Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2023 Jul 12.
Published in final edited form as: Cell Rep. 2022 Dec 13;41(11):111783. doi: 10.1016/j.celrep.2022.111783

Mucosal exposure to non-tuberculous mycobacteria elicits B-cell mediated immunity against pulmonary tuberculosis

Taru S Dutt 1,*, Burton R Karger 1, Amy Fox 1, Nathan Youssef 3, Rhythm Dadhwal 2, Malik Zohaib Ali 1,5, Johnathan Patterson 1, Elizabeth Creissen 1, Elisa Rampacci 4, Sarah Cooper 1, Brendan K Podell 1, Mercedes Gonzalez-Juarrero 1, Andres Obregon-Henao 1, Marcela Henao-Tamayo 1,*
PMCID: PMC10337594  NIHMSID: NIHMS1904314  PMID: 36516760

Summary

Bacille Calmette Guerin (BCG) is the only licensed vaccine against Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB) disease. However, BCG has limited efficacy, necessitating development of better vaccines. Non-tuberculous mycobacteria (NTM), ubiquitously present in the environment, can be opportunistic pathogens. Individuals in TB endemic countries experience higher exposure to NTM, but previous studies have not elucidated the relationship between NTM exposure and BCG efficacy against TB. Therefore, we developed a mouse model (BCG+NTM) to simulate human BCG immunization regime and continuous NTM exposure. BCG+NTM mice exhibit superior and prolonged protection against pulmonary TB, with increase in B-cell influx and anti-Mtb antibodies in serum and airways, compared to BCG alone. Notably, spatial transcriptomics and immunohistochemistry reveal that BCG+NTM mice form B-cell aggregates with features of germinal center development, which correlates with reduced Mtb burden. Our studies suggest a direct relationship between NTM exposure and TB protection, with B-cells playing a crucial role.

Keywords: Non-tuberculous mycobacteria, BCG, tuberculosis, B-cell, antibodies, tertiary lymphoid structures, germinal centers

Introduction

Non-tuberculous mycobacteria (NTM) comprise a diverse group of mycobacterial species that do not cause tuberculosis (TB), but they can cause lung disease in individuals with local or systemic immune suppression1,2. These opportunistic pathogens3,4 are ubiquitous in the environment. Hence, humans are continuously exposed to NTM, most likely starting early in life, via mucosal routes such as the gastrointestinal tract (water/food), skin (wounds), or airways (inhalation). High exposure to NTM occurs in some regions of the world due to a combination of environmental, sanitary, and socio-economic reasons. Notably, TB endemicity overlaps areas of the world with high NTM exposure. NTM share a large number of antigens with Mtb and BCG; thus, it has been postulated that NTM modulate vaccine-mediated protection and immunity against TB 5-7. However, the effect of pre-exposure to NTM on subsequent Mtb infection is still debatable, although highly studied.

TB is a severe public health problem, affecting a quarter of the world's population8. Approximately 1.5 million people die of TB annually, including HIV-associated deaths. Despite concerted efforts, it continues to spread worldwide 9-11. TB is a treatable disease requiring six to nine months of multi-antibiotic treatment12. However, long-term therapy may lead to drug-induced toxicity, human microbiota dysbiosis, and patient non-compliance, resulting in other secondary infections, a weakened immune system, and the emergence of antibiotic-resistant TB, respectively 13-16. Current efforts to fight TB by improving diagnosis and developing better anti-TB vaccines and drugs are falling short 17-19. Furthermore, the COVID-19 pandemic crippled the World Health Organization's TB guidelines, such as early patient diagnosis, treatment follow-up, and compliance 20,21, erasing much of the last decade’s progress against TB and other infectious diseases. As a result, TB continues to cause immense human misery and considerable economic loss, and it is one of the leading causes of global inequity.

BCG is the only licensed vaccine against TB22. Unfortunately, its efficacy varies greatly depending on the country and demographics23,24. BCG’s variable protection against TB has been attributed, amongst others, to genetic divergence of BCG strains used for vaccination25,26, high virulence of clinical Mtb strains19,27,28, host genetics29,30, and interference by NTM7,31,32. Data from animal models exploring the influence of NTM on Mtb pathogenesis and BCG vaccine responses vary between studies. While early research demonstrated that NTM infection (without BCG vaccination) protects animals against Mtb infection33-35, others reported that mice pre-exposed to NTM with subsequent BCG vaccination appear to mask the overall protection provided by BCG vaccination6,36. Such discrepancies could be attributed to a multitude of variables in experimental designs, including NTM concentration, duration, and route of NTM exposure, as well as NTM strain. Considering the preceding and to elucidate if and how NTM impacts BCG efficacy against TB, we developed a murine model closely simulating the natural history of human exposure to mycobacteria, including: 1) early BCG vaccination; 2) exposure to viable NTM via drinking water [1000 CFU/mL for high-income countries (HIC), 1.0x105 CFU/mL for low to middle-income countries (LMIC)]; and 3) maintaining continuous NTM exposure even throughout Mtb infection.

Compared to BCG-only vaccinated mice, a high dose of NTM delivered via drinking water to BCG-vaccinated mice (BCG+NTMhigh) provided more robust and long-lasting protection against Mtb infection 120 days post-infection (dpi), with lower Mtb burden and reduced pulmonary inflammation. Immunological studies showed significantly higher numbers of B220hiMHC-IIhi B-cells and CX3CR1hiMHC-IIhi macrophages in Peyer’s patches (PPs) of NTM exposed mice, regardless of BCG vaccination status before Mtb infection. In the lungs, BCG+NTMhigh mice showed a significant increase in B-cells, CXCR5+ T-follicular helper cells (TFH), and Perforin+Granzyme-B+ natural killer (NK) cells post-infection, strongly correlating with reduced Mtb burden. Additionally, mice exposed to high doses of NTM, irrespective of BCG vaccination status, had considerably higher titers of anti-mycobacterial IgG and IgA antibodies in serum and bronchoalveolar lavage fluid (BALF), both pre-and post-Mtb infection. Surprisingly, BCG+NTMhigh mice also developed follicle-like structures in the lungs. Immunohistochemistry and spatial transcriptomics confirmed B-cell predominance in those structures, along with markers associated with B-cell activation, somatic hypermutation, and germinal center (GC) formation. These data suggest that NTM provide protective immunity against pulmonary TB and that B-cells and GC are crucial in this process.

Results

Oral NTM exposure boosts BCG-mediated protection against pulmonary tuberculosis

Initially, to establish a mouse model of NTM exposure (BCG+NTM mice) representative of the natural history of human exposure to mycobacteria, three-week-old C57BL/6, C3HeB/FeJ, and C3H/HeOuJ mice were vaccinated with BCG (1x105 CFU/mouse). Fifteen days later and throughout the remainder of the experiment, mice were continuously exposed via the drinking water to 1000 CFU/ml Mycobacterium avium subsp. avium (MA), the most ubiquitous species of NTM37]. Water containing NTM was replaced weekly, and bacterial concentration and viability in drinking water were monitored throughout the study (Figure S1). Ninety days after initiating NTM exposure (and 104 days post-BCG vaccination), mice were infected via the aerosol route with Mtb HN878 (50-60 CFU/mouse). Mtb HN878 is a hypervirulent W-Beijing clinical strain with high infectivity38,39, reported to induce a granulomatous response in mice resembling the pathology observed in TB-infected human and non-human primates40. Control mice were immunized with BCG or saline or fed NTM alone (Figure 1A).

Figure 1: Murine model of NTM exposure via drinking water and bacterial burden.

Figure 1:

(A) Study-1: Experimental plan showing a timeline of BCG vaccination, NTM exposure, and Mtb infection using C3H/OuJ, C3HeB/FeJ, and C57BL/6 mice. Colony-forming units (CFUs) showing Mtb burden in the lungs of C3H/HeOuJ (B), C3HeB/FeJ (C), and C57BL/6 (D) mice at 30-, and 60 dpi). (E) Study-2: The experimental plan shows a timeline for BCG vaccination, NTM exposure to low (1000 CFU/mL) and high (1.0 x 105 CFU/mL) doses, and Mtb infection in C3H/HeOuJ mice. (F) Plots showing Mtb CFUs in the lungs of C3H/HeOuJ mice at 30-, 60-, and 120 dpi. (G) Pathology scores represent the proportion of lesion area over total lung area per stained tissue section. (H) Lung sections were stained with hematoxylin and eosin at 120 dpi. Biological replicates, n= 5, technical replicates, n=2. Statistical significance was calculated using ANOVA with the Tukey-HSD test, and p<0.05 was considered significant. *p<0.05, **p<0.005.

To determine the effect of NTM on BCG's efficacy against Mtb, the bacterial burden in the lungs was enumerated at 30 and 60 dpi. As expected, at 30 dpi, BCG vaccination protected all strains of mice against Mtb infection, as evidenced by a one-log reduction in lung CFUs. Interestingly, NTM exposure did not abrogate BCG-mediated protection against Mtb. BCG's protective efficacy, however, began to wane at 60 dpi in vaccinated mice, as previously reported for BCG-vaccinated mice infected with W-Beijing strains41,42. Strikingly, protection was preserved in BCG-vaccinated mice (C3H/HeOuJ and C57BL/6) continuously exposed to NTM via the oral route (Figure 1B-D).

To further investigate the long-term effect of NTM exposure and the impact of different NTM concentrations on BCG-mediated protection against TB (Figure 1E), C3H/HeOuJ mice (intermediate susceptibility to Mtb) were fed water containing 1000 (NTMlow) or 1x105 CFU/mL (NTMhigh) NTM, reported to be present in drinking water of HIC and LMIC countries43-45, respectively. Consistent with the findings above, BCG+NTMlow mice had lower bacterial burden in the lungs at 30 and 60 dpi with Mtb HN878 (Figure 1F). Significantly, protection against TB at 120 dpi was only observed for BCG+NTMhigh. Furthermore, a significant reduction in lung pathology was observed in BCG+NTMhigh mice up to 120 dpi (Figures 1G and 1H). Improved histopathology confirmed that exposure to viable NTM via drinking water did not compromise the protective efficacy of BCG against Mtb infection but instead provided a boost effect.

NTM colonize Peyer’s patches and activate B-cells

Given that mice were administered NTM orally, we assessed their presence and effect on the cellular composition of PPs, the main lymphoid structure associated with the gut. Using an anti-lipoarabinomannan (LAM) antibody 46, NTM were indeed shown to colonize PPs (Figure 2D), specifically the PPs interfollicular region (IFR) and follicles (high magnification insert, Figure 2D). The cellular composition of PPs was evaluated by flow cytometry (Table S1). One day before Mtb infection (−1 dpi), significantly increased numbers of B-cells (CD3B220+MHC-II and CD3B220+MHC-II+), phagocytic cells (CD3CX3CR1+MHC-II+), and CD4+ T cells (CD3+CD4+) were observed in PPs of BCG+NTMhigh mice (Figure 2A and 2B). Immune cell populations in PPs were also evaluated at 30, 60, and 120 dpi after Mtb infection (and continuously exposed to NTM) and correlated to the corresponding lung CFUs (Figure 2C). Mice exposed to high NTM concentrations, irrespective of their BCG vaccination status, showed increased B-cells in PPs, significantly (p<0.01) correlating to reduced Mtb burden in the lungs. Therefore, NTM colonization of PPs could prime the adaptive immune response against mycobacterial antigens.

Figure 2: Peyer's patches (PPs) immune profiling and immunohistochemistry.

Figure 2:

(A) Flow cytometry dot plots showing increased B-cells in BCG+NTMhigh mice. The bivariate plot here represents all mice concatenated together (n=5). (B) PPs cells were stained with B-cells, T-cells, macrophages, and dendritic cell markers and analyzed using flow cytometry. Data here represent total B-cell counts, CX3CR1+ macrophages, and T-cells on day-1 of Mtb infection. (C) Immune cell types present in PPs at days 30, 60, and 90 post-Mtb infection (heatmap) and the corresponding CFUs in the lung (barplot). (D) PPs sections were stained using the OPAL multiplex IHC (mIHC) staining technology and the Leica Bond III autostainer. The panel of antibodies used was anti-B220 (opal 570) antibody, anti-LAM antibody (opal 690), and GL-7 (opal 520) for B-cell, NTM, and GC visualization and location, respectively. Images were acquired using the multispectral camera of the Polaris System (Akoya bioscience) and analyzed using the inForm tissue Finder and phenochartsoftwares. Biological replicates, n= 5, technical replicates, n=3. Statistical significance was calculated using ANOVA with the Tukey-HSD test, and p<0.05 was considered significant. *p<0.05, **p<0.005.

Oral NTM exposure augments pulmonary B-cell responses and production of cross-reactive anti-mycobacterial IgG and IgA antibodies before Mtb infection

Exposure of BCG-vaccinated mice to NTM via the oral route significantly protected against pulmonary TB (reduced lung CFUs and pathology); thus, we investigated whether NTM induced memory immune responses that cross-react and protect against Mtb. Flow cytometric analysis of the lungs was performed to evaluate the immune response elicited by NTM before Mtb infection (antibody panels, Tables S2 and S3). Our findings indicate that NTMhigh and BCG+NTMhigh mice had significantly higher numbers of both T-independent (B1) and T-dependent (B2) B-cells in their lungs (Figure 3A). Interestingly, mice exposed to NTMhigh without BCG vaccination also had increased pulmonary influx of B1 memory (CD19+B220CD44+CD62LMHC-II+CD34+CD73+), B2 memory (CD19+B220+MHC-II+CD4+CD62LCD73+), and progenitor B-cells (CD19+B220CD44+CD62LMHC-II+CD34+), corroborating that NTM generates memory B-cell responses. Additionally, we observed a significant increase in TFH in the lungs of NTMhigh and BCG+NTMhigh mice (Figure 3B).

Figure 3: Immune response in lungs on day-1 before Mtb infection.

Figure 3:

Graphs showing total cell numbers of each cell type present in lungs on day-1 before infection (A and B) Indirect ELISA targeting IgA and IgG antibodies against Mtb whole cell lysate (C). Biological replicates, n= 5, technical replicates, n=3. Statistical significance was calculated using ANOVA with the Tukey-HSD test, and p<0.05 was considered significant. *p<0.05, **p<0.005, ***p<0.0005.

Induction of cross-reactive anti-mycobacterial antibodies in mice orally exposed to NTM —yet not infected with Mtb— was evaluated in serum and BALF via an indirect enzyme-linked immunosorbent assay (ELISA) against Mtb HN878 whole cell lysate. A significantly higher titer of anti-mycobacterial IgA and IgG antibodies in BALF and serum was observed in mice exposed to a high NTM concentration regardless of BCG vaccination status than in those exposed to NTMlow or unexposed (Figure 3C). When combined with flow cytometry results presented above, these findings suggest that NTM exposure results in the generation of memory B-cells and cross-reactive antibodies against Mtb antigens.

Increased B-cells and anti-Mtb antibodies in lungs correlate with enhanced protection upon TB infection

To further investigate the possible immune mechanism(s) of protection, flow cytometry analysis was conducted on leukocytes obtained from lungs of Mtb infected mice at 30, 60, and 120 dpi (Figure 4A, Table S2, and S3). BCG-vaccinated mice not exposed to NTM had higher total T cell counts at 30 dpi, which declined significantly at 60 and 120 dpi, perhaps related to loss of protection at later time points. In contrast, NTMhigh mice had significantly higher numbers of B cells (B220+/CD19+ cells), effector CD8 T cells (CD3+CD8+CD44+Granzyme-B+Perforin+), and NK cells (CD3NKp46+Perforin+Granzyme-B+). A correlation analysis between lung CFUs and various immune cell populations was performed to evaluate the cell types associated with reduced bacterial burden in the lungs (Figure 4B and Figure S2). We found that at 30 dpi, the primary immune cells associated with protection (R-value > −0.5, and p-value < 0.05) were B1 and B2 B-cells, memory B-cells, effector NK cells, TFH, and effector T cells. In contrast, at 120 dpi, NK cells and different B-cell subpopulations (B2, memory, and MHC-II+ B-cells) were significantly correlated with reduced bacterial burden (rather than B1 B-cells and effector T cells). Total B-cells also significantly correlated (p = 0.0037, R = −0.52) with lower lung CFUs at 30-, 60-, and 120-days post-infection (Figure 4C). Our results point to a crucial association between B-cells and NK cells and decreased Mtb burden in the lungs of BCG+NTMhigh mice.

Figure 4: Comprehensive analysis of innate and adaptive immune cell populations at days 30-, 60-, and 120-days post Mtb infection.

Figure 4:

(A) Immune cell populations in the lungs were stained using flow cytometry and analyzed using Cytotypr. Data here show the total cell numbers of each population converted to z-score and represented as a heatmap. (B) Spearman correlation analysis of various immune cell types with the bacterial burden (CFUs) in the lung at 30-, 60-, and 120-dpi. Significance was calculated using the Wilcoxon test with Benjamini-Hochberg correction. (C) Correlation analysis between total B-cells at all time points and lung CFUs using the linear regression model. R-value represents regression value. Statistical significance is calculated using a t-test. (D) Indirect ELISA showing the presence of anti-Mtb IgG and IgA antibodies in the bronchoalveolar lavage fluid (BALF) and serum of mice at 30, 60, and 120 days post-Mtb infection. Values represent the area under the curve (AUC) calculated using three different dilutions (1/50, 1/250, and 1/1250). Biological replicates: n=5, technical replicates for flow cytometry and CFU: n=2; technical replicates for ELISA: n=3. All graphs were created using ggplot2 in RStudio, and significance was calculated using R package stats. *p<0.05, **p<0.005, ***p<0.0005.

The presence of anti-Mtb antibodies in BALF and serum was evaluated by ELISA (Figure 4D), and results were expressed as the area under the curve (AUC). At day 120 post-Mtb infection, we found significantly higher IgA and IgG levels in the BALF of BCG+NTMlow and BCG+NTMhigh mice than in all other groups. However, no significant differences were observed in serum IgA or IgG levels. These findings suggest that NTM confers enhanced protection via B-cell and antibody-mediated mechanisms and that B-cell activation and antibody generation occur within the lung.

Mice exposed to NTM develop B-cell aggregates in the lungs

Immunohistochemistry results demonstrated the presence of pulmonary B cell aggregates (B220+ cells forming clusters), which were very clear at 120 dpi, predominantly in BCG+NTMhigh mice (Figure 5A). Notoriously, B cell aggregates (shown in yellow) localized around the lesion area and were surrounded by Mtb (shown in red). The number of B cell aggregates in the lungs of BCG+NTMhigh mice (Figure 5B) inversely correlated with lower Mtb burden (R=−0.72, p <0.001, Figure 5C).

Figure 5: Immunohistochemistry showing the presence of B-cell aggregates in the lungs.

Figure 5:

Five μm lung sections from saline, NTMlow, NTMhigh, BCG, BCG+NTMlow, and BCG+NTMhigh were stained using the OPAL multiplex IHC (mIHC) staining technology using the LabSat® Research autostainer (Lunaphore platform; Epedria). Slides were scanned using the multispectral camera of the Polaris System (Akoya bioscience) and analyzed with the inForm tissue Finder and phenoptr softwares. The panel of antibodies used included anti-LAM antibody (opal 690), anti-mouse B220 antibody (opal 570), and anti-mouse CD4 antibody (opal 520) (A) B-cell aggregates were calculated in each mouse's lung lobe by drawing a boundary around each B-cell aggregate, teaching the software to count the number of follicles (B) Correlation analysis was performed between CFUs and B-cell aggregates in the lung using the Spearman rank correlation method (C). Biological replicates: n=5, technical replicates: n=3. Statistical significance was calculated using R package stats. *p<0.05, **p<0.005.

To further corroborate our histopathological findings and determine the cell types and gene signatures associated with these structures, we performed spatial transcriptomics using 10X Genomics Visium kit for FFPE tissues. Lung histology images and cell-type distribution of one mouse from each group are shown in Figure 6A (the remaining images are shown in Figure S3). Spatial transcriptomics analysis revealed that NTMhigh and BCG+NTMhigh mice had a significantly higher frequency of pulmonary B cells and dendritic cells than other groups (Figures 6B and C). In contrast, higher macrophage but lower B cell influx was observed in the lungs of saline mice and mice only vaccinated with BCG.

Figure 6: Spatial transcriptome profiling of lung sections at 120-day post-Mtb infection.

Figure 6:

Spatial transcriptomics was performed using 10X visium kit for Formalin-fixed paraffin-embedded tissues on eight lung sections (two sections from each group from two separate mice). (A) H&E-stained lung sections from each group and corresponding cell types at each spot (second panel). (B) The cell type ratio in saline, NTMhigh, BCG, and BCG+NTMhigh mice. (C) Principal component analysis and representative UMAP embedding of single-cell data. (D) SpatialFeaturePlot showing marker expression of top variable features in different groups. Expression ranges from blue to red (lowest to highest expression, respectively).

Notably, mice exposed to NTM exhibited an increased stromal (CD45LinSca-1+CD29+CD105+) cell density in the lungs. Thus, we evaluated markers associated with follicle structures in the lungs of NTM-exposed mice and compared them to the saline and BCG groups (Figure 6D). The follicle structures in the lungs of mice exposed to NTM expressed Cd19 (a B-cell marker), Bcl-6 (a GC B-cell marker), Ltb (lymphotoxin-B, a marker associated with the formation of lymphoid tissue), Mki67 (a proliferation marker), and Cxcr5 (a marker of follicular B and T cells).

Gene ontology (GO) and gene set enrichment (GSE) analysis were performed with clusterProfiler to identify up or down-regulated pathways in each group. The top 25 biological pathways activated in saline, NTMhigh, BCG, and BCG+NTMhigh mice are depicted in Figure S4. The GSE analysis showed that a significant number of genes (p<0.05) activated in saline mice are involved in response to bacterial lipoproteins, removal of superoxide radicals, nitric oxide production, granulocyte migration, and chronic inflammatory response (Figure S4-A). Additionally, the top pathways activated in the BCG group revealed an inflammatory progression of infection, including interferon-alpha and beta responses and wound healing (Figure S4-B). However, in NTM mice, there was a significant upregulation (p<0.05) of pathways associated with humoral immune responses, B-cell activation, IgA, and complement activation (Figure S4-C). Interestingly, the top 25 pathways (p<0.05, gene involvement < 30) activated in BCG+NTMhigh mice are involved in GC formation, B-cell activation and differentiation, somatic recombination, isotype-switching, immunoglobulin complex formation, IgA activation, and NK cell differentiation (Figure S4-D). Collectively, our findings suggest the formation of a lymphoid-like structure containing GC in the lungs of BCG+NTMhigh mice, which may be involved in B-cell mediated immunity.

Transcriptional profiling revealed activation of memory B-cells and LT-LTBR signaling in BCG+NTMhigh mice.

Using DESeq2, we investigated differentially expressed genes (DEGs) in each cluster and identified specific signatures for some groups. For instance, in TB-infected saline control mice, an inflammatory response characterized by a significant upregulation of Nos2, Thbs1, Cxcl10, and Tgfbi was observed (Figure 7A). Conversely, Cd38 activation (a marker for switched memory B-cells) was only observed in B-cells from BCG+NTMhigh mice, a further indication of GC formation in the lungs of these mice (Figure 7B). We also found significantly (p<0.05) increased Ccr6 expression in macrophages, epithelial cells, and fibroblasts in BCG+NTMhigh mice. It is worth noting that MKi67, a major cell proliferation marker, was highly upregulated in neutrophils and macrophages from the saline group. In contrast, its expression in BCG+NTMhigh mice was significantly higher (p<0.01) in B-cells and dendritic cells.

Figure 7: Transcriptomic profile and cell-cell interaction among groups.

Figure 7:

Differential gene expression of each cell type in each group was evaluated using the FindMarkers function in Seurat. (A) Selected markers highly differential between saline and NTMhigh, BCG, and BCG+NTMhigh groups are represented as Feature plots. Scale varies from grey to purple (least to high expression). (B) Violin plots showing marker expression of Cd38, Ccr6, Mki67, and Tnfrsf13c. Markers were selected based on significantly higher expression (p<0.01) between BCG+NTMhigh groups compared to all. (C) Cell-cell interaction analysis using CellChat. Circle plots show the number of interactions between each cell type in an individual group. (D) Barplot indicates the number of total interactions between all cell types and their interaction strength. Values were calculated using the "compareInteractions" function in CellChat. (E) Heatmap showing signaling pathways upregulated in different groups. Red: activated; blue: not activated. (F) Dot plot shows the receptor-ligand interactions of the specific pathways activated in saline, NTMhigh, BCG, and BCG+NTMhigh and the cell types involved with those pathways. The circle's color represents communication probability (red: highest; blue: lowest), and the size of the circle represent the significance level. p<0.05 was considered significant.

Cellular interactions were interrogated using CellChat47 (Figure 7). Figure 7C depicts significant networks of cellular interactions between all cell types within each group. The plots demonstrate that NTMhigh and BCG+NTMhigh mice had a significantly higher number of inferred interactions (1391 and 1805, respectively) between different cell types than saline (692) and BCG (731) mice (Figure 7D). The strength of these interactions also correlated with the number of interactions as follows: BCG+NTMhigh (89.34 %) > NTMhigh (62.45 %) > BCG (23.78 %) > saline (19.65 %). In BCG+NTMhigh mice, B-cells strongly interacted with macrophages, dendritic and stromal cells (Figure 7C). We then determined which signaling pathways were up or downregulated in different groups. Seven signaling pathways were identified to be activated exclusively in BCG+NTMhigh mice, including WNT (Wingless-related integration site), PROS (PIK3CA-Related Overgrowth Spectrum), TWEAK (TNF-related weak inducer of apoptosis), ALCAM (Activated Leukocyte Cell Adhesion Molecule), CD6, CSF (Colony Stimulation Factor), and TNF (Tumor Necrosis Factor). In saline mice, we discovered major interactions between Thbs1 (thrombospondin1) and Sdc1 (Syndecan-1), Sdc4 (Syndecan-4), and Cd36 (Figure 7E). Additionally, the saline group exhibited Notch1-Dll4 interaction in monocytes, indicating tuberculosis progression48. BCG mice also showed similar cellular interactions to saline, including the Thbs1 interaction with Cd47 and Spp1, Icam, and Il16 gene interactions with their ligands. In NTMhigh and BCG+NTMhigh mice, Lta (lymphotoxin) —characteristic of secondary lymphoid organs and involved in lymphoid tissue development—contributed significant receptor-ligand interactions with Tnfrsf1b, Tnfrsf1a, Tnfrsf14, Lta-(Ltb Ltbr). Our results indicate that Lt signaling interaction was predominant between B-cells and dendritic and stromal cells (Figure 7F, panels 2 and 4). We also noticed that Lt and Ccl interaction probability with their respective receptors was more significant in BCG+NTMhigh group than in NTMhigh group. Interestingly, we also observed an interaction between Xcl1 (lymphotactin) and Xcr1(lymphotactin receptor) in DCs, but only in the NTMhigh group. Cross-presenting DCs are known to express Xcr1, which binds to its ligand Xcl1 (present on effector T-cells/NK cells) and induces a cytotoxic immune response mediated by DCs49,50. In comparison, the BCG+NTMhigh group exhibited significant interactions (p<0.01) between Wnt4 and its receptors, and Tnf-Tnfrsf1a, Tnf-Tnfrsf1b, Pros1-Ax1, and Cd6-Alcam. These signaling pathways are required for immune cell maintenance, renewal, activation, proliferation, and effector/memory function regulation51-53. These findings indicate that the BCG+NTMhigh group significantly upregulates signaling pathways involved in B-cell activation and maintenance, B-cell-DC interaction, and Lt-Ltbr signaling, all of which may be involved in the lymphoid follicle and GC formation.

Discussion

Our findings suggest that exposure to viable NTM via drinking water boosts BCG protection against TB by activating B-cells and humoral immunity, a branch of the immune system not effectively induced by BCG. Using three different murine strains varying in susceptibility against TB (C3H/HeOuJ, C3HeB/FeJ, and C57BL/6), we observed substantially decreased Mtb burden and pathology in lungs of mice chronically exposed to NTM upon BCG vaccination. We developed a model to specifically address several shortcomings of previous publications, including: 1) administering heat-killed NTM intraperitoneally31, subcutaneously6, or via oral gavage31,54; 2) ceasing animal exposure to NTM upon Mtb infection6,36; 3) inconsistent time of NTM initiation, i.e., before or after BCG immunization55-59; 4) animals exposed to an unrealistic concentration of NTM (sometimes reaching 1x108 CFU/mL). These variables do not recapitulate the natural history of human-mycobacteria interaction, which we attempted to do by: 1) animals were BCG-vaccinated at an early age, before NTM exposure (humans are routinely BCG vaccinated within 48 hours of birth); 2) exposure to live, viable NTM via drinking water (without trauma), at concentrations previously reported for HIC (1000 CFU/mL) and LMIC (1.0x105 CFU/mL)44,45,60-62, and 3) sustaining continuous NTM exposure throughout Mtb infection. Our results showing the potent mucosal response induced by NTM could provide a roadmap for future TB vaccines.

Collectively, our results suggest the following scenario (Figure S5): 1) parenteral BCG elicits T-cell mediated immunity (Figure 4); 2) PPs in the gut are colonized by live NTM given orally (Figure 2); 3) within PPs, NTM potently induce memory mucosal immunity when uptaken and presented by CX3CR1+ APCs (Figure 2), activating TFH (Figure 2) and stimulating B-cell proliferation and differentiation, antibody production and isotype switching (Figure 2 and 3); 4) mucosal immunity primed in the gut seeds other mucosae including the lung, leading to antibody (IgA and IgG) production locally in BALF preceding Mtb infection (Figure 3 and 4 ); 5) upon infection with Mtb, sharing similar antigens with NTM, ectopic pulmonary GC are generated to support the expansion of memory mucosal immunity, with lymphotoxin playing an important role (Figure 5, 6 and 7); and 6) effector mechanisms including NK antibody-dependent cell-mediated cytotoxicity (ADCC) (Figure S2 and S6), control Mtb replication and limit inflammation.

While B-cells are critical in the control of infectious diseases, research on TB has mainly focused on cell-mediated immune responses39,69-79. A growing body of evidence now appreciates a protective role for B-cells and antibodies against Mtb infection80-87. Data from human studies have demonstrated the presence of anti-Mtb antibodies in the serum of latently infected and active TB patients88. Notably, antibodies and B-cells have also been observed in follicular-like formations in the lungs of Mtb-infected humans and animals89-91, displaying characteristics of B-cell aggregates and GC90,92,93, as we found in the lungs of BCG+NTMhigh mice at 120 days post-Mtb infection (Figure 5 and 6). GCs are specialized structures in which B-cells undergo somatic hypermutation and clonal selection to generate long-lived and high-affinity antibody-secreting plasma cells, as well as memory B-cells96,97. GCs have been reported in Mtb-infected mice, non-human primates, and humans over the past decade and are associated with improved Mtb containment in the lungs98. However, the involvement of NTM exposure and cross-protective B-cells in the development of GCs is yet unknown.

In our study, we found that B-cells significantly correlated (R=−0.52, p=0.0037) with reduced bacterial burden in the lungs throughout the course of infection (Figure 4C). We also found the presence of B-cell aggregates in the lungs of BCG+NTMhigh mice at 120 dpi, correlating with decreased bacterial load and pathology (Figure 5). Our DEGs and pathway analysis (spatial transcriptomics) suggest GC formation in the lungs is associated with highly upregulated genes, including Cd19, lymphotoxin alpha and beta (Lt⍺ and Ltβ), Mki67, Bcl6, Cxcr5, Cd83, Cd38, Igha, and Tnfrsf13 (a lymphotoxin beta receptor) (Figure 6 and 7). In secondary lymphoid organs, Lt⍺ and Ltβ produced by lymphoid tissue-inducer cells bind to the lymphotoxin receptor on lymphoid tissue-organizer cells, initiating a signaling cascade that generates GCs99,100. The interaction between Ltα/Ltβ on B cells and Tnfrsf13 on DCs, endothelial cells, and stromal cells using CellChat analysis suggests a possible role of lymphotoxin-mediated signaling in the development of GCs in the lungs and sheds light on the cellular and molecular processes leading to their induction.

Other cell types (CD3+ T-cells and myeloid cells) increased at 60 and 120 dpi; however, they were not directly associated with protection (Figure S7-S9). In fact, increased numbers of myeloid cells correlated with a higher bacterial burden in the lungs. While total CD3+ T cells were not directly associated with bacterial burden and pathology, subpopulations of CD3+ T cells, including CD3+CD4+CXCR5+ follicular helper T cells and CD3+CD8+CD44+Perforin+ effector T cells, significantly correlated with the reduced bacterial burden. Follicular helper T cells play a critical role in protective immunity, helping B-cells produce antibodies against foreign pathogens, and effector T cells participate in ADCC. Correlation analysis also showed a significant association of anti-Mtb IgA/IgG antibodies with reduced bacterial burden in the lungs (Figure S10). These findings suggest a major contribution of B-cells, antibodies, effector T cells, and NK cells in reducing bacterial burden in the lungs. To further investigate the mechanism(s) of protection, we conducted a GSE analysis and pathway enrichment of lung transcriptome 120 dpi (Figure S6). Results indicated that a high fraction of genes are involved in the "Natural Killer Cell-Mediated Cytotoxicity" pathway, suggesting that ADCC may act as a protective mechanism.

In conclusion, our results demonstrate that exposure to NTM via drinking water (similar to human exposure to NTM) does not diminish the efficiency of the BCG vaccine against TB but rather confers additional protection to BCG-vaccinated mice. BCG has been known to induce TB-specific cell-mediated immune responses for a long time, and our data suggest that NTM elicits a complementary, potent humoral immune response. Consequently, an optimal balance of mucosal and systemic immune responses protects against TB and disease progression. It has been postulated that mucosal immunity plays a crucial role in respiratory infection, including Mtb101-103. Mucosal antibodies, including IgA, have been extensively studied and shown to be important in protecting the host from Mtb infection104,105. However, most conventional vaccinations are intended for systemic administration, which boosts systemic immunity but induces a weaker immune response at the mucosal site of pathogen entry. Interestingly, a 2018 study by Hoft et al. reported similar findings to the ones we described above. Specifically, oral vs. intradermal BCG vaccination in humans resulted in significant IgA-mediated mucosal immunity and Th1 immunity, respectively106. Our joint findings underscore the need to develop TB vaccines that elicit systemic and mucosal protection. Based on our results, NTM could be engineered as a mucosal TB vaccine as they reach, colonize, and prime immunological responses in mucosal induction site (PPs). Further research is warranted.

Limitations of the study:

We were not able to completely rule out the possibility that NTM given orally reached the lungs, explaining the presence of memory B cells and anti-mycobacterial IgA and IgG in BALF of mice before Mtb infection. Our results using conventional microbiological techniques (plating on agar) and immunofluorescence with anti-LAM suggested otherwise (Figure S11). However, their sensitivity might not suffice to identify pauci-NTM colonization of the lungs or NTM in a non-culturable state. Our hypothesis that NTM colonization of PPs plays a key role is based on previous reports that PPs —but not spleen or lymph nodes— constitute an optimal niche conducive to the generation of higher affinity IgA-producing B-cells63-65, which then migrate and populate other mucosal sites66,67. To confirm this hypothesis, further experiments should have been performed where B cells from the PPs or lungs from BCG+NTMhigh group should be adoptively transferred to saline (control) mice and evaluate their migration to the mucosal site (including lungs) and their protective efficacy. However, due to the limitation of the samples, we could not perform this experiment.

The association between B-cells, antibodies, and protection against TB is primarily correlative and represents the second limitation of our study. Transferring protection against TB to naïve animals with serum or sorted memory B-cells obtained from NTM-exposed mice would conclusively prove this.

Star methods

RESOURCE AVAILABILITY

Lead Contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by Dr. Marcela Henao-Tamayo (Marcela.Henao_Tamayo@ColoState.EDU)

Materials availability

This study did not generate new unique reagents.

Data and code availability

Spatial transcriptomics data are available at the GEO database, and the accession number is GSE203037.

All original codes will be deposited to Github and will be publicly available as of the date of publication.

EXPERIMENTAL MODEL AND SUBJECT DETAILS

All animal studies complied with NIH guidelines and were approved by the Animal Use and Care Committee of Colorado State University. Three-week-old female C3H/HeOuJ mice were purchased from the Jackson Laboratories (Bar Harbor, ME). Animals were maintained in a BSL-3 facility at Colorado State University and had ad libitum access to water and chow.

METHOD DETAILS

Bacterial strains and culture

Mycobacterium tuberculosis HN878 was grown at 37°C in Middlebrook 7H9 liquid medium (Sigma Aldrich, cat#M0178) supplemented with 10% oleic acid-albumin-dextrose-catalase (OADC), 0.5% glycerol, and 0.05% tween 80. Mycobacterium bovis Bacille Calmette Guerin (BCG) Pasteur was cultured in a Middle brook 7H9 liquid medium. Mycobacterium avium subsp. avium (TMC 724) was purchased from ATCC (cat# 25292) and was cultured according to the manufacturer's guidelines. Briefly, the lyophilized pellet was rehydrated in 0.5 mL of 7H9 broth, then transferred in a 5 mL tube of 7H9 broth aseptically. 400uL of the 7H9 containing lyophilized pellet was plated onto 7H11 plate and incubated at 37˚C for 21 days. Single colonies were inoculated in 10 mL of 7H9 media and incubated at 37°C with constant shaking. Optical density (OD) was measured at regular intervals. When reaching an OD of 0.8, cultures were aliquoted and kept at −80˚C until used

Experimental vaccination and infections

Study 1: Study 1 was performed with three mice strains, C3H/HeOuJ, C3HeB/FeJ, and C57BL/6, using 1000 CFU/mL NTM in drinking water. Study 2: Study 2 was performed with C3H/HeOuJ mice using 1000 CFU/mL and 1.0 x 105 CFU/mL NTM in drinking water.

BCG, NTM, and Mtb regimen:

For BCG vaccination, four-week-old female C3H/HeOuJ, C3HeB/FeJ, and C57BL/6 mice were immunized with 1x105 CFU of BCG Pasteur subcutaneously. For NTM exposure, we selected a Mycobacterium avium strain (MA) in our study because of its highest prevalence among all NTM 107. Two separate water stocks containing MA at 1000 CFU/mL and 1x105 CFU/mL were prepared and administered to the mice via drinking water. Water was changed every week to maintain consistency and plated on 7H11 plates to evaluate the correct concentration (Figure S1). For low-dose Mtb aerosol infections, one mL Mtb HN878 was diluted in 7.25 ml of sterile PBS to 2 × 106 CFU/ml and placed in a nebulizer attached to an airborne infection system (Glass-Col, Terre Haute, IN, USA). Mice were exposed to an aerosol infection to deposit approximately 50-60 CFU in the lungs of each mouse. After infection, five mice were euthanized, and whole lungs were plated onto 7H11 plates to evaluate the CFUs in the lungs.

Enumeration of Mycobacterial burden in lungs.

The lungs of mice were homogenized using a Blender homogenizer (Next Advance, cat# BT24M) set with a speed of eight for four minutes, with 500 μL of PBS (Corning, cat# MT21040CV) containing four 3.2 mm stainless steel beads (Next Advance, cat# SSB32). To ensure complete cell lysis, initial organ homogenates were further homogenized by transferring 100 μL of the initial homogenate to new 2.0 mL conical screw cap (Cole Parmer, cat# EW-06245-51) tubes with 400 μL of 0.05% Tween80 (Sigma Aldrich, cat# P5188), PBS (Corning cat# MT21040CV) and with 2.0 mm zirconium beads (Next Advance, cat# ZROB20) using a homogenizer (Benchmark Scientific Inc., cat# D1030) for ten seconds at 2800 rpm. After complete homogenization, serial dilutions were performed in 96 well plates (Falcon, cat# 353227) with a dilution factor of 5, and 100 μL of each dilution was plated on 7H11 agar plates. CFUs were enumerated by counting the colonies after 28 days of incubation at 37°C.

Isolation of cells and preparation of single-cell suspension

Mice were euthanized by CO2 asphyxiation, the thoracic cavity was opened, and whole blood was collected via cardiac puncture. An equal volume of PBS was added to the blood and centrifuged at 800× g for 10 min at 25˚C with the brake-off. The buffy coat was collected, washed, and erythrocytes were lysed using 1× Miltenyi RBC lysis buffer (Miltenyi, CA, USA). Cells were washed and resuspended in one mL of freezing media and saved at −80˚C. Bronchoalveolar lavage (BAL) was harvested and centrifuged at 1300 x g for 10 minutes, and supernatant and cell (pellets) were saved separately at −80˚C without and with freezing media, respectively. Subsequently, the lungs were isolated 108 and treated with DNase IV (500 units/mL, cat# D5025) solution and Liberase (0.5 mg/mL, cat# 5401127001) for 30 min at 37˚C to dissociate and digest pulmonary collagen. Cells were homogenized, passed through a 70 μm filter, and erythrocytes were lysed using Gey's RBC lysis buffer (Sigma Aldrich, cat#R7757). Lung cells were resuspended in 1 mL of Dulbecco's modified Eagle's minimal essential medium (Corning, cat#15-017-CV). For Peyer's patches (PPs) isolation, the abdominal cavity was opened, and the whole small intestine and large intestine were removed. The intestine was placed in a 12mm round plate with complete media. The PPs were harvested from the intestines and placed into tubes for flow cytometry and histopathology/immunofluorescence staining. Each mouse generally has 5-6 PPs each. For flow cytometry, PPs cells were incubated with DNase IV (500 units/mL) and Liberase (0.5 mg/mL) for 30 min at 37˚C and homogenized using a syringe plunger and passed through a 70 μm filter to prepare a single-cell suspension.

Flow cytometry

Lungs and PPs cells were counted using the method described previously by 108. Cells were seeded at 1 x 106/well of a 96-well V-bottom plate (Greiner Bio-One, cat#651-180). For panels including surface and intracellular markers, cells were incubated with Brefeldin A (Biolegend, cat# 420601) (1x prepared in complete media) for 6 hours in a CO2 incubator (VWR) at 37˚C. For surface markers staining, cells were incubated with 100 μl of Zombie NIR (Biolegend, cat# 423106) viability dye (1:2000 dilution prepared in PBS) for 15 minutes in the dark. Fc receptors were blocked using an anti-mouse CD16/32 antibody (Biolegend, cat# 156604) (1:100 dilution) and stained with predetermined optimal concentrations of specific antibodies (Tables S1 and S2) for 30 minutes in the dark at 4˚C. For intracellular cytokines and transcription factors staining, cells were further incubated with 1x Foxp3 Perm/Fix buffer (eBiosciences, cat# 00-5523-00) for 1 hour at 37°C, washed with 1x permeabilization buffer (eBiosciences, cat#00-5523-00), and incubated with intracellular antibody cocktail (prepared in 1x permeabilization buffer) overnight at 4˚C. The next day, cells were washed twice, resuspended in 300 μL of 1x permeabilization buffer, and acquired using a Cytek Aurora spectral flow cytometer (Cytek Biosciences), where 50,000 events of PPs and 100,000 events of lungs were recorded. Data were analyzed using manual gating and cyto-feature engineering pipeline 109.

Enzyme-linked Immunosorbent assay:

An enzyme-linked immunosorbent assay (ELISA) was performed to evaluate the presence of anti-Mtb antibodies in the serum and BAL. High binding 96-half-well microplates (Corning Life Sciences, cat#3690) were coated with 100 ng of Mtb HN878 lysate (BEI, cat# NR-14824) prepared in PBS and incubated overnight at 4˚C. The next day, plates were washed five times with 180 μL of wash buffer (PBS + 0.05% Tween-20), and non-specific interactions were blocked using 180 μL of blocking buffer (PBS + 0.05% Tween-20 + 2% BSA + 2% normal goat serum [Jackson ImmunoResearch Inc., cat#005-000-121 West Grove, PA, USA]). After 2 hours, plates were washed, and different dilutions of serum and BAL prepared in blocking buffer were added to the wells and incubated for 1 hour. Plates were then washed and incubated for 1 hour with horseradish peroxidase (HRP)-conjugated anti-mouse IgG (Jackson ImmunoResearch Inc, cat# 115-035-003) or anti-mouse IgA secondary antibodies (Southern Biotech, cat# 1040-05) prepared in blocking buffer. The colorimetric substrate was developed with the addition of 100 μl of TMB substrate (Thermo Fisher Scientific, Rockford, cat# ENN301), and the reaction was stopped by adding 50 μl of 1 M sulphuric acid. Absorbance was measured at 450 nm using a BioTek Synergy 2 plate reader (BioTek Instruments Inc., Winooski, VT, USA).

Histopathological evaluation and lesion scoring

The extent of lesion burden was analyzed by lung histopathology and expressed as the proportion of lesion area over total lung area per stained tissue section. Stained slides were blinded and analyzed by a veterinary pathologist (Dr. Brendan Podell, Colorado State University). H&E-stained sections were scanned at 20X magnification using an Olympus VS120 microscope, Hamamatsu ORCA-R2 camera, and Olympus VS-ASW 2.9 software. Visiopharm software was used for image analysis. For each tissue section, a region of interest (ROI) was generated at low magnification with a custom tissue detecting algorithm using decision forest training and classification to differentiate tissue versus background based on color and area. Lesions were identified within tissue ROIs at high magnification with an additional custom-made algorithm using decision forest training and classification based on staining intensity, color normalization and deconvolution, area, and morphological features. Percent lesion calculations were integrated into the same algorithm and calculated from tissue area and lesion area as designated by the ROI and lesions detected. Lesion identification and quantification were then reviewed and edited by a pathologist as needed.

Multiplex Immunohistochemistry

For multiplexed immunohistochemistry (mIHC), formalin-fixed and paraffin-embedded (FFPE) sections were cut at 5 μm and stained using the Opal Polaris 4 Color Kit (Akoya Biosciences Inc. cat# NEL830001KT) with 4-plex staining. Five μm lung sections were stained using the OPAL multiplex IHC (mIHC) staining technology using the LabSat Research Automated staining instrument (Lunaphore Technologies SA, Epedria). Slides were scanned using the multispectral camera of the Polaris System (Akoya bioscience) and analyzed using the inForm tissue Finder and Phenochart softwares. The panel of antibodies used was anti-LAM antibody (opal 690), anti-mouse B220 antibody (opal 570), and anti-mouse CD4 antibody (opal 520). mIHC were finally stained using the OPAL multiplex staining technology and the Leica Bond III autostainer by Imaging Core at the University of Colorado, Anschutz Medical Campus, Denver. Images were acquired using the multispectral camera of the Polaris System (Akoya Biosciences Inc.) and analyzed using the inForm tissue Finder and phenochart softwares.

Sample preparation, cDNA amplification, and sequencing for spatial transcriptomics

Sectioning and tissue placement:

FFPE lung tissue blocks from saline, NTMhigh, BCG, and BCG+NTMhigh groups were soaked in ice-cold water for 2 hours, and 5 μm sections were cut using Leica microtome. Another 10 μm section was subjected to RNA quality assessment using Qiagen RNAeasy FFPE kit and evaluating the DV200 value. Sections were collected in the water bath and allowed to float on the water surface until they were flat and free from wrinkles. Sections were then placed within the frames of capture areas on Visium spatial slides. Slides were then dried at 42°C in an oven and left in a desiccator overnight to ensure complete dehydration.

Deparaffinization, histological staining, imaging, and decrosslinking:

Slides were retrieved from the desiccator and placed at 60°C in the oven for 2 hours, cooled, and deparaffinized using xylene. Slides were washed using 100%, 96%, and 70% alcohol, respectively, and stained with Hematoxylin (Leica, cat# 3801570) and Eosin (Leica, cat# 3801570). Imaging of the sections was performed using a KEYENCE BZ-X710 microscope (KEYENCE). For decrosslinking of the sections, slides were washed with 0.1N HCl, added 100uL of TE buffer (pH 9.0), and incubated at 70°C in a thermocycler for 1 hour.

Probe hybridization, probe ligation, and probe extension and release:

After decrosslinking, probes for the mouse whole transcriptome, consisting of a pair of specific probes for each targeted gene, were hybridized to the sections at 50°C overnight in a thermocycler. Slides were washed, and a ligation step was performed to seal the junction between the probe pairs hybridized to RNA by adding probe ligation enzyme and incubating at 37°C for 1 hour in a thermocycler. The tissue sections were then enzymatically permeabilized for 40 minutes at 37°C, and probes were extended by adding UMI, spatial barcode, and partial read 1.

Library preparation and sequencing:

Released ligated probe products were harvested from the slides and further processed to generate cDNA libraries for sequencing. To determine the sample index PCR cycle number, a qPCR was performed using Kappa Sybr master mix and primers (Roche, cat# 07959389001). The cDNA was then indexed and amplified. The indexed libraries were then cleaned, and size selected using SPRIselect (Beckman Coulter, cat# B23318). The average size of the indexed cDNA libraries was determined with an Agilent HS D1000 tape (Agilent, cat# 5067-5584) using Agilent tapestation (Agilent, cat# G2991BA). The molarity and concentrations of each library were measured using a qPCR, and libraries were pooled and diluted to 4 nM. Paired-end sequencing was performed on the Illumina NextSeq500 platform at 1.8pM loading concentration, with 29 bases from read 1 and 51 bases from read 2. A total of 80-100 million read pairs per sample were generated.

Spatial transcriptomics computational analysis

Processing raw reads and demultiplexing was performed using 10X Genomics’ Space Ranger (version 1.31.1) mkfastq. Demultiplexed samples were mapped to the annotated mouse whole transcriptome (mm10) targeted gene expression analysis using Space Ranger count pipeline. Filtered reads of individual samples were converted to Seurat object and normalized using SCTransform function 110. Each section was analyzed separately and as a merged object to evaluate the differences between the two. Variable features of integrated data were assessed, and principal component analyses were performed. Clusters were identified using FindNeighbors and FindClusters functions and represented as Uniform Manifold Approximation and Projection (UMAP). Differentially expressed genes (DEGs) in each cluster were identified using FindMarkers from the Seurat package 111, and cell types were assigned via SingleR package 112 using ImmGenData reference in the Celldex package. Cell-cell interaction for each group was performed using CellChat, and DEGs in each group were evaluated using DESeq2. Gene enrichment analysis of DEGs of each group was performed using clusterprofiler. We extracted the gene symbols of each group and subjected them to KEGG pathway analysis using mus musculus data. The top 25 pathways were visualized, and enrichment scores were represented as the positive and negative log10 values of the corrected p-value.

Quantification and Statistical Analyses

The experiment was performed on three separate occasions, the last repetition used C3H/HeOuJ only. The sample size was calculated by differentiation of proportions of 0.9 vs. 0.1 at a power of 80% (Fisher's exact test). Group means (n = 5) CFU, total cell counts of different populations, and ELISA were analyzed using Tukey-HSD two-way ANOVA analysis to analyze differences between groups. Data were considered significant if p < 0.05. Data analysis and plotting were performed using R studio (version 4.1.2). Mean (n = 5) subjective pathology scores were compared between groups using the Kruskal-Wallis test for non-parametric data with an alpha of 0.05.

Supplementary Material

Supplementary data

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
Anti-mouse CD11c Alexa-fluor 532 ThermoFisher Cat# 58-0116-41; RRID: AB_11218876
Anti-mouse A4b7 integrin PE Biolegend Cat# 120606; RRID: AB_493267
Anti-mouse CD62L PE/Dazzle 594 Biolegend Cat# 104448; RRID: AB_2566163
Anti-mouse CXCR5 PerCP-eFluor 710 ThermoFisher Cat# 46-7185-82; RRID: AB_2573837
Anti-mouse B220 PE/Cy7 BD Biosciences Cat# 552772; RRID: AB_394458
Anti-mouse CD73 PerCP-Cy5.5 Biolegend Cat# 127214; RRID: AB_11219403
Anti-mouse CD34 BV421 Biolegend Cat# 119321; RRID: AB_10900980
Anti-mouse CD11b Pacific Blue Biolegend Cat# 101224; RRID: AB_755986
Anti-mouse MHC-II BV480 BD Biosciences Cat# 566088; RRID: AB_2869739
Anti-mouse CD3 BV510 Biolegend Cat# 100234; RRID: AB_2562555
Anti-mouse CD45 BV570 Biolegend Cat# 103136; RRID: AB_2562612
Anti-mouse CX3CR1 BV605 Biolegend Cat# 149027; RRID: AB_2565937
Anti-mouse CD8a BV650 Biolegend Cat# 100742; RRID: AB_2563056
Anti-mouse CD138 BV785 Biolegend Cat# 142534; RRID: AB_2814047
Anti-mouse NKp46 BV711 Biolegend Cat# 137621; RRID: AB_2563289
Anti-mouse NKM-16-2-4 APC Miltenyi Biotec Cat# 130-102-149; RRID: AB_2660295
Anti-mouse CCR9 AF-647 Biolegend Cat# 129710; RRID: AB_2073247
Anti-mouse CD44 APC/Fire 750 Biolegend Cat# 103062; RRID: AB_2616727
Anti-mouse CD4 AF-700 Biolegend Cat# 100430; RRID: AB_4936999
Anti-mouse Ly6G PerCP Biolegend Cat# 127654; RRID: AB_2616999
Anti-mouse CD27 BV650 Biolegend Cat# 124233; RRID: AB_2687192
Anti-mouse CCR2 BV711 Biolegend Cat# 357232; RRID: AB_2800970
Anti-mouse CD19 BV605 Biolegend Cat# 115540; RRID: AB_2563067
Anti-mouse Ly6C AF700 Biolegend Cat# 128024; RRID: AB_10643270
Anti-mouse IL-10 FITC Biolegend Cat# 505006; RRID: AB_315360
Anti-mouse CD4 AF-532 ThermoFisher Cat# 58-0042-82; RRID: AB_11218891
Anti-mouse Perforin PE Biolegend Cat# 154406; RRID: AB_2721641
Anti-mouse PD-1 PE/Cy7 Biolegend Cat# 109110; RRID: AB_572017
Anti-mouse IL-12 PerCP-Cy5.5 Biolegend Cat# 505212; RRID: AB_2566225
Anti-mouse IFNγ Pacific Blue Biolegend Cat# 505818; RRID: AB_893526
Anti-mouse CD8 BV570 Biolegend Cat# 100740; RRID: AB_2563055
Anti-mouse IL-4 BV605 Biolegend Cat# 504126; RRID: AB_2686971
Anti-mouse CD3 BV605 Biolegend Cat# 100237; RRID: AB_2562039
Anti-mouse KLRG1 BV786 Biolegend Cat# 138429; RRID: AB_2629749
Anti-mouse TGFbeta BV421 Biolegend Cat# 141408; RRID: AB_2650898
Anti-mouse Granzyme-B AF-647 Biolegend Cat# 515406; RRID: AB_2566333
Anti-mouse Bcl-6 APC Biolegend Cat# 358506; RRID: AB_2562472
Peroxidase AffiniPure Goat Anti-Mouse IgG (H+L) Jackson Immunoresearch Cat# 115-035-003
Goat anti-mouse IgA HRP Southern Biotech Cat# 1040-05
Anti-rat IgG, HRP-linked Antibody CellSignaling/Fisher Cat# 7077
Anti-mouse B220 (mIHC) BDPharma Cat# 550286; RRID: AB_393581
Anti-mouse CD4 (mIHC) Invitrogen Cat# 14-9766-82; RRID: AB_2573008
Anti-mouse GL7 (mIHC) Invitrogen Cat# 14-5902-82; RRID: AB_467715
Bacterial and virus strains
Mycobacterium tuberculosis HN878 In house Legacy of Dr. Ian Orme’s lab
Mycobacterium bovis, BCG Pasteur TMC#1011 A kind gift from Dr. Angelo Izzo Bickett et. al., 2020113
Mycobacterium avium (TMC 724) ATCC Cat# 25292
Mycobacterium tuberculosis HN878 whole cell lysate NIH BEI Cat# NR-14824
Chemicals, peptides, and recombinant proteins
TMB Thermo Fisher Scientific cat# ENN301
Hematoxylin Leica Biosystems Cat# 3801570
Eosin Leica Biosystems Cat# 3801615
Opal Polaris 4 Color Kit Akoya Biosciences Cat# NEL830001KT
Deposited data
Spatial Transcriptomics 10X Genomics NCBI GEO database Accession number: GSE203037
Experimental models: Organisms/strains
C3H/HeOuJ The Jackson Laboratory JAX:000635
C57BL/6 The Jackson Laboratory JAX: 000664
C3HeB/FeJ The Jackson Laboratory JAX: 000658
Software and algorithms
FlowJo BD Biosciences RRID: SCR_009520
Rstudio (Version 4.1.2) Open Software RRID: SCR_000432
Cytotypr In-house remotes::install_github("aef1004/cytotypr")
Spatial transcriptomics analyzed data Seurat, SingleR, CellChat, and in house codes https://github.com/tarudutt/NTM_manuscript_data_analysis DOI: 10.5281/zenodo.7242921
Phenochart Akoya Biosciences Version 1.0.12 www.akoyabio.com
inForm® Akoya Biosciences Version 2.4.8 www.akoyabio.com

Acknowledgment:

We want to thank the Otsuka Pharmaceutical CO. Ltd for providing us with anti-LAM antibody (OTB). We would also like to thank CSU Flow Cytometry Core (RRID: SCR_022000) and Anschutz Research Histology service, University of Colorado, School of Medicine. We are grateful to Dr. Mark Stenglein for providing access to the CSU's cluster server and guiding us through essential aspects of RNA-seq data analysis. We would also like to thank Marylee Kapuscinski for providing training in sequencing.

Funding:

This research was supported by NIH grant R01AI127475, R21AI121099 (NIH-NIAID), and Colorado State University's Microbiology, Immunology, and Pathology funding. This research was also supported by grant S10OD016226 by the Office of the Director of the National Institutes of Health.

Footnotes

Declaration of interests: The authors declare no competing interests.

References

  • 1.Ratnatunga CN, Lutzky VP, Kupz A, Doolan DL, Reid DW, Field M, Bell SC, Thomson RM, and Miles JJ (2020). The Rise of Non-Tuberculosis Mycobacterial Lung Disease. Front. Immunol 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Desai AN, and Hurtado R (2021). Nontuberculous Mycobacterial Infections. JAMA 325, 1574. 10.1001/jama.2020.19062. [DOI] [PubMed] [Google Scholar]
  • 3.Porvaznik I, Solovič I, and Mokrý J (2017). Non-Tuberculous Mycobacteria: Classification, Diagnostics, and Therapy. Adv. Exp. Med. Biol 944, 19–25. 10.1007/5584_2016_45. [DOI] [PubMed] [Google Scholar]
  • 4.To K, Cao R, Yegiazaryan A, Owens J, and Venketaraman V (2020). General Overview of Nontuberculous Mycobacteria Opportunistic Pathogens: Mycobacterium avium and Mycobacterium abscessus. J. Clin. Med 9, 2541. 10.3390/jcm9082541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Andersen P, and Doherty TM (2005). The success and failure of BCG — implications for a novel tuberculosis vaccine. Nat. Rev. Microbiol 3, 656–662. 10.1038/nrmicro1211. [DOI] [PubMed] [Google Scholar]
  • 6.Brandt L, Feino Cunha J, Weinreich Olsen A, Chilima B, Hirsch P, Appelberg R, and Andersen P (2002). Failure of the Mycobacterium bovis BCG Vaccine: Some Species of Environmental Mycobacteria Block Multiplication of BCG and Induction of Protective Immunity to Tuberculosis. Infect. Immun 70, 672–678. 10.1128/IAI.70.2.672-678.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Shah JA, Lindestam Arlehamn CS, Horne DJ, Sette A, and Hawn TR (2019). Nontuberculous Mycobacteria and Heterologous Immunity to Tuberculosis. J. Infect. Dis 220, 1091–1098. 10.1093/infdis/jiz285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Global Tuberculosis Report. 2021 https://www.who.int/teams/global-tuberculosis-programme/tb-reports/global-tuberculosis-report-2021. [Google Scholar]
  • 9.Chakaya J, Khan M, Ntoumi F, Aklillu E, Fatima R, Mwaba P, Kapata N, Mfinanga S, Hasnain SE, Katoto PDMC, et al. (2021). Global Tuberculosis Report 2020 – Reflections on the Global TB burden, treatment and prevention efforts. Int. J. Infect. Dis 113, S7–S12. 10.1016/j.ijid.2021.02.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Kaufmann SHE (2021). Vaccine Development Against Tuberculosis Over the Last 140 Years: Failure as Part of Success. Front. Microbiol 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Matteelli A, Rendon A, Tiberi S, Al-Abri S, Voniatis C, Carvalho ACC, Centis R, D’Ambrosio L, Visca D, Spanevello A, et al. (2018). Tuberculosis elimination: where are we now? Eur. Respir. Rev 27. 10.1183/16000617.0035-2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Sotgiu G, Centis R, D’ambrosio L, and Migliori GB (2015). Tuberculosis Treatment and Drug Regimens. Cold Spring Harb. Perspect. Med 5, a017822. 10.1101/cshperspect.a017822. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Madhav B, Iyer A, and Jayalakshmi TK (2015). Side effect profile of 2nd line drugs in multi drug resistant (MDR) and extensively drug resistant (XDR) tuberculosis. Eur. Respir. J 46. 10.1183/13993003.congress-2015.PA2708. [DOI] [Google Scholar]
  • 14.Marais BJ, Chakaya J, Swaminathan S, Fox GJ, Ehtesham NZ, Ntoumi F, Zijenah L, Maurer M, and Zumla A (2020). Tackling long-term morbidity and mortality after successful tuberculosis treatment. Lancet Infect. Dis 20, 641–642. 10.1016/S1473-3099(20)30167-5. [DOI] [PubMed] [Google Scholar]
  • 15.Wang J, Xiong K, Zhao S, Zhang C, Zhang J, Xu L, and Ma A (2020). Long-Term Effects of Multi-Drug-Resistant Tuberculosis Treatment on Gut Microbiota and Its Health Consequences. Front. Microbiol 11, 53. 10.3389/fmicb.2020.00053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wipperman MF, Fitzgerald DW, Juste MAJ, Taur Y, Namasivayam S, Sher A, Bean JM, Bucci V, and Glickman MS (2017). Antibiotic treatment for Tuberculosis induces a profound dysbiosis of the microbiome that persists long after therapy is completed. Sci. Rep 7, 10767. 10.1038/s41598-017-10346-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Dalberto PF, de Souza EV, Abbadi BL, Neves CE, Rambo RS, Ramos AS, Macchi FS, Machado P, Bizarro CV, and Basso LA (2020). Handling the Hurdles on the Way to Anti-tuberculosis Drug Development. Front. Chem 8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Dockrell HM, and Smith SG (2017). What Have We Learnt about BCG Vaccination in the Last 20 Years? Front. Immunol 8. 10.3389/fimmu.2017.01134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Henao-Tamayo M, Shanley CA, Verma D, Zilavy A, Stapleton MC, Furney SK, Podell B, and Orme IM (2015). The Efficacy of the BCG Vaccine against Newly Emerging Clinical Strains of Mycobacterium tuberculosis. PLOS ONE 10, e0136500. 10.1371/journal.pone.0136500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Dheda K, Perumal T, Moultrie H, Perumal R, Esmail A, Scott AJ, Udwadia Z, Chang KC, Peter J, Pooran A, et al. (2022). The intersecting pandemics of tuberculosis and COVID-19: population-level and patient-level impact, clinical presentation, and corrective interventions. Lancet Respir. Med 0. 10.1016/S2213-2600(22)00092–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Pai M, Kasaeva T, and Swaminathan S (2022). Covid-19’s Devastating Effect on Tuberculosis Care — A Path to Recovery. N. Engl. J. Med 0, null. 10.1056/NEJMp2118145. [DOI] [PubMed] [Google Scholar]
  • 22.CDCTB (2021). Tuberculosis (TB)- Vaccine. Cent. Dis. Control Prev https://www.cdc.gov/tb/topic/basics/vaccines.htm. [Google Scholar]
  • 23.Davenne T, and McShane H (2016). Why don’t we have an effective tuberculosis vaccine yet? Expert Rev. Vaccines 15, 1009–1013. 10.1586/14760584.2016.1170599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Prentice S, and Dockrell HM (2020). Antituberculosis BCG vaccination: more reasons for varying innate and adaptive immune responses. J. Clin. Invest 130, 5121–5123. 10.1172/JCI141317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ritz N, Hanekom WA, Robins-Browne R, Britton WJ, and Curtis N (2008). Influence of BCG vaccine strain on the immune response and protection against tuberculosis. FEMS Microbiol. Rev 32, 821–841. 10.1111/j.1574-6976.2008.00118.x. [DOI] [PubMed] [Google Scholar]
  • 26.Zhang L, Ru H-W, Chen F-Z, Jin C-Y, Sun R-F, Fan X-Y, Guo M, Mai J-T, Xu W-X, Lin Q-X, et al. (2016). Variable Virulence and Efficacy of BCG Vaccine Strains in Mice and Correlation With Genome Polymorphisms. Mol. Ther. J. Am. Soc. Gene Ther 24, 398–405. 10.1038/mt.2015.216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Keyser A, Troudt JM, Taylor JL, and Izzo AA (2011). BCG sub-strains induce variable protection against virulent pulmonary Mycobacterium tuberculosis infection, with the capacity to drive Th2 immunity. Vaccine 29, 9308–9315. 10.1016/j.vaccine.2011.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ordway DJ, Shang S, Henao-Tamayo M, Obregon-Henao A, Nold L, Caraway M, Shanley CA, Basaraba RJ, Duncan CG, and Orme IM (2011). Mycobacterium bovis BCG-Mediated Protection against W-Beijing Strains of Mycobacterium tuberculosis Is Diminished Concomitant with the Emergence of Regulatory T Cells▿. Clin. Vaccine Immunol. CVI 18, 1527–1535. 10.1128/CVI.05127-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Nikonenko BV, Logunova NN, Sterzhanova NV, Kayukova SI, and Apt AS (2021). Efficacy of BCG Vaccination Depends on Host Genetics. Bull. Exp. Biol. Med 171, 445–448. 10.1007/s10517-021-05246-9. [DOI] [PubMed] [Google Scholar]
  • 30.Smith CM, Proulx MK, Olive AJ, Laddy D, Mishra BB, Moss C, Gutierrez NM, Bellerose MM, Barreira-Silva P, Phuah JY, et al. (2016). Tuberculosis Susceptibility and Vaccine Protection Are Independently Controlled by Host Genotype. mBio 7, e01516–16. 10.1128/mBio.01516-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Poyntz HC, Stylianou E, Griffiths KL, Marsay L, Checkley AM, and McShane H (2014). Non-tuberculous mycobacteria have diverse effects on BCG efficacy against Mycobacterium tuberculosis. Tuberculosis 94, 226–237. 10.1016/j.tube.2013.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Price DN, Kusewitt DF, Lino CA, McBride AA, and Muttil P (2016). Oral Tolerance to Environmental Mycobacteria Interferes with Intradermal, but Not Pulmonary, Immunization against Tuberculosis. PLOS Pathog. 12, e1005614. 10.1371/journal.ppat.1005614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Orme IM, and Collins FM (1984). Efficacy of Mycobacterium bovis BCG vaccination in mice undergoing prior pulmonary infection with atypical mycobacteria. Infect. Immun 44, 28–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Orme IM, Roberts AR, and Collins FM (1986). Lack of evidence for a reduction in the efficacy of subcutaneous BCG vaccination in mice infected with nontuberculous mycobacteria. Tubercle 67, 41–46. 10.1016/0041-3879(86)90030-9. [DOI] [PubMed] [Google Scholar]
  • 35.Palmer CE, and Long MW (1966). Effects of infection with atypical mycobacteria on BCG vaccination and tuberculosis. Am. Rev. Respir. Dis 94, 553–568. 10.1164/arrd.1966.94.4.553. [DOI] [PubMed] [Google Scholar]
  • 36.Flaherty DK, Vesosky B, Beamer GL, Stromberg P, and Turner J (2006). Exposure to Mycobacterium avium can modulate established immunity against Mycobacterium tuberculosis infection generated by Mycobacterium bovis BCG vaccination. J. Leukoc. Biol 80, 1262–1271. 10.1189/jlb.0606407. [DOI] [PubMed] [Google Scholar]
  • 37.Dowdell K, Haig S-J, Caverly LJ, Shen Y, LiPuma JJ, and Raskin L (2019). Nontuberculous Mycobacteria in Drinking Water Systems – The Challenges of Characterization and Risk Mitigation. Curr. Opin. Biotechnol 57, 127–136. 10.1016/j.copbio.2019.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Manca C, Tsenova L, Bergtold A, Freeman S, Tovey M, Musser JM, Barry CE, Freedman VH, and Kaplan G (2001). Virulence of a Mycobacterium tuberculosis clinical isolate in mice is determined by failure to induce Th1 type immunity and is associated with induction of IFN-α/β. Proc. Natl. Acad. Sci 98, 5752–5757. 10.1073/pnas.091096998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ordway D, Henao-Tamayo M, Harton M, Palanisamy G, Troudt J, Shanley C, Basaraba RJ, and Orme IM (2007). The Hypervirulent Mycobacterium tuberculosis Strain HN878 Induces a Potent TH1 Response followed by Rapid Down-Regulation. J. Immunol 179, 522–531. 10.4049/jimmunol.179.1.522. [DOI] [PubMed] [Google Scholar]
  • 40.Bobba S, Choreño-Parra JA, Rangel-Moreno J, Ahmed M, Mehra S, Rosa B, Martin J, Mitreva M, Kaushal D, Zúñiga J, et al. (2020). Mice infected with the hypervirulent Mycobacterium tuberculosis HN878 strain develop lung lesions resembling human tubercle granulomas. J. Immunol 204, 156.20–156.20. [Google Scholar]
  • 41.Henao-Tamayo M, Obregón-Henao A, Creissen E, Shanley C, Orme I, and Ordway DJ (2015). Differential Mycobacterium bovis BCG Vaccine-Derived Efficacy in C3Heb/FeJ and C3H/HeOuJ Mice Exposed to a Clinical Strain of Mycobacterium tuberculosis. Clin. Vaccine Immunol. CVI 22, 91–98. 10.1128/CVI.00466-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ordway DJ, Shang S, Henao-Tamayo M, Obregon-Henao A, Nold L, Caraway M, Shanley CA, Basaraba RJ, Duncan CG, and Orme IM (2011). Mycobacterium bovis BCG-Mediated Protection against W-Beijing Strains of Mycobacterium tuberculosis Is Diminished Concomitant with the Emergence of Regulatory T Cells▿. Clin. Vaccine Immunol. CVI 18, 1527–1535. 10.1128/CVI.05127-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Honda JR, Virdi R, and Chan ED (2018). Global Environmental Nontuberculous Mycobacteria and Their Contemporaneous Man-Made and Natural Niches. Front. Microbiol 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Spaulding AB, Lai YL, Zelazny AM, Olivier KN, Kadri SS, Prevots DR, and Adjemian J (2017). Geographic Distribution of Nontuberculous Mycobacterial Species Identified among Clinical Isolates in the United States, 2009–2013. Ann. Am. Thorac. Soc 14, 1655–1661. 10.1513/AnnalsATS.201611-860OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zulu M, Monde N, Nkhoma P, Malama S, and Munyeme M (2021). Nontuberculous Mycobacteria in Humans, Animals, and Water in Zambia: A Systematic Review. Front. Trop. Dis 2. [Google Scholar]
  • 46.Sigal GB, Pinter A, Lowary TL, Kawasaki M, Li A, Mathew A, Tsionsky M, Zheng RB, Plisova T, Shen K, et al. (2018). A Novel Sensitive Immunoassay Targeting the 5-Methylthio-d-Xylofuranose-Lipoarabinomannan Epitope Meets the WHO’s Performance Target for Tuberculosis Diagnosis. J. Clin. Microbiol 56, e01338–18. 10.1128/JCM.01338-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jin S, Guerrero-Juarez CF, Zhang L, Chang I, Ramos R, Kuan C-H, Myung P, Plikus MV, and Nie Q (2021). Inference and analysis of cell-cell communication using CellChat. Nat. Commun 12, 1088. 10.1038/s41467-021-21246-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Castro RC, Zambuzi FA, Fontanari C, de Morais FR, Bollela VR, Kunkel SL, Schaller MA, and Frantz FG (2020). NOTCH1 and DLL4 are involved in the human tuberculosis progression and immune response activation. Tuberc. Edinb. Scotl 124, 101980. 10.1016/j.tube.2020.101980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kroczek R, and Henn V (2012). The Role of XCR1 and its Ligand XCL1 in Antigen Cross-Presentation by Murine and Human Dendritic Cells. Front. Immunol 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ordway D, Higgins DM, Sanchez-Campillo J, Spencer JS, Henao-Tamayo M, Harton M, Orme IM, and Gonzalez Juarrero M (2007). XCL1 (lymphotactin) chemokine produced by activated CD8 T cells during the chronic stage of infection with Mycobacterium tuberculosis negatively affects production of IFN-gamma by CD4 T cells and participates in granuloma stability. J. Leukoc. Biol 82, 1221–1229. 10.1189/jlb.0607426. [DOI] [PubMed] [Google Scholar]
  • 51.Ampudia J, Young-Greenwald WW, Badrani J, Gatto S, Pavlicek A, Doherty T, Connelly S, and Ng CT (2020). CD6-ALCAM signaling regulates multiple effector/memory T cell functions. J. Immunol 204, 150.13–150.13. [Google Scholar]
  • 52.Wajant H, and Siegmund D (2019). TNFR1 and TNFR2 in the Control of the Life and Death Balance of Macrophages. Front. Cell Dev. Biol 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zhang Q, Pan Y, Ji J, Xu Y, Zhang Q, and Qin L (2021). Roles and action mechanisms of WNT4 in cell differentiation and human diseases: a review. Cell Death Discov. 7, 1–10. 10.1038/s41420-021-00668-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Weir RE, Black GF, Nazareth B, Floyd S, Stenson S, Stanley C, Branson K, Sichali L, Chaguluka SD, Donovan L, et al. (2006). The influence of previous exposure to environmental mycobacteria on the interferon-gamma response to bacille Calmette–Guérin vaccination in southern England and northern Malawi. Clin. Exp. Immunol 146, 390–399. 10.1111/j.1365-2249.2006.03222.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Buddle BM, Wards BJ, Aldwell FE, Collins DM, and de Lisle GW (2002). Influence of sensitisation to environmental mycobacteria on subsequent vaccination against bovine tuberculosis. Vaccine 20, 1126–1133. 10.1016/s0264-410x(01)00436-4. [DOI] [PubMed] [Google Scholar]
  • 56.Demangel C, Garnier T, Rosenkrands I, and Cole ST (2005). Differential effects of prior exposure to environmental mycobacteria on vaccination with Mycobacterium bovis BCG or a recombinant BCG strain expressing RD1 antigens. Infect. Immun 73, 2190–2196. 10.1128/IAI.73.4.2190-2196.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Howard CJ, Kwong LS, Villarreal-Ramos B, Sopp P, and Hope JC (2002). Exposure to Mycobacterium avium primes the immune system of calves for vaccination with Mycobacterium bovis BCG. Clin. Exp. Immunol 130, 190–195. 10.1046/j.1365-2249.2002.01987.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Orme IM, and Collins FM (1984). Efficacy of Mycobacterium bovis BCG vaccination in mice undergoing prior pulmonary infection with atypical mycobacteria. Infect. Immun 44, 28–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Thom M, Howard C, Villarreal-Ramos B, Mead E, Vordermeier M, and Hope J (2008). Consequence of prior exposure to environmental mycobacteria on BCG vaccination and diagnosis of tuberculosis infection. Tuberc. Edinb. Scotl 88, 324–334. 10.1016/j.tube.2007.12.002. [DOI] [PubMed] [Google Scholar]
  • 60.Arfaatabar M, Karami P, and Khaledi A (2021). An update on prevalence of slow-growing mycobacteria and rapid-growing mycobacteria retrieved from hospital water sources in Iran – a systematic review. Germs 11, 97–104. 10.18683/germs.2021.1245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Donohue MJ, Mistry JH, Donohue JM, O’Connell K, King D, Byran J, Covert T, and Pfaller S (2015). Increased Frequency of Nontuberculous Mycobacteria Detection at Potable Water Taps within the United States. Environ. Sci. Technol 49, 6127–6133. 10.1021/acs.est.5b00496. [DOI] [PubMed] [Google Scholar]
  • 62.Monde N, Munyeme M, Muwonge A, Muma JB, and Malama S (2018). Characterization of non-tuberculous mycobacterium from humans and water in an Agropastoral area in Zambia. BMC Infect. Dis 18, 20. 10.1186/s12879-017-2939-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Chen H, Zhang Y, Ye AY, Du Z, Xu M, Lee C-S, Hwang JK, Kyritsis N, Ba Z, Neuberg D, et al. (2020). BCR selection and affinity maturation in Peyer’s patch germinal centres. Nature 582, 421–425. 10.1038/s41586-020-2262-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Lycke NY, and Bemark M (2017). The regulation of gut mucosal IgA B-cell responses: recent developments. Mucosal Immunol. 10, 1361–1374. 10.1038/mi.2017.62. [DOI] [PubMed] [Google Scholar]
  • 65.Reboldi A, and Cyster JG (2016). Peyer’s patches: Organizing B cell responses at the intestinal frontier. Immunol. Rev 271, 230–245. 10.1111/imr.12400. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Corthesy B (2013). Multi-Faceted Functions of Secretory IgA at Mucosal Surfaces. Front. Immunol 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Li Y, Jin L, and Chen T (2020). The Effects of Secretory IgA in the Mucosal Immune System. BioMed Res. Int 2020, e2032057. 10.1155/2020/2032057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Mantis NJ, Rol N, and Corthésy B (2011). Secretory IgA’s Complex Roles in Immunity and Mucosal Homeostasis in the Gut. Mucosal Immunol. 4, 603–611. 10.1038/mi.2011.41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Ancelet L, and Kirman J (2012). Shaping the CD4+ memory immune response against tuberculosis: the role of antigen persistence, location and multi-functionality. Biomol. Concepts 3, 13–20. 10.1515/bmc.2011.051. [DOI] [PubMed] [Google Scholar]
  • 70.Cooper AM (2017). Defining the Kinetics, Phenotype, and Function of T Cells Induced by Mycobacterium tuberculosis: Pillar of Immunity to Tuberculosis. J. Immunol 199, 831–832. 10.4049/jimmunol.1700854. [DOI] [PubMed] [Google Scholar]
  • 71.Gern BH, Adams KN, Plumlee CR, Stoltzfus CR, Shehata L, Moguche AO, Busman-Sahay K, Hansen SG, Axthelm MK, Picker LJ, et al. (2021). TGFβ restricts expansion, survival, and function of T cells within the tuberculous granuloma. Cell Host Microbe 29, 594–606.e6. 10.1016/j.chom.2021.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Henao-Tamayo MI, Ordway DJ, Irwin SM, Shang S, Shanley C, and Orme IM (2010). Phenotypic Definition of Effector and Memory T-Lymphocyte Subsets in Mice Chronically Infected with Mycobacterium tuberculosis. Clin. Vaccine Immunol 17, 618–625. 10.1128/CVI.00368-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Henao-Tamayo MI, Obregón-Henao A, Arnett K, Shanley CA, Podell B, Orme IM, and Ordway DJ (2016). Effect of bacillus Calmette-Guérin vaccination on CD4+Foxp3+ T cells during acquired immune response to Mycobacterium tuberculosis infection. J. Leukoc. Biol 99, 605–617. 10.1189/jlb.4A0614-308RR. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Jasenosky LD, Scriba TJ, Hanekom WA, and Goldfeld AE (2015). T cells and adaptive immunity to Mycobacterium tuberculosis in humans. Immunol. Rev 264, 74–87. 10.1111/imr.12274. [DOI] [PubMed] [Google Scholar]
  • 75.McShane H, Brookes R, Gilbert SC, and Hill AV (2001). Enhanced immunogenicity of CD4+ T-cell responses and protective efficacy of a DNA-modified vaccinia virus Ankara prime-boost vaccination regimen for murine tuberculosis. Infect. Immun 69, 681–686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Moguche AO, Musvosvi M, Penn-Nicholson A, Plumlee CR, Mearns H, Geldenhuys H, Smit E, Abrahams D, Rozot V, Dintwe O, et al. (2017). Antigen Availability Shapes T Cell Differentiation and Function during Tuberculosis. Cell Host Microbe 21, 695–706.e5. 10.1016/j.chom.2017.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Pathan AA, Wilkinson KA, Klenerman P, McShane H, Davidson RN, Pasvol G, Hill AV, and Lalvani A (2001). Direct ex vivo analysis of antigen-specific IFN-γ-secreting CD4 T cells in Mycobacterium tuberculosis-infected individuals: associations with clinical disease state and effect of treatment. J. Immunol 167, 5217–5225. [DOI] [PubMed] [Google Scholar]
  • 78.Torrado E, Robinson RT, and Cooper AM (2011). The cellular response to mycobacteria: balancing protection and pathology. Trends Immunol. 32, 66–72. 10.1016/j.it.2010.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Yang JD, Mott D, Sutiwisesak R, Lu Y-J, Raso F, Stowell B, Babunovic GH, Lee J, Carpenter SM, Way SS, et al. (2018). Mycobacterium tuberculosis-specific CD4+ and CD8+ T cells differ in their capacity to recognize infected macrophages. PLOS Pathog. 14, e1007060. 10.1371/journal.ppat.1007060. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Chan J, Mehta S, Bharrhan S, Chen Y, Achkar JM, Casadevall A, and Flynn J (2014). The role of B cells and humoral immunity in Mycobacterium tuberculosis infection. Semin. Immunol 26, 588–600. 10.1016/j.smim.2014.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Kozakiewicz L, Chen Y, Xu J, Wang Y, Dunussi-Joannopoulos K, Ou Q, Flynn JL, Porcelli SA, Jacobs WR, and Chan J (2013). B cells regulate neutrophilia during Mycobacterium tuberculosis infection and BCG vaccination by modulating the interleukin-17 response. PLoS Pathog. 9, e1003472. 10.1371/journal.ppat.1003472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Lu LL, Chung AW, Rosebrock T, Ghebremichael M, Yu WH, Grace PS, Schoen MK, Tafesse F, Martin C, Leung V, et al. (2016). A functional role for antibodies in tuberculosis. Cell 167, 433–443.e14. 10.1016/j.cell.2016.08.072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Maglione PJ, Xu J, and Chan J (2007). B Cells Moderate Inflammatory Progression and Enhance Bacterial Containment upon Pulmonary Challenge with Mycobacterium tuberculosis. J. Immunol 178, 7222–7234. 10.4049/jimmunol.178.11.7222. [DOI] [PubMed] [Google Scholar]
  • 84.Phuah JY, Mattila JT, Lin PL, and Flynn JL (2012). Activated B Cells in the Granulomas of Nonhuman Primates Infected with Mycobacterium tuberculosis. Am. J. Pathol 181, 508–514. 10.1016/j.ajpath.2012.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Rao M, Valentini D, Poiret T, Dodoo E, Parida S, Zumla A, Brighenti S, and Maeurer M (2015). B in TB: B Cells as Mediators of Clinically Relevant Immune Responses in Tuberculosis. Clin. Infect. Dis 61, S225–S234. 10.1093/cid/civ614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Rijnink WF, Ottenhoff THM, and Joosten SA (2021). B-Cells and Antibodies as Contributors to Effector Immune Responses in Tuberculosis. Front. Immunol 12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Tiwari S, Dutt TS, Chen B, Chen M, Kim J, Dai AZ, Lukose R, Shanley C, Fox A, Karger BR, et al. (2020). BCG-Prime and boost with Esx-5 secretion system deletion mutant leads to better protection against clinical strains of Mycobacterium tuberculosis. Vaccine 38, 7156–7165. 10.1016/j.vaccine.2020.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Maekura R, Kitada S, Osada-Oka M, Tateishi Y, Ozeki Y, Fujicawa T, Miki M, Jyunnko O, Mori M, and Matsumoto S (2019). Serum antibody profiles in individuals with latent Mycobacterium tuberculosis infection. Microbiol. Immunol 63, 130–138. 10.1111/1348-0421.12674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Linge I, Dyatlov A, Kondratieva E, Avdienko V, Apt A, and Kondratieva T (2017). B-lymphocytes forming follicle-like structures in the lung tissue of tuberculosis-infected mice: Dynamics, phenotypes and functional activity. Tuberc. Edinb. Scotl 102, 16–23. 10.1016/j.tube.2016.11.005. [DOI] [PubMed] [Google Scholar]
  • 90.Maglione PJ, Xu J, and Chan J (2007). B cells moderate inflammatory progression and enhance bacterial containment upon pulmonary challenge with Mycobacterium tuberculosis. J. Immunol. Baltim Md 1950 178, 7222–7234. 10.4049/jimmunol.178.11.7222. [DOI] [PubMed] [Google Scholar]
  • 91.Phuah JY, Mattila JT, Lin PL, and Flynn JL (2012). Activated B cells in the granulomas of nonhuman primates infected with Mycobacterium tuberculosis. Am. J. Pathol 181, 508–514. 10.1016/j.ajpath.2012.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kahnert A, Höpken UE, Stein M, Bandermann S, Lipp M, and Kaufmann SHE (2007). Mycobacterium tuberculosis Triggers Formation of Lymphoid Structure in Murine Lungs. J. Infect. Dis 195, 46–54. 10.1086/508894. [DOI] [PubMed] [Google Scholar]
  • 93.Maglione PJ, and Chan J (2009). How B cells shape the immune response against Mycobacterium tuberculosis. Eur. J. Immunol 39, 676–686. 10.1002/eji.200839148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Choreño-Parra JA, Bobba S, Rangel-Moreno J, Ahmed M, Mehra S, Rosa B, Martin J, Mitreva M, Kaushal D, Zúñiga J, et al. (2020). Mycobacterium tuberculosis HN878 Infection Induces Human-Like B-Cell Follicles in Mice. J. Infect. Dis 221, 1636–1646. 10.1093/infdis/jiz663. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Linge I, Dyatlov A, Kondratieva E, Avdienko V, Apt A, and Kondratieva T (2017). B-lymphocytes forming follicle-like structures in the lung tissue of tuberculosis-infected mice: Dynamics, phenotypes and functional activity. Tuberc. Edinb. Scotl 102, 16–23. 10.1016/j.tube.2016.11.005. [DOI] [PubMed] [Google Scholar]
  • 96.Allen CDC, Okada T, and Cyster JG (2007). Germinal-center organization and cellular dynamics. Immunity 27, 190–202. 10.1016/j.immuni.2007.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Mesin L, Ersching J, and Victora GD (2016). GERMINAL CENTER B CELL DYNAMICS. Immunity 45, 471–482. 10.1016/j.immuni.2016.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Achkar JM, Chan J, and Casadevall A (2015). B cells and antibodies in the defense against Mycobacterium tuberculosis infection. Immunol. Rev 264, 167–181. 10.1111/imr.12276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Browning JL, Allaire N, Ngam-Ek A, Notidis E, Hunt J, Perrin S, and Fava RA (2005). Lymphotoxin-beta receptor signaling is required for the homeostatic control of HEV differentiation and function. Immunity 23, 539–550. 10.1016/j.immuni.2005.10.002. [DOI] [PubMed] [Google Scholar]
  • 100.Lu T, and Browning J (2014). Role of the Lymphotoxin/LIGHT System in the Development and Maintenance of Reticular Networks and Vasculature in Lymphoid Tissues. Front. Immunol 5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Lai R, Afkhami S, Haddadi S, Jeyanathan M, and Xing Z (2015). Mucosal immunity and novel tuberculosis vaccine strategies: route of immunisation-determined T-cell homing to restricted lung mucosal compartments. Eur. Respir. Rev 24, 356–360. 10.1183/16000617.00002515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Li W, Deng G, Li M, Liu X, and Wang Y (2012). Roles of Mucosal Immunity against Mycobacterium tuberculosis Infection. Tuberc. Res. Treat 2012, 791728. 10.1155/2012/791728. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Todoroff J, Lemaire MM, Fillee C, Jurion F, Renauld J-C, Huygen K, and Vanbever R (2013). Mucosal and Systemic Immune Responses to Mycobacterium tuberculosis Antigen 85A following Its Co-Delivery with CpG, MPLA or LTB to the Lungs in Mice. PLOS ONE 8, e63344. 10.1371/journal.pone.0063344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Abebe F, and Bjune G (2009). The protective role of antibody responses during Mycobacterium tuberculosis infection. Clin. Exp. Immunol 157, 235–243. 10.1111/j.1365-2249.2009.03967.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Reljic R, Williams A, and Ivanyi J (2006). Mucosal immunotherapy of tuberculosis: is there a value in passive IgA? Tuberc. Edinb. Scotl 86, 179–190. 10.1016/j.tube.2006.01.011. [DOI] [PubMed] [Google Scholar]
  • 106.Hoft DF, Xia M, Zhang GL, Blazevic A, Tennant J, Kaplan C, Matuschak G, Dube TJ, Hill H, Schlesinger LS, et al. (2018). PO and ID BCG vaccination in humans induce distinct mucosal and systemic immune responses and CD4+ T cell transcriptomal molecular signatures. Mucosal Immunol. 11, 486–495. 10.1038/mi.2017.67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Hoefsloot W, van Ingen J, Andrejak C, Ängeby K, Bauriaud R, Bemer P, Beylis N, Boeree MJ, Cacho J, Chihota V, et al. (2013). The geographic diversity of nontuberculous mycobacteria isolated from pulmonary samples: an NTM-NET collaborative study. Eur. Respir. J 42, 1604–1613. 10.1183/09031936.00149212. [DOI] [PubMed] [Google Scholar]
  • 108.Fox A, Dutt TS, Karger B, Obregón-Henao A, Anderson GB, and Henao-Tamayo M (2020). Acquisition of High-Quality Spectral Flow Cytometry Data. Curr. Protoc. Cytom 93, e74. 10.1002/cpcy.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Fox A, Dutt TS, Karger B, Rojas M, Obregón-Henao A, Anderson GB, and Henao-Tamayo M (2020). Cyto-Feature Engineering: A Pipeline for Flow Cytometry Analysis to Uncover Immune Populations and Associations with Disease. Sci. Rep 10, 7651. 10.1038/s41598-020-64516-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Hafemeister C, and Satija R (2019). Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296. 10.1186/s13059-019-1874-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Stuart T, and Satija R (2019). Integrative single-cell analysis. Nat. Rev. Genet 20, 257–272. 10.1038/s41576-019-0093-7. [DOI] [PubMed] [Google Scholar]
  • 112.Aran D, Looney AP, Liu L, Wu E, Fong V, Hsu A, Chak S, Naikawadi RP, Wolters PJ, Abate AR, et al. (2019). Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol 20, 163–172. 10.1038/s41590-018-0276-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Bickett TE, McLean J, Creissen E, Izzo L, Hagan C, Izzo AJ, Silva Angulo F, and Izzo AA (2020). Characterizing the BCGinduced macrophage and neutrophil mechanisms for defense against Mycobacterium tuberculosis. Front. Immunol 11, 1202. [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

Supplementary data

Data Availability Statement

Spatial transcriptomics data are available at the GEO database, and the accession number is GSE203037.

All original codes will be deposited to Github and will be publicly available as of the date of publication.

RESOURCES