1. Introduction
Tuberculosis (TB) is a severe respiratory infectious disease caused by the pathogen
Mycobacterium tuberculosis (M. tb). It remains the second leading cause of mortality from a single infectious agent worldwide [
1]. Presently, the Bacillus Calmette–Guérin (BCG) vaccine is the only licensed vaccine for TB prevention. However, its efficacy is limited, providing protection for only 10–20 years and presenting notable safety concerns for immunocompromised individuals [
2,
3]. Consequently, there is an imperative need for the development of vaccines that ensure enhanced safety and improved protective efficacy.
In mycobacteria, lipoproteins, situated within the bacterial plasma membrane or outer membrane, serve as crucial surface proteins constituting the bacterial envelope [
4]. During infection, these lipoproteins facilitate adhesion, colonization, evasion of the host immune system, and modulation of immune responses [
5]. Approximately 99 lipoproteins have been identified in M. tb, each fulfilling specific functional roles [
5]. Among these, LppX is notable; the deletion of
lppX diminishes M. tb’s virulence, a reduction attributed to a unique hydrophobic cavity within its protein structure that mediates the transport of phthiocerol dimycocerosates (PDIMs) to the outer cell membrane [
6,
7]. Another significant lipoprotein, RpfB, one of the resuscitation-promoting factors, facilitates M. tb’s reactivation from dormancy by cleaving the glycosidic bond between N-acetylglucosamine and N-acetylmuramic acid, thereby compromising cell wall integrity [
8]. Certain lipoproteins, such as LpqH, directly influence the host immune system. LpqH downregulates MHC class II expression, aiding M. tb’s immune evasion, and disrupts macrophage cytokine production [
9].
In recent years, lipoproteins with diverse functions have been explored as potential vaccine candidates against M. tb, capitalizing on their immunological properties (
Table 1). Many lipoprotein-based vaccines have demonstrated considerable efficacy in combating M. tb. For instance, the M. tbΔ
lpqS attenuated live vaccine exhibits comparable effectiveness to BCG in guinea pig models [
10]. In mouse models, the RpfB protein vaccine reduces the bacterial load in the lungs and spleens by 2 log
10 and 1 log
10, respectively [
11]. Similarly, the PstS-3 DNA vaccine achieves a 1.5 log
10 reduction in the lung bacterial burden in mice [
9]. Collectively, these findings position M. tb lipoproteins as promising antigens for anti-M. tb vaccine development.
LprO (possible lipoprotein LprO, Rv0179c) is one such lipoprotein, found predominantly in pathogenic mycobacteria. In this study, LprO was utilized as an antigen to construct two vaccine types: the pcDNA-lprO DNA vaccine and the BCG Japan::pNBV1-lprO live vaccine. Acting as a vaccine antigen, LprO amplifies Th1-type immune responses, leading to a reduction in the M. tb burden in murine lungs. These findings suggest that LprO could be a valuable antigen for M. tb, presenting a novel target for vaccine research and a potential strategy for tuberculosis prevention.
2. Methods and Materials
2.1. Prediction of LprO Epitopes
To predict CD4+ T cell epitopes for the target gene, the NetMHC II 2.3 Server (
http://www.cbs.dtu.dk/services/NetMHCII/, accessed on 8 April 2025) was utilized. The FASTA file containing the amino acid sequence of the gene (retrievable from the NCBI website:
https://www.ncbi.nlm.nih.gov/gene/886796, accessed on 8 April 2025) was imported. The amino acid length was set to 15, and the IC50 threshold was defined as <50 nmol/L. The sequence was analyzed using the 25 alleles available on the server, with the results sorted by affinity before submission.
For CD8
+ T cell epitope prediction, the NetMHCcons 1.1 Server (
http://www.cbs.dtu.dk/services/NetMHCcons/, accessed on 8 April 2025) was employed. The FASTA file of the amino acid sequence was imported, setting the peptide length to between 8 and 11 mers and the IC50 threshold to <50 nmol/L. The analysis was performed using the server’s 25 alleles, with the results sorted by affinity prior to submission.
B cell epitope prediction was conducted using the IEDB online tool (
http://tools.iedb.org/bcell/, accessed on 8 April 2025, Vesrion 2.0), wherein the amino acid sequence of LprO was inputted. The BepiPred Linear Epitope Prediction 2.0 tool was selected to identify B cell epitopes for the LprO protein [
16].
2.2. Structural Prediction of LprO Protein
The AlphaFold 3 online platform (Version 3.0) was used to predict and analyze the three-dimensional structure of the LprO protein based on its amino acid sequence. The pLDDT score, ranging from 0 to 100, was employed to assess the reliability of the predicted structure, with higher scores indicating greater confidence [
17]. The resultant LprO protein structure was annotated and visualized using PyMOL (Version 3.1.1).
2.3. Strains Cultivation and Growth Conditions
Mycobacterium bovis BCG (ATCC 927) and Mycobacterium tuberculosis H37Rv were cultured in Middlebrook 7H9 broth (Franklin Lakes, BD, USA), supplemented with 0.2% glycerol, 10% OADC (oleic acid, bovine serum albumin, dextrose, and catalase; Difco), and 0.5% Tween 80, or on 7H11 agar, containing 0.2% glycerol and 10% OADC, at 37 °C. Escherichia coli DH5α and BL21(DE3) were grown in Luria–Bertani medium (Sangon Biotech, Shanghai, China) or on agar for cloning and expression purposes.
2.4. Molecular Cloning
To construct pcDNA-lprO, primers were designed based on the genomes of M. tb. The lprO gene was amplified via PCR (Vazyme, Nanjing, China). During the construction process, the signal peptide sequence of the LprO protein was removed, a Kozak sequence was appended to the 5′ end of the lprO gene, and a Flag tag was incorporated at the 3′ end. The resulting pcDNA-lprO constructs were sequenced by Shanghai Qingke Biotechnology Co., Ltd., (Qingke, Shanghai, China) and analyzed using SnapGene software (Version 6.0.2) to verify the accuracy of the gene sequence.
2.5. Intracellular Expression Level Analysis
Following the manufacturer’s protocol for the Lipo8000™ (Beyotime, Shanghai, China) transfection reagent, DNA was transfected into HeLa cells. After 48 h, the cell culture supernatant was discarded, and the cells were washed with 1 mL of sterile PBS, which was subsequently removed. Each well was treated with 100 μL of RIPA buffer (Sangon Biotech, Shanghai, China) for cell lysis. Following thorough lysis, the lysate was collected and centrifuged at 12,000 rpm for 30 min at 4 °C. The resulting supernatant was mixed with 5× protein loading buffer and boiled for 15 min in a metal bath to ensure complete protein denaturation. A second centrifugation at 12,000 rpm for 10 min was performed before loading the total protein onto an SDS-PAGE gel, which was run at 120 V for 60 min. The proteins were transferred from the SDS-PAGE gel to a PVDF membrane using the eBlot™ L1 Rapid Wet Transfer System(GenScript, Nanjing, China) for Western blot analysis. The LprO protein was detected using an anti-Flag antibody, while GAPDH was identified using an anti-GAPDH antibody (Sangon Biotech, Shanghai, China). The PVDF membrane was incubated with the appropriate secondary antibodies and developed using a chemiluminescent substrate. Imaging was conducted using the LAS4000 imaging system.
2.6. Construction of Recombinant Strains and qRT-PCR Analysis
The vector construction strategy involved inserting the lprO gene, along with its 300 bp upstream sequence and a Flag tag, into the Hind III restriction site of the pNBV1 plasmid. The pNBV1-lprO construct was sequenced by Shanghai Qingke Biotechnology Co., Ltd. Upon sequence verification, the vector was electroporated into BCG Japan, with pNBV1 alone introduced as a control strain. Streptomycin-resistant colonies of BCG Japan::pNBV1 and BCG Japan::pNBV1-lprO were isolated. Following colony expansion, RNA was extracted using Trizol (Invitrogen, Carlsbad, CA, USA), and the total RNA was reverse transcribed into cDNA using the TAKARA reverse transcription kit. A quantitative real-time PCR (qRT-PCR) was then conducted, following the Qiagen qRT-PCR detection kit instructions to assess the transcriptional levels of lprO.
2.7. Detection of Secreted Proteins in BCG Culture Supernatant
Activated BCG Japan::pNBV1 and BCG Japan::pNBV1-lprO strains were inoculated in 100 mL of streptomycin-supplemented 7H9 medium and incubated at 37 °C until the optical density at 600 nm (OD600) reached 0.8–1.2. The cultures were centrifuged at 4000 rpm for 10 min to pellet the bacteria, which were then transferred into 50 mL centrifuge tubes. The pellets were resuspended in 50 mL of Sauton’s medium, centrifuged again at 4000 rpm for 10 min, and the supernatant discarded. The bacterial pellets were fully resuspended in fresh Sauton’s medium within sterile conical flasks, adjusting the OD600 to 0.6. The cultures were incubated at 37 °C in a shaking incubator until they reached the logarithmic growth phase (OD600 1.0–1.2). The bacterial pellets were collected through centrifugation at 4000 rpm for 10 min, and the supernatant was carefully collected. This supernatant was filtered through a 0.22 μm membrane to eliminate any residual bacterial cells and concentrated 50-fold using a 3000 Da ultrafiltration tube. The concentrated supernatant was utilized to prepare protein samples for the Western blot analysis. GroEL2, an intracellularly expressed protein in Mycobacterium (1:500 antibody dilution), served as a negative control, while Ag85B, a known secreted protein (1:500 antibody dilution), functioned as a positive control.
2.8. Macrophage Infection and Cell Viability Assessment
Raw264.7 macrophages were seeded in a 96-well plate, at a density of 1 × 10⁴ cells per well, and cultured in 100 μL of 1640 cell culture medium for 20 h prior to infection. For each time point, a separate 96-well plate was utilized. The outermost wells contained the control cells (Raw264.7 cells + 1640 medium), while the wells with only 1640 medium served as blanks. The cells were infected at multiplicities of infection (MOIs) of 1 and 50. After 4 h of incubation, the culture supernatant was discarded, and the cells were washed twice with sterile PBS. Subsequently, 100 μL of 1640 medium containing 50 μg/mL of gentamicin was added, followed by a further two-hour incubation period. The medium was then discarded, the cells were washed twice with PBS, and fresh medium containing 100 units/mL of penicillin and 100 μg/mL of streptomycin was added for continued incubation. Cell viability was assessed at 4, 24, 48, 72, and 96 h post-infection. At each time point, the culture medium was removed, and 100 μL of 1640 medium was added to each well, followed by 10 μL of CCK-8 solution (Beyotime, Shanghai, China). The plate was gently shaken and incubated at 37 °C for 1–4 h until an orange color developed. Absorbance was measured at 450 nm to quantify cell viability.
2.9. Analysis of BCG Virulence in SCID Mice
Six-to-eight-week-old female SCID mice were obtained from Shanghai Slake Biological Technology Co., Ltd. (Slake, Shanghai, China) and maintained in a specific pathogen-free (SPF) grade animal facility. Following an acclimatization period of approximately one week, the mice were used for the experimental procedures. BCG Pasteur served as the positive control, while BCG Japan::pNBV1 functioned as the negative control. BCG Japan::pNBV1-lprO (with two biological replicates) was used as the experimental group, and sterile PBS acted as the blank control. Each mouse received a 100 μL tail vein injection of either bacterial suspension (containing an OD600 of 0.5) or sterile PBS, with 15 mice assigned per group. On day one and at week four post-injection, one mouse from each group, excluding the PBS control, was euthanized. The lungs and spleens were harvested and placed into grinding tubes containing 1 mL of sterile PBS, supplemented with 10% glycerol. The tissues were homogenized at 5000 rpm for 20 s, repeated three times, using a tissue homogenizer. The homogenate was serially diluted and plated, followed by incubation at 37 °C for four weeks to determine the actual inoculation dose. The mice were weighed weekly to monitor body weight fluctuations. Survival was recorded daily, and survival curves were plotted to evaluate the mortality rates across all the groups.
2.10. Immunogenicity Assessment
For the immunogenicity analysis, BCG-immunized C57BL/6J mice received subcutaneous injections of ~106 CFU of either BCG Japan::pNBV1 or BCG Japan::pNBV1-lprO. DNA-immunized C57BL/6J mice were intramuscularly injected with 100 μg/100 μL of either pcDNA or pcDNA-lprO, administered in three immunizations at two-week intervals. The blank control group received 100 μL of sterile PBS. Three weeks after the final DNA immunization, spleen lymphocytes were isolated from all the mice. The cells were plated at a density of 2 × 106 cells/well in 48-well plates, with each sample divided into two groups: one stimulated with PPD and the other left unstimulated. After 30 h of incubation, 4 μL of Monensin Solution (diluted 10-fold) was added for an additional 6 h of stimulation. For the positive control group, 50 ng/mL of PMA and 500 ng/mL of ionomycin were added, alongside the Monensin Solution for 5 h of stimulation. Post-stimulation, the cells were collected and subjected to flow cytometry analysis. The cells were transferred into tubes and centrifuged at 4 °C, 1700 rpm for 5 min. The pellet was washed with 1 mL of Staining Buffer, centrifuged again, and the supernatant discarded. The cells were resuspended in 100 μL of Staining Buffer, followed by the addition of 4 μL of TruStain FcX™ (anti-mouse CD16/32, Biolegend, San Diego, CA, USA) and incubated at 4 °C in the dark for 15 min. Next, 1 μL of Zombie Aqua™ Fixable Viability Kit (Biolegend)dye was added, and the cells were incubated at 4 °C in the dark for another 15 min. After washing with Staining Buffer and centrifugation, the cells were resuspended in 100 μL of Staining Buffer and stained sequentially with APC/Fire™ 750 anti-mouse CD3 (Biolegend), Alexa Fluor® 700 anti-mouse CD4 (Biolegend), PerCP/Cyanine5.5 anti-mouse CD8a (Biolegend), PE anti-mouse/human CD44 (Biolegend), and FITC anti-mouse CD62L (Biolegend), following the manufacturer’s instructions.
Incubate the cells at 4 °C in the dark for 20 min. Wash the cells with 1 mL of Staining Buffer, followed by centrifugation at 4 °C, 1700 rpm for 5 min, and discard the supernatant. Add 500 μL of fixation buffer (Biolegend), gently mix through pipetting, and incubate again at 4 °C in the dark for 20 min. Centrifuge at 4 °C, 1700 rpm for 5 min, discard the supernatant, and add 500 μL of 1× perm/wash buffer. Gently mix through pipetting, centrifuge at 4 °C, 1700 rpm for 5 min, and discard the supernatant. Repeat this washing step twice. Resuspend the cells in 100 μL of Staining Buffer and sequentially add APC anti-mouse TNF-α (Biolegend), PE/Dazzle™ 594 anti-mouse IFN-γ (Biolegend), and Brilliant Violet 421™ anti-mouse IL-2 (Biolegend), following the manufacturer’s instructions. Incubate at 4 °C in the dark for 20 min. Wash the cells with 1 mL of 1× perm/wash buffer (Biolegend), centrifuge at 4 °C, 1700 rpm for 5 min, and discard the supernatant. Perform an additional wash using 1 mL of Staining Buffer, centrifuge in identical conditions, and discard the supernatant. Finally, resuspend the cells in 300 μL of Staining Buffer and proceed with flow cytometry analysis.
2.11. Animal Protection Studies
To evaluate the anti-tuberculosis protective efficacy of the vaccine using a zebrafish (Danio rerio) M. marinum infection model, AB strain zebrafish aged 3–4 months were sourced from Nanjing Yishuli Hua and acclimated for one week before experimentation. The zebrafish were anesthetized with 500 mg/L of tricaine, which was injected into the dorsal muscle using a microinjection syringe. Each fish received 2 μL of solution, containing a total DNA dose of 6 μg. The blank control group was injected with an equivalent volume of sterile PBS. Immediately following injection, electroporation was performed using six 50 V pulses, each lasting 5 ms. The pcDNA-immunized group served as the negative control, while the experimental groups were immunized with pcDNA-lprO. Immunizations were administered twice at two-week intervals. Two weeks after the final immunization, the zebrafish were infected with 200 CFU of M. marinum 535 per fish via intraperitoneal injection, and survival was monitored post-infection.
To further assess the vaccine’s protective efficacy, a mouse BCG Pasteur infection model was employed. Four-week-old female BALB/c mice were purchased from Shanghai Slake Biological Technology Co., Ltd. and housed in an SPF animal facility. After one week of acclimatization, the mice were used for experimentation. Each mouse received a subcutaneous injection of ~106 CFU of either BCG Japan::pNBV1 or BCG Japan::pNBV1-lprO. The blank control group was injected with 100 μL of sterile PBS. Eight weeks post-injection, the mice were challenged via tail vein injection with ~107 CFU of BCG Pasteur. Three weeks following the challenge, lung and spleen tissues were harvested from the mice to determine the bacterial load.
To assess the protective efficacy of the vaccine, a mouse M. tb infection model was employed. Four-week-old female BALB/c or C57BL/6J mice were procured from Jiangsu Jicui Pharmaceutical Biotechnology Co., Ltd. (GemPharmatech, Nanjing, China) and housed in SPF facilities for one week prior to experimentation. The mice in the BCG immunization groups received subcutaneous injections of ~10⁶ CFU of either BCG Japan::pNBV1 or BCG Japan::pNBV1-lprO. The mice in the DNA immunization groups were intramuscularly injected with 100 μg/100 μL of either pcDNA or pcDNA-lprO, with three immunizations administered at two-week intervals. The blank control group was injected with 100 μL of sterile PBS. Three weeks following the final DNA immunization, all the mice were infected with approximately 150 CFU of M. tb H37Rv via aerosol exposure. Four weeks post-infection, lung and spleen tissues were harvested to evaluate the bacterial load.
2.12. Histopathological Analysis
The zebrafish were anesthetized with tricaine and, subsequently, euthanized. Following euthanasia, the specimens were rinsed once with sterile PBS and fixed in 4% paraformaldehyde for one week. They were then rinsed overnight with running water. Decalcification was carried out by immersing the zebrafish in a 10% EDTA solution for one week. The samples were then dehydrated using a graded ethanol series and cleared with xylene. Tissue infiltration was performed using paraffin. The paraffin was filtered to remove impurities, and the zebrafish were immersed in paraffin at 60 °C for 2 h, followed by being transferred to fresh paraffin for overnight incubation to eliminate residual xylene. Embedding was completed by placing the zebrafish in molds containing molten paraffin, ensuring the removal of air bubbles. After natural solidification, the blocks were cooled at −30 °C for 3–5 min, demolded, and sent to Shanghai Ruibaohe Biotechnology Co., Ltd. for sectioning. The returned sections underwent deparaffinization. The sections were immersed in hematoxylin for 10 min, rinsed with running water until clear, and differentiated in 1% hydrochloric acid for 2 s, followed by further rinsing.
They were then immersed in eosin for 5 s and rinsed thoroughly with running water. After drying, the sections were cleared in Van-Clear I (Servicebio, Wuhan, China) for 10 min and Van-Clear II for 5 min. Once dried, the sections were mounted with coverslips and examined microscopically. Sections containing intact zebrafish viscera were selected for acid-fast staining. After deparaffinization, sections were immersed in carbol fuchsin solution for 20 min, rinsed with tap water, and differentiated in acid alcohol for 2–5 s. Decolorization was repeated until red, rod-shaped bacteria were visible under the microscope. Sections were then counterstained with methylene blue for 3–5 s, rinsed, dried, mounted with coverslips, and examined microscopically.
4. Discussion
TB, caused by M. tb infection, is a chronic respiratory disease that has persisted for over 2000 years. Over the past two centuries, TB has been responsible for approximately one billion deaths and continues to pose a significant global health threat [
18]. The BCG vaccine, first introduced for human TB prevention in 1921, remains the only licensed vaccine against TB [
19]. As one of the longest used and most widely administered vaccines globally, BCG has demonstrated a robust safety profile. However, its efficacy as an anti-tuberculosis vaccine remains limited, offering only partial protection against pulmonary TB in children and inconsistent protection in adults (ranging from 0% to 80%), with its effectiveness diminishing with age [
20].
Various strategies are being explored worldwide to develop new vaccines against tuberculosis: (1) the construction of recombinant BCG (rBCG) vaccines, (2) the development of subunit vaccines, and (3) the creation of vaccines based on alternative bacterial strains [
21]. In this study, the potential of LprO as an antigen was investigated through the construction of recombinant BCG and DNA subunit vaccines. The
lprO gene was overexpressed in BCG Japan to produce a live recombinant BCG vaccine (designated BCG Japan::pNBV1-
lprO). Additionally, a DNA subunit vaccine encoding
lprO was constructed using the pcDNA 3.1 vector (designated pcDNA-
lprO). Using a
M. marinum-infected zebrafish model, immunization with pcDNA-
lprO significantly extended the survival of infected zebrafish. Furthermore, in a M. tb-infected mouse model, both LprO-based vaccines effectively reduced the lung bacterial burdens compared to their respective controls, although the differences compared to the control were not statistically significant for mice immunized with the recombinant BCG vaccine. Notably, pcDNA-
lprO, as a DNA vaccine encoding a single antigen, achieved a substantial reduction in the lung bacterial load (by 0.28–0.69 log
10 compared to the pcDNA group), highlighting the promising protective potential of LprO against tuberculosis.
The immunogenicity analyses of the two vaccine forms revealed that the recombinant BCG vaccine elicited a more robust CD4
+ Th1 response, characterized by elevated levels of IFN-γ, TNF-α, and IL-2, as well as an increase in dual and triple cytokine-positive cells, alongside a modest CD8
+ IFN-γ
+ TNF-α
+ T and CD8
+ IFN-γ
+ TNF-α
+ IL-2
+ T cell response. In contrast, the pcDNA-
lprO vaccine predominantly induced a stronger antigen-specific CD8
+ TNF-α
+ T cell response. In the case of the recombinant BCG vaccine, overexpression of
lprO resulted in alterations within the BCG strain itself, leading to changes in the expression of the genes beyond
lprO, potentially contributing to immune responses distinct from the parental strain. This phenomenon is consistent with observations in terms of different BCG strains, which, due to varying deletion clusters, exhibit divergent immune profiles [
22]. Sang et al. reported that overexpression of
phoP-phoR in recombinant BCG Japan enhanced its protective capacity against tuberculosis. Transcriptomic analysis of the recombinant BCG demonstrated that
phoP-phoR overexpression modified the expression of numerous proteins involved in lipid metabolism, potentially accounting for its enhanced immunogenicity [
23]. Both LprO vaccine forms demonstrated comparable immunogenicity, eliciting significantly elevated levels of M. tb and LprO-specific CD8
+ TNF-α
+ T cell responses relative to the control group. These findings suggest that the anti-tubercular efficacy of the LprO single antigen is primarily mediated through the induction of CD8
+ TNF-α
+ T cell responses. Previous studies have established that CD8
+ T cells possess multifunctional properties, enabling them to release cytokines or cytotoxic molecules upon activation, resulting in the apoptosis of target cells [
24]. During M. tb infection, the host induces apoptosis in infected cells, encapsulating the bacteria within apoptotic bodies, which are subsequently phagocytosed by uninfected macrophages, thereby limiting bacterial dissemination [
25]. Notably, prior research has shown that the M. tb lipoprotein Mpt83 can induce apoptosis in infected macrophages, suggesting that the mycobacterium lipoprotein, LprO, may exhibit similar capabilities [
13,
26].
Currently, 15 anti-tuberculosis vaccines are undergoing clinical trials, including nine subunit vaccines (encompassing viral vector, RNA, and protein-adjuvant platforms). Of these, all but two RNA vaccines (BNT164a1 and BNT164b1), for which the antigenic details remain undisclosed, incorporate multiple antigens. In this study, the DNA vaccine targeting tuberculosis contained only the LprO antigen, potentially limiting its protective efficacy. Given the resilience of M. tb, which is either eradicated by the host immune system or contained within granulomas leading to latent infection, incomplete clearance or control can result in active disease progression [
27]. Future research should explore the integration of additional antigens to construct fusion proteins capable of eliciting a broader anti-tuberculosis immune response. Moreover, alternative subunit vaccine formats, such as protein–adjuvant vaccines, merit further investigation. For example, studies suggest that the protein–adjuvant form of the mycobacterium lipoprotein LppX induces higher IFN-γ levels compared to its DNA vaccine counterpart [
14,
28].