Skip to main content

Establishment of an in vitro model of monocyte-like THP-1 cells for trained immunity induced by bacillus Calmette-Guérin

Abstract

Background

Mycobacteria bloodstream infections are common in immunocompromised people and usually have disastrous consequences. As the primary phagocytes in the bloodstream, monocytes and neutrophils play critical roles in the fight against bloodstream mycobacteria infections. In contrast to macrophages, the responses of monocytes infected with the mycobacteria have been less investigated.

Results

In this study, we first established a protocol for infection of non-adherent monocyte-like THP-1 cells (i.e. without the differentiation induced by phorbol 12-myristate 13-acetate (PMA) by bacillus Calmette-Guérin (BCG). Via the protocol, we were then capable of exploring the global transcriptomic profiles of non-adherent THP-1 cells infected with BCG, and found that NF-κB, MAPK and PI3K-Akt signaling pathways were enhanced, as well as some inflammatory chemokine/cytokine genes (e.g. CCL4, CXCL10, TNF and IL-1β) were up-regulated. Surprisingly, the Akt-HIF-mTOR signaling pathway was also activated, which induces trained immunity. In this in vitro infection model, increased cytokine responses to lipopolysaccharides (LPS) restimulation, higher cell viability, and decreased Candida albicans loads were observed.

Conclusions

We have first characterized the transcriptomic profiles of BCG-infected non-adherent THP-1 cells, and first developed a trained immunity in vitro model of the cells.

Peer Review reports

Background

Tuberculosis (TB), caused primarily by Mycobacterium tuberculosis (Mtb), remains the world’s leading lethal infectious disease [1]. When aerosols containing Mtb are inhaled into the lower lungs, the bacteria first encounter lung-resident alveolar macrophages patrolling the air-lung epithelium interface [2,3,4]. The phagocytized bacteria are then translocated from the site of infection to nearby lymph nodes, facilitating the presentation of antigens and triggering of adaptive immune responses [2]. However, there is a risk of transmitting the bacteria to other organs via bloodstream infection (BSI), resulting in various forms of extrapulmonary TB (EPTB) [5]. Upon infection, there is a selective recruitment of mycobacteria-permissive monocytes via the CCL2 - CCR2 chemokine axis and the establishment of a growth-permissive niche [6]. Furthermore, mycobacteria can bypass macrophage defenses and escape into monocytes or neutrophils, where they can survive [2, 4, 7]. This is particularly the case when human immunodeficiency virus (HIV) infection impairs the ability of macrophages to control Mtb [8].

Monocytes and neutrophils, the major phagocytes in human blood, account for respectively 3–10% and 40–60% of leukocytes [9]. Subsets of monocytes have different functions and their proportions change during Mtb infection, disease progression, and treatment [10,11,12,13], reflecting the important role of these cells in the host’s fight against Mtb. Monocytes (and neutrophils) acquire epigenetic modifications and metabolic reprogramming by in vivo exposure to bacillus Calmette-Guérin (BCG) or Candida albicans [14, 15]. The modifications result in the acquisition of innate immune memory, termed trained immunity, which confers nonspecific protection against heterologous pathogens by enhancing secondary immune responses [14,15,16]. Therefore, an exploration of the interaction between mycobacteria and circulating monocytes directly in vitro could help in gaining a better understanding of this key stage. Heterogeneous primary monocytes or relatively pure sub-populations of interest can be isolated from peripheral blood via sophisticated and time-consuming methods such as magnetic/fluorescence activated cell sorting (M/FACS). Alternatively, the non-adherent monocyte-like cell line THP-1 could be an option.

Due to the non-adherent nature of some monocytes [17], as well as their much lower phagocytic capacities than macrophages [18], thus it is challenging to infect non-adherent monocytes by bacteria (e.g. mycobacteria). It is also difficult to remove free extracellular bacteria from non-adherent (infected) monocytes post infection. Therefore, we aimed to employ non-adherent THP-1 cells and BCG to establish an improved infection protocol and, on the basis of this, investigate the response of THP-1 cells post BCG infection.

Results

Infection of non-adherent THP-1

Non-adherent cells and bacteria are in a state of stochastic movement, which is not conducive to the phagocytosis of bacteria by host cells [19, 20]. To establish an efficient infection protocol, constant number of THP-1 cells (i.e. 2 × 105 cells/well of 96-well plate), and then bacteria in differing numbers, were sequentially sedimented onto the bottom of wells in cell culture plates. Centrifugation increased the infection efficacy significantly (Fig. 1a), and no additional cell death was observed (Fig. 1b). The infection efficacy of centrifugation for 15 min at 200 × g was significantly higher than centrifugation for 5 mins at 200 × g (Fig. 1c). A plateau in infection efficiency (approximately 50% of cells infected) could be reached by co-culture for 1 h after centrifugation (Fig. 1d). The infection efficacy was MOI-dependent, and plateaued approximately at an MOI of 40 (Fig. 1e, Figure S1b and c).

Fig. 1
figure 1

Optimization of parameters for the in vitro infection of non-adherent THP-1 cells by BCG. a Centrifugation increases infection efficacy. THP-1 cells (2 × 105 cells/100 µL R10/well) were seeded into 96-well plates, and infected with BCG-EGFP (MOI = 10) with or without centrifugation (200 × g, 15 min), and co-cultured for 1 h. EGFP positive cells (FL1-H::FITC) were detected by flow cytometry. b No enhanced cell death after centrifugation (200 × g, 15 min). Positive control: 2% paraformaldehyde (PFA). c The effects of centrifugation and co-culture on infection efficacy. THP-1 cells were seeded into 96-well plates, and infected with BCG-EGFP (MOI = 20) with or without co-culture for 1 h after centrifugation at 200 × g for 5 or 15 min. No infection: THP-1 cells without BCG-EGFP infection; Mix pre-FACS: mixing THP-1 cells and BCG-EGFP prior to flow cytometry; 15 min C_W/O 1 h C: centrifugation for 15 min without subsequent 1 h co-culture; 15 min C_with 1 h C: centrifugation for 15 min followed with 1 h co-culture. d The effect of co-culture durations on infection efficacy. THP-1 cells were seeded into 96-well plates, and infected with BCG-EGFP (MOI = 20) with different co-culture durations after centrifugation (200 × g, 15 min). e The effect of MOIs on infection efficacy. THP-1 cells were seeded into 96-well plates and infected with BCG at various MOIs and co-cultured for 1 h after 200 × g for 15 min. * P < 0.05; ** P < 0.01; # P < 0.0001. Data shown are the mean ± SD, representative of at least twice repeated experiment

Removal of non-adherent extracellular bacteria

MACS enables cells of interest to be selectively separated. CD32 expresses on the membrane of THP-1 cells [21]. Therefore, we used biotin-labeled anti-human CD32 antibody and anti-biotin magnetic beads to sort out THP-1 cells. The results showed that our MACS strategy could effectively sort out THP-1 cells and remove free extracellular BCG-EGFP (Fig. 2a and Figure S1a). Thus, an efficient infection model/protocol of non-adherent THP-1 cells was established (Fig. 2b).

Fig. 2
figure 2

Removal of extracellular BCG by MACS. a The removal of extracellular BCG-EGFP (white arrow) by MACS. Left panel: without MACS; middle panel: one round of MACS; right panel: two rounds of MACSs. b Schematic of infecting non-adherent THP-1 cells with BCG and MACS for sorting out THP-1 cells from free extracellular BCG

The transcriptome of BCG-infected THP-1 cells

RNA-seq was used for transcriptome analysis of BCG-infected non-adherent THP-1 cells. There were significant differences between the BCG group and the BLANK/MOCK groups (Figure S2a). Compared with the BLANK group, the BCG group had 1528 DEGs (|LogFC| ≥ 1, FDR < 0.05), including 1056 up-regulated genes and 472 down-regulated genes (Figure S2b). After BCG infection, the expression of monocyte surface antigens of CD14, CD36, CD40, CD80 and CD86 increased (Table S1), implying cell maturation and activation. Chemokines are critical molecules that recruit immune cells by chemotaxis and activate leukocytes during mycobacterial diseases [22], as seen in the presence of CCL2–5, CCL7–8, CCL13, CCL20, CXCL1–3 and CXCL8–14 in the up-regulated DEGs. Chemokines bind to their receptors and initiate immune cell migration as well as activation [23].

Gene ontology (GO) enrichment analyses showed that the down-regulated DEGs were leaded with response to inorganic substance (GO:0010035) and regulation of ion transport (GO:0043269) (Figure S2c); the up-regulated DEGs were leaded with inflammatory response (GO:0006954), positive regulation of cytokine production (GO:0001819) and cellular response to cytokine stimulus (GO:0071345) (Fig. 3a and Figure S2c). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of DEGs showed that the IL-17, NF-kappa B, PI3K-AKt, TNF, Toll-like receptor, NOD-like receptor and MAPK signaling pathways were up-regulated, while Hippo, Rap1 signaling pathways were down-regulated (Fig. 3b and Table S2). GSEA (using hallmark gene sets) analysis of all genes revealed that the inflammatory response (e.g. TNF, IL-6 and IFN-γ, etc.), PI3K-AKT-mTOR pathway, glycolysis, reactive oxygen species pathway, fatty acid metabolism and cholesterol metabolism pathway were activated (Fig. 3c, Figure S2d and Table S3). Glycolysis and cholesterol metabolism have been shown to be involved in trained immunity [24, 25]. The PI3k-Akt-HIF1a-mTOR signaling pathway axis has been shown to be the molecular basis for the induction of trained immunity in monocytes [25]. Therefore, we used this model to study the induction of trained immunity in non-adherent THP-1 cells.

Fig. 3
figure 3

Transcriptomic analysis of BCG-EGFP-infected non-adherent THP-1 cells. a The network of GO terms. Top 16 clusters with their representative enriched terms (one per cluster). The “count” is the number of genes present in the given ontology term, colored by cluster ID, where nodes that share the same cluster ID are typically close to each other. b The bubble chart of top 10 KEGG pathways. Enriched pathways of up-regulated and down-regulated DEGs were ranked by gene ratio (i.e. mapped gene number/pathway gene number). c The GSEA plots associated with trained immunity. Genes are ranked by Signal2Noise in GSEA plot. Gene expression levels decreased from left (red) to right (blue). The matching gene expression heatmaps were shown beneath. Gene expression values were normalized to z-score

Short-term BCG-induced trained immunity

The induction of trained immunity by BCG was carried out according to the schematic diagram showed in Fig. 4a. Compared with the control group, BCG training increased IL-6 secretion, and this trend was MOI-dependent (Fig. 4b). The BCG-trained THP-1 cells had increased cell viability than the control cells, even after (lipopolysaccharide) LPS stimulation (Fig. 4d). Consistently, there was decreased apoptosis in THP-1 cells after BCG training (Fig. 4c and Figure S3). Importantly, the trained non-adherent THP-1 cells have lower C. albicans loads compared to untrained cells at 3 h post infection (Fig. 4e).

Fig. 4
figure 4

BCG-induced trained immunity in non-adherent THP-1 cells. a Schematic of short-term trained immunity. b The concentration of IL-6 secreted by trained THP-1 cells after LPS restimulation. c The apoptosis measured by flow cytometry 24 h after LPS stimulation. d The cell viability of BCG-trained THP-1 cells before and 24 h after LPS stimulation. Pre-stim: before LPS stimulation; post-stim: 24 h post LPS stimulation. e The CFUs of C. albicans (3 h post infection) after infecting BCG-trained THP-1 cells. ns P > 0.05; * P < 0.05; ** P < 0.01; *** P < 0.001; # P < 0.0001. Data shown are the mean ± SD, representative of at least twice repeated experiments

Discussion

Mtb is one of the most common pathogens causing BSI among HIV-infected patients living in high TB-burden settings with a prevalence of nearly 20% and a case-fatality rate approaching 50% [26,27,28,29]. Monocytes are the major cell type known to be infected with Mtb and play a vital role in immune response against Mtb infection [30]. THP-1 cells were widely used to investigate the functions of monocytes and macrophages [31, 32].

The BCG pellets resuspended in R10 were settled to the bottom of the centrifuge tubes after centrifugation at low speeds of 200 × g and 500 × g for 5 min (Figure S4a). The different buoyant densities of monocytes and mycobacteria have been reported [33, 34]. When using density gradient centrifugation, it was found that both THP-1 cells and BCG located at the same interface between the medium and the Ficoll (Figure S4b). These results indicated that differential density gradient centrifugation was not feasible for removing free extracellular mycobacteria from suspending cells. Finally, we developed a low-speed centrifugation and MACS-based in vitro infection protocol for non-adherent THP-1 cells (Fig. 2b). The model/protocol (Fig. 2b) has the advantages of high infection efficacy, ease of operation, and potentially broad utility in exploring the interaction between other non-adherent cells and pathogenic bacteria.

Although this was the first work to analyze the transcriptome of non-adherent THP-1 cells following BCG infection, the transcriptome of mycobacteria-infected THP-1 cells differentiated (by PMA) macrophages has been previously reported [35,36,37]. TNF-α, IL-1β, CXCL1-2, and NFKBIA were found to be up-regulated after BCG infection [35], and the NOD-like signaling pathway and cytokine-cytokine receptor interaction were enriched in DEGs [36]. The expression levels of genes encoding inflammatory cytokines or cytokine receptors such as TNF, IL-1β, IL-32, IFNGR and TNFR were also significantly up-regulated in BCG-infected non-adherent THP-1 cells (Table S1). These gene products play important immunomodulatory roles in the early stages of mycobacterial infection [23, 38]. However, the expression level of IL-10 also increased 5-fold (Table S1). IL-10 can suppress the presentation of antigens, as well as the production of pro-inflammatory cytokines, chemokines, co-stimulatory molecules and adhesion molecules in macrophages and other cell types [39, 40]. Further gene enrichment analysis showed that BCG-infected THP-1 cells were up-regulated in the cytokine-cytokine receptor interaction, NOD-like receptor signaling pathway, chemotaxis, TNF, MAPK, NF-κB, IL-17 and other pathways (Table S2) involved in mycobacteria infection [41, 42]. The NOD-like receptor signaling pathway was also found to be enriched in phorbol 12-myristate 13-acetate (PMA)-differentiated THP-1 macrophages after BCG infection [36].

Intriguingly, the Akt-mTOR-HIF-1a pathway, which could induce aerobic glycolysis as the metabolic basis of trained immunity [43], was also up-regulated in BCG-infected non-adherent THP-1 cells (Fig. 3b). Consistent with our findings (Fig. 4c and d), BCG immunization has been shown to inhibit monocyte apoptosis and increase cell viability [44,45,46]. The β-glucan-induced trained immunity to protect mice from sepsis has been reported [47], and our results also suggested that the BCG-trained non-adherent THP-1 cells have lower C. albicans loads compared to untrained cells (Fig. 4e).

Trained immunity models using primary human blood monocytes have been established, of which the cells are isolated (via CD14-mediated MACS or plastic adhesion), cultured (adherent) and trained in vitro [48, 49], and do not mimic the characteristic suspending/non-adherent state in vivo. It has been shown that shear flow in dynamic culture reduced pro-inflammatory signaling while increasing secretion of the anti-inflammatory cytokine IL-10 and enhancing migration of monocytes [50]. Furthermore, currently used isolation methods affect monocyte function [51,52,53,54,55], and adherent culture also activates monocyte differentiation to macrophages [56]. However, there are no data on BCG-trained immunity using suspending primary monocytes. To the best of our knowledge, all current trained immunity studies involving THP-1 cells have been performed using PMA-differentiated THP-1 macrophages [57,58,59,60] rather than using non-adherent THP-1 cells. In this study, we firstly established a trained immunity model of suspending monocytes based on non-adherent THP-1 cells. Although there are studies (albeit limited) reporting differences between non-adherent THP-1 cells and primary monocytes [61, 62], it would be intriguing to study trained immunity and infection of suspending primary monocytes using this model later.

Conclusions

In summary, an efficient in vitro model/protocol for infecting non-adherent THP-1 cells with BCG by combining low-speed centrifugation, co-culture, and MACS is established, of which the principle is also applicable to other non-adherent cells and bacteria. In addition, we addressed the transcriptomic profiles of non-adherent THP-1 cells infected with BCG, which had not been explored previously to our knowledge. Finally, we used this in vitro model to test the trained immunity of THP-1 cells induced by BCG, which holds in vivo hint.

Materials and methods

Bacterial strains and growth conditions

Mycobacterium bovis BCG Pasteur was kindly gifted by Xiao-ming Zhang’s lab of Institut Pasteur of Shanghai Chinese Academy of Sciences, China. The cognate BCG-EGFP was constructed via electroporating BCG Pasteur with an EGFP-expressing plasmid (i.e. EGFP was cloned into the plasmid of pMFA41) [63] and being selected on Middlebrook 7H11 agar with kanamycin (final concentration: 50 µg/mL). BCG was grown at 37 °C in liquid Middlebrook 7H9 broth (BD Difco, USA) supplemented with 10% (v/v) oleic acid-albumin-dextrose-catalase enrichment (OADC; BD Difco, USA), 0.5% glycerol and 0.05% Tween-80. The complete medium is referred to as 7H9 hereafter. When necessary, kanamycin was added to a final concentration of 50 µg/mL. Candida (C.) albicans was grown in Sabouraud broth medium (SDB: 1% peptone, 4% dextrose) or on Sabouraud agar medium (SDA: 1% peptone, 4% dextrose, 2% agar) at 37 °C.

Cell culture

THP-1 cells (ATCC) were grown at 37 °C with 5% CO2 in RPMI 1640 (Biological Industries, USA) with 10% fetal bovine serum (FBS; Biological Industries, USA). This complete medium is referred to as R10 hereafter.

Cell infection

THP-1 cells (2 × 105 cells/100 µL R10/well) were seeded into 96-well plates. Plates were centrifuged (200 × g, 5 mins) to sediment cells. Then the BCG-EGFP strain suspended in 100 µL R10 was gently/carefully added into each well without disturbing THP-1 cells. BCG pellets were added at a range of multiplicities of infection (MOI; BCG-EGFP:THP-1 or BCG:THP-1 = 0, 1, 5, 10, 20, 40 or 80). Then the plates were centrifuged once again (200 × g, 15 min) to sediment the bacteria. Following that, plates were carefully transferred into a cell culture cabinet and incubated at 37 °C for 1 h or other times required.

Testing the protocols for removing non-adherent extracellular bacteria

Protocol 1

Testing if BCG and THP-1 cells could be physically separated by low-speed centrifugation. BCG cultured in 7H9 was sedimented at 1800 × g for 5 min. Then the supernatant was discarded and BCG pellets were gently re-suspended in R10 and sedimented at 500 × g or 200 × g for 5 min.

Protocol 2

Testing if BCG and THP-1 cells could be physically separated by Ficoll density gradient centrifugation. One mL of Ficoll (TBD, China) was added into a 1.5-mL tube, then gently overlaid with 0.5 mL R10 containing THP-1 cells or BCG. After centrifugation at 400 × g for 20 min (acceleration = 5, deceleration = 4). Then the vertical positions of THP-1 cells and BCG layers were visually assessed.

Protocol 3

Testing if BCG and THP-1 cells could be physically separated by MACS. Infected THP-1 cells were suspended in 50 µL R10 containing 1 µL CD32-biotin antibody (Miltenyi Biotec, USA), and incubated at 4℃ for 30 min. Then 150 µL R10 was added and sedimented cells. Cell pellets were washed once with 200 µL R10 and sedimented cells. Cell pellets were re-suspended in 80 µL R10, and the suspension was further added with 20 µL anti-biotin MicroBeads (Miltenyi Biotec). After incubating at 4℃ for 15 min, 2 mL R10 was added and sedimented at 300 × g for 10 min. Cell pellets were re-suspended in 500 µL R10 and subjected to MACS according to the user manual. The separated cells were visually observed under fluorescence (confocal) microscopy. R10 was exclusively used across protocol 3 (including staining and MACS) rather than other buffers to avoid undesirable induction of serum-starvation-relevant autophagy [64].

Analysis of RNA-seq data

Non-adherent THP-1 cells were infected with BCG-EGFP (MOI = 20) as described above, or with a null infection. There were three groups (three repeats/group): group 1/BLANK, null infection and null MACS; group 2/MOCK, null infection and MACS. Group 3/BCG, BCG-EGFP infection and MACS. The three groups were operated identically during the cell/bacteria interaction phase. Subsequently the three groups were cultured further for 24 h. RNA was extracted using RNAiso Plus (TaKaRa, Japan). The subsequent RNA quantification, mRNA capture, and RNA sequencing were performed by GENEWIZ Inc. (Suzhou, China) using a 150 base pairs (bp) paired-end sequencing strategy (roughly 6 Giga bp/sample; Novaseq, Illumina, USA). Image analysis and base calling were conducted using the NovaSeq Control Software (HCS) + OLB + GAPipeline-1.6 (Illumina).

Cutadapt (v1.9.1) was used to remove adapters, bases with phred < 20 and being N, and short reads (i.e. < 75 bp) [65]. Hisat2 (v2.0.1) was used to index reference genome sequences downloaded from ENSEMBL (i.e. GRCh38), and to align the cleaned reads to the indexed genome [66]. DESeq2 Bioconductor package (v1.6.3) was used for differential expression analysis (|LogFC| ≥ 1, FDR < 0.05) [67]. Pathway enrichment analysis was performed using Metascape (https://metascape.org/) [68].

Genes were ranked by Signal2Noise, and used for Gene Set Enrichment Analysis (GSEA) using GSEA v4.2.3 (http://www.broadinstitute.org/gsea) [69]. A hallmark gene set with a positive normalized enrichment score (NES) indicates that the gene set is over-represented at the top of a ranked gene list, and thus indicates a positive correlation (up-regulated expression); whereas a negative NES indicates a negative correlation (down-regulation). |NES| > 1 and P value < 0.05 were considered as significant enrichment. The cognate gene expression levels were scaled by Z-score and presented as heatmaps (http://www.bioinformatics.com.cn).

Short-term trained immunity

THP-1 cells were infected as described above and rested for 4 days. On day 4, THP-1 cells were collected and seeded into 96-well plates (2 × 105 cells/well). The cell viability was detected by the CCK-8 kit (Yeasen, China) before and after stimulation with R10 containing 25 ng/mL LPS (eBioscience, USA) for 24 h. The stimulated cells were collected for an apoptosis assay with the Cell Cycle and Apoptosis Analysis Kit (Yeasen, China), and IL-6 in supernatant was assayed by ELISA (Invitrogen, USA). All tests were performed in accordance with the manufacturers’ instructions. In the nonspecific protection assay, C. albicans pellets were collected and re-suspended in PBS, then diluted to 0.8–1 × 104 colony forming units (CFUs)/mL with R10, and 50 µL was added to each well. The plates were centrifuged at 600 × g for 5 mins to facilitate the bacteria-cells contact, and incubated at 37 °C for 3 h. Next, cells were sedimented at 600 × g for 5 mins, and then 0.1 mL of sterile water was added for 10 min to lyse the cells. A series of dilutions were plated on SDA and colonies were enumerated after incubating at 37 °C for 48 h.

Statistical analysis

Students’ t test (unpaired, two-tailed) and one-way ANOVA with Tukey’s multiple comparisons were used to assess statistical significance using GraphPad Prism (v8.0.1) (https://www.graphpad.com/).

Availability of data and materials

The RNA-seq data of this study have been deposited in the NCBI Sequence Read Archive with the SRA accession number PRJNA905923.

References

  1. World Health Organization. Global tuberculosis report 2022. 2022.

    Google Scholar 

  2. Cohen SB, Gern BH, Delahaye JL, Adams KN, Plumlee CR, Winkler JK, Sherman DR, Gerner MY, Urdahl KB. Alveolar macrophages provide an early Mycobacterium tuberculosis Niche and initiate dissemination. Cell Host Microbe. 2018;24(3):439-446e434.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Philips JA, Ernst JD. Tuberculosis pathogenesis and immunity. Annu Rev Pathol. 2012;7:353–84.

    Article  CAS  PubMed  Google Scholar 

  4. Srivastava S, Ernst JD, Desvignes L. Beyond macrophages: the diversity of mononuclear cells in tuberculosis. Immunol Rev. 2014;262(1):179–92.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Krishnan N, Robertson BD, Thwaites G. The mechanisms and consequences of the extra-pulmonary dissemination of mycobacterium tuberculosis. Tuberculosis (Edinb). 2010;90(6):361–6.

    Article  CAS  PubMed  Google Scholar 

  6. Cambier CJ, O’Leary SM, O’Sullivan MP, Keane J, Ramakrishnan L. Phenolic glycolipid facilitates mycobacterial escape from microbicidal tissue-resident macrophages. Immunity. 2017;47(3):552-565e554.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Zhai W, Wu F, Zhang Y, Fu Y, Liu Z. The immune escape mechanisms of mycobacterium tuberculosis. Int J Mol Sci. 2019;20(2):340.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Auld SC, Staitieh BS. HIV and the tuberculosis set point: how HIV impairs alveolar macrophage responses to tuberculosis and sets the stage for progressive disease. Retrovirology. 2020;17(1):32.

    Article  PubMed  PubMed Central  Google Scholar 

  9. Dale DC, Boxer L, Liles WC. The phagocytes: neutrophils and monocytes. Blood. 2008;112(4):935–45.

    Article  CAS  PubMed  Google Scholar 

  10. Castaño D, García LF, Rojas M. Increased frequency and cell death of CD16 + monocytes with mycobacterium tuberculosis infection. Tuberculosis (Edinb). 2011;91(5):348–60.

    Article  PubMed  Google Scholar 

  11. Sánchez MD, García Y, Montes C, París SC, Rojas M, Barrera LF, Arias MA, García LF. Functional and phenotypic changes in monocytes from patients with tuberculosis are reversed with treatment. Microbes Infect. 2006;8(9–10):2492–500.

    Article  PubMed  Google Scholar 

  12. Balboa L, Barrios-Payan J, González-Domínguez E, Lastrucci C, Lugo-Villarino G, Mata-Espinoza D, Schierloh P, Kviatcovsky D, Neyrolles O, Maridonneau-Parini I, et al. Diverging biological roles among human monocyte subsets in the context of tuberculosis infection. Clin Sci (Lond). 2015;129(4):319–30.

    Article  CAS  PubMed  Google Scholar 

  13. Balboa L, Romero MM, Basile JI, Sabio y García CA, Schierloh P, Yokobori N, Geffner L, Musella RM, Castagnino J, Abbate E, et al. Paradoxical role of CD16 + CCR2 + CCR5 + monocytes in tuberculosis: efficient APC in pleural effusion but also mark disease severity in blood. J Leukoc Biol. 2011;90(1):69–75.

    Article  CAS  PubMed  Google Scholar 

  14. Kleinnijenhuis J, Quintin J, Preijers F, Joosten LA, Ifrim DC, Saeed S, Jacobs C, van Loenhout J, de Jong D, Stunnenberg HG, et al. Bacille Calmette-Guerin induces NOD2-dependent nonspecific protection from reinfection via epigenetic reprogramming of monocytes. Proc Natl Acad Sci U S A. 2012;109(43):17537–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Quintin J, Saeed S, Martens JHA, Giamarellos-Bourboulis EJ, Ifrim DC, Logie C, Jacobs L, Jansen T, Kullberg BJ, Wijmenga C, et al. Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes. Cell Host Microbe. 2012;12(2):223–32.

    Article  CAS  PubMed  Google Scholar 

  16. Moorlag S, Rodriguez-Rosales YA, Gillard J, Fanucchi S, Theunissen K, Novakovic B, de Bont CM, Negishi Y, Fok ET, Kalafati L, et al. BCG vaccination induces long-term functional reprogramming of human neutrophils. Cell Rep. 2020;33(7):108387.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Klinder A, Markhoff J, Jonitz-Heincke A, Sterna P, Salamon A, Bader R. Comparison of different cell culture plates for the enrichment of non-adherent human mononuclear cells. Exp Ther Med. 2019;17(3):2004–12.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Nagl M, Kacani L, Müllauer B, Lemberger EM, Stoiber H, Sprinzl GM, Schennach H, Dierich MP. Phagocytosis and killing of bacteria by professional phagocytes and dendritic cells. Clin Diagn Lab Immunol. 2002;9(6):1165–8.

    PubMed  PubMed Central  Google Scholar 

  19. Razin S. Adherence of pathogenic mycoplasmas to host cells. Biosci Rep. 1999;19(5):367–72.

    Article  CAS  PubMed  Google Scholar 

  20. Di Martino P. Bacterial adherence: much more than a bond. AIMS Microbiol. 2018;4(3):563–6.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Forrester MA, Wassall HJ, Hall LS, Cao H, Wilson HM, Barker RN, Vickers MA. Similarities and differences in surface receptor expression by THP-1 monocytes and differentiated macrophages polarized using seven different conditioning regimens. Cell Immunol. 2018;332:58–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Shey MS, Balfour A, Masina N, Bekiswa A, Schutz C, Goliath R, Dielle R, Katoto PD, Wilkinson KA, Lewinsohn D, et al. Mycobacterial-specific secretion of cytokines and chemokines in healthcare workers with apparent resistance to infection with mycobacterium tuberculosis. Front Immunol. 2023;14:1176615.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Domingo-Gonzalez R, Prince O, Cooper A, Khader SA. Cytokines and chemokines in Mycobacterium tuberculosis infection. Microbiol Spectr. 2016;4(5):10.

  24. Bekkering S, Arts RJW, Novakovic B, Kourtzelis I, van der Heijden C, Li Y, Popa CD, Ter Horst R, van Tuijl J, Netea-Maier RT, et al. Metabolic induction of trained immunity through the mevalonate pathway. Cell. 2018;172(1–2):135-146e139.

    Article  CAS  PubMed  Google Scholar 

  25. Cheng SC, Quintin J, Cramer RA, Shepardson KM, Saeed S, Kumar V, Giamarellos-Bourboulis EJ, Martens JH, Rao NA, Aghajanirefah A, et al. mTOR- and HIF-1α-mediated aerobic glycolysis as metabolic basis for trained immunity. Science. 2014;345(6204):1250684.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Crump JA, Ramadhani HO, Morrissey AB, Saganda W, Mwako MS, Yang LY, Chow SC, Njau BN, Mushi GS, Maro VP, et al. Bacteremic disseminated tuberculosis in sub-saharan Africa: a prospective cohort study. Clin Infect Dis. 2012;55(2):242–50.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Jacob ST, Pavlinac PB, Nakiyingi L, Banura P, Baeten JM, Morgan K, Magaret A, Manabe Y, Reynolds SJ, Liles WC, et al. Mycobacterium tuberculosis bacteremia in a cohort of hiv-infected patients hospitalized with severe sepsis in Uganda–high frequency, low clinical suspicion [corrected] and derivation of a clinical prediction score. PLoS One. 2013;8(8):e70305.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Muchemwa L, Shabir L, Andrews B, Bwalya M. High prevalence of mycobacterium tuberculosis bacteraemia among a cohort of HIV-infected patients with severe sepsis in Lusaka, Zambia. Int J STD AIDS. 2017;28(6):584–93.

    Article  PubMed  Google Scholar 

  29. Barr DA, Lewis JM, Feasey N, Schutz C, Kerkhoff AD, Jacob ST, Andrews B, Kelly P, Lakhi S, Muchemwa L, et al. Mycobacterium tuberculosis bloodstream infection prevalence, diagnosis, and mortality risk in seriously ill adults with HIV: a systematic review and meta-analysis of individual patient data. Lancet Infect Dis. 2020;20(6):742–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Ganbat D, Seehase S, Richter E, Vollmer E, Reiling N, Fellenberg K, Gaede KI, Kugler C, Goldmann T. Mycobacteria infect different cell types in the human lung and cause species dependent cellular changes in infected cells. BMC Pulm Med. 2016;16:19.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Chanput W, Mes JJ, Wichers HJ. THP-1 cell line: an in vitro cell model for immune modulation approach. Int Immunopharmacol. 2014;23(1):37–45.

    Article  CAS  PubMed  Google Scholar 

  32. Shah PT, Tufail M, Wu C, Xing L. THP-1 cell line model for tuberculosis: a platform for in vitro macrophage manipulation. Tuberculosis (Edinb). 2022;136:102243.

    Article  CAS  PubMed  Google Scholar 

  33. den Hertog AL, Klatser PR, Anthony RM. Buoyant density of mycobacterium tuberculosis: implications for sputum processing. Int J Tuberc Lung Dis. 2009;13(4):466–71.

    Google Scholar 

  34. Ulmer AJ, Scholz W, Ernst M, Brandt E, Flad HD. Isolation and subfractionation of human peripheral blood mononuclear cells (PBMC) by density gradient centrifugation on Percoll. Immunobiology. 1984;166(3):238–50.

    Article  CAS  PubMed  Google Scholar 

  35. Huante MB, Gupta S, Calderon VC, Koo SJ, Sinha M, Luxon BA, Garg NJ, Endsley JJ. Differential inflammasome activation signatures following intracellular infection of human macrophages with Mycobacterium bovis BCG or Trypanosoma cruzi. Tuberculosis (Edinb). 2016;101s:S35-s44.

    Article  CAS  PubMed  Google Scholar 

  36. Pu W, Zhao C, Wazir J, Su Z, Niu M, Song S, Wei L, Li L, Zhang X, Shi X, et al. Comparative transcriptomic analysis of THP-1-derived macrophages infected with Mycobacterium tuberculosis H37Rv, H37Ra and BCG. J Cell Mol Med. 2021;25(22):10504–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Wu K, Dong D, Fang H, Levillain F, Jin W, Mei J, Gicquel B, Du Y, Wang K, Gao Q, et al. An interferon-related signature in the transcriptional core response of human macrophages to Mycobacterium tuberculosis infection. PLoS One. 2012;7(6):e38367.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Bai X, Kim SH, Azam T, McGibney MT, Huang H, Dinarello CA, Chan ED. IL-32 is a host protective cytokine against Mycobacterium tuberculosis in differentiated THP-1 human macrophages. J Immunol. 2010;184(7):3830–40.

    Article  CAS  PubMed  Google Scholar 

  39. Ouyang W, O’Garra A. IL-10 family cytokines IL-10 and IL-22: from basic science to clinical translation. Immunity. 2019;50(4):871–91.

    Article  CAS  PubMed  Google Scholar 

  40. Mittal SK, Roche PA. Suppression of antigen presentation by IL-10. Curr Opin Immunol. 2015;34:22–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Tchou-Wong KM, Tanabe O, Chi C, Yie TA, Rom WN. Activation of NF-kappaB in mycobacterium tuberculosis- induced interleukin-2 receptor expression in mononuclear phagocytes. Am J Respir Crit Care Med. 1999;159(4 Pt 1):1323–9.

    Article  CAS  PubMed  Google Scholar 

  42. Schorey JS, Cooper AM. Macrophage signalling upon mycobacterial infection: the MAP kinases lead the way. Cell Microbiol. 2003;5(3):133–42.

    Article  CAS  PubMed  Google Scholar 

  43. Cheng S-C, Quintin J, Cramer RA, Shepardson KM, Saeed S, Kumar V, Giamarellos-Bourboulis EJ, Martens JHA, Rao NA, Aghajanirefah A, et al. mTOR- and HIF-1 alpha-mediated aerobic glycolysis as metabolic basis for trained immunity. Science. 2014;345(6204):1579.

    Article  CAS  Google Scholar 

  44. Chávez-Galán L, Vesin D, Martinvalet D, Garcia I. Low dose BCG infection as a model for macrophage activation maintaining cell viability. J Immunol Res. 2016;2016:4048235.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Kremer L, Estaquier J, Brandt E, Ameisen JC, Locht C. Mycobacterium bovis bacillus calmette Guérin infection prevents apoptosis of resting human monocytes. Eur J Immunol. 1997;27(9):2450–6.

    Article  CAS  PubMed  Google Scholar 

  46. Sepulveda RL, Arredondo S, Rodriguez E, Gonzalez B, Leiva LE, Sorensen RU. Effect of human newborn BCG immunization on monocyte viability and function at 3 months of age. Int J Tuberc Lung Dis. 1997;1(2):122–7.

    CAS  PubMed  Google Scholar 

  47. Harriett AJ, Esher Righi S, Lilly EA, Fidel P Jr, Noverr MC. Efficacy of Candida dubliniensis and fungal β-Glucans in Inducing trained innate immune protection against inducers of sepsis. Front Cell Infect Microbiol. 2022;12:898030.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bekkering S, Blok BA, Joosten LA, Riksen NP, van Crevel R, Netea MG. In vitro experimental model of trained innate immunity in human primary monocytes. Clin Vaccine Immunol. 2016;23(12):926–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Domínguez-Andrés J, Arts RJW, Bekkering S, Bahrar H, Blok BA, de Bree LCJ, Bruno M, Bulut Ö, Debisarun PA, Dijkstra H, et al. In vitro induction of trained immunity in adherent human monocytes. STAR Protocols. 2021;2(1):100365.

    Article  PubMed  PubMed Central  Google Scholar 

  50. Wirthgen E, Hornschuh M, Wrobel IM, Manteuffel C, Däbritz J. Mimicking of blood flow results in a distinct functional phenotype in human non-adherent classical monocytes. Biology. 2021;10(8):748.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Otto NA, Butler JM, Ramirez-Moral I, van Weeghel M, van Heijst JWJ, Scicluna BP, Houtkooper RH, de Vos AF, van der Poll T. Adherence affects Monocyte Innate Immune function and metabolic reprogramming after lipopolysaccharide stimulation in vitro. J Immunol. 2021;206(4):827–38.

    Article  CAS  PubMed  Google Scholar 

  52. Kim D, Kim JY. Anti-CD14 antibody reduces LPS responsiveness via TLR4 internalization in human monocytes. Mol Immunol. 2014;57(2):210–5.

    Article  CAS  PubMed  Google Scholar 

  53. Elkord E, Williams PE, Kynaston H, Rowbottom AW. Human monocyte isolation methods influence cytokine production from in vitro generated dendritic cells. Immunology. 2005;114(2):204–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Nielsen MC, Andersen MN, Møller HJ. Monocyte isolation techniques significantly impact the phenotype of both isolated monocytes and derived macrophages in vitro. Immunology. 2020;159(1):63–74.

    Article  CAS  PubMed  Google Scholar 

  55. Hornschuh M, Haas V, Winkel PP, Gökyildirim MY, Mullins CS, Wrobel IM, Manteuffel C, Wirthgen E. Negative magnetic sorting preserves the functionality of ex vivo cultivated non-adherent human monocytes. Biology. 2022;11(11):1583.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kelley JL, Rozek MM, Suenram CA, Schwartz CJ. Activation of human blood monocytes by adherence to tissue culture plastic surfaces. Exp Mol Pathol. 1987;46(3):266–78.

    Article  CAS  PubMed  Google Scholar 

  57. de Homdedeu M, Sanchez-Moral L, Violán C, Ràfols N, Ouchi D, Martín B, Peinado MA, Rodríguez-Cortés A, Arch-Sisquella M, Perez-Zsolt D, et al. Mycobacterium manresensis induces trained immunity in vitro. iScience. 2023;26(6):106873.

    Article  PubMed  PubMed Central  Google Scholar 

  58. van Dijk A, Anten J, Bakker A, Evers N, Hoekstra AT, Chang JC, Scheenstra MR, Veldhuizen EJA, Netea MG, Berkers CR, et al. Innate immune training of human macrophages by cathelicidin analogs. Front Immunol. 2022;13:777530.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Huijser E, van Helden-Meeuwsen CG, Grashof DGB, Tarn JR, Brkic Z, Huisman JMA, Wahadat MJ, van de Werken HJG, Lopes AP, van Roon JAG, et al. Trained immunity in primary Sjögren’s syndrome: linking type I interferons to a pro-atherogenic phenotype. Front Immunol. 2022;13:840751.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Pan W, Hao S, Zheng M, Lin D, Jiang P, Zhao J, Shi H, Yang X, Li X, Yu Y. Oat-derived β-glucans induced trained immunity through metabolic reprogramming. Inflammation. 2020;43(4):1323–36.

    Article  CAS  PubMed  Google Scholar 

  61. Schildberger A, Rossmanith E, Eichhorn T, Strassl K, Weber V. Monocytes, peripheral blood mononuclear cells, and THP-1 cells exhibit different cytokine expression patterns following stimulation with lipopolysaccharide. Mediators Inflamm. 2013;2013:697972.

    Article  PubMed  PubMed Central  Google Scholar 

  62. Liu Y, Li H, Czajkowsky DM, Shao Z. Monocytic THP-1 cells diverge significantly from their primary counterparts: a comparative examination of the chromosomal conformations and transcriptomes. Hereditas. 2021;158(1):43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Fan XY, Ma H, Guo J, Li ZM, Cheng ZH, Guo SQ, Zhao GP. A novel differential expression system for gene modulation in Mycobacteria. Plasmid. 2009;61(1):39–46.

    Article  CAS  PubMed  Google Scholar 

  64. Gutierrez MG, Master SS, Singh SB, Taylor GA, Colombo MI, Deretic V. Autophagy is a defense mechanism inhibiting BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell. 2004;119(6):753–66.

  65. Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 2011;17(1):10–12.

  66. Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12(4):357–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Varet H, Brillet-Guéguen L, Coppée JY, Dillies MA. SARTools: a DESeq2- and EdgeR-Based R pipeline for comprehensive differential analysis of RNA-Seq data. PLoS One. 2016;11(6):e0157022.

    Article  PubMed  PubMed Central  Google Scholar 

  68. Zhou Y, Zhou B, Pache L, Chang M, Khodabakhshi AH, Tanaseichuk O, Benner C, Chanda SK. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat Commun. 2019;10(1):1523.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Mootha VK, Lindgren CM, Eriksson KF, Subramanian A, Sihag S, Lehar J, Puigserver P, Carlsson E, Ridderstråle M, Laurila E, et al. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet. 2003;34(3):267–73.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are thankful to Dr. Douglas B. Lowrie for his critical reading and improvement on this draft.

Funding

This study was supported by the National Key Research and Development Program of China (2022YFC2302900, 2021YFC2301503), National Natural and Science Foundation of China (82171815, 82171739), Shanghai Municipal Health Bureau (2022XD060) and Shanghai Science and Technology Commission ( 19XD1403100).

Author information

Authors and Affiliations

Authors

Contributions

Project concept conceived, data analysis and manuscript writing (X.-Y Fan, J.-C, Xu, and K. Wu), experiments performed (J.-C Xu, K. Wu, R.-Q Ma, J.-H. Li, and J. Tao), paper revision (J.-C Xu, K. Wu, Z. Hu, X.-Y Fan).

Corresponding author

Correspondence to Xiao-Yong Fan.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Additional file 1:

 Table S1. Inflammation-related genes (BCG vs Blank). Table S2. KEGG pathways enrichment analysis of DEGs. Table S3. GSEA analysis of hallmark gene sets. Figure S1. In vitro infection of non-adherent THP-1 cells by BCG-EGFP. (a) The confocal images of THP-1 cells infected with BCG-EGFP. (b) The gating strategy of BCG-infected THP-1 cells. (c) The histogram of THP-1 cells infected with BCG-EGFP with different MOIs. Figure S2. Transcriptomic analysis of BCG-infected THP-1 cells. (a) The venn diagram of DEGs among BLANK, MOCK and BCG groups. (b) The volcano plot of DEGs (BCG vs. BLANK). (c) The top 20 GO biological processes (BCG vs. BLANK). (d) The GSEA plots of inflammation-related pathways. Figure S3. BCG-induced trained immunity in non-adherent THP-1 cells. (a) The gating strategy of apoptotic THP-1 cells.  (b) The pseudocolor plot of apoptotic and live THP-1 cells. (c) The histogram of apoptotic THP-1 cells infected with BCG. Figure S4. The removal of extracellular BCG. (a) Centrifugation could not separate THP-1 cells and BCG. BCG pellets were re-suspended in R10 and sedimented to the bottom of centrifuge tube at 200 or 500 × g for 5 mins. (b) Density gradient centrifugation using Ficoll could not separate THP-1 cells and BCG. The centrifugation parameter was 400 × g, 20 mins (acceleration = 5, deceleration = 4).

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xu, JC., Wu, K., Ma, Rq. et al. Establishment of an in vitro model of monocyte-like THP-1 cells for trained immunity induced by bacillus Calmette-Guérin. BMC Microbiol 24, 130 (2024). https://doi.org/10.1186/s12866-024-03191-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12866-024-03191-x

Keywords