent in the initial stage of infection, but its survival was reduced in the chronic phase. This suggests that induction of microbial genes encoding Hsps might provide a novel strategy to boost the immune response of individuals with latent infections [39].

How A. phagocytophilum interacts with the mammalian immune system is still unclear. Both T and B cells have been shown to play important roles in the control and clearance of A. phagocytophilum [40, 41]. CD4+ T cells and T-helper 1 (Th1) play a key role in the immune response to the infection of A. phagocytophilum [30, 42]. IFNγ, IL-12, and IL-18 also play important roles in the early clearance of A. phagocytophilum [30, 43]. Well-known anti-bacterial innate immune detection system such as TLR2, TLR4, and their adaptor MyD88 appear to play no role in the immune response to A. phagocytophilum infection [41]. Some studies suggest that signaling through the Nod Like Receptor (NLR) family member IPAF (NLRC4), its adaptor ASC, and Caspase-1 is critical for the control of A. phagocytophilum infection during the early phase of infection [30].

Rip2 has been previously shown to play an essential role in the immunity against various intracellular pathogens including Mycobacterium tuberculosis [44]. Rip2 also plays an important role in the control of A. phagocytophilum infection [44]. A. phagocytophilum infection upregulates Rip2, the adaptor molecule of the cytoplasmic pattern recognition receptor Nod1 and 2 in immune cells [45]. Following peptidoglycan detection, Nod1/Nod2 recruit and associate with the adaptor protein Rip2, triggering proinflammatory signaling pathways via NF-κB and the mitogen-activated protein (MAP) kinases p38, JNK, and ERK [46]. IL-8, a major inflammatory chemokine, is heavily induced during A. phagocytophilum infection in humans [47]. Trafficking of neutrophils to the sites of infection is induced by this chemokine and Rip2, which appears to play an important role in neutrophil recruitment in vivo [48]. IFN-γ, another inflammatory cytokine, plays a major role in the immune pathology and early clearance of A. phagocytophilum infection [43]. It is also known that an adaptive CD4+ T cell mediated response is critical for the complete clearance of A. phagocytophilum infection [42]. Previous reports have shown the importance of natural killer (NK) cells, NKT cells [49] and CD4 + T cells [42] in the IFNγ production and host defense to A. phagocytophilum infection (Figure 1).

Figure 1.

A. phagocytophilum and M. tuberculosis employ common strategies but different mechanisms to infect host cells: Nod proteins activate NF-κB through the serine–threonine kinase Rip2. NF-κB is translocated to the nucleus and stimulates cytokine expression. Secretion of IL-8 stimulates neutrophil and T cell migration. Stat1 participates in the IFN-γ signaling of intracellular bacteria. (A). Neutrophil infected with A. phagocytophilum: IFN-γ contributes to tissue injury and disease. AnkA is secreted by the bacteria and translocates to the nucleus of infected cells regulating host cell transcription facilitating intracellular bacterial survival and growth. (B). Macrophage infected with M. tuberculosis: IFN-γ activates macrophage phagocytosis killing intracellular bacteria. TNF-α attracts macrophages and lymphocytes at the site of infection and promotes granuloma formation thus resulting in limited antigen presentation.

Using oligonucleotide array technology [50], it was observed that genes involved in the immune response were modulated in neutrophils infected with A. phagocytophilum. Among the genes that were most upregulated in the early transcriptional response to infection in neutrophils were cytokines, chemokines, and their receptors (e.g., CCL3, CCL3L3, IL-8, IL-1β, and CXCR4).

The major adipocyte lipid droplet-associated phosphoprotein perilipin (PLIN) is upregulated in HL60 infected cells. Both protein and mRNA levels were higher in infected cells and the over expression of PLIN was parallel with bacterial infection levels [51]. Furthermore, PLIN knockdown resulted in a reduction of A. phagocytophilum infection in HL60 cells, suggesting the bacteria modulate host lipid metabolism to infect and multiply in the host cell [51].

In THP-1 cells, A, phagocytophilum infection displays an upregulation of histone deacetylates 1 and 2 (HDAC1 and HDAC2), while protein levels exhibit a similar kinetic pattern for both HDACs. Moreover, pharmacological inhibition of HDAC and HDAC1 silencing reduced the level of bacterial infection in THP-1 cells [52]. Mycobacterial infection of THP-1 cells specifically inhibits HLA-DR gene expression by a pathway involving HDAC complex formation at the HLA-DR promoter, resulting in histone deacetylation and gene silencing [53].

Proteins secreted by bacteria are involved in many important tasks and they account for many of the virulence factors of pathogens. Outer membrane protein A (OmpA), also known as peptidoglycan-associated lipoprotein, is conserved among most Gram-negative bacteria and interacts with peptidoglycan to maintain outer membrane integrity [54]. The expression of OmpA increases in the early stages of infection. OmpA is presented on the pathogen’s surface and is upregulated during invasion of HL-60 cells. Sera from HGA patients and experimentally infected mice recognize recombinant OmpA. Pretreatment of A. phagocytophilum organisms with OmpA antiserum reduces their ability to infect HL-60 cells [54].

A. phagocytophilum uses surface proteins invasins OmpA, Asp14, and AipA to bind and infect mammalian hosts [55, 56]. OmpA interacts with α1,3-fucose, which is critical for the bacteria to bind host cell surfaces and invade them. OmpA, Asp14, and AipA play essential roles in the A. phagocytophilum lifecycle [54, 55, 57]. Directing the immune response to their binding domains could enhance protective efficacy. It has been observed that an antibody cocktail specific for the OmpA, Asp14, and AipA binding domains blocked A. phagocytophilum infection of host cells [56]. This finding could help the development of an anti-multi-invasin vaccine to protect against human and veterinary granulocytic anaplasmosis or even against other obligate intracellular pathogens such as Mycobacterium spp. since they also use multiple invasins to enter host cells [58].

MSP1a and MSP1b from Anaplasma marginale have been shown to be adhesins for host cells [59]. Studies on the immunogenicity of recombinant BCG expressing the MSP1a antigen suggested that the immune responses were influenced by the level of antigen expression [60]. These results indicated that recombinant M. bovis BCG expressing MSP1a could be used to test for protective antibody production for the control of anaplasmosis.

The A. phagocytophilum genome encodes a type four secretion system (T4SS) that may facilitate intracellular survival by translocation of virulence factors that appear to be important for the manipulation of the host cell. Ankirin A (AnkA) is a translocated virulence factor that is tyrosine-phosphorylated by host cell kinases upon translocation into the host cell early during infection [16, 61].

Anaplasma translocation substrate 1 (Ats-1) protein belongs to T4SS of Anaplasma, which is secreted and localizes into the mitochondria of human neutrophils and HL60 cells. Ats-1 translocates to the host cell mitochondria matrix via the translocase of the outer mitochondrial membrane (TOM) complex. Transfection assays with RF/6A and yeast cells demonstrated that Ats-1 inhibits etoposide and Bax-induced apoptosis respectively [62] thus facilitating pathogen survival.

In mammalian cells, A. phagocytophilum activates extracellular signal-regulated kinase (Erk)1/2, a key protein of the MAP kinase pathway [50, 63]. AptA (Anaplasma phagocytophilum toxin A, formerly named APH_0233) stimulates Erk1/2 phosphorylation in HL60 and HEK293 cells [64]. Furthermore, AptA interacts with vimentin, and gene silencing and inhibitory enzymatic assays in HL60 cells and neutrophils respectively, demonstrated that vimentin is necessary for Erk1/2 activation and Anaplasma infection [64].

Anaplasma inclusions have a double-lipid bilayer membrane, and induce autophagosome formation in the host cell. Also, beclin 1 and light chain 3 (LC3) proteins that play a central role in autophagy are colocalized bacterial replicative inclusions. Furthermore, assays of inhibition and induction of this catabolic mechanism in HL60 infected cells demonstrated that autophagy benefits infection, rather than elimination [65]. Ref. [66] described that induction of autophagy in host cells is mediated trough beclin 1 (Becn1) that binds Ats-1 to supply nutrients for pathogen growth. Additionally, gene silencing of Becn1 inhibits infection in mammalian cells [66].

Advertisement

3. The Mycobacterium tuberculosis complex, a global burden for human and animal health

Macrophages play a central role in the first line of defense against pathogenic microorganisms, however, they are also the key target cells for mycobacteria. The bacteria can live and replicate inside the macrophages, thus evading the innate immune response against infection of the host through immunosuppression and immune evasion [53]. Co-evolution of M. tuberculosis with its hosts has enabled the pathogen to develop host immune evasion strategies that interfere with both innate and adaptive immunity. These include the manipulation of their phagosome within host macrophages, the avoidance of pattern recognition receptors, the modulation of host cytokine production, and the manipulation of antigen presentation to prevent or alter the quality of T-cell responses [67]. Other mechanisms include interference with phagosomal acidification and trafficking, blocking autophagy and apoptosis-mediated killing, perturbing calcium signaling, and inhibiting the inflammasome activation in order to modulate the host immune responses. Manipulation of these host pathways is achieved by bacterial components such as cell wall lipids, serine threonine kinases, phosphatases and proteases, and using specialized secretion systems.

The outcome of infection with Mycobacterium tuberculosis depends on the ability of the immune response to clear or contain the infection. When this fails, the bacterium replicates, disseminates within the host, and elicits a pathologic inflammatory response. Individuals infected with M. tuberculosis develop mainly CD4 T cell responses to protein components of M. tuberculosis, an immune response that can persist for years. Infections with mycobacteria are characterized by their chronic course and, even with an adequate immune response, they can persist inside macrophages. Progression to active disease is possible even decades after exposure [68] and is typically triggered by immune compromise. Although the bacteria are concealed within the infected macrophage, B cells and antibodies also play a role on the immune response to intracellular bacteria and are likely to be important in the control of M. tuberculosis [69]. In addition, B cells are a major cellular component of the granuloma (an important mechanism of host defense against tuberculosis) where they can process and present antigen to T cells, secrete antibodies, and modulate inflammation through the production of IL-10 [70]. In vitro human B cells have been shown to ingest mycobacteria, produce IgM, and upregulate the expression of the costimulatory molecules CD80 and CD86 and the chemokine CXCL10 [71]. The human CD4 T cell response exhibits Th1-response characteristics [72].

Mycobacteria have a distinct secretion system, named type VII (T7SS or ESX), which is associated with virulence and pathogenesis, including growth in macrophages [73] and antigen presentation. This system is encoded by a locus that is deleted in attenuated strains of M. bovis (bacille Calmette–Guérin (BCG) strains), which are used to vaccinate against tuberculosis [74].

IL-8 is a chemokine with a significant role in regulating leukocyte influx in TB. In vivo studies have shown that pre-treatment with anti-IL-8 alone inhibits mycobacterial granuloma formation [75]. IL-8 is involved in attracting neutrophils and T cells and in monocyte recruitment [76]. Targeting IL-8 secretion during inflammation could be the subject for new therapeutic approaches [77].

Mycobacterial components can activate MAP kinase signaling cascades but this activation varies depending on the species of Mycobacterium. For instance, it appears to be diminished in macrophages infected with pathogenic strains of M. avium [78].

IFN-γ is a Th1 cytokine that plays a vital role in the protective immune response against M. tuberculosis infection [2]. In cattle, IFN-γ is produced predominately by activated CD4 T cells following presentation of M. bovis antigens on the surface of antigen presenting cells (APCs) [79]. IL-10 is released following phagocytosis of pathogenic mycobacteria [80] and has been shown to inhibit the pro-inflammatory cytokine response through down regulation of IL-12 and IFN-γ [81, 82]. Increased IL-10 levels appear to correlate with progression of infection in a bovine tuberculosis model [83] (Figure 1).

Nod proteins and their adaptor molecule Rip2 are key components of a family of cytosolic innate immune pattern recognition receptors [84]. Nod2 triggers cytokine production by dendritic cells in response to live M. tuberculosis, but is not essential to control infection [85].

M. tuberculosis can use the TLR2 pathway to modify the host environment [86]. The adaptor molecule myeloid differentiation factor-88 (MyD88) appears to play a significant role in the pathogenesis of Mycobacterium. Mice lacking MyD88 are highly susceptible to M. tuberculosis infection, with a mean time to death of approximately 42 days.

Glycolipids are one of the most common cell surface components of macrophages and dendritic cells. They interact with intracellular bacteria, estimulating the host immune response [87]. LprG (Rv1411c), a cell membrane lipoprotein essential for M. tuberculosis virulence, binds to the acyl groups of lipoglycan [88]. In murine macrophage cells (RAW 264.7), LprG is essential for macrophage entry and inhibition of phagosome—lysosome fusion. Also, it has been described that LprG has a significant role in the production of lipoglycan lipoarabinomannan (LAM), one of the major cell surface components of M. tuberculosis [87, 89]. LprI is another lipoprotein used by M. tuberculosis to bind and inhibit the lytic activity of lysosomes [90].

Mycolic acids are major components of the outer membrane of M. tuberculosis. HadC (Rv0637) contributes to mycolic acids biosynthesis and its mutation or silencing is directly related to the loss of M. tuberculosis virulence [91].

M. tuberculosis encodes the serine protease Rv2224c (Hip1) that is present on the cellular membrane [92]. In primary macrophages, silencing of Hip1 notably decreased mycobacterial growth compared to the wild type bacteria. Moreover, levels of cytokines (TNF-α, IL-1β, IL-6) were increased in macrophages infected with wild-type M. tuberculosis compared to the mutant Hip1 M. tuberculosis [93]. The stress-induced protein GroEL2 is a substrate for Hip1 [92]. Hip1 appears to limits dendritic cells cytokine secretion and through under modulation of CD40 and CD86, it could affect dendritic cell maturation, and decrease antigen presentation to CD4 T cells [94].

Transcriptional assays have shown that M. tuberculosis infection of human monocytes activate the MAPK pathway to promote over expression of IL-23, that is involved in the modulation of Th1/Th17 cells [95]. In addition, it has been reported that the bacteria may suppress the differentiation of monocytes into dendritic cells through the release of IL-10 [96].

Intracellular bacteria can manipulate host gene expression through epigenetic modifications to help infection and survival inside the host cell. Ghorpade et al. [97] described that M. bovis bacillus Calmette-Guérin (BCG) modify epigenetically nitric oxide and KLF4 to restrain the class II transactivator (CIITA) and MHC-II expression thereby eluding immune surveillance. Evidence supports that M. tuberculosis infection in THP1 cells induces overexpression of HDAC1, which is implicated in the downregulation of IL-12B that plays a key role in the Th1 response [98].

Lsr2 is a M. tuberculosis protein with histone-like features, including the ability to regulate a variety of transcriptional responses in mycobacteria. Lsr2 protects mycobacteria against reactive oxygen intermediates (ROI) in vitro and during macrophage infection shielding bacterial DNA by binding to it [99] suggesting it could be a good candidate as a drug target.

Advertisement

4. Conclusions

Intracellular bacteria such as Anaplasma and Mycobacterium use similar mechanisms to infect vertebrate host cells. These strategies include manipulation of the immune response, subversion of phagocyte cells and the use of proteins for infection and manipulation of host gene expression. Nevertheless, different pathogens have evolved specific strategies when infecting their hosts. Abundantly expressed proteins are often the primary targets of research, however, less prominently expressed antigens may have equally good or even superior vaccine potential. Research into the antigen catalog available for immune recognition of infected cells could provide new directions for antigen discovery and vaccine development.

Advertisement

Acknowledgments

VL was funded by a grant of the Universidad Autónoma de Sinaloa, Mexico.

References

  1. 1. de la Fuente J, Villar M, Cabezas-Cruz A, Estrada-Peña A, Ayllón N, Alberdi P. Tick-host-pathogen interactions: Conflict and cooperation. PLoS Pathogens. 2016;12(4):e1005488. DOI: 10.1371/journal.ppat.1005488
  2. 2. Scriba TJ, Coussens AK, Fletcher HA. Human immunology of tuberculosis. Microbiology Spectrum. 2016;4(4):1-23
  3. 3. Rikihisa Y. Anaplasma phagocytophilum and Ehrlichia chaffeensis: Subversive manipulators of host cells. Nature Reviews. Microbiology. 2010;8(5):328-339 Available from: http://dx.doi.org/10.1038/nrmicro2318%5Cnhttp://www.nature.com/doifinder/10.1038/nrmicro2318
  4. 4. Galindo RC, Ayllón N, Carta T, Vicente J, Kocan KM, Gortazar C, et al. Characterization of pathogen-specific expression of host immune response genes in Anaplasma and Mycobacterium species infected ruminants. Comparative Immunology, Microbiology and Infectious Diseases. 2010;33(6):e133-e142
  5. 5. Jongejan F, Uilenberg G. The global importance of ticks. Parasitology. 2005;129(7):S3. Available from: http://www.journals.cambridge.org/abstract_S0031182004005967; [cited 2014 Jul 11]
  6. 6. de la Fuente J. Overview: Ticks as vectors of pathogens that cause disease in humans and animals. Frontiers in Bioscience. 2008;13:6938. Available from: http://www.bioscience.org/2008/v13/af/3200/list.htm
  7. 7. Ayllón N, Villar M, Galindo RC, Kocan KM, Šíma R, López JA, et al. Systems biology of tissue-specific response to Anaplasma phagocytophilum reveals differentiated apoptosis in the tick vector Ixodes scapularis. PLoS Genetics. 2015;11(3):1-29
  8. 8. Severo MS, Pedra JHF, Ayllón N, Kocan KM, de la Fuente J. Anaplasma. Molecular Medical Microbiology. 2015;3:2033-2042. Available from: http://linkinghub.elsevier.com/retrieve/pii/B9780123971692001104
  9. 9. Dumler JS, Bakken JS. Erhlichial diseases of humans: Emerging tick-borne infections. Clinical Infectious Diseases. 1995;20:1102-1110
  10. 10. de la Fuente J, Estrada-Peña A, Cabezas-Cruz A, Kocan KM. Anaplasma phagocytophilum uses common strategies for infection of ticks and vertebrate hosts. Trends in Microbiology. 2016;24(3):173-180. Available from: http://dx.doi.org/10.1016/j.tim.2015.12.001
  11. 11. Severo MS, Stephens KD, Kotsyfakis M, Pedra JHF. Anaplasma phagocytophilum: Deceptively simple or simply deceptive? Future Microbiology. 2012;7:719-731
  12. 12. Rikihisa Y, Lin M. Anaplasma phagocytophilum and Ehrlichia chaffeensis type IV secretion and Ank proteins. Current Opinion in Microbiology. 2010 Feb;13(1):59-66. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3251840&tool=pmcentrez&rendertype=abstract; [cited 2014 Jul 23]
  13. 13. de la Fuente J, Ayoubi P, Blouin EF, Almazán C, Naranjo V, Kocan KM. Gene expression profiling of human promyelocytic cells in response to infection with Anaplasma phagocytophilum. Cellular Microbiology. 2005 Apr;7(4):549-559. Available from http://www.ncbi.nlm.nih.gov/pubmed/15760455; [cited 2014 Jul 23]
  14. 14. Lee HC, Kioi M, Han J, Puri RK, Goodman JL. Anaplasma phagocytophilum-induced gene expression in both human neutrophils and HL-60 cells. Genomics. 2008 Sep;92(3):144-151. Available from http://www.ncbi.nlm.nih.gov/pubmed/18603403; [cited 2014 Jul 23]
  15. 15. Pruneau L, Moumène A, Meyer DF, Marcelino I, Lefrançois T, VachiÃcry N. Understanding Anaplasmataceae pathogenesis using “Omics”; approaches. Frontiers in Cellular and Infection Microbiology. 2014;4(July):1-7. Available from: http://journal.frontiersin.org/article/10.3389/fcimb.2014.00086/abstract
  16. 16. IJdo JW, Carlson AC, Kennedy EL. Anaplasma phagocytophilum AnkA is tyrosine-phosphorylated at EPIYA motifs and recruits SHP-1 during early infection. Cellular Microbiology. 2007 May;9(5):1284-1296. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17250594; [cited 2014 Jul 23]
  17. 17. Yoshiie K, Kim HY, Mott J, Rikihisa Y. Intracellular infection by the human granulocytic Ehrlichiosis agent inhibits human neutrophil apoptosis. Infection and Immunity. 2000;68(3):1125-1133. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=97257&tool=pmcentrez&rendertype=abstract
  18. 18. Gulia-Nuss M, Nuss AB, Meyer JM, Sonenshine DE, Roe RM, Waterhouse RM, et al. Genomic insights into the Ixodes scapularis tick vector of Lyme disease. Nature Communications. 2016;7(May 2015):10507. Available from: http://www.nature.com/doifinder/10.1038/ncomms10507
  19. 19. Park J, Choi KS, Dumler JS. Major surface protein 2 of Anaplasma phagocytophilum facilitates adherence to granulocytes. Infection and Immunity. 2003;71(7):4018-4025
  20. 20. Nelson CM, Herron MJ, Felsheim RF, Schloeder BR, Grindle SM, Chavez AO, et al. Whole genome transcription profiling of Anaplasma phagocytophilum in human and tick host cells by tiling array analysis. BMC Genomics. 2008;16:1-16
  21. 21. Granquist EG, Stuen S, Crosby L, Lundgren AM, Alleman AR, Barbet AF. Variant-specific and diminishing immune responses towards the highly variable MSP2(P44) outer membrane protein of Anaplasma phagocytophilum during persistent infection in lambs. Veterinary Immunology and Immunopathology. 2010;133(15):2-4
  22. 22. Choi K, Scorpio DG, Dumler JS. Stat1 negatively regulates immune-mediated injury with Anaplasma phagocytophilum infection. Journal of Immunology. 2014;15(10):5088-5098
  23. 23. Rauch I, Müller M, Decker T. The regulation of inflammation by interferons and their STATs. JAK-STAT. 2013;2(1):e23820. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24058799%5Cnhttp://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=PMC3670275
  24. 24. Choi K, Dumler JS. Anaplasma phagocytophilum, interferon gamma production and Stat1 signaling. Microbiology and Immunology. 2013;57(3):207-212
  25. 25. Qu HQ, Fisher-Hoch SP, McCormick JB. Molecular immunity to mycobacteria: Knowledge from the mutation and phenotype spectrum analysis of Mendelian susceptibility to mycobacterial diseases. International Journal of Infectious Diseases. 2011;15(5):1-19
  26. 26. Bussmeyer U, Sarkar A, Broszat K, Lüdemann T, Möller S, van Zandbergen G, et al. Impairment of gamma interferon signaling in human neutrophils infected with Anaplasma phagocytophilum. Infection and Immunity. 2010 Jan;78(1):358-363. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2798180&tool=pmcentrez&rendertype=abstract; [cited 2014 Jul 23]
  27. 27. de la Fuente J, Gortazar C. Wild boars as hosts of human-pathogenic Anaplasma phagocytophilum variants [letter]. Emerging Infectious Diseases 2012;18(12):2094-2095
  28. 28. Galindo RC, Ayllón N, Smrdel KS, Boadella M, Beltrán-Beck B, Mazariegos M, et al. Gene expression profile suggests that pigs (Sus Scrofa) are susceptible to Anaplasma phagocytophilum but control infection. Parasites & Vectors. 2012 Jan;5:181. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3453518&tool=pmcentrez&rendertype=abstract; [cited 2014 Jul 23]
  29. 29. Wang X, Shaw DK, Hammond HL, Sutterwala FS, Rayamajhi M, Shirey KA, et al. The prostaglandin E2-EP3 receptor axis regulates Anaplasma phagocytophilum-mediated NLRC4 inflammasome activation. PLoS Pathogens. 2016;12(8):1-23
  30. 30. Pedra JH, Sutterwala FS, Sukumaran B, Ogura Y, Qian F, Montgomery RR, et al. ASC/PYCARD and caspase-1 regulate the IL-18/IFN-gamma axis during Anaplasma phagocytophilum infection. Journal of Immunology. 2007;179(7):4783-4791. Available from: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=17878377%5Cnhttp://www.jimmunol.org/cgi/reprint/179/7/4783.pdf
  31. 31. Chen G, Wang X, Severo MS, Sakhon OS, Sohail M, Brown LJ, et al. The tick salivary protein sialostatin L2 inhibits caspase-1-mediated inflammation during Anaplasma phagocytophilum infection. Infection and Immunity. 2014;82(6):2553-2564
  32. 32. Dumler JS, Barat NC, Barat CE, Bakken JS. Human granulocytic anaplasmosis and macrophage activation. Clinical Infectious Diseases. 2007 Jul 15;45(2):199-204. Available from http://www.ncbi.nlm.nih.gov/pubmed/17578779; [cited 2014 Jul 23]
  33. 33. Villar M, Ayllón N, Kocan KM, Bonzón-Kulichenko E, Alberdi P, Blouin EF, et al. Identification and characterization of Anaplasma phagocytophilum proteins involved in infection of the tick vector, Ixodes scapularis. PLoS One. 2015;10(9):1-26
  34. 34. Contreras M, Alberdi P, Mateos-Hernández L, Fernández de Mera IG, García-Pérez AL, Vancová M, et al. Anaplasma phagocytophilum MSP4 and HSP70 proteins are involved in interactions with host cells during pathogen infection. Frontiers in Cellular and Infection Microbiology. 2017;7(July):307. Available from: http://www.ncbi.nlm.nih.gov/pubmed/28725639%0Ahttp://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=PMC5496961
  35. 35. Schlesinger MJ. Heat shock proteins: Minireview. The Journal of Biological Chemistry. 1990;265(21 (July 25)):12111-12114
  36. 36. Whitley D, Goldberg SP, Jordan WD. Heat shock proteins: A review of the molecular chaperones. Journal of Vascular Surgery. 1999;29(4):748-751. Available from: http://www.sciencedirect.com/science/article/pii/S0741521499703290
  37. 37. Yu A, Li P, Tang T, Wang J, Chen Y, Liu L. Roles of Hsp70s in stress responses of microorganisms, plants, and animals. BioMed Research International. 2015;2015:510319. DOI: 10.1155/2015/510319
  38. 38. Vabulas RM, Wagner H, Schild H. Heat shock proteins as ligands of toll-like receptors. In: Beutler B, Wagner H, editors. Toll-like Receptor Family Members and their Ligands. Berlin, Heidelberg: Springer Berlin Heidelberg; 2002. pp. 169-184. Available from: https://doi.org/10.1007/978-3-642-59430-4_11
  39. 39. Stewart GR, Snewin VA, Walzl G, Hussell T, Tormay P, O’Gaora P, et al. Overexpression of heat-shock proteins reduces survival of Mycobacterium tuberculosis in the chronic phase of infection. Nature. 2001;7:732-737
  40. 40. Telford SR 3rd, Dawson JE, Katavolos P, Warner CK, Kolbert CP, Persing DH. Perpetuation of the agent of human granulocytic ehrlichiosis in a deer tick-rodent cycle. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(12):6209-6214
  41. 41. von Loewenich FD, Scorpio DG, Reischl U, Dumler JS, Bogdan C. Frontline: Control of Anaplasma phagocytophilum, an obligate intracellular pathogen, in the absence of inducible nitric oxide synthase, phagocyte NADPH oxidase, tumor necrosis factor, toll-like receptor (TLR)2 and TLR4, or the TLR adaptor molecule MyD88. European Journal of Immunology. 2004 Jul;34(7):1789-1797. Available from http://www.ncbi.nlm.nih.gov/pubmed/15214027; [cited 2014 Jul 23]
  42. 42. Birkner K, Steiner B, Rinkler C, Kern Y, Aichele P, Bogdan C, et al. The elimination of Anaplasma phagocytophilum requires CD4+ T cells, but is independent of Th1 cytokines and a wide spectrum of effector mechanisms. European Journal of Immunology. 2008;38(12):3395-3410
  43. 43. Akkoyunlu M, Fikrig E. Gamma interferon dominates the murine cytokine response to the agent of human granulocytic ehrlichiosis and helps to control the degree of early rickettsemia. Infection and Immunity. 2000;68(4):1827-1833
  44. 44. Sukumaran B, Ogura Y, Pedra JHF, Kobayashi K, Flavell RA, Fikrig E. Rip2 contributes to host defense against Anaplasma phagocytophilum infection. FEMS Immunology and Medical Microbiology. 2012;66(22):211-219
  45. 45. Sukumaran B, Carlyon JA, Cai J, Berliner N, Fikrig E. Early transcriptional response of human neutrophils to Anaplasma phagocytophilum infection. Infection and Immunity. 2005;73(12):8089-8099
  46. 46. Kobayashi K, Inohara N, Hernandez LD, Galán JE, Núñez G, Janeway CA, et al. RICK/Rip2/CARDIAK mediates signalling for receptors of the innate and adaptive immune systems. Nature. 2002;416(6877):194-199. Available from: http://www.nature.com/doifinder/10.1038/416194a
  47. 47. Akkoyunlu M, Malawista SE, Anguita J, Fikrig E. Exploitation of interleukin-8-induced neutrophil chemotaxis by the agent of human granulocytic ehrlichiosis. Infection and Immunity. 2001;69(9):5577-5588
  48. 48. Magalhaes JG, Lee J, Geddes K, Rubino S, Philpott DJ, Girardin SE. Essential role of Rip2 in the modulation of innate and adaptive immunity triggered by Nod1 and Nod2 ligands. European Journal of Immunology. 2011;41(5):1445-1455
  49. 49. Choi K, Webb T, Oelke M, Scorpio DG, Dumler JS. Differential innate immune cell activation and Proinflammatory response in Anaplasma phagocytophilum infection. Infection and Immunity. 2007;75(6):3124-3130
  50. 50. Lee HC, Kioi M, Han J, Puri RK, Goodman JL. Anaplasma phagocytophilum-induced gene expression in both human neutrophils and HL-60 cells. Genomics. 2008;92(3):144-151
  51. 51. Manzano-Roman R, Almazán C, Naranjo V, Blouin EF, Kocan KM, de la Fuente J. Expression of perilipin in human promyelocytic cells in response to Anaplasma phagocytophilum infection results in modified lipid metabolism. Journal of Medical Microbiology. 2008 Feb;57(Pt 2):159-163. Available from: http://www.ncbi.nlm.nih.gov/pubmed/18201980; [cited 2014 Jul 23]
  52. 52. Garcia-Garcia JC, Barat NC, Trembley SJ, Dumler JS. Epigenetic silencing of host cell defense genes enhances intracellular survival of the rickettsial pathogen Anaplasma phagocytophilum. PLoS Pathogens. 2009 Jun;5(6):e1000488. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2694362&tool=pmcentrez&rendertype=abstract; [cited 2014 Jul 23]
  53. 53. Wang X, Zhang J, Liang J, Zhang Y, Teng X, Yuan X, et al. Protection against Mycobacterium tuberculosis infection offered by a new multistage subunit vaccine correlates with increased number of IFN-γ+IL-2+ CD4+and IFN-γ+ CD8+ T cells. PLoS One. 2015;10(3):1-18
  54. 54. Ojogun N, Kahlon A, Ragland SA, Troese MJ, Mastronunzio JE, Walker NJ, et al. Anaplasma phagocytophilum outer membrane protein A interacts with sialylated glycoproteins to promote infection of mammalian host cells. Infection and Immunity. 2012;80(11):3748-3760
  55. 55. Kahlon A, Ojogun N, Ragland SA, Seidman D, Troese MJ, Ottens AK, et al. Anaplasma phagocytophilum Asp14 is an invasin that interacts with mammalian host cells via its C terminus to facilitate infection. Infection and Immunity. 2013;81(1):65-79
  56. 56. Seidman D, Hebert KS, Truchan HK, Miller DP, Tegels BK, Marconi RT, et al. Essential domains of Anaplasma phagocytophilum invasins utilized to infect mammalian host cells. PLoS Pathogens. 2015;11(2):1-29. Available from: http://dx.doi.org/10.1371/journal.ppat.1004669
  57. 57. Seidman D, Ojogun N, Walker NJ, Mastronunzio J, Kahlon A, Hebert KS, et al. Anaplasma phagocytophilum surface protein AipA mediates invasion of mammalian host cells. Cellular Microbiology. 2014;16(8):1133-1145
  58. 58. Govender VS, Ramsugit S, Pillay M. Mycobacterium tuberculosis adhesins: Potential biomarkers as anti-tuberculosis therapeutic and diagnostic targets. Microbiology (United Kingdom). 2014;160(Part 9):1821-1831
  59. 59. de La Fuente J, Garcia-Garcia JC, Blouin EF, Rodríguez SD, García MA, Kocan KM. Evolution and function of tandem repeats in the major surface protein 1a of the ehrlichial pathogen Anaplasma marginale. Animal Health Research Reviews. 2001;2(2):163-173
  60. 60. Michelon A, Conceição FR, Binsfeld PC, da Cunha CW, Moreira ÂN, Argondizzo AP, et al. Immunogenicity of Mycobacterium bovis BCG expressing Anaplasma marginale MSP1a antigen. Vaccine. 2006;24(37-39):6332-6339
  61. 61. Lin M, den Dulk-Ras A, Hooykaas PJJ, Rikihisa Y. Anaplasma phagocytophilum AnkA secreted by type IV secretion system is tyrosine phosphorylated by Abl-1 to facilitate infection. Cellular Microbiology. 2007 Nov;9(11):2644-2657. Available from http://www.ncbi.nlm.nih.gov/pubmed/17587335
  62. 62. Niu H, Kozjak-Pavlovic V, Rudel T, Rikihisa Y. Anaplasma phagocytophilum Ats-1 is imported into host cell mitochondria and interferes with apoptosis induction. PLoS Pathogens. 2010 Feb;6(2):e1000774. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=2824752&tool=pmcentrez&rendertype=abstract; [cited 2014 Jul 23]
  63. 63. Xiong Q, Lin M, Rikihisa Y. Cholesterol-dependent Anaplasma phagocytophilum exploits the low-density lipoprotein uptake pathway. PLoS Pathogens. 2009;5(3):1-13
  64. 64. Sukumaran B, Mastronunzio JE, Narasimhan S, Fankhauser S, Uchil PD, Levy R, et al. Anaplasma phagocytophilum AptA modulates Erk1/2 signalling. Cellular Microbiology. 2011;13(1):47-61
  65. 65. Niu H, Yamaguchi M, Rikihisa Y. Subversion of cellular autophagy by Anaplasma phagocytophilum. Cellular Microbiology. 2008 Mar;10(3):593-605. Available from: http://www.ncbi.nlm.nih.gov/pubmed/17979984; [cited 2014 Jul 23]
  66. 66. Niu H, Xiong Q, Yamamoto A, Hayashi-Nishino M, Rikihisa Y. Autophagosomes induced by a bacterial Beclin 1 binding protein facilitate obligatory intracellular infection. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(51):20800-20807. Available from: http://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=3529060&tool=pmcentrez&rendertype=abstract
  67. 67. Goldberg MF, Saini NK, Porcelli SA. Evasion of innate and adaptive immunity by Mycobacterium tuberculosis. Microbiology Spectrum. May 2017;2014:1-24
  68. 68. Wiker HG, Mustafa T, Bjune Ga, Harboe M. Evidence for waning of latency in a cohort study of tuberculosis. BMC Infectious Diseases. 2010;10:37
  69. 69. Chan J, Mehta S, Bharrhan S, Chen Y, Achkar JM, Casadevall A, et al. The role of B cells and humoral immunity in Mycobacterium tuberculosis infection. Seminars in Immunology. 2014;26(6):588-600. Available from: http://dx.doi.org/10.1016/j.smim.2014.10.005
  70. 70. Rao M, Valentini D, Poiret T, Dodoo E, Parida S, Zumla A, et al. B in tb: B cells as mediators of clinically relevant immune responses in tuberculosis. Clinical Infectious Diseases. 2015;61(Suppl 3):S225-S234
  71. 71. Hoff ST, Salman AM, Ruhwald M, Ravn P, Brock I, Elsheikh N, et al. Human B cells produce chemokine CXCL10 in the presence of Mycobacterium tuberculosis specific T cells. Tuberculosis. 2015;95(1):40-47. Available from: http://dx.doi.org/10.1016/j.tube.2014.10.005
  72. 72. Lindestam Arlehamn CS, Gerasimova A, Mele F, Henderson R, Swann J, Greenbaum JA, et al. Memory T cells in latent Mycobacterium tuberculosis infection are directed against three antigenic islands and largely contained in a CXCR3+CCR6+ Th1 subset. PLoS Pathogens. 2013;9(1):e1003130. DOI: 10.1371/journal.ppat.1003130
  73. 73. Stanley SA, Raghavan S, Hwang WW, Cox JS. Acute infection and macrophage subversion by Mycobacterium tuberculosis require a specialized secretion system. Proceedings of the National Academy of Sciences of the United States of America. 2003;100(22):13001-13006. Available from: http://www.pnas.org/cgi/doi/10.1073/pnas.2235593100
  74. 74. Sreejit G, Ahmed A, Parveen N, Jha V, Valluri VL, Ghosh S, et al. The ESAT-6 protein of Mycobacterium tuberculosis interacts with beta-2-microglobulin (β2M) affecting antigen presentation function of macrophage. PLoS Pathogens. 2014;10(10):e1004446. DOI: 10.1371/journal.ppat.1004446
  75. 75. Larsen CG, Thomsen MK, Gesser B, Thomsen PD, Deleuran BW, Nowak J, et al. The delayed-type hypersensitivity reaction is dependent on IL-8. Inhibition of a tuberculin skin reaction by an anti-IL-8 monoclonal antibody. Journal of Immunology. 1995;155(4):2151-2157. Available from: http://www.jimmunol.org/content/155/4/2151.abstract
  76. 76. Mukaida N, Harada A, Matsushima K. Interleukin-8 (IL-8) and monocyte chemotactic and activating factor (MCAF/MCP-1), chemokines essentially involved in inflammatory and immune reactions. Cytokine & Growth Factor Reviews. 1998;9(1):9-23
  77. 77. Yang X, Corvalan JRF, Wang P, Roy CM, Davis CG. Fully human anti-interleukin-8 monoclonal antibodies: potential therapeutics for the treatment of inflammatory disease states. Journal of Leukocyte Biology. 1999;66(September):401-410
  78. 78. Roach SK, Schorey JS. Differential regulation of the mitogen-activated protein kinases by pathogenic and nonpathogenic mycobacteria. Infection and Immunity. 2002;70(6):3040-3052
  79. 79. Liébana E, Girvin RM, Welsh M, Neill D, Pollock JM, Neill SD. Generation of CD8 + T-cell responses to Mycobacterium bovis and mycobacterial antigen in experimental bovine tuberculosis. Infection and Immunity. 1999;67(3):1034-1044
  80. 80. Shaw TC, Thomas LH, Friedland JS. Regulation of Il-10 secretion after phagocytosis of Mycobacterium tuberculosis by human monocytic cells. Cytokine. 2000;12(5):483-486. Available from: http://linkinghub.elsevier.com/retrieve/pii/S1043466699905860
  81. 81. Gong J, Zhang M, Modlin RL, Linsley PS, Iyer D, Lin Y, et al. Interleukin-10 downregulates Mycobacterium tuberculosis induced Th1 responses and CTLA-4 expression. Infection and Immunity. 1996;64(3):913-918
  82. 82. Fulton Sa, Cross JV, Toossi ZT, Boom WH. Regulation of interleukin-12 by interleukin-10, transforming growth factor-beta, tumor necrosis factor-alpha, and interferon-gamma in human monocytes infected with Mycobacterium tuberculosis H37Ra. The Journal of Infectious Diseases. 1998;178(4):1105-1114
  83. 83. Widdison S, Schreuder LJ, Villarreal-Ramos B, Howard CJ, Watson M, Coffey TJ. Cytokine expression profiles of bovine lymph nodes: Effects of Mycobacterium bovis infection and bacille Calmette-Guérin vaccination. Clinical and Experimental Immunology. 2007;75(11):5127-5134
  84. 84. Chin AI, Dempsey PW, Bruhn K, Miller JF, Xu Y, Cheng G. Involvement of receptor-interacting protein 2 in innate and adaptive immune responses. Nature. 2002;416(6877):190-194. Available from: http://www.nature.com/doifinder/10.1038/416190a
  85. 85. Gandotra S, Jang S, Murray PJ, Salgame P, Ehrt S. Nucleotide-binding oligomerization domain protein 2-deficient mice control infection with Mycobacterium tuberculosis. Infection and Immunity. 2007;75(11):5127-5134
  86. 86. Scanga CA, Bafica A, Feng CG, Allen W, Hieny S, Sher A, et al. MyD88-deficient mice display a profound loss in resistance to Mycobacterium tuberculosis associated with partially impaired Th1 cytokine and nitric oxide synthase 2 expression. Infection and Immunity. 2004;72(4):2400-2404
  87. 87. Källenius G, Correia-Neves M, Buteme H, Hamasur B, Svenson SB. Lipoarabinomannan, and its related glycolipids, induce divergent and opposing immune responses to Mycobacterium tuberculosis depending on structural diversity and experimental variations. Tuberculosis. 2016;96:120-130
  88. 88. Drage MG, Tsai H-C, Pecora ND, Cheng T-Y, Arida AR, Shukla S, et al. Mycobacterium tuberculosis lipoprotein LprG (Rv1411c) binds triacylated glycolipid agonists of toll-like receptor 2. Nature Structural & Molecular Biology. 2010;17(99):1088-1095. Available from: http://www.nature.com/doifinder/10.1038/nsmb.1869
  89. 89. Gaur RL, Ren K, Blumenthal A, Bhamidi S, Gibbs S, Jackson M, et al. LPRG-mediated surface expression of lipoarabinomannan is essential for virulence of Mycobacterium tuberculosis. PLoS Pathogens. 2014;10(9):e1004376. DOI: 10.1371/journal.ppat.1004376
  90. 90. Sethi D, Mahajan S, Singh C, Lama A, Hade MD, Gupta P, et al. Lipoprotein LprI of Mycobacterium tuberculosis acts as a lysozyme inhibitor. The Journal of Biological Chemistry. 2016;291(6):2938-2953
  91. 91. Slama N, Jamet S, Frigui W, Pawlik A, Bottai D, Laval F, et al. The changes in mycolic acid structures caused by hadC mutation have a dramatic effect on the virulence of Mycobacterium tuberculosis. Molecular Microbiology. 2016;99(4):794-807
  92. 92. Naffin-Olivos JL, Georgieva M, Goldfarb N, Madan-Lala R, Dong L, Bizzell E, et al. Mycobacterium tuberculosis Hip1 modulates macrophage responses through proteolysis of GroEL2. PLoS Pathogens. 2014;10(5):e1004132. DOI: 10.1371/journal.ppat.1004132
  93. 93. Rengarajan J, Murphy E, Park A, Krone CL, Hett EC, Bloom BR, et al. Mycobacterium tuberculosis Rv2224c modulates innate immune responses. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(1):264-269
  94. 94. Madan-Lala R, Sia JK, King R, Adekambi T, Monin L, Khader SA, et al. Mycobacterium tuberculosis impairs dendritic cell functions through the serine hydrolase Hip1. Journal of Immunology. 2014;192(9):4263-4272. Available from: http://www.jimmunol.org/cgi/doi/10.4049/jimmunol.1303185
  95. 95. Elena G, Elena RME, Scandurra M, Gafa V, Pardini M, et al. Expression of proinflammatory and regulatory cytokines via NF-κB and MAPK-dependent and IFN regulatory factor-3-independent mechanisms in human primary monocytes infected by Mycobacterium tuberculosis. Clinical & Developmental Immunology. 2011;2011:841346. DOI: 10.1155/2011/841346
  96. 96. Remoli ME, Giacomini E, Petruccioli E, Gafa V, Severa M, Gagliardi MC, et al. Bystander inhibition of dendritic cell differentiation by Mycobacterium tuberculosis -induced IL-10. Immunology and Cell Biology. 2011;89(3):437-446 Available from: http://www.ncbi.nlm.nih.gov/pubmed/20805841
  97. 97. Ghorpade DS, Holla S, Sinha AY, Alagesan SK, Balaji KN. Nitric oxide and KLF4 protein epigenetically modify class II transactivator to repress major histocompatibility complex II expression during Mycobacterium bovis Bacillus Calmette-Guerin infection. The Journal of Biological Chemistry. 2013;288(28):20592-20606
  98. 98. Chandran A, Antony C, Jose L, Mundayoor S, Natarajan K, Kumar RA. Mycobacterium tuberculosis infection induces HDAC1-mediated suppression of IL-12B gene expression in macrophages. Frontiers in Cellular and Infection Microbiology. 2015;5(December):1-14. Available from: http://journal.frontiersin.org/Article/10.3389/fcimb.2015.00090/abstract
  99. 99. Colangeli R, Haq A, Arcus VL, Summers E, Magliozzo RS, McBride A, et al. The multifunctional histone-like protein Lsr2 protects mycobacteria against reactive oxygen intermediates. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(11):4414-4418

Written By

Vladimir López, Pilar Alberdi and José de la Fuente

Submitted: 19 June 2017 Reviewed: 06 October 2017 Published: 26 September 2018