Back to Journals » International Journal of Nanomedicine » Volume 20
Vancomycin-Loaded Isogenous Membrane Vesicles for Macrophage Activation and Intracellular Methicillin-Resistant Staphylococcus aureus Elimination
Authors Dou J, Shang W, Peng H, Yang Y, Chen J, Rao Y, Tan L, Hu Z, Wang Y, Huang X, Yang Y, Wu J, Hu Q, Xiao C, Rao X
Received 22 February 2025
Accepted for publication 7 June 2025
Published 17 June 2025 Volume 2025:20 Pages 7637—7651
DOI https://doi.org/10.2147/IJN.S524445
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 3
Editor who approved publication: Prof. Dr. Anderson Oliveira Lobo
Jianxiong Dou,1,* Weilong Shang,1,* Huagang Peng,1 Yi Yang,1 Juan Chen,2 Yifan Rao,3 Li Tan,1 Zhen Hu,1 Yuting Wang,1 Xiaonan Huang,1 Yuhua Yang,1 Jianghong Wu,1 Qiwen Hu,1 Chuan Xiao,1 Xiancai Rao1,3
1Department of Microbiology, College of Basic Medical Sciences, Army Medical University, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Chongqing, 400038, People’s Republic of China; 2Department of Pharmacy, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People’s Republic of China; 3Department of Emergency Medicine, Xinqiao Hospital, Army Medical University, Chongqing, 400037, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Xiancai Rao, Email [email protected]; [email protected] Chuan Xiao, Email [email protected]
Introduction: Methicillin-resistant Staphylococcus aureus (MRSA), a notorious multidrug-resistant (MDR) pathogen, frequently resides and proliferates within macrophages, contributing to refractory and recurrent infections. Conventional antibiotics exhibit limited efficacy against intracellular MRSA due to poor cellular penetration.
Methods: Vancomycin (VAN) was encapsulated into membrane vesicles (ΔagrAMVs) derived from the attenuated S. aureus strain RN4220ΔagrA, generating VAN-loaded nanoparticles (ΔagrAMV-VAN). In vitro and in vivo experiments were performed to test the efficacy of ΔagrAMV-VAN in intracellular MRSA clearance.
Results: ΔagrAMV-VAN demonstrated sustained VAN release and efficient extracellular MRSA eradication. Moreover, macrophages actively internalized ΔagrAMV-VAN, leading to VAN accumulation in intracellular compartments and M1 macrophage polarization, which increased MRSA killing. In vivo animal experiments revealed that ΔagrAMV-VAN was safe and effectively eliminated intracellular MRSA in abdominal infections.
Conclusion: Our findings propose a nanotherapeutic strategy that uses bacterial-derived vesicles for targeted antibiotic delivery, overcoming the intrinsic limitations of conventional therapies against intracellular MDR pathogens.
Keywords: MRSA, ΔagrAMV-VAN nanoparticles, macrophage activation, intracellular infections, intracellularly bacterial clearance
Graphical Abstract:
Introduction
Infectious diseases caused by bacteria, particularly multidrug-resistant (MDR) strains, pose a serious threat to human health.1 Methicillin-resistant Staphylococcus aureus (MRSA) is a prominent MDR pathogen known for its rapid spread and ability to cause various infections, ranging from common wound infections to lethal pneumonia, osteomyelitis, and sepsis.2 The resistance of MRSA to multiple clinically relevant drugs, such as methicillin, erythromycin, and clindamycin, complicates treatment and control efforts.3 Structure-based virtual screening has identified potential inhibitors targeting key S. aureus determinants, including the glycopeptide-resistance regulator GraR,4 methicillin-resistance-associated FmtA,5 GTPase YsxC,6 and lipophilic membrane,7 which may serve as leads for novel anti-MRSA agents. Notably, MRSA can subvert the innate immune system of macrophages and proliferate intracellularly, promoting chronic and recurrent infections.8,9 Vancomycin (VAN) is considered as a last-resort drug for severe MRSA infections.10 However, the poor cellular penetration and rapid macrophage clearance of VAN limit its efficacy against intracellular pathogens.9,11,12 Therefore, enhancing drug delivery to achieve high intracellular accumulation represents a promising strategy for eradicating persistent MRSA infections.
Nanodrug carriers substantially improve transmembrane permeability, intracellular accumulation, and antimicrobial activity of encapsulated drugs against intracellular infections caused by MDR pathogens.13,14 A variety of nanodrug carriers, such as liposomes,15 poly(lactic-co-glycolic acid) particles,16 exosomes,17 and inorganic nanoparticles,18 have been developed to enhance drug delivery efficiency to macrophages. Among these, bacterial membrane vesicles (MVs), lipid-bound nanoparticles (20–400 nm in diameter) spontaneously released during bacterial growth, have emerged as attractive nanodrug carriers.19,20 These MVs are enriched with proteins, peptidoglycans, DNA, and RNA, most of which are pathogen-associated molecular patterns (PAMPs) capable of targeting and activating immune cells, including macrophages, to kill intracellular pathogens.19 While MVs derived from Gram-negative bacteria such as Escherichia coli have been widely explored for antibiotic delivery,18 they frequently contain lipopolysaccharide (LPS), a membrane–bound endotoxin that contributes to bacterial pathogenicity.20 In contrast, Gram-positive bacteria such as S. aureus produce LPS-free MVs with superior biocompatibility.20 Additionally, S. aureus MVs demonstrate high macrophage uptake efficiency.21 Notably, multiple studies have revealed that MVs are favorably internalized by their homologous bacteria,22–24 implying an intrinsic targeting capability of bacterial MVs.
In our previous study, we constructed an attenuated S. aureus strain, RN4220ΔagrA, and demonstrated the hypotoxicity of ΔagrAMVs to macrophages.25 Herein, we hypothesized that isogenic ΔagrAMVs encapsulated with VAN would be favorably targeted and taken up by MRSA-infected macrophages, enhancing intracellular bacterial killing. To evaluate the nanodrug carrier potential of S. aureus MVs, we prepared VAN-loaded ΔagrAMVs (ΔagrAMV-VAN). These nanoparticles exhibited a sustained-release profile and were sufficiently internalized by macrophages. Moreover, ΔagrAMV treatment promoted macrophage polarization toward the M1 phenotype, which facilitated the clearance of intracellular MRSA. ΔagrAMV-VAN demonstrated excellent safety, and treatment with ΔagrAMV-VAN effectively killed MRSA in macrophages in both in vitro and in vivo models. Collectively, this study provides a new design for isogenic nanomedicines that combines efficient drug delivery with potent antimicrobial activity against intracellular infections.
Materials and Methods
Bacterial Strains and Culture
The S. aureus strain RN4220ΔagrA was generated previously,25 and the strain USA300 (FPR3757) was kindly provided by Dr. Min Li (Shanghai Jiao Tong University, China). S. aureus strains were cultured in brain heart infusion (BHI) medium (Oxoid, UK) or on BHI agar (BHIA) at 37 °C.
Preparation of ΔagrAMVs and ΔagrAMV-VAN
ΔagrAMVs were prepared from the culture supernatant of S. aureus strain RN4220ΔagrA as previously described.23,25 Briefly, a single colony of RN4220ΔagrA grown on BHIA was picked, inoculated into fresh BHI broth, and cultivated at 37 °C for 12 h with agitation. Subsequently, the culture was diluted 1:100 in 300 mL of fresh BHI broth and incubated at 37 °C for another 12 h. The supernatant containing ΔagrAMVs was collected by centrifugation at 6000 ×g for 10 min, followed by centrifugation at 10,000 ×g for 10 min, and filtered through a 0.22 μm membrane filter (Merck Millipore, USA) to remove dead cells and cellular debris. The resulting solution was concentrated by ultrafiltration using a 100 kDa hollow fiber membrane column (GE Healthcare, USA) to enrich ΔagrAMVs. After ultracentrifugation at 200,000 ×g with a rotor (HITACHI, Japan) for 3 h, the ΔagrAMV pellets were resuspended in 4 mL of 50% (v/v) Optiprep density gradient solution (Alere Technologies AS, Norway), followed by sequential layering with 2 mL of 40% Optiprep, 2 mL of 20% Optiprep, and 1.5 mL of 10% Optiprep. After centrifugation at 200,000 ×g for 3 h at 4°C, the solution was divided into five fractions from top to bottom, transferred into sterile tubes, and analyzed by SDS-PAGE (Servicebio, China). The ΔagrAMVs located between the 20% and 40% Optiprep layers were carefully extracted, concentrated using an ultrafiltration tube (Millipore), resuspended in phosphate-buffered saline (PBS), and stored at −80 °C until use.
VAN-loaded ΔagrAMVs (ΔagrAMV-VAN) were prepared as previously described.26 In brief, VAN (Sigma-Aldrich, USA) and ΔagrAMVs were mixed at mass ratios of 1:1, 1:2, or 2:1 and subjected to sonication using a UP-50H Ultrasonic cell disruptor (Hielscher, Germany) at 30% power for six cycles (4 s pulse, 2 s pause). The mixture was then incubated at 37 °C for 1 h to allow ΔagrAMVs reformation. Free VAN was removed by ultrafiltration using an Amicon® Ultra-centrifugal filter (MWCO = 100 kDa, GE Healthcare, UK). The purified ΔagrAMV-VAN was aseptically filtered through a 0.22 μm syringe filter (Millipore) and stored at −80°C.
Characterization of ΔagrAMVs and ΔagrAMV-VAN
ΔagrAMV samples were dropped onto copper grids, allowed to sediment naturally for15 min,negatively stained with 2% (v/v) phosphotungstic acid for 15 s, and air-dried for 1 h.20 The morphology of ΔagrAMVs and ΔagrAMV-VAN was observed using transmission electron microscopy (TEM; HT7700, Hitachi, Japan). The particle size distribution and zeta potential of ΔagrAMVs and ΔagrAMV-VAN were measured by dynamic light scattering (DLS) using a Nanoparticle Size Analyzer (Zatasizer Nano ZSP, Malvern, USA).
VAN Release
High-performance liquid chromatography (HPLC) was performed to quantify ΔagrAMV-VAN as previously reported.26 Briefly, freshly prepared ΔagrAMV-VAN was enclosed in a dialysis bag (MWCO:100 kDa; Ruiswbio, USA) and incubated at 37 °C with shaking. At each time point, 1 mL aliquots were collected and analyzed by HPLC. Each aliquot was mixed with an appropriate volume of acetonitrile in a centrifuge tube. After sonication, the supernatant was collected by centrifugation at 18,000 ×g for 10 min, filtered through a 0.22 μm syringe filter, and transferred to an HPLC auto-sampler. Approximately 1 mL of the sample was injected into the HPLC system (Agilent 1260, Agilent Technologies, CA) equipped with a C18 column (extended-C18, 250 mm × 4.6 mm, 5 μm, 100 Å, Agilent). The mobile phase (KH2PO4 :acetonitrile, 90.5: 9.5, v/v, pH = 3.2) was used at 30 °C with a flow rate of 1 mL/min. VAN elution was monitored at 236 nm, and a standard curve was established using VAN concentrations ranging from 3.125 to 50 µg/mL with OpenLAB CDS ChemStation Edition software. The release profile of VAN from ΔagrAMV-VAN was evaluated in PBS (pH 7.4).
Direct Killing of MRSA by ΔagrAMV-VAN
The bactericidal activity of ΔagrAMV-VAN against planktonic MRSA was determined as previously described.27 Briefly, overnight cultured MRSA USA300 was diluted with PBS to 1×106 colony-forming units per milliliter (CFU/mL), and 100 μL of the bacterial suspension was added to each well of a 96-well plate. Next, 100 μL of PBS, VAN (5 μg/mL), ΔagrAMVs (25 μg/mL), ΔagrAMVs (25 μg/mL) plus VAN (5 μg/mL), or ΔagrAMV-VAN (25 μg/mL) was added to each well. Afterward, the plates were incubated at 37 °C for 24 h. At 0, 2, 6, 12, and 24 h post-incubation, 10 μL aliquots were collected from each well for CFU enumeration. After 24 h of treatment, bacterial cells were collected and stained using a LIVE/DEAD BacLight Bacterial Viability kit (APExBIO, USA). Cell viability was assessed using confocal laser scanning microscopy (LSM880, Zeiss, Germany).
In vitro Cellular Uptake of ΔagrAMVs and ΔagrAMV-FITC-VAN
Mouse RAW264.7 macrophages (HaiStar Biotech, Beijing, China; Cat. No. TCM-C766) were cultured in Dulbecco’s Modified Eagle Medium (DMEM; Zeta Life, USA) supplemented with 10% fetal bovine serum (FBS; Thermo Fisher Scientific, USA) and 1% (v/v) penicillin-streptomycin solution (Zeta Life, USA) at 37 °C with 5% (v/v) CO2. For cell uptake experiments, RAW264.7 cells (1 × 105 cells/well) were seeded in confocal dishes (In Vitro Scientific, China) and incubated overnight at 37 °C. ΔagrAMVs, liposomes (Abnova, China), and ΔagrAMVs pretreated with proteinase K (2 μg/mL; Beyotime, China) were labeled with 30 μM DiO dye (Dalian Meilun Biotech, China) at 37 °C for 90 min. Cells were then treated with DiO-labeled ΔagrAMVs, liposomes, or proteinase K-treated ΔagrAMVs (PK; 5 μg per dish). After 90 min of incubation at 37 °C with 5% (v/v) CO2, the culture medium was removed, and cells were washed twice with PBS. Cells were fixed with 500 μL of 4% paraformaldehyde (Biosharp, China) at room temperature for 15 min, followed by nuclear staining with 200 μL of 4’,6-diamidino-2-phenylindole (DAPI; Dowobio, China) for 10 min. After washing twice with PBS (4 min each), cells were observed under a super-resolution laser scanning confocal microscope (LSM880, Zeiss, Germany). DiO (green fluorescence) and DAPI exhibited maximum excitation wavelengths of 484 nm and 358 nm, respectively.
FITC-labeled VAN (FITC-VAN; Ruixibio, Xi’an, China) was used to prepare ΔagrAMV-FITC-VAN formulation at a 1:2 mass ratio. For uptake experiment, RAW264.7 macrophages were treated with either FITC-VAN (3 μg/dish) or ΔagrAMV-FITC-VAN (15 μg/dish). Fluorescence intensity was detected by confocal laser scanning microscopy (CLSM).20
Inflammatory Factor Detection
RAW264.7 macrophages were seeded in 24-well plates at a density of 1×105 cells/well and incubated overnight at 37 °C with 5% (v/v) CO2. The following day, cells were treated with VAN (5 μg/mL), liposomes (5 μg/mL), or ΔagrAMVs (5 μg/mL). An equal volume of PBS was used as a negative control. After 12 h of treatment, culture supernatants were collected, and the levels of inflammatory factors TNF-α and IL-6 were detected using an enzyme-linked immunosorbent assay (ELISA) kit (MEIMIAN, China).20
Macrophage Polarization Analysis
RAW264.7 macrophages cultured in confocal dishes were treated with ΔagrAMVs (5 μg/mL) for 12 or 24 h. Untreated cells were used as blank controls, while PBS-treated cells served as negative controls. After fixation with 4% (v/v) paraformaldehyde, macrophage polarization was assessed using anti-mouse CD86 antibodies (Multisciences, China). CD86+ cells, representing M1-polarized macrophages, were analyzed by flow cytometry (Challenbio, China) and CLSM (LSM880) as described.28,29
MRSA Killing by ΔagrAMV-Activated Macrophages
Methicillin-resistant S. aureus (MRSA) infection was performed as previously described.30 For assessing ΔagrAMV-mediated macrophage killing of MRSA, RAW264.7 cells were seeded in 24-well plates (1 × 105 cells/well) and cultured overnight at 37 °C with 5% CO2. Cells were then infected with MRSA USA300 at a multiplicity of infection (MOI) of 15 (approximately 5×106 CFU/well). After 2 h of infection, extracellular bacteria were eliminated by treatment with 20 μg/mL lysostaphin (Sigma-Aldrich, USA) for 20 min. Afterward, fresh DMEM medium supplemented with VAN (2.5 μg/mL), liposomes (2.5 μg/mL), or ΔagrAMVs (2.5 μg/mL) was added, followed by incubation for an additional 12 h. PBS was used as a negative control. Intracellular bacterial counts were determined by plate assay.18 In brief, infected cells were collected by centrifugation at 6000 ×g for 10 min at 4 °C. Cell pellets were treated with 15 µg/mL lysostaphin for 20 min at room temperature, washed thrice with PBS, and lysed with Hank’s Balanced Salt Solution (Servicebio, China) containing 0.2% (v/v) Triton X-100 for 10 min. Subsequently, intracellular CFUs were quantified using BHI agar plate dilution assays.
For evaluating ΔagrAMV-activated macrophage killing of MRSA, RAW264.7 cells were pretreated for 2 h with VAN, liposomes, or ΔagrAMVs at various concentrations (0, 2.5, 5.0, and 10.0 μg/mL). Following treatment, MRSA infection was conducted as above. After lysostaphin treatment (20 μg/mL) to eliminate extracellular bacteria and replacement with fresh DMEM, intracellular MRSA counts were detected 12 and 24 h post-infection.
Intracellular MRSA Killing by ΔagrAMV-VAN
RAW264.7 cells were cultured in 24-well plates and infected with MRSA USA300 at an MOI of 20 as described above. The infected cells were then cultured in fresh DMEM supplemented with ΔagrAMV-VAN (25 μg/mL). Control groups included MRSA-infected cells treated with PBS, VAN alone (5 μg/mL), ΔagrAMVs alone (25 μg/mL), and ΔagrAMV (25 μg/mL) + VAN (5 μg/mL). Intracellular bacterial loads were counted at 12 and 24 h post-infection.
In vivo Anti-MRSA Effect of ΔagrAMV-VAN
Female BALB/c mice (6–8 weeks old) were obtained from Chongqing Byrness Weil Biotechnology Co., Ltd. (Chongqing, China). All animal experiments were approved by the Laboratory Animal Welfare and Ethics Committee of Army Medical University (Approval No. AMUWEC2020735).
A murine abdominal infection model was established as previously described.18 Briefly, 20 BALB/c mice were intraperitoneally inoculated with 5×107 CFU of MRSA USA300 and randomly allocated into five groups: (1) PBS (negative control), (2) VAN (10 mg/kg; positive control), (3) ΔagrAMVs (6 mg/kg), (4) ΔagrAMV+VAN (equivalent individual doses), and (5) ΔagrAMV-VAN (6 mg/kg). After 24 h of treatment, mice were anesthetized with 1% (w/v) pentobarbital sodium, followed by peritoneal lavage with 3 mL sterile PBS. All animals were euthanized on day 2 post-infection. Peritoneal lavage fluid was centrifuged at 6000 ×g for 10 min at 4 °C to pellet cells. Bacterial loads in both the supernatant (extracellular) and cell lysates (intracellular) were counted separately.
Safety Assessment
RAW264.7 cells were used to assess the cytotoxicity of ΔagrAMV-VAN using the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay as previously described.18 In brief, RAW264.7 macrophages were seeded in 96-well plates (2 × 103 cells/well) and cultured for 24 h at 37 °C with 5% (v/v) CO2. The culture medium was replaced with fresh medium supplemented with different concentrations of ΔagrAMVs (0, 25, 50, 100, and 200 μg/mL). Cell viability was detected 2, 6, 12, and 24 h after treatment using a Cell Counting Kit (Zeta Life).
A mouse abdominal infection model was generated (n = 3 per group) and treated with PBS, VAN (10 mg/kg), ΔagrAMVs (6 mg/kg), or ΔagrAMV-VAN (6 mg/kg). After 24 h of treatment, MRSA-infected mice were sacrificed, and organs (including the heart, liver, spleen, lungs, and kidneys) were harvested and fixed with 4% (v/v) paraformaldehyde. Histopathological examination was then performed on organ sections using hematoxylin and eosin (H&E) staining. Serum IL-6 and TNF-α levels at 6 h post-treatment were determined using an ELISA kit (MEIMIAN).
Statistical Analysis
All experiments were repeated at least three times, and the data are expressed as the mean ± standard deviation (SD) or standard error of the mean (SEM). Statistical comparisons were performed using GraphPad Prism 9.5 program (GraphPad Software, USA). The significance of differences between groups was assessed using Student’s t-test, one-way analysis of variance (ANOVA), or two-way ANOVA. A P-value of < 0.05 was considered statistically significant.
Results and Discussion
Preparation and Characterization of ΔagrAMVs and ΔagrAMV-VAN Particles
The quorum-sensing accessory gene regulator (agr) is a major factor that controls S. aureus virulence.31,32 We previously showed that agr deletion remarkably decreased the virulence of MVs in S. aureus strain RN4220.20 Therefore, we used the engineered RN4220ΔagrA as the starting strain.25 ΔagrAMVs were prepared from the culture supernatant of RN4220ΔagrA. After density gradient centrifugation, the ΔagrAMVs was verified by SDS-PAGE, and were found to be mainly located between 20% and 40% Optiprep (Figure S1). TEM analysis exhibited the nanoscale vesicles of ΔagrAMVs (Figure 1a). The protein concentration of ΔagrAMVs was determined (Figure S2), and VAN-loaded ΔagrAMVs were prepared using an ultrasonic method as previously described.18 VAN-loaded liposomal formulations often have low encapsulation efficiencies, ranging from 0.1% to 9%.11 To achieve high drug-loading efficiency, ΔagrAMVs:VAN mass ratios of 1:1, 1:2, and 2:1 were used. The results indicated that the ΔagrAMVs:VAN ratio of 1:2 achieved the highest VAN loading ratio of 21.6%, which was significantly higher than those at 1:1 (6.3%) and 2:1 (2.0%; P < 0.001; Figure 1b). TEM observations demonstrated that ΔagrAMV-VAN maintained its spherical shape (Figure 1c). DLS measurements indicated that the mean size of ΔagrAMVs was 71 nm, whereas the particle size of ΔagrAMV-VAN increased to 101 nm (Figure 1d). Xie et al18 found that the size of VAN-loaded and polydopamine-encapsulated mesoporous SiO2 nanoparticles increased from 106 to 122 nm after coating with E. coli MVs. This phenomenon indicated the versatile potential of bacterial MVs as drug carriers.
Polydispersity index (PDI) determination revealed that ΔagrAMV-VAN had a decreased PDI from 0.6 (ΔagrAMVs) to 0.38 (Figure 1e), along with an increase in the absolute value of the membrane surface potential (Figure 1f). These data indicate that the uniformity of ΔagrAMV particles improved after loading VAN under our experimental conditions. HPLC was performed to determine the VAN content in ΔagrAMV-VAN according to a standard curve (Figure S3). The results showed that approximately 80% of the VAN was released from ΔagrAMV-VAN within 8 h under neutral conditions (Figure 1g). Overall, the prepared ΔagrAMV-VAN maintained a spherical, nanosized shape with relative homogeneity compared to ΔagrAMVs, and the VAN-loaded ΔagrAMV particles exhibited a sustained release effect.
MRSA Killing Effect of ΔagrAMV-VAN In Vitro
To address the resistance of S. aureus, many structure-based virtual screening methods have been developed to target antibiotic-resistance-associated determinants such as GraR and FmtA.4,5 The screened compounds or inhibitors are potential antimicrobial agents against MRSA after molecular dynamics, pharmacophore modeling, and binding free energy analyses.4–7 However, the application of these identified molecules against MRSA infections can only be realized after further experimental evaluation. To assess the anti-staphylococcal effect of ΔagrAMV-VAN, agar plate counting was performed as previously described.10 As shown in Figure 2a, the minimal inhibitory concentration (MIC) of ΔagrAMV-VAN against MRSA USA300 was 12.5 μg/mL. Approximately 1×106 CFUs of S. aureus USA300 cells were treated with 10 μg/mL VAN, 50 μg/mL ΔagrAMVs, ΔagrAMV+VAN (10 μg/mL VAN + 50 μg/mL ΔagrAMVs), or 50 μg/mL ΔagrAMV-VAN. Plate counting was conducted at 0, 2, 6, 12, and 24 h post-culture, and the results indicated that ΔagrAMVs exhibited no bacteriostatic activity, showing bacterial counts similar to those of the PBS control (Figure 2b). However, the VAN-loaded ΔagrAMV formulation (ΔagrAMV-VAN) showed antimicrobial activity comparable to that of the free VAN and the ΔagrAMV+VAN combination, except at the 6-h time point, where the ΔagrAMV-VAN treatment group showed a slightly higher survival rate than the free VAN and ΔagrAMV+VAN groups (Figure 2c). We hypothesized that the slow-release effect of ΔagrAMV-VAN might account for this phenomenon. Notably, complete elimination of MRSA USA300 at 12 h post-incubation was achieved in the VAN, ΔagrAMV+VAN, and ΔagrAMV-VAN groups (Figure 2b and c).
CLSM, equipped with a LIVE/DEAD cell staining kit, allowed direct visualization of bacterial inactivation, where live bacteria (labeled with calcein-AM) appeared green, and dead cells (stained with propidium iodide, PI) appeared red.33 As shown in Figure 2d, treatment with VAN, ΔagrAMV+VAN, and ΔagrAMV-VAN considerably increased red fluorescence (dead cells) compared to the PBS and ΔagrAMV groups. This finding was consistent with the plate counting results (Figure 2b), indicating that the encapsulation of VAN in S. aureus ΔagrAMVs did not compromise the antibacterial activity of VAN.
Cellular Uptake of ΔagrAMV Nanoparticle by Macrophages in vitro
Studies have shown that MVs derived from S. aureus can be efficiently taken up by macrophages, a process mediated by motor proteins on the MV surface.33 While primary macrophages derived from human peripheral blood mononuclear cells may offer greater physiological relevance for modeling intracellular infections,34 stable macrophage cell lines such as human THP-1 and murine RAW264.7 have been widely utilized in developing intracellular infection models.35–37 To evaluate the potential of ΔagrAMVs as a vehicle for intracellular drug delivery, we used the RAW264.7 cell line to assess the uptake efficiency of ΔagrAMVs by macrophages. Commercial liposomes, which are established drug delivery carriers,15 served as controls. DiO-labeled ΔagrAMVs or liposomes were co-incubated with RAW264.7 cells, and the green fluorescence intensity within macrophages was visualized using CLSM (Figure 3a). The fluorescence intensity was significantly higher in the ΔagrAMV group than in the liposome group (P < 0.01; Figure 3b). However, uptake was inhibited when ΔagrAMVs were pretreated with proteinase K (PK) for 1 h (ΔagrAMVs (PK)), suggesting that this phenomenon may be attributed to the degradation of surface-associated proteins on ΔagrAMVs.33
To determine whether VAN loading affected ΔagrAMV uptake efficiency, we examined the internalization of ΔagrAMV-VAN nanoparticles by RAW264.7 cells. Fluorescein isothiocyanate (FITC)-labeled VAN (FITC-VAN) was used to generate the nanomaterial ΔagrAMV-FITC-VAN. CLSM revealed that the uptake of ΔagrAMV-FITC-VAN by RAW264.7 macrophages was remarkably enhanced compared to free FITC-VAN (Figure 3c). Quantitative fluorescence analysis confirmed that the ΔagrAMV-FITC-VAN formulation significantly improved VAN uptake efficiency (P < 0.001; Figure 3d). Overall, these results indicate that ΔagrAMV-VAN can efficiently deliver VAN to macrophages. Given that high intracellular drug concentrations are crucial for killing intracellular pathogens,12 the enhanced internalization of VAN mediated by ΔagrAMVs may facilitate the clearance of intracellular MRSA within macrophages.
Effect of ΔagrAMVs on Macrophage Polarization
Macrophages exhibit two activation phenotypes in response to external stimuli: M1 and M2, which are closely associated with inflammatory responses.28,38 M2 macrophages characterized by an altered metabolic state or impaired bactericidal activity, can provide a niche for the long-term survival of intracellular bacteria, contributing to persistent and recurrent infections.3,39 The MVs secreted by bacteria contain diverse biomolecules, such as proteins, lipids, nucleic acids, and metabolites, which play crucial roles in bacterial physiology like resistance transfer.19,40 Studies have shown that bacterial MVs containing various PAMPs can modulate macrophage activation.41–43 To determine the effect of S. aureus RN4220ΔagrA-derived ΔagrAMVs on macrophage activation, we treated RAW264.7 cells with ΔagrAMVs for 12 h. As shown in Figure 4a and b, ΔagrAMV treatment considerably increased TNF-α and IL-6 levels compared to the PBS, VAN, and liposome groups, which displayed similar cytokine profiles. The increased TNF-α and IL-6 levels suggest M1 polarization of macrophages, a phenotype associated with enhanced bacterial clearance.38,44
CD86, a hallmark of M1 macrophages,2 was analyzed by flow cytometry. Treatment with S. aureus ΔagrAMVs for 12 or 24 h substantially increased the proportion of CD86+ macrophages compared to the untreated and PBS control groups (Figure 4c and d), further supporting M1 polarization of RAW264.7 cells. This finding was further confirmed by immunofluorescence staining (Figure 4e and f). Collectively, these results demonstrate that S. aureus RN4220ΔagrA-derived ΔagrAMVs induce M1 macrophage polarization, thereby facilitating intracellular bacterial clearance.
Effect of ΔagrAMVs on MRSA Killing by Macrophages
Since M1 macrophages promote pathogen clearance,2,38 we investigated the effect of ΔagrAMVs on macrophage antibacterial activity. RAW264.7 macrophages were infected with MRSA USA300 at an MOI of 15. After 2 h of infection, cells were treated with PBS, VAN, liposomes, or ΔagrAMVs for 12 h. Plate counting revealed a remarkable reduction in intracellular MRSA load in macrophages treated with ΔagrAMVs compared to the PBS control (Figure 5a). Although VAN treatment also considerably reduced MRSA burden in macrophages, liposomes did not contribute to MRSA killing by macrophages. These data indicate that ΔagrAMVs promote macrophage-mediated clearance of intracellular MRSA.
Next, we determined whether ΔagrAMVs could prevent MRSA infection in macrophages. RAW264.7 cells were pretreated with PBS, VAN, liposomes, or ΔagrAMVs for 2 h prior to MRSA infection. After 12 h of infection, intracellular bacteria were counted using a plate assay.45 ΔagrAMV pretreatment significantly decreased intracellular MRSA burden compared to PBS (P < 0.001; Figure 5b), with dose-dependent protection observed (Figure 5c). In contrast, neither VAN nor liposomes conferred prophylactic effects against MRSA infection (Figure 5b). These results indicate that S. aureus ΔagrAMVs protect macrophages from MRSA infection and that ΔagrAMV-activated macrophages efficiently eliminate intracellular MRSA.
Effect of ΔagrAMV-VAN on Intracellular MRSA Killing
Given the macrophage-activating effect of S. aureus ΔagrAMVs, we determined whether ΔagrAMV encapsulation enhances the efficacy of VAN against intracellular MRSA in macrophages. RAW264.7 cells were seeded at 1×105 cells/well and cultured overnight in 5% CO2. Cells were then infected with S. aureus USA300 at an MOI of 20 for 2 h, followed by treatment with PBS (control), VAN alone, ΔagrAMVs alone, ΔagrAMV+VAN, or ΔagrAMV-VAN for 12 or 24 h. As shown in Figure 6a–c, ΔagrAMV-VAN treatment considerably reduced intracellular MRSA number compared to VAN alone at both time points (P < 0.01 or 0.001). While ΔagrAMVs and VAN combination also remarkably decreased intracellular MRSA burden, the ΔagrAMV-VAN group showed the lowest bacterial survival rate (Figure 6b and c), indicating that VAN encapsulation in ΔagrAMVs synergistically promotes bacterial clearance. This effect may be attributed to either ΔagrAMV-induced macrophage activation or improved intracellular VAN delivery via ΔagrAMVs, both of which could augment bactericidal activity against intracellular MRSA.18,38 Notably, the specific mechanism of MRSA neutralization was not elucidated. A recent study revealed that E. coli-derived MVs are preferentially internalized by E. coli and other Gram-negative bacteria like Klebsiella pneumoniae, but not by Gram-positive bacteria like S. aureus,32 highlighting the homotypic targeting capability of bacterial MVs.31 We hypothesize that VAN-loaded isogenous MVs may selectively target MRSA-infected macrophages, thereby improving intracellular bacterial clearance. Overall, the ΔagrAMV-VAN formulation showed potent intracellular bactericidal activity against MRSA, indicating significant potential as a therapeutic strategy for MRSA-refractory infections.
In vivo Anti-MRSA Efficacy of ΔagrAMV-VAN
The persistent intracellular presence of S. aureus is a major contributor to recurrent infections,5 primarily due to the limited ability of antibiotics to penetrate host cells.12 Our results demonstrated that encapsulation of VAN in ΔagrAMVs increased its cellular internalization (Figure 3c and d) and promoted intracellular MRSA clearance by RAW264.7 macrophages (Figure 6a–c). To evaluate the in vivo antibacterial efficacy of ΔagrAMV- VAN, we established a mouse model of abdominal infection as previously described.17 Peritoneal fluid samples were collected 24 h post-treatment with PBS, VAN, ΔagrAMVs, ΔagrAMV+VAN, or ΔagrAMV-VAN (n = 4 per group). As shown in Figure 6d, both VAN and ΔagrAMV+VAN treatments substantially reduced extracellular bacterial loads compared to PBS (P < 0.001). While ΔagrAMV-VAN also decreased extracellular bacteria (P < 0.01), its efficacy was lower than VAN alone or the physical mixture (Figure 6e). Notably, the ΔagrAMV+VAN combination displayed superior intracellular bactericidal effects compared to VAN alone, consistent with in vitro results (Figure 6a–c). Although statistical difference was not reached relative to the combination group, ΔagrAMV-VAN treatment exhibited the lowest intracellular MRSA burden among all groups (Figure 6f). Overall, these data indicate that the ΔagrAMV-VAN formulation efficiently eradicates intracellular MRSA in vivo, highlighting its therapeutic potential for persistent infections.
Safety Assessment
Compared to MVs secreted by the wild-type RN4220 strain, ΔagrAMVs derived from S. aureus RN4220ΔagrA exhibit lower toxicity.25 To assess the biocompatibility of ΔagrAMV-VAN, RAW264.7 cells were treated with various concentrations (0, 25, 50,100, and 200 µg/mL) of ΔagrAMV-VAN for different durations (2, 6, 12, and 24 h). Cell viability remained unaffected within the first 12 h of treatment (Figure 7a). Interestingly, a 24 h treatment with ΔagrAMV-VAN remarkably increased cell proliferation compared to untreated controls. This effect may be attributed to uncertain proliferative molecules involved in ΔagrAMV nanoparticles, though the precise mechanism warrants further investigation.
We next investigated the in vivo toxicity of ΔagrAMV-VAN in a murine infection model. BALB/c mice were intraperitoneally injected with PBS, VAN, ΔagrAMVs, or ΔagrAMV-VAN. After 24 h post-injection, mouse organs (heart, liver, spleen, lungs, and kidneys) were harvested and sectioned for analysis. H&E staining revealed normal histomorphology in all all treatment groups, with no pathological alterations observed (Figure 7b). Additionally, ΔagrAMV-VAN treatment did not affect body weight or systemic levels of IL-6 and TNF-α compared to controls (Figure 7c–e). Collectively, these findings demonstrate that the ΔagrAMV-VAN formulation is biocompatible in vitro and non-toxic in vivo, supporting its potential as a safe and effective therapeutic for persistent MRSA infections.
Conclusion
We developed ΔagrAMV-VAN to achieve intracellular uptake and accumulation of therapeutic agents by MRSA-infected macrophages, enabling sustained elimination of intracellular MRSA. However, this study has several limitations. First, although multiple mass ratios (1:1, 1:2, and 2:1) were determined, the VAN loading efficiency in ΔagrAMV-VAN remained limited (21.6%). Novel preparation methods, such as using hybrid cellular MVs,27,46 may improve VAN loading capacity and warrant further exploration. Second, while MRSA can infect virtually all human organs, the tissue-targeting capability of ΔagrAMV-VAN was not evaluated. Additional infectious models, such as endocarditis and meningitis, are needed to conduct the therapeutic efficacy of ΔagrAMV-VAN in diverse pathological contexts. Third, although bacterial MVs contain certain PAMPs that may modulate macrophage activation,41–43 the specific PAMPs in ΔagrAMVs responsible for M1 macrophage polarization remain unidentified. Future studies employing proteomic analysis and genetic screening could elucidate the mechanistic basis of ΔagrAMVs-mediated macrophage activation. In summary, this study provides a promising strategy against intracellular MDR infections and offers a rational framework for designing exogenous antibacterial nanomaterials.
Data Sharing Statement
Additional information is available from the correspondence author based on reasonable request.
Ethics Approval
All animal experiments were performed in accordance with the Regulations for the Administration of Affairs Concerning Experimental Animals approved by the State Council of the People’s Republic of China, and animal procedures were reviewed and approved by the Animal Use and Care Administrative Advisory Committee of the Army Medical University (protocol no. AMUWEC2020735).
Acknowledgments
This study was supported by the National Natural Science Foundation of China (82071857 and 82272341).
Disclosure
The authors declare no conflicts of interest in this work.
References
1. Murray CJL, Ikuta KS, Sharara F, et al. Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet. 2022;399(10325):629–655. doi:10.1016/S0140-6736(21)02724-0
2. Abebe AA, Birhanu AG. Methicillin resistant Staphylococcus aureus: molecular mechanisms underlying drug resistance development and novel strategies to combat. Infect Drug Resist. 2023;16:7641–7662. doi:10.2147/IDR.S428103
3. Yu YJ, Yan JH, Chen QW, et al. Polymeric nano-system for macrophage reprogramming and intracellular MRSA eradication. J Control Release. 2023;353:591–610. doi:10.1016/j.jconrel.2022.12.014
4. Dhankhar P, Dalal V, Kotra DG, Kumar P. In-silico approach to identify novel potent inhibitors against GraR of S. aureus. Front Biosci. 2020;25(7):1337–1360. doi:10.2741/4859
5. Dalal V, Dhankhar P, Singh V, et al. Structure-based identification of potential drugs against FmtA of Staphylococcus aureus: virtual screening, molecular dynamics, MM-GBSA, and QM/MM. Protein J. 2021;40(2):148–165. doi:10.1007/s10930-020-09953-6
6. Kumari R, Rathi R, Pathak SR, Dalal V. Structural-based virtual screening and identification of novel potent antimicrobial compounds against YsxC of Staphylococcus aureus. J Mol Struct. 2022;1255:132476. doi:10.1016/j.molstruc.2022.132476
7. Kumari R, Dalal V. Identification of potential inhibitors for LLM of Staphylococcus aureus: structure-based pharmacophore modeling, molecular dynamics, and binding free energy studies. J Biomol Struct Dyn. 2022;40(20):9833–9847. doi:10.1080/07391102.2021.1936179
8. Turner NA, Sharma-Kuinkel BK, Maskarinec SA, et al. Methicillin-resistant Staphylococcus aureus: an overview of basic and clinical research. Nat Rev Microbiol. 2019;17(4):203–218. doi:10.1038/s41579-018-0147-4
9. Jorch SK, Surewaard BG, Hossain M, et al. Peritoneal GATA6+ macrophages function as a portal for Staphylococcus aureus dissemination. J Clin Invest. 2019;129(11):4643–4656. doi:10.1172/JCI127286
10. Rao Y, Peng H, Shang W, et al. A vancomycin resistance-associated WalK(S221P) mutation attenuates the virulence of vancomycin-intermediate Staphylococcus aureus. J Adv Res. 2022;40:167–178. doi:10.1016/j.jare.2021.11.015
11. al-Nawas B, Shah PM. Intracellular activity of vancomycin and Ly333328, a new semisynthetic glycopeptide, against methicillin-resistant Staphylococcus aureus. Infection. 1998;26(3):165–167. doi:10.1007/BF02771843
12. Xie S, Tao Y, Pan Y, et al. Biodegradable nanoparticles for intracellular delivery of antimicrobial agents. J Control Release. 2014;187:101–117. doi:10.1016/j.jconrel.2014.05.034
13. Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol. 2021;16(7):748–759. doi:10.1038/s41565-021-00931-2
14. Zhou K, Li C, Chen D, et al. A review on nanosystems as an effective approach against infections of Staphylococcus aureus. Int J Nanomed. 2018;13:7333–7347. doi:10.2147/IJN.S169935
15. Hulme J. Application of nanomaterials in the prevention, detection, and treatment of methicillin-resistant Staphylococcus aureus (MRSA). Pharmaceutics. 2022;14(4):805. doi:10.3390/pharmaceutics14040805
16. Lin X, He J, Li W, et al. Lung-targeting lysostaphin microspheres for methicillin-resistant Staphylococcus aureus pneumonia treatment and prevention. ACS Nano. 2021;15(10):16625–16641. doi:10.1021/acsnano.1c06460
17. Yang X, Xie B, Peng H, et al. Eradicating intracellular MRSA via targeted delivery of lysostaphin and vancomycin with mannose-modified exosomes. J Control Release. 2021;329:454–467. doi:10.1016/j.jconrel.2020.11.045
18. Xie B, Zhao H, Zhang R, et al. Bacteria-mimetic nanomedicine for targeted eradication of intracellular MRSA. J Control Release. 2023;357:371–378. doi:10.1016/j.jconrel.2023.03.053
19. Jahromi LP, Fuhrmann G. Bacterial extracellular vesicles: understanding biology promotes applications as nanopharmaceuticals. Adv Drug Deliv Rev. 2021;173:125–140. doi:10.1016/j.addr.2021.03.012
20. Yuan J, Yang J, Hu Z, et al. Safe staphylococcal platform for the development of multivalent nanoscale vesicles against viral infections. Nano Lett. 2018;18(2):725–733. doi:10.1021/acs.nanolett.7b03893
21. Wang X, Eagen WJ, Lee JC. Orchestration of human macrophage NLRP3 inflammasome activation by Staphylococcus aureus extracellular vesicles. Proc Natl Acad Sci U S A. 2020;117(6):3174–3184. doi:10.1073/pnas.1915829117
22. Ding C, He R, Cheng T, et al. Bacterial outer membrane-based biomimetic immune adaptors: mild immunomodulatory and bacterial targeted delivery strategy against implant-related infections. Adv Funct Mater. 2023;33(42):2304168. doi:10.1002/adfm.202304168
23. Peng X, Chen J, Gan Y, et al. Biofunctional lipid nanoparticles for precision treatment and prophylaxis of bacterial infections. Sci Adv. 2024;10(14):eadk9754. doi:10.1126/sciadv.adk9754
24. Wu S, Huang Y, Yan J, et al. Bacterial outer membrane-coated mesoporous silica nanoparticles for targeted delivery of antibiotic rifampicin against Gram-negative bacterial infection in vivo. Adv Funct Mater. 2021;31(35):2103442. doi:10.1002/adfm.202103442
25. Chen J, Lv Y, Shang W, et al. Loaded delta-hemolysin shapes the properties of Staphylococcus aureus membrane vesicles. Front Microbiol. 2023;14:1254367. doi:10.3389/fmicb.2023.1254367
26. Yang X, Shi G, Guo J, Wang C, He Y. Exosome-encapsulated antibiotic against intracellular infections of methicillin-resistant Staphylococcus aureus. Int J Nanomed. 2018;13:8095–8104. doi:10.2147/IJN.S179380
27. Huang R, Cai GQ, Li J, et al. Platelet membrane-camouflaged silver metal-organic framework drug system against infections caused by methicillin-resistant Staphylococcus aureus. J Nanobiotechnology. 2021;19(1):229. doi:10.1186/s12951-021-00978-2
28. Yunna C, Mengru H, Lei W, Weidong C. Macrophage M1/M2 polarization. Eur J Pharmacol. 2020;877:173090. doi:10.1016/j.ejphar.2020.173090
29. Benoit M, Desnues B, Mege JL. Macrophage polarization in bacterial infections. J Immunol. 2008;181(6):3733–3739. doi:10.4049/jimmunol.181.6.3733
30. Lehar SM, Pillow T, Xu M, et al. Novel antibody-antibiotic conjugate eliminates intracellular S. aureus. Nature. 2015;527(7578):323–328. doi:10.1038/nature16057
31. Podkowik M, Perault AI, Putzel G, et al. Quorum-sensing agr system of Staphylococcus aureus primes gene expression for protection from lethal oxidative stress. Elife. 2024;12:RP89098. doi:10.7554/eLife.89098
32. Jacob KM, Hernández-Villamizar S, Hammer ND, Reguera G. Mucin-induced surface dispersal of Staphylococcus aureus and Staphylococcus epidermidis via quorum-sensing dependent and independent mechanisms. mBio. 2024;15(8):e0156224. doi:10.1128/mbio.01562-24
33. Haney MJ, Klyachko NL, Zhao Y, et al. Exosomes as drug delivery vehicles for Parkinson’s disease therapy. J Control Release. 2015;207:18–30. doi:10.1016/j.jconrel.2015.03.033
34. Moldovan A, Wagner F, Schumacher F, et al. Chlamydia trachomatis exploits sphingolipid metabolic pathways during infection of phagocytes. mBio. 2025;18(5):e0398124. doi:10.1128/mbio.03981-24
35. Wang ZJ, Zhu YY, Bai L-Y. Bai LY, et al.A new therapeutic strategy for infectious diseases against intracellular multidrug-resistant bacteria. J Control Release. 2024;375:467–477. doi:10.1016/j.jconrel.2024.09.028
36. Yang X, Shi G, Lin Z, et al. Pathogen-targeting biomineralized bacterial outer membrane vesicles for eradicating both intracellular and extracellular Staphylococcus aureus. J Control Release. 2025;382:113702. doi:10.1016/j.jconrel.2025.113702
37. Turner AB, Giraldo-Osorno PM, Douest Y, et al. Race for the surface between THP-1 macrophages and Staphylococcus aureus on various titanium implants with well-defined topography and wettability. Acta Biomater. 2025;191:113–139. doi:10.1016/j.actbio.2024.11.013
38. Mège JL, Mehraj V, Capo C. Macrophage polarization and bacterial infections. Curr Opin Infect Dis. 2011;24(3):230. doi:10.1097/QCO.0b013e328344b73e
39. Eisele NA, Ruby T, Jacobson A, et al. Salmonella require the fatty acid regulator PPARdelta for the establishment of a metabolic environment essential for long-term persistence. Cell Host Microbe. 2013;14(2):171–182. doi:10.1016/j.chom.2013.07.010
40. Lee AR, Park SB, Kim SW, et al. Membrane vesicles from antibiotic-resistant Staphylococcus aureus transfer antibiotic-resistance to antibiotic-susceptible Escherichia coli. J Appl Microbiol. 2022;132(4):2746–2759. doi:10.1111/jam.15449
41. Kaparakis-Liaskos M, Ferrero RL. Immune modulation by bacterial outer membrane vesicles. Nat Rev Immunol. 2015;15(6):375–387. doi:10.1038/nri3837
42. Mehanny M, Koch M, Lehr CM, Fuhrmann G. Streptococcal extracellular membrane vesicles are rapidly internalized by immune cells and alter their cytokine release. Front Immunol. 2020;11:80. doi:10.3389/fimmu.2020.00080
43. Bitto NJ, Cheng L, Johnston EL, et al. Staphylococcus aureus membrane vesicles contain immunostimulatory DNA, RNA and peptidoglycan that activate innate immune receptors and induce autophagy. J Extracell Vesicles. 2021;10(6):e12080. doi:10.1002/jev2.12080
44. Kadomoto S, Izumi K, Mizokami A. Macrophage polarity and disease control. Int J Mol Sci. 2021;23(1):144. doi:10.3390/ijms23010144
45. Liu H, Wei X, Wang Z, et al. LysSYL: a broad-spectrum phage endolysin targeting Staphylococcus species and eradicating S. aureus biofilms. Microb Cell Fact. 2024;23(1):89. doi:10.1186/s12934-024-02359-4
46. Zhang S, Chen T, Lu W, et al. Hybrid cell membrane-engineered nanocarrier for triple-action strategy to address Pseudomonas aeruginosa infection. Adv Sci. 2025;12(6):e2411261. doi:10.1002/advs.202411261
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.