Summary
The humanized mouse model has been developed as a model to identify and characterize human immune responses to human pathogens and has been used to better identify vaccine candidates. In the current studies, the humanized mouse was used to determine the ability of a vaccine to affect the immune response to infection with Mycobacterium tuberculosis. Both human CD4+ and CD8+ T cells responded to infection in humanized mice as a result of infection. In humanized mice vaccinated with either BCG or with CpG‐C, a liposome‐based formulation containing the M. tuberculosis antigen ESAT‐6, both CD4 and CD8 T cells secreted cytokines that are known to be required for induction of protective immunity. In comparison to the C57BL/6 mouse model and Hartley guinea pig model of tuberculosis, data obtained from humanized mice complemented the data observed in the former models and provided further evidence that a vaccine can induce a human T‐cell response. Humanized mice provide a crucial pre‐clinical platform for evaluating human T‐cell immune responses in vaccine development against M. tuberculosis.
Keywords: cytokines, rodent, T cell, tuberculosis, vaccination
Introduction
Mice with reconstituted human immune system components (humanized mice) are increasingly being explored for efficient and cost‐effective evaluation of disease pathogenesis, therapeutic responses in vivo, and for the development of new vaccines.1 Humanized mice are better suited to study infection with human‐restricted pathogens and the immune responses against them than other small animal models such as mice and rats, which have been traditionally used as hosts to study disease pathogenesis.2, 3 Accordingly several human‐trophic viruses including human immunodeficiency virus type‐1, Epstein–Barr virus, Kaposi's sarcoma‐associated herpesvirus, herpes simplex virus, human cytomegalovirus, hepatitis C virus and Dengue virus, as well as bacterial pathogens such as Mycobacterium tuberculosis and Salmonella typhi, have been studied in humanized mice primarily to investigate disease pathogenesis and the innate and adaptive immune responses that these pathogens evoke post infection.4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 Additionally, a variety of antigens and human vaccine preparations have been shown to induce innate and adaptive T‐cell‐mediated immune responses in humanized mice, including dinitrophenyl, keyhole limpet haemocyanin antigen, toxic shock syndrome toxin 1, tetanus toxoid, DTaP vaccine and hepatitis B virus vaccine.21, 22, 23 Therefore, vaccine development that aims to primarily evaluate antigen plus adjuvant candidates for improved T‐cell immunity can be advanced in humanized mice.
The immunopathogenesis of disease caused by M. tuberculosis has been examined in the humanized mouse model and has gained interest because the model may provide a better understanding of the infectious process and characterization of the response of the human immune system. These studies, one using Mycobacterium bovis bacillus Calmette–Guérin (BCG) and M. tuberculosis Harlingen,24 and the other using M. tuberculosis H37Rv,25 demonstrated human T cells within granulomas. Another study using BCG suggested that the T cells were anergic to M. tuberculosis antigens, but responded to mitogen stimulation.26 Interestingly, infection in all these studies resulted in relatively high numbers of mycobacteria in multiple organs regardless of the virulence of the strain. It is not clear why these mice have difficulty in limiting infection, despite the presence of effector immune cells. None of these previous studies examined the effect of vaccination on infection with M. tuberculosis.
C57BL/6 mice are used to test vaccine candidate immunogenicity and protective immunity, during early vaccine development, whereas Hartley guinea pigs provide information about a vaccine candidate to prevent disease.27 These commonly used models have provided valuable information about tuberculosis immunopathogenesis, but shortcomings have been reported in their ability to model all aspects of tuberculosis in humans.28
The objective of the current studies was to assess the lymphocyte response to vaccination against M. tuberculosis in the humanized mouse using the standard BCG vaccine and a vaccine that contained CpG‐C as a molecular adjuvant and determine if vaccination of the humanized mouse could induce a cytokine response predictive of what is seen in humans. A comparison was made with the humanized mouse model and two commonly used models for testing novel tuberculosis vaccines, the C57BL/6 mouse model and the Hartley guinea pig model.
Materials and methods
Generation of humanized mice
Humanized mice were constructed and validated as per standard operating procedures by HuMurine Technologies (La Verne, CA). Briefly, human CD34+ hematopoietic progenitor cells (HPCs) were isolated and enriched by using a commercially available kit (Miltenyi Biotech, San Diego, CA) as per the manufacturer's instructions. Isolated cells were cryopreserved in freezing media containing DMSO and then stored in liquid nitrogen. Newborn NOG pups (NOD.Cg‐PrkdcscidIl2rgtm1sug/JicTac; Taconic Biosciences, Hudson, NY) were irradiated using a γ irradiator within 96 hr of birth with one dose of 100 cGy and immediately after were injected intra‐hepatically with 1 × 105–5 × 105 thawed HPCs in 50 μl of PBS. All engrafted mice were bled at 12 weeks post‐engraftment and peripheral blood was analysed for human leucocyte reconstitution by assessment of the ratio of human CD45+ to mouse CD45+ cells. Mice with human CD45 levels > 30% in the peripheral blood were selected for experiments.
Animals
Pathogen‐free, female, 6‐ to 8‐week‐old C57BL/6 mice (Jackson Laboratory, Bar Harbor, ME) and out‐bred Hartley guinea pigs, weighing 450–500 g (Charles River Laboratory, Wilmington, MA) were maintained in the Animal Biosafety Level 3 facility at Colorado State University with sterile chow and water ad libitum. The pathogen‐free nature of the mice was determined by routine screening of sentinel mice. Colorado State University Animal Care and Use Committee approved all experimental procedures.
Reagents
1,2‐Dioleoyl‐3‐trimethylammonium‐propane (chloride salt) (DOTAP; Avanti Polar Lipid Inc, Alabaster, AL) was used to prepare liposomes for the vaccine formulation. The Toll‐like receptor 9 agonist, CpG‐C DNA (ODN 2395), Human/Mouse (Hycult Biotech Inc., Plymouth Meeting, PA) was also used for vaccine formulation. Recombinant ESAT‐6 (6 kDa early secretory antigenic target of M. tuberculosis) was obtained from BEI Resources (Manassas, MD).
Mycobacteria and infection
Mycobacterium tuberculosis H37Rv (TMCC#102) was grown as a pellicle on Proskauer and Beck (P&B) medium then passaged three times in P&B medium containing 0·05% Tween‐80 to mid‐log phase and vials of working stocks were frozen at −80° until used. Mycobacterium bovis BCG Pasteur (TMCC#1011) was grown in P&B medium with 0·01% Tween‐80 to mid‐log phase. Aliquots were stored at −80° and thawed before use. Mice were infected with virulent M. tuberculosis H37Rv through the aerosol route using the Middlebrook Aerosol Exposure Chamber (Glas‐Col, Terre Haute, IN) using the standard exposure protocol to deliver approximately 100 CFU of bacilli per mouse.29 Guinea pigs were exposed through the respiratory route to 10–20 CFU of virulent M. tuberculosis H37Rv using a Madison Aerosol Chamber (Madison, WI).29 Post‐infection, guinea pigs were monitored daily for body temperature, health conditions and weighed weekly until euthanasia was required due to disease progression. Humane end‐point criteria, specified and approved by the Institutional Animal Care and Use Committee, were applied to determine the time of euthanasia.
Determination of colony‐forming units
Lungs from infected mice were homogenized in sterile saline and plated in 10‐fold serial dilutions onto 7H11 agar to determine the colony‐forming units (CFU). Plates were incubated at 37° for 21 days, after which colonies were counted. In mice vaccinated with BCG and then infected with M. tuberculosis, organ homogenates were also plated on 7H11 agar supplemented with thiophene‐2‐carboxylic acid hydrazide to inhibit the growth of BCG. When compared with organ homogenates plated on 7H11 agar, there were no differences in the CFU, indicating that only M. tuberculosis was being detected.
Vaccination protocols
Mice were inoculated subcutaneously with 5 × 104 CFU M. bovis BCG and guinea pigs intradermally with 103 CFU M. bovis BCG. For intranasal inoculations, a formulation consisting of 1 mm DOTAP, liposome, 20 μg CpG‐C ODN and 2 μg recombinant ESAT‐6 protein in a total volume of 0·02 ml was administered three times at 2‐week intervals.
Primary cell isolation and flow cytometry
Single‐cell suspensions from lungs and spleens of mice were obtained as previously described.30 For surface staining, cells were resuspended to 107 cells/ml in FACS buffer and incubated with primary antibody at the manufacturer's recommended concentration. Monoclonal antibodies, conjugated to their respective fluorochromes were purchased from BD Biosciences (San Jose, CA) and eBioscience (San Diego, CA). For surface phenotype characterization, cells were incubated with CD3‐phycoerythrin (PE) (Clone: HIT3a), CD4‐FITC (Clone: SK3), CD8‐V500 (Clone: RPT‐T8), CD45RA‐PE (Clone: HI100), CD45RO‐allophycocyanin (APC) (Clone: UCLH1). For intracellular cytokine staining interferon‐γ (IFN‐γ) ‐APC PE‐Cyanine7 (Clone: 4S.B3), interleukin‐2 (IL‐2) ‐APC (Clone: MQ1‐17H12), tumour necrosis factor‐α (TNF‐α) ‐FITC (Clone: MAb11), Granzyme B‐V450 (Clone: GB11), granulysin‐Alexa Fluor® 488 (Clone: RB1) and perforin‐FITC (Clone: dG9) were used. Cells were analysed on a FACSCanto II flow cytometer (BD Biosciences). Data were analysed using flowjo software (TreeStar, Ashland, OR). Analysis and presentation of distributions of CD4+ and CD8+ T cells for cytokine expression were performed using spice version 5·1, downloaded from http://exon.niaid.nih.gov.31
ELISpot assay
To determine the frequency of human granzyme B secreting cells, splenocytes from infected mice were cultured at 5 × 105/ml in RPMI‐1640 containing 10% fetal bovine serum (Atlas Biologicals, Fort Collins, CO) and penicillin and streptomycin with antigen for 24 hr at 37° in 5% CO2. The human granzyme B ELISpot assay (BD Biosciences) was used according to the manufacturer's protocol. For IFN‐γ and IL‐2 in the C57BL/6 mouse model, splenocytes were isolated as described above and the number of spot‐forming units was detected using the IFN‐ γ and IL‐2 ELISpot assays (eBioscience) according to the manufacturer's protocol. The number of spot‐forming cells was determined using a Series 5 UV‐Immunospot Analyzer (Cellular Technology, Ltd., Shaker Heights, OH).
Analysis of data
The log10‐transformed data were used to determine significant differences between groups. Data were assessed for normal distribution before analysis. For analysis between multiple groups a one‐way analysis of variance with a Bonferroni t‐test was used for all pairwise multiple comparisons.
Results
Development and validation of the humanized immune system mouse
HuMurine's humanized NOG (Hu‐M™) mice are individually constructed by injecting pre‐qualified human CD34+ HPCs into newborn non‐obese diabetic NOD/SCID/γ c R−/− (NOG) mice resulting in stable multi‐lineage human haematopoiesis at 12–32 weeks post engraftment (Fig. 1a). The Hu‐M™ mouse develops and sustains humanization levels of > 50%, as determined by the ratio of human lymphocytes to murine leucocytes in the peripheral blood at 12 and 32 weeks’ post engraftment. These mice also display human T cells (CD3+) and B cells (CD19+) (Fig. 1a), as well as lower levels of human natural killer cells (CD56+) and macrophages (CD14+) (data not shown) in the peripheral blood and can sustain enhanced differentiation and maturation of human CD4+ (T helper type 1 cells), CD8+ T cytotoxic T cells for up to 32 weeks post engraftment (Fig. 1b). These mice can generate CD4+ and CD8+ naive (CD45RA+) and memory (CD45RO+) T cells, both of which are thought to be major drivers of immunity against tuberculosis through the production of effector molecules such as IFN‐γ, TNF‐α and interleukin IL‐2 (Fig. 1c).32, 33 Given the emerging role of the B‐cell‐mediated immune response against tuberculosis, we also established the presence of various B‐cell subsets in these mice including immature (CD38+), memory (CD27+), plasma (CD138+) and IgM+ cells (Fig. 1d).34 As the Hu‐M™ mouse supports the development and maturation of the immune cells implicated to play an important role in immunity against tuberculosis, we decided to implement this platform to understand the immunological parameters that determine long‐term protective immunity against tuberculosis.
Figure 1.

Profile of human lymphoid cell reconstitution in peripheral blood and spleen of the Hu‐M™. At 12 and 31–32 weeks, post‐engraftment, peripheral blood (PB) was collected, processed and examined for human CD45, CD3, CD4, CD8 and CD19 expression (a and b). All events were first gated on human CD45 expression and subsequently examined for T‐ and B‐cell‐specific markers (n = 11). Representative dot plot for the expression of (c) human T‐cell subsets including CD4, CD8, CD45RA and CD45RO (d) and human B‐cell subsets including CD27, CD38, CD138 and IgM in the spleen of Hu‐M™ mice at 25 weeks’ post engraftment.
Cellular phenotype analysis after low‐dose aerosol infection with M. tuberculosis
To determine the human T‐cell response to infection, we examined the distribution of CD45RA (naive) and CD45RO (memory) expressing CD4+ and CD8+ T cells in the lungs and spleens of Hu‐M™ mice at days 7, 22 and 34 post‐infection. The phenotype of these cells was identified based on the gating strategy outlined in Fig. 2(a). At day 0 post‐infection the lungs of naive mice contained a significantly greater number of human CD4+ CD45RA− CD45RO+ T cells compared with CD4+ CD45RA+ CD45RO− T cells, suggesting the presence of activated memory T cells before infection (Fig. 2b). No difference was observed between cell phenotypes at days 7 and 22 post‐infection until day 34, at which point there was a significant increase in CD4+ CD45RA− CD45RO+ T cells, coupled with a decline in CD4+ CD45RA+ CD45RO− T cells (Fig. 2b). In the spleen, the number of CD4+ CD45RA+ CD45RO− and CD4+ CD45RA− CD45RO+ T cells declined from the onset of infection until day 22, after which at day 34 there was a significantly greater number of activated CD4+ CD45RA− CD45RO+ T cells. The response profile observed with the human CD8+ T cells reflected the CD4+ T‐cell response observed in the lungs and spleens (Fig. 2b), suggesting a multi‐subpopulation T‐cell response including higher levels of effector and memory cells in Hu‐M™ mice infected with M. tuberculosis.
Figure 2.

Human CD4+ and CD8+ T‐cell phenotypes in the lungs and spleen of Hu‐M™ mice after infection with a low‐dose aerosol of Mycobacterium tuberculosis H37Rv at 7, 22 and 34 days of infection. (a) Cells were analysed, initially by gating for the lymphocyte population using forward scatter (FSC) versus side scatter (SSC) parameters, then gating for CD3+ T cells and then for the CD4+ or CD8+ subsets, and then for phenotypic expression of CD45RA and CD45RO (representative gating strategy). (b) The lungs and spleens from infected animals were excised and examined for the presence of naive (CD45RA) and effector (CD45RO) T cells by flow cytometry after pulmonary infection. ***P<0·001.
Post‐infection cytokine expression
Cytokines such as IFN‐γ, IL‐2, TNF‐α, IL‐12 and chemokines are important for recruitment, differentiation, proliferation and activation of cells within the granuloma.35, 36, 37, 38, 39 Of interest in tuberculosis are the expression of IFN‐γ, TNF‐α and IL‐2, which have all been shown to be necessary for adaptive immunity to intracellular pathogens.32, 33 Therefore, we wanted to examine the expression of IL‐2, TNF‐α and IFN‐γ by human CD4+ and CD8+ T cells in response to infection. In the lungs, IL‐2 secretion was not detected for either CD4+ or CD8+ T cells at day 0 (Fig. 3a), despite the presence of elevated numbers of CD45RA− CD45RO+ suggesting the possibility that these cells were a pool of activated T cells resulting from engraftment, that had stopped proliferating at the time of infection. By day 7, the number of CD4+ IL‐2+ T cells increased significantly in this organ, when compared with day 0 (P < 0·001), but waned at day 22, only to increase again at day 34 (P < 0·001), suggesting waves of proliferation in response to infection. Similar kinetics of expression for CD4+ IL‐2+ T cells was observed in the spleen, with a peak at day 7 (Fig. 3b). CD4+ IFN‐γ + and CD4+ TNF‐α + T cells increased significantly at day 34 in the lungs, compared with all other time‐points (P < 0·05). Splenic CD4+ IFN‐γ + and CD4+ TNF‐α + T‐cell numbers were reduced at day 21 compared with all other time‐points (P < 0·01), before a significant increase at day 34 (P < 0·01).
Figure 3.

Human CD4+ and CD8+ T‐cell responses in the lungs and spleens of mice to pulmonary infection after low‐dose aerosol infection with Mycobacterium tuberculosis H37Rv. Lung (a) and spleen (b) CD4+ and CD8+ T cells were analysed for expression of interferon‐γ (IFN‐γ), interleukin‐2 (IL‐2) and tumour necrosis factor‐α (TNF‐α), and for the expression of cytolytic proteins in CD8+ T cells at days 7, 22 and 34 post infection (c). The flow cytometric gating strategy represented in (d) was used to obtained cell numbers. n = 5 or n = 6 mice per group per time‐point. *P<0·05, **P<0·01, ***P<0·001.
CD8+ IL‐2+ T‐cell numbers increased significantly in the lungs (Fig. 3a) at all time‐points after infection (P < 0·001), but only at day 7 in the spleen (P < 0·05) (Fig. 3b). CD8+ IFN‐γ + and CD8+ TNF‐α + T cells were elevated significantly in the lungs at day 34 post‐infection compared with all other time‐points (P < 0·001), suggesting the recruitment of effector CD8+ T cells to the lung in the latter stages of infection. In the spleen, the number of CD8+ IFN‐γ + T cells was significantly greater at day 34 compared with days 0 and 22 (P < 0·01) and at day 7 compared with day 22 (P < 0·05). The number of CD8+ TNF‐α + T cells was elevated at day 0 and subsequently declined as infection progressed in the spleen. Analysis of CD8+ T‐cell cytolytic proteins in the lung and spleen showed elevated number of cells expressing Granzyme B from days 7 to 34 of infection, whereas expression of perforin and granulysin increased at day 34 (Fig. 3c). These data suggest that there was a significant human T‐cell response during pulmonary infection with M. tuberculosis. Furthermore, these cells were able to undergo differentiation to an effector state and produce cytokines that are induced during the human infection.
Vaccination alters the immune response to infection in Hu‐M™ mice
To determine if the cellular response to infection was influenced by previous vaccination, lung CD4+ and CD8+ T cells from humanized mice vaccinated with either BCG or CpG‐C/liposome/ESAT‐6 and subsequently infected with M. tuberculosis were assessed for expression of IFN‐γ, IL‐2 and TNF‐α. For vaccination, BCG was used for one group and a second group received intranasal CpG‐C formulated with liposomes and ESAT‐6, and the response was monitored at day 30 post‐infection. Day 30 post‐infection was chosen for analysis for two reasons: first, because based on our initial infection, the day 34 time‐point demonstrated a significant difference between infected and non‐infected mice with regards to the presence of effector T cells (Fig. 2) and second, because day 30 coincided with the C57BL/6 mouse model for testing vaccine candidates, in which mice are analysed for CFU and immunological status.27 It has been our experience that at this time‐point a significant difference between vaccinated and non‐vaccinated groups can be observed. Studies with this vaccine formulation and regimen had previously shown that it was effective in significantly reducing the mycobacterial burden in M. tuberculosis‐infected mice and limiting disease in infected guinea pigs (c.f. Fig. 7). Therefore, it was chosen for the Hu‐M™ mouse study to compare responses in the different models. Analysis of cytokine expression in CD4+ and CD8+ T cells in the lung (Fig. 4) and spleen (see Supplementary material, Fig. S1) by flow cytometry, based on the gating strategy in Fig. 4(a), showed that the quality of immune response varied between the treatment groups in response to pulmonary infection, suggesting that vaccination as well as the type of vaccine, both affected the phenotype of the immune response. All groups responded to infection by induction of CD4+ IFN‐γ + IL‐2+ TNF‐α + T cells, although infection alone (Dextrose 5% in Water (D5W)‐treated mice) resulted in a significantly greater number of these cells compared with the vaccinated groups (P < 0·05). The diversity in the CD4+ T‐cell response to infection was greater for the D5W group (diluent used to prepare the CpG‐C‐formulated vaccine), whereas BCG vaccination caused the response to infection to be focused, with fewer numbers of CD4+ IFN‐γ + IL‐2− TNF‐α + and CD4+ IFN‐γ + IL‐2− TNF‐α − T cells when compared with the D5W group, although this was not statistically significant. For CD4+ IFN‐γ − IL‐2+ TNF‐α − and CD4+ IFN‐γ − IL‐2+ TNF‐α + T cells, BCG vaccination caused a significant reduction in cell numbers after infection (P < 0·05) indicating the possibility of decline in the expansion of antigen‐specific T cells and moving into the effector phase, with cells producing macrophage‐activating factors. There was no statistical difference in CD4+ IFN‐γ − IL‐2− TNF‐α + T cells between groups. In the spleen, the CD4+ T‐cell response to infection was focused predominantly on cells that were IFN‐γ + IL‐2− TNF‐α − (see Supplementary material, Fig. S1b) and may reflect the diverse requirement for effector T‐cell function in different organs. The breadth of the response in lungs of infected mice was broader for CD8+ T cells than for CD4+ T cells (Fig. 4b). Although CD8+ IFN‐γ + IL‐2+ TNF‐α + T cells were observed in all groups, the predominant response consisted of CD8+ IFN‐γ + IL‐2+ TNF‐α – T cells and CD8+ IFN‐γ – IL‐2+ TNF‐α – T cells, although this was not statistically different (Fig. 4b). CD8+ T cells were also examined for their ability to produce granzyme and perforin in response to infection (Fig. 4c). A significant reduction in CD8+ granyme+ perforin+ T cells was observed in BCG‐vaccinated mice when compared with D5W‐treated mice (P < 0·05), possibly indicating the skewing of the CD8+ T‐cell response to cytokine production, rather than cytolytic activity. In the spleen, CD8+ T cells displayed a greater diversity in their response to infection, when compared with the lung, without significant differences among groups, except for the BCG group, in which there were significantly fewer IFN‐γ + IL‐2‐ TNF‐α +‐expressing cells in comparison with D5W‐treated mice (see Supplementary material, Fig. S1b, P < 0·01).
Figure 7.

(a) C57BL/6 mouse model, CFU in the lungs of mice and interferon‐γ (IFN‐γ) and interleukin‐2 (IL‐2) response in spleens of infected mice. Mice were vaccinated, rested for 30 days and then infected with a low‐dose aerosol of Mycobacterium tuberculosis H37Rv and CFU and ELISpot performed at day 30 post‐infection (n = 4 or n = 5 mice per group). (b) Hartley guinea pigs were vaccinated, rested for 10 weeks and then infected with a low‐dose aerosol of M. tuberculosis H37Rv and monitored for disease development (n = 10 guinea pigs per group). *P < 0·05, ***P < 0·001, when compared with the saline‐treated group.
Figure 4.

Multi‐parameter analysis of lung CD4+ and CD8+ T cells in mice vaccinated with either BCG or CpG‐C/liposome/ESAT‐6 and then infected with Mycobacterium tuberculosis H37Rv at day 30 post‐infection. (a) Selection of the final multi‐parameter populations was based on the identification of lymphocyte cellular morphology from the forward scatter (FSC) and side scatter (SSC) characteristics. The CD4+ and CD8+ T‐cell populations expressing interferon‐γ (IFN‐γ) or not was selected for further classification of the expression of tumour necrosis factor‐α (TNF‐α) and interleukin‐2 (IL‐2). (b) Pie charts showing the percentage of the relative distribution for each treatment group of the subsets of CD4+ and CD8+ T cells expressing IFN‐γ, IL‐2 and TNF‐α. (c) Lung cells were also analysed for the presence of Granzyme+ Perforin+ CD8+ T cell. n = 4 to n = 6 mice per group. Statistical analysis was performed for each treatment group for a cytokine combination in comparison with the D5W‐treated group. *P < 0·05.
Lung cells from vaccinated mice were examined for expression of IFN‐γ, IL‐2 and granzyme B by ELISpot assays (see Supplementary material, Fig. S2). These results supported the flow cytometry data showing that human immune cells express key anti‐mycobacterial molecules in response to infection after vaccination.
We next examined the CFU and extent of pathology in the lungs of mice after infection to determine if vaccination affected the infection outcome. Figure 5 shows data from a representative experiment of the log10 CFU in the lungs of the vaccinated groups, compared with the log10 CFU in the lungs of the D5W‐treated group. There was no significant difference in the log10 CFU between groups, but in contrast there was a range of approximately 2 log10 CFU between mice within each group. Examination of lung sections revealed that in each group, infection was focused into defined granulomatous areas. On closer inspection (Fig. 6), mice in the D5W and CpG‐C/liposome‐treated groups had multiple aggregates of cellular accumulations containing cellular debris. In contrast, mice vaccinated with BCG or CpG‐C/liposome/ESAT‐6 granulomatous lesions consisted of cellular organized aggregates. These data suggest that vaccination resulted in the accumulation of cellular aggregates in the lungs of infected mice that were able to respond to infection.
Figure 5.

Representative study of log10 CFU in the lungs of Hu‐M™ mice vaccinated with either BCG or CpG‐C/liposome/ESAT‐6 compared with mice treated with D5W. CFU was determined at day 30 post low‐dose aerosol Mycobacterium tuberculosis infection. Day 30 was chosen based on the mouse model used for testing vaccine candidates, at which time the vaccine‐induced immunity will have induced an effector response. n = 4 to n = 6 mice per group.
Figure 6.

Representative photomicrographs of histopathology from lung sections taken from Hu‐M™ mice infected with a low‐dose aerosol of Mycobacterium tuberculosis H37Rv. Mice were treated with D5W (diluent for CpG‐C/liposome formulation), vaccinated with BCG, CpG‐C/liposome/ESAT‐6 or treated with CpG‐C/liposome. Lungs from day 30 post infection, were placed into formalin, embedded in paraffin and sections were stained with haematoxylin & eosin. All groups displayed focal lesions with necrosis, but vaccinated animals had organized cellular aggregates. Original magnification, 60×. Bar = 20 μm.
Comparison of Hu‐M™ mice to the C57BL/6 and guinea pig models of tuberculosis
The same vaccines used in the humanized mouse were used to inoculate C57BL/6 mice and Hartley guinea pigs. In the mouse model, the CpG‐C/liposome/ESAT‐6 formulation caused a significant reduction in CFU in the lung and spleen when compared with the saline‐treated group (Fig. 7a) that was associated with increased numbers of cells secreting IFN‐γ and IL‐2 when compared with the BCG‐vaccinated group (Fig. 7b). In the guinea pig, the vaccine formulation caused a significant increase in the survival time of guinea pigs when compared with the saline‐treated group (P < 0·05, Fig. 7c). These data suggest that in two animal models used for assessing the efficacy of vaccines, the CpG‐C‐based vaccine was able to stimulate a potent T‐cell response that caused a reduction in CFU in mice and prolonged the survival of guinea pigs after pulmonary infection. The three models provide a broad picture of the capacity of a vaccine to induce protective immunity in the standard mouse and guinea pig models and then its ability to generate human T cells that can respond to infection. In this instance, it was notable that in the humanized mouse the CpG‐G‐based vaccine induced a different type of immune response to infection from that of the attenuated vaccine strain, BCG.
Discussion
The immune response in humans to infection with M. tuberculosis has been well documented and requires the concerted involvement of multiple cellular compartments and cytokines working together, resulting, in the majority of cases, in a latent state in which the organism persists for many years in an immunologically primed host.40 It is thought that a better understanding of the immune response during infection in humans may provide better strategies for vaccine development, given that the current vaccine against tuberculosis, BCG, provides at best only partial protection.41 Early studies provided evidence for the importance of T‐cell‐mediated immunity as a key factor for protective immunity,42, 43 and although this remains true, subsequent studies have focused on innate immune cells that can dictate the course of T‐cell immunity, B cells and antibodies that may be involved in clearance of organisms, similar to their role in other infectious agents such as viruses.34, 44, 45, 46, 47 In general, the relative contribution of each of these components in the immune response may be debatable, but the fact remains that there are currently no new vaccines approved for use to combat mycobacterial infection. Animal model testing is an integral part of screening new vaccines.
A better mechanistic understanding of the workings of a vaccine will provide valuable data for improved vaccine development. The pipeline of animal models to precisely define vaccine‐induced immunity has proven to be both beneficial and problematic. Our current data confirm previous studies showing that the humanized mouse can be used to investigate the immunopathogenesis of tuberculosis infection,24, 25, 26 and extends these studies to show that it can also be used in examining vaccine‐mediated immunity. However, we now introduce the concept that the model can also be used to study novel vaccines to characterize the immune response generated by human immune cells in vivo. In the current study, we used two distinct vaccine formulations, one was the attenuated strain of M. bovis, BCG, which is widely used and the second, a CpG‐C‐based subunit vaccine expressing a single M. tuberculosis antigen. The Hu‐M™ model was able to discern the differences in immune responses to infection generated by these two classes of vaccines. Mice inoculated with the CpG‐C formulations displayed a range of responses, suggesting that the interaction with TLR9 was viable and that it induced T‐cell‐mediated immunity. Hu‐M™ mice also developed effector T‐cell phenotypes similar to those reported by others in the C57BL/6 mouse,48 however, it is difficult to directly assess T‐cell function in the guinea pig model due to the limited availability of validated reagents.
Similar to previous studies in humanized mice infected with M. tuberculosis, we confirmed the induction of human CD4+, CD8+ T cells and the expression of cytolytic proteins.25 Our data differ from previous results in that inoculation of BCG into humanized mice induced an immune response to infection, but infection was not controlled. Based on our organ homogenate plating protocol, using thiophene‐2‐carboxylic acid hydrazide‐supplemented growth media there was no indication that BCG grew to the levels reported.26 This may be due to the different route of inoculation used in the current study. Our intention was to use BCG as a vaccine and therefore, as with the majority of mouse vaccine studies using BCG, it was administered subcutaneously, whereas the former study infected mice with a much higher dose via the intravenous route. Immediate dissemination of a high dose of BCG may have overwhelmed the immune system leading to higher bacterial loads in humanized mice. This outcome is reminiscent of experiments in which SCID mice infected with a high intravenous dose of BCG showed progressive growth of the organism, in the absence of adaptive immunity.49 Indeed, our observations indicated that aerosol infection with virulent M. tuberculosis resulted in relatively higher CFU when compared with the C57BL/6 mouse model. These differences may be due to multiple factors, possibly related to differences between the capacity of human and mouse cells to control pulmonary infection. In addition, there are substantial differences in the relative frequency and distribution of T and B cells between the inbred immunocompetent C57BL/6 and the humanized Hu‐M™ mice. The distribution of human T and B lymphocytes in the Hu‐M™ mouse is similar to what had been observed and reported for humanized NOG mice.13, 50 Development of the humanized Hu‐M™ mouse model to more efficiently control infection is a future goal, and will provide a better understanding of the mechanisms involved in anti‐tuberculosis protective immunity.
Humanized mouse models have been previously used to study tuberculosis pathogenesis.24, 25 The bone marrow, liver and thymus (BLT) reconstituted mice were shown to have CD4+ and CD8+ T cells with proliferative capacity to pan stimulation using anti‐CD3/CD28.25 A previous study using human CD34+ haematopoietic stem cells provided similar evidence of polyclonal stimulation of CD4+ and CD8+ T cells,24 and both studies demonstrated that these cells were localized to lung granulomas. An additional study demonstrated a lack of antigen‐specific human T‐cell responses,26 whereas the current study provides evidence that both CD4+ and CD8+ T cells undergo T‐cell receptor rearrangement that human T cells identify and respond to M. tuberculosis antigenic stimulation. We have for the first time demonstrated the utility of this platform for understanding the efficacy of novel vaccine candidates against tuberculosis in contrast to earlier studies using BLT and Hu‐NSG mice, which were primarily employed to recapitulate the pathogenesis of the TB‐induced lung pathology. As observed, T cells from vaccinated and infected humanized mice were able to respond to infection by expanding and producing higher levels of cytokines than non‐vaccinated infected mice when stimulated ex vivo.
Although effector cells were detected in the infected mice, it is difficult to make a statement about memory T cells, given the bacterial burden. In the Hu‐M™, 4 weeks after the final vaccination, T cells were able to respond to infection, suggesting the presence of memory T cells. Studies in C57BL/6 mice using CpG‐C formulated with liposomes and ESAT‐6 have demonstrated that this formulation does induce a memory T‐cell response (AAI, manuscript in preparation). Further studies in post‐vaccinated Hu‐M™ mice will need to determine whether these mice can develop memory T cells similar to those observed in the C57BL/6 mice and ultimately in humans.51
In human T‐cell responses during tuberculosis infection, other groups have demonstrated that CD4+ T cells producing multiple cytokines combinations are present in subjects with active tuberculosis disease, although no defined pattern was associated with protection.52 Similar observations were reported in patients from another study showing that CD4+ IFN‐γ + IL‐2+ TNF‐α + T cells were increased during disease as well as single IL‐2 and TNF‐α single‐positive CD8+ T cells.53 Although the previous observations were derived from human peripheral blood cells, the data from our studies using lung and spleen cells from Hu‐M™ mice demonstrated a more restricted cytokine CD4+ T‐cell response, such as splenic CD4+ T‐cell populations secreting predominantly IFN‐γ, regardless of the pre‐existing immune status, whereas CD8+ T cells displayed a greater diversity in cytokine secretion. In general, no dominant hierarchy in response was observed for the combination of cytokines, suggesting the fact that multiple cell secreting phenotypes may be required for combating M. tuberculosis infection. The current study also examined CD8+ T‐cell granzyme B/perforin expression, demonstrating the presence of elevated numbers of cells during infection, whereas studies using the BLT mouse had only examined granulysin expression.25 Given that CD8+ granulysin/perforin expression has been shown to be produced during chronic tuberculosis,54 it would be of great interest to determine if there was a relationship with chronic tuberculosis and granulysin in the CD34+ HPC model, requiring the study to be extended beyond 30 days. In addition, as in humans, where potent T‐cell responses were observed in the face of active disease, the infected Hu‐M™ mice displayed high levels of infection, possibly reflecting the complex interaction between the pathogen and human immune cells. Further studies will be required to better understand the immune response in the Hu‐M™ mouse to determine how closely the model reflects human disease. Indeed, we observed a wide response to infection, as reflected in the CFU reduction even from mice within a given group, possibly reflecting the diversity in response to vaccination, similar to that seen in humans.55
With respect to cytokine‐secreting cells, the Hu‐M™ mice vaccinated with CpG‐C/liposome/ESAT‐6 developed relatively similar numbers of CD4+ and CD8+ T cells when compared with BCG‐vaccinated animals. In contrast, in the C57BL/6 mouse model, although both vaccination protocols resulted in a significant reduction in CFU, significant differences in the number of cytokine‐secreting cells between the two groups were observed. This may be a specific phenomenon associated with the mouse model or may reflect the different immune mechanisms induced by the vaccines to induce protective immunity. In addition, the reason there are fewer IFN‐γ‐secreting and IL‐2‐secreting cells in the vaccinated groups compared with the saline and adjuvant only treated groups is that the recall immune response in the vaccinated groups has resulted in a reduction in CFU and therefore in the contraction phase. Based on the guinea pig model, CpG‐C/liposome/ESAT‐6 formulation was not as efficient as BCG in preventing disease and prolonging survival, again suggesting possible different mechanisms of protective immunity. These data are subsequently supported by the Hu‐M™ model, which showed a diversity in immune response to infection, depending on the type of vaccine employed.
The question is where does the humanized mouse model fit into the scheme of animal modelling for human tuberculosis and what value does it add to the testing of vaccines? Although no model perfectly recapitulates the human infection, the humanized mouse can help in identifying critical components in the human immune response that can guide future vaccine development, particularly those that are not represented in animals. The non‐human primate is becoming more widely used as the model of choice for testing vaccines;56, 57, 58 however, the limitations associated with purchase and care make this model suitable for end‐stage vaccine development, rather than an initial screen for new vaccines. Development of the humanized mouse for M. tuberculosis infection is required and warranted as an additional preclinical tool in studying various aspects of tuberculosis disease.59, 60, 61 Future employment of the humanized mouse model will allow us to better identify factors associated with human immunity against infection and induced by vaccines at an early stage of development. The use of this model provides important information with regards to other infectious pathogens and with further refinement of the engrafted cells can lead to a human immune response that closely reflects the human responses.
Disclosures
The authors declare that we have no conflict of interest.
Supporting information
Figure S1. Multi‐parameter analysis of spleen CD4+ and CD8+ T cells in mice vaccinated with either BCG or CpG‐C/liposome/ESAT‐6 and then infected with Mycobacterium tuberculosis H37Rv based on the gating strategy to identify CD4+ and CD8+ T cells secreting interferon‐γ, interleukin‐2 and tumour necrosis factor‐α.
Figure S2. ELISpot analysis of lung cells from vaccinated Hu‐M™ mice, infected with a low‐dose aerosol infection of Mycobacterium tuberculosis H37Rv at day 30 post infection for interferon‐γ, interleukin‐2 and granzyme B.
Acknowledgements
This work was supported by the NIH/NIAID programme Advanced Small Animal Models for the Testing of Candidate Therapeutic and Preventative Interventions against Mycobacteria (HHSN272201000009I‐003, task order 12) at Colorado State University (AAI).
References
- 1. Leung C, Chijioke O, Gujer C, Chatterjee B, Antsiferova O, Landtwing V et al Infectious diseases in humanized mice. Eur J Immunol 2013; 43:2246–54. [DOI] [PubMed] [Google Scholar]
- 2. Baumler A, Fang FC. Host specificity of bacterial pathogens. Cold Spring Harb Perspect Med 2013; 3:a010041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Wolfe ND, Dunavan CP, Diamond J. Origins of major human infectious diseases. Nature 2007; 447:279–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Wu W, Vieira J, Fiore N, Banerjee P, Sieburg M, Rochford R et al KSHV/HHV‐8 infection of human hematopoietic progenitor (CD34+) cells: persistence of infection during hematopoiesis in vitro and in vivo . Blood 2006; 108:141–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Aldrovandi GM, Feuer G, Gao L, Jamieson B, Kristeva M, Chen IS et al The SCID‐hu mouse as a model for HIV‐1 infection. Nature 1993; 363:732–6. [DOI] [PubMed] [Google Scholar]
- 6. Smith MS, Goldman DC, Bailey AS, Pfaffle DL, Kreklywich CN, Spencer DB et al Granulocyte‐colony stimulating factor reactivates human cytomegalovirus in a latently infected humanized mouse model. Cell Host Microbe 2010; 8:284–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Thomas S, Klobuch S, Podlech J, Plachter B, Hoffmann P, Renzaho A et al Evaluating human T‐cell therapy of cytomegalovirus organ disease in HLA‐transgenic mice. PLoS Pathog 2015; 11:e1005049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Washburn ML, Bility MT, Zhang L, Kovalev GI, Buntzman A, Frelinger JA et al A humanized mouse model to study hepatitis C virus infection, immune response, and liver disease. Gastroenterology 2011; 140:1334–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Firoz Mian M, Pek EA, Chenoweth MJ, Ashkar AA. Humanized mice are susceptible to Salmonella typhi infection. Cell Mol Immunol 2011; 8:83–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Strowig T, Gurer C, Ploss A, Liu YF, Arrey F, Sashihara J et al Priming of protective T cell responses against virus‐induced tumors in mice with human immune system components. J Exp Med 2009; 206:1423–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Yajima M, Imadome K, Nakagawa A, Watanabe S, Terashima K, Nakamura H et al T cell‐mediated control of Epstein–Barr virus infection in humanized mice. J Infect Dis 2009; 200:1611–5. [DOI] [PubMed] [Google Scholar]
- 12. Jaiswal S, Pearson T, Friberg H, Shultz LD, Greiner DL, Rothman AL et al Dengue virus infection and virus‐specific HLA‐A2 restricted immune responses in humanized NOD‐scid IL2rγnull mice. PLoS One 2009; 4:e7251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Sato K, Nie C, Misawa N, Tanaka Y, Ito M, Koyanagi Y. Dynamics of memory and naive CD8+ T lymphocytes in humanized NOD/SCID/IL‐2Rγnull mice infected with CCR5‐tropic HIV‐1. Vaccine 2010; 28(Suppl. 2):B32–7. [DOI] [PubMed] [Google Scholar]
- 14. Jiang Q, Zhang L, Wang R, Jeffrey J, Washburn ML, Brouwer D et al FoxP3+ CD4+ regulatory T cells play an important role in acute HIV‐1 infection in humanized Rag2–/– γC–/– mice in vivo . Blood 2008; 112:2858–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Gorantla S, Makarov E, Finke‐Dwyer J, Gebhart CL, Domm W, Dewhurst S et al CD8+ cell depletion accelerates HIV‐1 immunopathology in humanized mice. J Immunol 2010; 184:7082–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Arnold L, Tyagi RK, Meija P, Swetman C, Gleeson J, Perignon JL et al Further improvements of the P. falciparum humanized mouse model. PLoS One 2011; 6:e18045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Kwant‐Mitchell A, Ashkar AA, Rosenthal KL. Mucosal innate and adaptive immune responses against herpes simplex virus type 2 in a humanized mouse model. J Virol 2009; 83:10664–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Libby SJ, Brehm MA, Greiner DL, Shultz LD, McClelland M, Smith KD et al Humanized nonobese diabetic‐scid IL2rγnull mice are susceptible to lethal Salmonella Typhi infection. Proc Natl Acad Sci USA 2010; 107:15589–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Parsons CH, Adang LA, Overdevest J, O'Connor CM, Taylor JR Jr, Camerini D et al KSHV targets multiple leukocyte lineages during long‐term productive infection in NOD/SCID mice. J Clin Invest 2006; 116:1963–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Wege AK, Florian C, Ernst W, Zimara N, Schleicher U, Hanses F et al Leishmania major infection in humanized mice induces systemic infection and provokes a nonprotective human immune response. PLoS Negl Trop Dis 2012; 6:e1741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Becker PD, Legrand N, van Geelen CM, Noerder M, Huntington ND, Lim A et al Generation of human antigen‐specific monoclonal IgM antibodies using vaccinated “human immune system” mice. PLoS One 2010; 5:pii: e13137. doi: 10.1371/journal.pone.0013137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Lang J, Kelly M, Freed BM, McCarter MD, Kedl RM, Torres RM et al Studies of lymphocyte reconstitution in a humanized mouse model reveal a requirement of T cells for human B cell maturation. J Immunol 2013; 190:2090–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Tonomura N, Habiro K, Shimizu A, Sykes M, Yang YG. Antigen‐specific human T‐cell responses and T cell‐dependent production of human antibodies in a humanized mouse model. Blood 2008; 111:4293–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Heuts F, Gavier‐Widen D, Carow B, Juarez J, Wigzell H, Rottenberg ME. CD4+ cell‐dependent granuloma formation in humanized mice infected with mycobacteria. Proc Natl Acad Sci USA 2013; 110:6482–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Calderon VE, Valbuena G, Goez Y, Judy BM, Huante MB, Sutjita P et al A humanized mouse model of tuberculosis. PLoS One 2013; 8:e63331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Lee J, Brehm MA, Greiner D, Shultz LD, Kornfeld H. Engrafted human cells generate adaptive immune responses to Mycobacterium bovis BCG infection in humanized mice. BMC Immunol 2013; 14:53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Izzo A, Brandt L, Lasco T, Kipnis AP, Orme I. NIH pre‐clinical screening program: overview and current status. Tuberculosis 2005; 85:25–8. [DOI] [PubMed] [Google Scholar]
- 28. Cardona PJ, Williams A. Experimental animal modelling for TB vaccine development. Int J Infect Dis 2017; 56:268–73. [DOI] [PubMed] [Google Scholar]
- 29. Keyser A, Troudt JM, Taylor JL, Izzo AA. BCG sub‐strains induce variable protection against virulent pulmonary Mycobacterium tuberculosis infection, with the capacity to drive Th2 immunity. Vaccine 2011; 29:9308–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Taylor JL, Wieczorek A, Keyser AR, Grover A, Flinkstrom R, Karls RK et al HspX‐mediated protection against tuberculosis depends on its chaperoning of a mycobacterial molecule. Immunol Cell Biol 2012; 90:945–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Roederer M, Nozzi JL, Nason MC. SPICE: exploration and analysis of post‐cytometric complex multivariate datasets. Cytometry A 2011; 79:167–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Wille‐Reece U, Flynn BJ, Lore K, Koup RA, Kedl RM, Mattapallil JJ et al HIV Gag protein conjugated to a Toll‐like receptor 7/8 agonist improves the magnitude and quality of Th1 and CD8+ T cell responses in nonhuman primates. Proc Natl Acad Sci USA 2005; 102:15190–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Wille‐Reece U, Flynn BJ, Lore K, Koup RA, Miles AP, Saul A et al Toll‐like receptor agonists influence the magnitude and quality of memory T cell responses after prime‐boost immunization in nonhuman primates. J Exp Med 2006; 203:1249–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. 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. Clin Infect Dis 2015; 61(Suppl. 3):S225–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Cooper AM, Dalton DK, Stewart TA, Griffin JP, Russell DG, Orme IM. Disseminated tuberculosis in interferon γ gene‐disrupted mice. J Exp Med 1993; 178:2243–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Cooper AM, Roberts AD, Rhoades ER, Callahan JE, Getzy DM, Orme IM. The role of interleukin‐12 in acquired immunity to Mycobacterium tuberculosis infection. Immunology 1995; 84:423–32. [PMC free article] [PubMed] [Google Scholar]
- 37. Flynn JL, Chan J, Triebold KJ, Dalton DK, Stewart TA, Bloom BR. An essential role for interferon γ in resistance to Mycobacterium tuberculosis infection. J Exp Med 1993; 178:2249–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Flynn JL, Goldstein MM, Chan J, Triebold KJ, Pfeffer K, Lowenstein CJ et al Tumor necrosis factor‐α is required in the protective immune response against Mycobacterium tuberculosis in mice. Immunity 1995; 2:561–72. [DOI] [PubMed] [Google Scholar]
- 39. Flynn JL, Goldstein MM, Triebold KJ, Sypek J, Wolf S, Bloom BR. IL‐12 increases resistance of BALB/c mice to Mycobacterium tuberculosis infection. J Immunol 1995; 155:2515–24. [PubMed] [Google Scholar]
- 40. Lipworth S, Hammond RJ, Baron VO, Hu Y, Coates A, Gillespie SH. Defining dormancy in mycobacterial disease. Tuberculosis 2016; 99:131–42. [DOI] [PubMed] [Google Scholar]
- 41. Andersen P, Doherty TM. The success and failure of BCG – implications for a novel tuberculosis vaccine. Nat Rev Microbiol 2005; 3:656–62. [DOI] [PubMed] [Google Scholar]
- 42. Lefford MJ. Transfer of adoptive immunity to tuberculosis in mice. Infect Immun 1975; 11:1174–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Ravn P, Boesen H, Pedersen BK, Andersen P. Human T cell responses induced by vaccination with Mycobacterium bovis bacillus Calmette–Guérin. J Immunol 1997; 158:1949–55. [PubMed] [Google Scholar]
- 44. van Crevel R, Ottenhoff TH, van der Meer JW. Innate immunity to Mycobacterium tuberculosis . Clin Microbiol Rev 2002; 15:294–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Lerner TR, Borel S, Gutierrez MG. The innate immune response in human tuberculosis. Cell Microbiol 2015; 17:1277–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Achkar JM, Casadevall A. Antibody‐mediated immunity against tuberculosis: implications for vaccine development. Cell Host Microbe 2013; 13:250–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Maglione PJ, Chan J. How B cells shape the immune response against Mycobacterium tuberculosis . Eur J Immunol 2009; 39:676–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Junqueira‐Kipnis AP, Turner J, Gonzalez‐Juarrero M, Turner OC, Orme IM. Stable T‐cell population expressing an effector cell surface phenotype in the lungs of mice chronically infected with Mycobacterium tuberculosis . Infect Immun 2004; 72:570–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. North RJ, Izzo AA. Mycobacterial virulence. Virulent strains of Mycobacteria tuberculosis have faster in vivo doubling times and are better equipped to resist growth‐inhibiting functions of macrophages in the presence and absence of specific immunity. J Exp Med 1993; 177:1723–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Ito M, Hiramatsu H, Kobayashi K, Suzue K, Kawahata M, Hioki K et al NOD/SCID/γ(c)(null) mouse: an excellent recipient mouse model for engraftment of human cells. Blood 2002; 100:3175–82. [DOI] [PubMed] [Google Scholar]
- 51. Korsholm KS, Hansen J, Karlsen K, Filskov J, Mikkelsen M, Lindenstrom T et al Induction of CD8+ T‐cell responses against subunit antigens by the novel cationic liposomal CAF09 adjuvant. Vaccine 2014; 32:3927–35. [DOI] [PubMed] [Google Scholar]
- 52. Caccamo N, Guggino G, Joosten SA, Gelsomino G, Di Carlo P, Titone L et al Multifunctional CD4+ T cells correlate with active Mycobacterium tuberculosis infection. Eur J Immunol 2010; 40:2211–20. [DOI] [PubMed] [Google Scholar]
- 53. Sutherland JS, Adetifa IM, Hill PC, Adegbola RA, Ota MO. Pattern and diversity of cytokine production differentiates between Mycobacterium tuberculosis infection and disease. Eur J Immunol 2009; 39:723–9. [DOI] [PubMed] [Google Scholar]
- 54. Andersson J, Samarina A, Fink J, Rahman S, Grundstrom S. Impaired expression of perforin and granulysin in CD8+ T cells at the site of infection in human chronic pulmonary tuberculosis. Infect Immun 2007; 75:5210–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Ndlovu H, Marakalala MJ. Granulomas and inflammation: host‐directed therapies for tuberculosis. Front Immunol 2016; 7:434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Pena JC, Ho WZ. Non‐human primate models of tuberculosis. Microbiol Spectr 2016; 4:doi:10.1128/microbiolspec.TBTB2‐0007‐2016. [DOI] [PubMed] [Google Scholar]
- 57. McMurray DN. A nonhuman primate model for preclinical testing of new tuberculosis vaccines. Clin Infect Dis 2000; 30(Suppl 3):S210–2. [DOI] [PubMed] [Google Scholar]
- 58. Bushmitz M, Lecu A, Verreck F, Preussing E, Rensing S, Matz‐Rensing K et al Guidelines for the prevention and control of tuberculosis in non‐human primates: recommendations of the European primate veterinary association working group on tuberculosis. J Med Primatol 2009; 38:59–69. [DOI] [PubMed] [Google Scholar]
- 59. Walsh GP, Tan EV, dela Cruz EC, Abalos RM, Villahermosa LG, Young LJ et al The Philippine cynomolgus monkey (Macaca fascicularis) provides a new nonhuman primate model of tuberculosis that resembles human disease. Nat Med 1996; 2:430–6. [DOI] [PubMed] [Google Scholar]
- 60. Dutta NK, McLachlan J, Mehra S, Kaushal D. Humoral and lung immune responses to Mycobacterium tuberculosis infection in a primate model of protection. Trials Vaccinol 2014; 3:47–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Flynn JL, Gideon HP, Mattila JT, Lin PL. Immunology studies in non‐human primate models of tuberculosis. Immunol Rev 2015; 264:60–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Figure S1. Multi‐parameter analysis of spleen CD4+ and CD8+ T cells in mice vaccinated with either BCG or CpG‐C/liposome/ESAT‐6 and then infected with Mycobacterium tuberculosis H37Rv based on the gating strategy to identify CD4+ and CD8+ T cells secreting interferon‐γ, interleukin‐2 and tumour necrosis factor‐α.
Figure S2. ELISpot analysis of lung cells from vaccinated Hu‐M™ mice, infected with a low‐dose aerosol infection of Mycobacterium tuberculosis H37Rv at day 30 post infection for interferon‐γ, interleukin‐2 and granzyme B.
