1
|
Lin Y, Wang J, Bu F, Zhang R, Wang J, Wang Y, Huang M, Huang Y, Zheng L, Wang Q, Hu X. Bacterial extracellular vesicles in the initiation, progression and treatment of atherosclerosis. Gut Microbes 2025; 17:2452229. [PMID: 39840620 DOI: 10.1080/19490976.2025.2452229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/13/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular and cerebrovascular diseases. However, current anti-atherosclerosis drugs have shown conflicting therapeutic outcomes, thereby spurring the search for novel and effective treatments. Recent research indicates the crucial involvement of oral and gastrointestinal microbiota in atherosclerosis. While gut microbiota metabolites, such as choline derivatives, have been extensively studied and reviewed, emerging evidence suggests that bacterial extracellular vesicles (BEVs), which are membrane-derived lipid bilayers secreted by bacteria, also play a significant role in this process. However, the role of BEVs in host-microbiota interactions remains insufficiently explored. This review aims to elucidate the complex communication mediated by BEVs along the gut-heart axis. In this review, we summarize current knowledge on BEVs, with a specific focus on how pathogen-derived BEVs contribute to the promotion of atherosclerosis, as well as how BEVs from gut symbionts and probiotics may mitigate its progression. We also explore the potential and challenges associated with engineered BEVs in the prevention and treatment of atherosclerosis. Finally, we discuss the benefits and challenges of using BEVs in atherosclerosis diagnosis and treatment, and propose future research directions to address these issues.
Collapse
Affiliation(s)
- Yuling Lin
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyu Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Bu
- Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China
| | - Ruyi Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junhui Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yubing Wang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyi Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiumei Hu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Li Y, Guo T, He J, Liu D, Peng S, Xu A. SLC35A2-mediated bisected GlcNAc-modified extracellular vesicles enhance immune regulation in breast cancer lung metastasis. Int Immunopharmacol 2025; 154:114505. [PMID: 40157085 DOI: 10.1016/j.intimp.2025.114505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 04/01/2025]
Abstract
This study investigates the role of SLC35A2-mediated bisected GlcNAc-modified small extracellular vesicles (sEVs) in breast cancer (BC) lung metastasis. By modulating B3GALT1 expression, these sEVs regulate the pre-metastatic immune microenvironment, enhancing CD8+ T cell infiltration and reducing immune evasion. The use of β-peptide-loaded sEVs further amplifies anti-metastatic effects, as demonstrated in vivo mouse models and molecular analyses. These findings underscore the therapeutic potential of glycosylation-modified sEVs in enhancing immune responses and controlling BC metastasis.
Collapse
Affiliation(s)
- Yangyang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Tao Guo
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Juntong He
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Defeng Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Shihao Peng
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Department of General Surgery, Anhui Public Health Clinical Center, Hefei, Anhui, China
| | - Aman Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
3
|
Yuan Y, Cao K, Gao P, Wang Y, An W, Dong Y. Extracellular vesicles and bioactive peptides for regenerative medicine in cosmetology. Ageing Res Rev 2025; 107:102712. [PMID: 40032214 DOI: 10.1016/j.arr.2025.102712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/10/2025] [Accepted: 02/25/2025] [Indexed: 03/05/2025]
Abstract
As life quality improves and the life pressure increases, people's awareness of maintaining healthy skin and hair grows. However, the use of bioactive peptides in regenerative medical aesthetics is often constrained by the high molecular weight, which impedes skin penetration. In contrast, extracellular vesicles not only possess regenerative properties but also serve as effective carriers for bioactive peptides. Given their anti-inflammatory and bactericidal properties, capacity to promote angiogenesis, optimize collagen alignment, facilitate re-epithelialization and stimulate hair growth, extracellular vesicles become an emerging and promising solution for skin regeneration treatments. The combination of peptides and extracellular vesicles enhances therapeutic efficacy and improves the bioavailability of bioactive peptides. In this review, we summarize the functions of bioactive peptides and plant- and animal-derived extracellular vesicles in regenerative medicine with cosmetology, along with examples of their combined applications. Additionally, we provide an overview of peptides and extracellular vesicles currently available on the market and in clinical practice, discussing the challenges and solutions associated with their use.
Collapse
Affiliation(s)
- Yize Yuan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Kailu Cao
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Peifen Gao
- National Vaccine & Serum Institute, China National Biotech Group, Sinopharm Group, Beijing 101111, China
| | - Yinan Wang
- National Vaccine & Serum Institute, China National Biotech Group, Sinopharm Group, Beijing 101111, China
| | - Wenlin An
- National Vaccine & Serum Institute, China National Biotech Group, Sinopharm Group, Beijing 101111, China.
| | - Yiyang Dong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
4
|
Li Q, Ou Z, Lin J, Tang D, He B, Wu Y, Huang X, Huang X, Ru B, Wang Q, Yao W, Situ B, Zheng L. Specific labeling of outer membrane vesicles with antibiotic-conjugated probe reveals early bacterial infections in blood. Nat Commun 2025; 16:3535. [PMID: 40229269 PMCID: PMC11997070 DOI: 10.1038/s41467-025-58676-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/26/2025] [Indexed: 04/16/2025] Open
Abstract
Bacterial outer membrane vesicles (OMVs) are nano-sized structures derived from the outer membrane of Gram-negative bacteria, which have emerged as key players in host-pathogen interactions, yet their potential as biomarkers remains largely unexplored due to the difficulty of identification in complex biological samples. Here we show an approach for detecting and quantifying bacterial OMVs in blood using a Polymyxin B-fluorescein probe (PmBF), which targets bacterial lipopolysaccharides (LPS). The probe selectively labels OMVs, enabling their differentiation from host extracellular vesicles and quantitative analysis using nano-flow cytometry. In male mouse models of pneumonia, we observe elevated serum PmBF+ EVs as early as 6 h post-infection, preceding positive blood cultures. In clinical samples, PmBF+ EVs show superior performance for diagnosing bacterial infections and differentiate them from virus or mycoplasma infections. Our findings highlight circulating PmBF+ EVs as promising biomarkers of bacterial infections.
Collapse
Affiliation(s)
- Qianbei Li
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zihao Ou
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Jinduan Lin
- Department of Laboratory Medicine, Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People's Hospital), Qingyuan, 511500, China
| | - Die Tang
- Medical Laboratory of the Third Affiliated Hospital of Shenzhen University, Shenzhen, 518003, China
| | - Bairong He
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yuanyuan Wu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinyue Huang
- Prenatal Diagnosis Center, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xixin Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bingbing Ru
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qianwen Wang
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, 510260, China
| | - Weirong Yao
- The Second Hospital of Zhangzhou, Zhangzhou, 363000, China
| | - Bo Situ
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- State Key Laboratory of Multi-organ Injury Prevention and Treatment, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
5
|
Gillett DL, Selinidis M, Seamons T, George D, Igwe AN, Del Valle I, Egbert RG, Hofmockel KS, Johnson AL, Matthews KRW, Masiello CA, Stadler LB, Chappell J, Silberg JJ. A roadmap to understanding and anticipating microbial gene transfer in soil communities. Microbiol Mol Biol Rev 2025:e0022524. [PMID: 40197024 DOI: 10.1128/mmbr.00225-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
SUMMARYEngineered microbes are being programmed using synthetic DNA for applications in soil to overcome global challenges related to climate change, energy, food security, and pollution. However, we cannot yet predict gene transfer processes in soil to assess the frequency of unintentional transfer of engineered DNA to environmental microbes when applying synthetic biology technologies at scale. This challenge exists because of the complex and heterogeneous characteristics of soils, which contribute to the fitness and transport of cells and the exchange of genetic material within communities. Here, we describe knowledge gaps about gene transfer across soil microbiomes. We propose strategies to improve our understanding of gene transfer across soil communities, highlight the need to benchmark the performance of biocontainment measures in situ, and discuss responsibly engaging community stakeholders. We highlight opportunities to address knowledge gaps, such as creating a set of soil standards for studying gene transfer across diverse soil types and measuring gene transfer host range across microbiomes using emerging technologies. By comparing gene transfer rates, host range, and persistence of engineered microbes across different soils, we posit that community-scale, environment-specific models can be built that anticipate biotechnology risks. Such studies will enable the design of safer biotechnologies that allow us to realize the benefits of synthetic biology and mitigate risks associated with the release of such technologies.
Collapse
Affiliation(s)
- David L Gillett
- Department of Biosciences, Rice University, Houston, Texas, USA
| | - Malyn Selinidis
- Department of Biosciences, Rice University, Houston, Texas, USA
| | - Travis Seamons
- Department of Biosciences, Rice University, Houston, Texas, USA
| | - Dalton George
- Department of Biosciences, Rice University, Houston, Texas, USA
- School for the Future of Innovation in Society, Arizona State University, Tempe, Arizona, USA
| | - Alexandria N Igwe
- Department of Biological Sciences, Virginia Tech, Blacksburg, Virginia, USA
| | - Ilenne Del Valle
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
| | - Robert G Egbert
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Kirsten S Hofmockel
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Alicia L Johnson
- Baker Institute for Public Policy, Rice University, Houston, Texas, USA
| | | | - Caroline A Masiello
- Department of Biosciences, Rice University, Houston, Texas, USA
- Department of Earth, Environmental and Planetary Sciences, Rice University, Houston, Texas, USA
| | - Lauren B Stadler
- Department of Civil and Environmental Engineering, Rice University, Houston, Texas, USA
| | - James Chappell
- Department of Biosciences, Rice University, Houston, Texas, USA
| | | |
Collapse
|
6
|
Li C, Zeng A, Li L, Zhao W. Emerging Roles of Plant-Derived Extracellular Vesicles in Biotherapeutics: Advances, Applications, and Future Perspectives. Adv Biol (Weinh) 2025:e2500008. [PMID: 40197701 DOI: 10.1002/adbi.202500008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/27/2025] [Indexed: 04/10/2025]
Abstract
Extracellular vesicles (EVs) are nanoscale luminal vesicles, which play an important role in intercellular communication through surface signaling and molecular cargo delivery (proteins, lipids, nucleic acids, etc.). Recently, plant-derived extracellular vesicles (PDVs) containing multiple biological activities have received increasing attention due to their better biocompatibility and lower cytotoxicity in healthy tissues. In the biomedical field, PDVs are employed as cargo delivery vehicles, enabling diverse functionalities through engineering modification techniques. Nonetheless, there are certain issues with the study of PDVs, such as the lack of standardization in the identification and isolation criteria. This review provides a quick overview of the biogenesis, physicochemical properties, isolation techniques, and biomedical applications of PDVs in current studies, while critically analyzing the current challenges and opportunities. This paper is expected to provide some theoretical guidance for the development of PDVs and further biomedical applications.
Collapse
Affiliation(s)
- Cheng Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
| | - Aoqiong Zeng
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
| | - Li Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
| | - Wei Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, 1800 Lihu Avenue, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
7
|
Liao X, Si H, Lai Y, Zhang X, Feng Y, Zhou T, Feng Y, Yu L. Porphyromonas gingivalis-OMVs promote the epithelial-mesenchymal transition of oral squamous cell carcinoma by inhibiting ferroptosis through the NF-κB pathway. J Oral Microbiol 2025; 17:2482924. [PMID: 40206095 PMCID: PMC11980236 DOI: 10.1080/20002297.2025.2482924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/10/2025] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
Background Recent studies reported the role of Porphyromonas gingivalis (P. g) in promoting oral squamous cell carcinoma (OSCC) progression. However, the molecular mechanism remains unclear. Materials and methods P. g-OMVs were isolated using ultracentrifugation method and characterized by transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). CCK-8, migration, invasion, Quantitative real-time Polymerase Chain Reaction (qRT-PCR) and immunocytochemistry assays were performed to evaluate the effect of P. g-OMVs on tumor cells' proliferation, migration, invasion, epithelial-mesenchymal transition (EMT), and ferroptosis in vitro. Western blot was performed to study the phosphorylation of transcription factor nuclear factor kappa B (NF-κB). In vivo, the effect of P. g-OMVs on the growth of OSCC was evaluated using a xenograft tumor model, followed by hematoxylin and eosin and immunohistochemistry staining. Results TEM and NTA demonstrated that P. g-OMVs have a vesicular structure with a particle size of around 118 nm. Compared to the control group, P. g-OMVs significantly enhance the proliferation, migration, and invasion of tumor cells. In addition, P. g-OMVs promote the EMT of OSCC cells, which can be attenuated by ferroptosis activator erastin. Moreover, P. g-OMVs inhibit feroptosis of OSCC by activating NF-κB signaling. In vivo, P. g-OMVs significantly enhance tumor growth of OSCC. Inhibition of NF-κB could significnatly reduce the growth of OSCC, which can be further rescued using ferroptosis inhibitor Ferrostain-1. Conclusions P. g-OMVs promote OSCC progression by modulating the ferroptosis-related EMT through NF-κB signaling.
Collapse
Affiliation(s)
- Xinyue Liao
- Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Hang Si
- Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Yongxian Lai
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Department of Preventive Health Care, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoyan Zhang
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Department of Periodontics & Oral Mucosal Diseases, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
| | - Yun Feng
- Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Tiejun Zhou
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yan Feng
- Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
| | - Li Yu
- Oral & Maxillofacial Reconstruction and Regeneration of Luzhou Key Laboratory, Luzhou, China
- School of Stomatology, Southwest Medical University, Luzhou, China
- Department of Periodontics & Oral Mucosal Diseases, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, China
| |
Collapse
|
8
|
Zhao Y, Xue Y, Wang C, Zhao Z, Cui R, Zhu B. Antibacterial poly(ethyl methacrylate) surfaces constructed by facile amination with polyethyleneimine of different architectures. Colloids Surf B Biointerfaces 2025; 248:114458. [PMID: 39724827 DOI: 10.1016/j.colsurfb.2024.114458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/05/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
Polymethacrylate and its derivatives are widely used in food industry and biomedical applications for their plasticity, biocompatibility and optical transparency. However, susceptibility to bacterial growth on their surfaces limits their applications. In this study, linear and branched polyethyleneimine (PEI) molecules were grafted onto poly(ethyl methacrylate) (PEMA) via aminolysis using a simple one-step method to enhance the antibacterial properties of PEMA films. PEI-modified PEMA films were characterized by ATR-FTIR, X-ray photoelectron spectroscopy (XPS), field emission scanning electron microscopy (FESEM) and thermal gravimetric analysis (TGA). The modified films exhibited optimal bactericidal efficiency of 98.0 % against Escherichia coli (E. coli) and over 99.9 % against Staphylococcus aureus (S. aureus). Furthermore, hydrolysis was found to contribute to anchoring PEI onto PEMA as well. Though branched PEI exhibited a higher grafting amount than the linear ones under same conditions, PEMA modified with linear PEI presented a similar or even higher antibacterial efficiency than those grafted with branched PEI. Overall, PEI-grafted PEMA films prepared with simple one-step method exhibit effective antibacterial properties and good biocompatibilities, making them promising candidates for biomedical devices and other applications.
Collapse
Affiliation(s)
- Yu Zhao
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yunyun Xue
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China; Center of Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312000, China
| | - Chuyao Wang
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Zihao Zhao
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Ronglu Cui
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Baoku Zhu
- Key Laboratory of Macromolecular Synthesis and Functionalization (Ministry of Education), Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China; Center of Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing 312000, China.
| |
Collapse
|
9
|
Fu Z, Lin S, Chen H, Guo H, Li J, Chen Y, Lu Y, Liu J, Huang W, Pang Y. Generating Self-Adjuvated Nanofiber Vaccines by Coating Bacterial Flagella with Antigens. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415887. [PMID: 39981905 DOI: 10.1002/adma.202415887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/28/2025] [Indexed: 02/22/2025]
Abstract
Bacteria-based vaccines have received increasing attention given the ability to induce strong systemic immune responses. However, the application of bacteria as therapeutic agents inevitably suffers from infection-associated side effects due to the living characteristics. Here, the use of bacteria-derived flagella is described to construct self-adjuvated nanofiber vaccines. With the help of charge-reversal mediated by decoration with cationic polymers, the flagella can be coated with negatively charged antigens through electrostatic interaction. By virtue of the large aspect ratio, the resulting nanofiber vaccines show prolonged retention at the injection site and increased uptake by dendritic cells and macrophages. Thanks to the innate immunogenicity, self-adjuvated flagella robustly promote dendritic cell maturation and macrophage polarization, resulting in the elicitation of antigen-specific T-cell and B-cell immune responses. In ovalbumin-overexpressing melanoma-bearing mice, immunization with ovalbumin-carried vaccines not only exhibits a favorable tolerance, but also displays superior inhibition efficacies on tumor growth and metastasis separately under the therapeutic and prophylactic settings. The flexibility of this approach is further demonstrated for vaccine fabrication by coating with the SARS-CoV-2 Spike protein S1 subunit. Bacterial flagella-based self-adjuvated nanofiber platform proposes a versatile strategy to develop various vaccines for disease prevention and treatment.
Collapse
Affiliation(s)
- Zhenzhen Fu
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Sisi Lin
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Huan Chen
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Haiyan Guo
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Juanjuan Li
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yanmei Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yue Lu
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei Huang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Xiangfu Laboratory, Jiaxing, Zhejiang, 314102, China
| | - Yan Pang
- Shanghai Frontiers Science Center of Drug Target ldentification and Delivery,School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
10
|
Abolhasani FS, Vaghefinanekaran N, Yarahmadi A, Akrami S, Mirmahdavi S, Yousefi MH, Afkhami H, Shafiei M. Outer membrane vesicles in gram-negative bacteria and its correlation with pathogenesis. Front Immunol 2025; 16:1541636. [PMID: 40236702 PMCID: PMC11996793 DOI: 10.3389/fimmu.2025.1541636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 03/11/2025] [Indexed: 04/17/2025] Open
Abstract
There is a widespread distribution of gram-negative bacteria worldwide, which are responsible for the deaths of numerous patients each year. The illnesses they cause can be localized and systemic, and these bacteria possess several key virulence factors that contribute to their pathogenicity. In recent years, several distinct mechanisms of pathogenesis have evolved that remain largely unknown to scientists and medical experts. Among these, outer membrane vesicles (OMVs) are undoubtedly one of the most significant factors influencing virulence. OMVs contain various bacterial compounds and can have diverse effects on host organisms and the immune system, potentially exacerbating disease and inflammation while evading immune responses. This review comprehensively examines the role of OMVs in bacterial pathogenesis, their interaction with host cells, and their potential biomedical applications. Understanding the molecular mechanisms governing OMV biogenesis and function could pave the way for novel antimicrobial strategies and therapeutic interventions.
Collapse
Affiliation(s)
- Fatemeh Sadat Abolhasani
- Department of Pathobiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Sousan Akrami
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Solmaz Mirmahdavi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Microbiology, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Hasan Yousefi
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Hamed Afkhami
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, School of Medicine, Shahed University, Tehran, Iran
| | - Morvarid Shafiei
- Department of Bacteriology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
11
|
Truchet S, Malet-Villemagne J, Tessier G, Vidic J. Visualization of the Biogenesis, Dynamics, and Host Interactions of Bacterial Extracellular Vesicles. CHEMICAL & BIOMEDICAL IMAGING 2025; 3:132-136. [PMID: 40151818 PMCID: PMC11937970 DOI: 10.1021/cbmi.5c00002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 01/22/2025] [Indexed: 03/29/2025]
Affiliation(s)
- Sandrine Truchet
- Université
Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Jeanne Malet-Villemagne
- Université
Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Gilles Tessier
- Sorbonne
Université, CNRS UMR7210, INSERM UMRS968, Institut de la Vision, 75012 Paris, France
| | - Jasmina Vidic
- Université
Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| |
Collapse
|
12
|
Müller GA. The Transformation Experiment of Frederick Griffith I: Its Narrowing and Potential for the Creation of Novel Microorganisms. Bioengineering (Basel) 2025; 12:324. [PMID: 40150788 PMCID: PMC11939280 DOI: 10.3390/bioengineering12030324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/12/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
The construction of artificial microorganisms often relies on the transfer of genomes from donor to acceptor cells. This synthetic biology approach has been considerably fostered by the J. Craig Venter Institute but apparently depends on the use of microorganisms, which are very closely related. One reason for this limitation of the "creative potential" of "classical" transformation is the requirement for adequate "fitting" of newly synthesized polypeptide components, directed by the donor genome, to interacting counterparts encoded by the pre-existing acceptor genome. Transformation was introduced in 1928 by Frederick Griffith in the course of the demonstration of the instability of pneumococci and their conversion from rough, non-pathogenic into smooth, virulent variants. Subsequently, this method turned out to be critical for the identification of DNA as the sole matter of inheritance. Importantly, the initial experimental design (1.0) also considered the inheritance of both structural (e.g., plasma membranes) and cybernetic information (e.g., metabolite fluxes), which, in cooperation, determine topological and cellular heredity, as well as fusion and blending of bacterial cells. In contrast, subsequent experimental designs (1.X) were focused on the use of whole-cell homogenates and, thereafter, of soluble and water-clear fractions deprived of all information and macromolecules other than those directing protein synthesis, including outer-membrane vesicles, bacterial prions, lipopolysaccharides, lipoproteins, cytoskeletal elements, and complexes thereof. Identification of the reasons for this narrowing may be helpful in understanding the potential of transformation for the creation of novel microorganisms.
Collapse
Affiliation(s)
- Günter A. Müller
- Biology and Technology Studies Institute Munich (BITSIM), 80939 Munich, Germany; ; Tel.: +49-151-25216987
- Institute of Media Sociology, Department of Cultural Sciences, University of Paderborn, 33104 Paderborn, Germany
| |
Collapse
|
13
|
Wu Y, Huang X, Li Q, Yang C, Huang X, Du H, Situ B, Zheng L, Ou Z. Reducing severity of inflammatory bowel disease through colonization of Lactiplantibacillus plantarum and its extracellular vesicles release. J Nanobiotechnology 2025; 23:227. [PMID: 40114208 PMCID: PMC11924789 DOI: 10.1186/s12951-025-03280-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by compromised intestinal barrier function and a lack of effective treatments. Probiotics have shown promise in managing IBD due to their ability to modulate the gut microbiota, enhance intestinal barrier function, and exert anti-inflammatory effects. However, the specific mechanisms through which probiotics exert these therapeutic effects in IBD treatment remain poorly understood. Our research revealed a significant reduction of Lactiplantibacillus plantarum (L. plantarum) in the gut microbiota of IBD patients. L. plantarum is a well-known probiotic strain in the list of edible probiotics, recognized for its beneficial effects on gut health, including its ability to strengthen the intestinal barrier and reduce inflammation. We demonstrated that supplementation with L. plantarum could alleviate IBD symptoms in mice, primarily by inhibiting apoptosis in intestinal epithelial cells through L. plantarum's bacterial extracellular vesicles (L. plant-EVs). This protective effect is dependent on the efficient uptake of L. plant-EVs by intestinal cells. Intriguingly, watermelon enhances L. plantarum colonization and L. plant-EVs release, further promoting intestinal barrier repair. Our findings contribute to the understanding of L. plant-EVs in the probiotic-based therapeutic approach for IBD, as they are promising candidates for nanoparticle-based therapeutic methods that are enhanced by natural diets such as watermelon. This study thereby offers a potential breakthrough in the management and treatment of IBD.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinyue Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qianbei Li
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaoqun Yang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Ganzhou Hospital-Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xixin Huang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hualongyue Du
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Bo Situ
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Zihao Ou
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
14
|
Hong MG, Song EJ, Yoon HJ, Chung WH, Seo HY, Kim D, Lee D, Seo JG, Lee H, Kim SI, Kim GJ, Kim KN, Lee SN, Kim KS, Nam YD. Clade-specific extracellular vesicles from Akkermansia muciniphila mediate competitive colonization via direct inhibition and immune stimulation. Nat Commun 2025; 16:2708. [PMID: 40108178 PMCID: PMC11923206 DOI: 10.1038/s41467-025-57631-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
Akkermansia muciniphila, a promising candidate for next-generation probiotics, exhibits significant genomic diversity, classified into several distinct clades (AmI to AmIV). Notably, a single Akkermansia clade tends to predominate within individual hosts, with co-occurrence of different clades being rare. The mechanisms driving such clade-specific exclusion remain unclear. Here, we show that extracellular vesicles (EVs) derived from AmII clade inhibit the growth of clade I (AmI), conferring a competitive advantage to AmII. Moreover, we observe clade-specific immunoglobulin A (IgA) responses, where AmII clade-specific IgAs, induced by EVs from AmII, facilitate niche occupancy and competitive exclusion of AmI. These findings provide insights into the competitive dynamics of A. muciniphila clades and suggest that future personalized microbiome interventions could be optimized by considering the clade composition of A. muciniphila in individual hosts.
Collapse
Affiliation(s)
- Moon-Gi Hong
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si, Republic of Korea
| | - Eun-Ji Song
- Research Group of Personalized Diet, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Hye Jin Yoon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Won-Hyong Chung
- Research Group of Personalized Diet, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Hae Yeong Seo
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Dohak Kim
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si, Republic of Korea
| | - Dokyung Lee
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si, Republic of Korea
| | - Jae-Gu Seo
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si, Republic of Korea
| | - Hayoung Lee
- Digital Omics Research Center, Korea Basic Science Institute, Cheongju, Republic of Korea
- Critical Diseases Diagnostics Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Seung Il Kim
- Digital Omics Research Center, Korea Basic Science Institute, Cheongju, Republic of Korea
| | - Gwang Joong Kim
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, Republic of Korea
| | - Kil-Nam Kim
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju, Republic of Korea
| | - Sang-Nam Lee
- R&D Center, Enterobiome Inc., 814 Siksa-dong, Ilsandong-gu, Goyang-si, Republic of Korea.
| | - Kwang Soon Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Republic of Korea.
| | - Young-Do Nam
- Research Group of Personalized Diet, Korea Food Research Institute, 245 Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun, Jeollabuk-do, Republic of Korea.
| |
Collapse
|
15
|
Nakao R, Yamaguchi T, Shibasaki H, Saeki J, Takahashi A, Tominaga R, Abe K, Akeda Y, Nakagawa-Nakamura T, Nishino T, Ishihara K, Jinno-Oue A, Inoue S. Assessment of periodontitis vaccine using three different bacterial outer membrane vesicles in canine model. mSphere 2025:e0103324. [PMID: 40099899 DOI: 10.1128/msphere.01033-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/05/2025] [Indexed: 03/20/2025] Open
Abstract
Canines frequently develop periodontitis, which is similar and relevant to immunopathology and microbiology of human periodontitis. The aim of this study was to investigate whether bacterial outer membrane vesicle (OMV)-based periodontal vaccines induced humoral immune response in canines from a human vaccine development perspective. Porphyromonas gingivalis (Pg) and Treponema denticola (Td), two major periodontal pathobionts, were chosen as vaccine targets. Intranasal (IN) immunization with Pg OMVs and Td OMVs strongly elicited humoral immune responses against the two respective species in preparative mouse experiments, particularly when adjuvanted with a probiotic Escherichia coli derivative (EcNΔflhD)-derived OMVs. However, in beagles, intranasal immunization with the same Pg/Td/EcNΔflhD OMV vaccine insufficiently elicits humoral immune responses. Nevertheless, the subcutaneous booster with the same OMVs dramatically improved antibody responses in both systemic blood circulation and mucosal sites such as eyes, oral cavity, and upper and lower respiratory tracts. Metagenomic analysis of salivary microbiota revealed that the OMV vaccine might change the microbial composition, while not reducing the number of any periodontal pathobionts at least during the timeframe of the present beagle study. In in vitro Pg growth inhibition assay, serum samples from OMV-immunized beagles significantly inhibited growth of the gingipain-deficient strain but not the gingipain-expressing wild-type strain. Taken together, our data offer the trivalent OMV vaccine strategy by IN-prime/SC-boost regimen, which could elicit robust mucosal immune responses, while suggesting the requirement of revised periodontal vaccine regimen toward achievement of sterilizing immunity in the oral cavity. IMPORTANCE Bacterial outer-membrane vesicles (OMVs) are attractive for use as novel nanoparticle adjuvants, as well as delivery platforms. Periodontal diseases are the most prevalent oral diseases in humans and have serious health and economic burdens, greatly reducing quality of life. The aim of this study is to investigate the humoral immune responses to an OMV-based periodontal disease vaccine in beagles. The vaccine elicited strong mucosal immune responses when administered to beagles by a four-dose heterologous immunization (IN-IN-IN prime and subcutaneous [SC] boost). The OMV vaccine significantly altered the composition of the microbial community in the oral cavity. These findings suggest the utility of the intranasal (IN) prime followed by the SC boost regimen as a rational option to elicit robust humoral immune responses in canines, and most probably in humans as well. We here discuss the outcomes of beagle experiments, the mechanism behind immunological escape of Pg from host immunity, and a rational perspective toward sterilizing immunity in the oral cavity.
Collapse
Affiliation(s)
- Ryoma Nakao
- Department of Bacteriology I, National Institute of Infectious Diseases, Toyama, Tokyo, Japan
| | - Takehiro Yamaguchi
- Department of Bacteriology I, National Institute of Infectious Diseases, Toyama, Tokyo, Japan
| | - Haruka Shibasaki
- Department of Bacteriology I, National Institute of Infectious Diseases, Toyama, Tokyo, Japan
- Department of Animal Sciences, Teikyo University of Science, Adachi, Tokyo, Japan
| | - Jun Saeki
- Department of Animal Sciences, Teikyo University of Science, Adachi, Tokyo, Japan
| | - Aoi Takahashi
- Department of Bacteriology I, National Institute of Infectious Diseases, Toyama, Tokyo, Japan
- School of Bioscience and Biotechnology, Tokyo University of Technology, Hachioji, Tokyo, Japan
| | - Ryunosuke Tominaga
- Department of Bacteriology I, National Institute of Infectious Diseases, Toyama, Tokyo, Japan
- School of Bioscience and Biotechnology, Tokyo University of Technology, Hachioji, Tokyo, Japan
| | - Kimihiro Abe
- Department of Bacteriology I, National Institute of Infectious Diseases, Toyama, Tokyo, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Toyama, Tokyo, Japan
| | - Yukihiro Akeda
- Department of Bacteriology I, National Institute of Infectious Diseases, Toyama, Tokyo, Japan
| | | | - Tomohiko Nishino
- School of Bioscience and Biotechnology, Tokyo University of Technology, Hachioji, Tokyo, Japan
| | - Kazuyuki Ishihara
- Department of Microbiology, Tokyo Dental College, Chiyoda, Tokyo, Japan
| | - Atsushi Jinno-Oue
- Bioresource Center, Gunma University Graduate School of Medicine, Maebashi, Gunma, Japan
| | - Satoshi Inoue
- Department of Veterinary Science, National Institute of Infectious Diseases, Toyama, Tokyo, Japan
| |
Collapse
|
16
|
Jing L, Wang HY, Zhang N, Zhang WJ, Chen Y, Deng DK, Li X, Chen FM, He XT. Critical roles of extracellular vesicles in periodontal disease and regeneration. Stem Cells Transl Med 2025; 14:szae092. [PMID: 39703170 PMCID: PMC11954511 DOI: 10.1093/stcltm/szae092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 10/30/2024] [Indexed: 12/21/2024] Open
Abstract
Extracellular vesicles (EVs) are evolutionarily conserved communication mediators that play key roles in the development of periodontal disease as well as in regeneration processes. This concise review first outlines the pathogenic mechanisms through which EVs derived from bacteria lead to the progression of periodontitis, with a focus on the enrichment of virulence factors, the amplification of immune responses, and the induction of bone destruction as key aspects influenced by bacterial EVs. This review aims to elucidate the positive effects of EVs derived from mesenchymal stem cells (MSC-EVs) on periodontal tissue regeneration. In particular, the anti-inflammatory properties of MSC-EVs and their impact on the intricate interplay between MSCs and various immune cells, including macrophages, dendritic cells, and T cells, are described. Moreover, recent advancements regarding the repair-promoting functions of MSC-EVs are detailed, highlighting the mechanisms underlying their ability to promote osteogenesis, cementogenesis, angiogenesis, and the homing of stem cells, thus contributing significantly to periodontal tissue regeneration. Furthermore, this review provides insights into the therapeutic efficacy of MSC-EVs in treating periodontitis within a clinical context. By summarizing the current knowledge, this review aims to provide a comprehensive understanding of how MSC-EVs can be harnessed for the treatment of periodontal diseases. Finally, a discussion is presented on the challenges that lie ahead and the potential practical implications for translating EV-based therapies into clinical practices for the treatment of periodontitis.
Collapse
Affiliation(s)
- Lin Jing
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, People’s Republic of China
| | - Hong-Yu Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, People’s Republic of China
| | - Ning Zhang
- Cadet Regiment, School of Basic Medical Sciences, Air Force Medical University, Xi’an 710032, People’s Republic of China
| | - Wen-Jie Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, People’s Republic of China
| | - Yuzhe Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, People’s Republic of China
| | - Dao-Kun Deng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, People’s Republic of China
| | - Xuan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, People’s Republic of China
| | - Fa-Ming Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, People’s Republic of China
| | - Xiao-Tao He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, People’s Republic of China
| |
Collapse
|
17
|
Helliwell E, Rauch I, Nice T, Merritt J, Kreth J. Immunostimulatory effects of Streptococcus sanguinis extracellular membrane vesicles protect oral gingival epithelial cells from periodontal pathobiont damage. Infect Immun 2025; 93:e0053524. [PMID: 39969179 PMCID: PMC11895462 DOI: 10.1128/iai.00535-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
The commensal Streptococcus sanguinis is highly prevalent in the oral cavity and characterized for its ability to inhibit growth of oral pathogens. Like many other cell types, streptococci produce extracellular membrane vesicles (EMVs), which contain specific molecular cargo and facilitate interactions with host cells. We previously demonstrated that EMVs from S. sanguinis are internalized by gingival epithelial cells (GECs) without causing cell death. Our aim is to characterize the effects of vesicles on eukaryotic cells. Microscopy studies of gingival epithelial cells inoculated with EMVs from wild-type and specific deletion mutants show differential uptake, with decreased uptake of ΔSSA1099 EMVs and increased uptake of ΔSSA1882 EMVs relative to SK36 EMVs. However, EMVs from wild-type and deletion mutants showed similar patterns in cytokine and chemokine secretion. Transcriptomic analysis of gingival epithelial cells inoculated with SK36 EMVs showed a downregulation of genes implicated in apoptosis as well as interferon signaling, while showing an upregulation of genes involved in cytokine production. Gelatin zymography results show that SK36 EMVs have a contrasting result on production of MMP2/9; MMP2 production is decreased while MMP9 is increased by 48 hours post-inoculation (hpi). Dual-inoculation studies demonstrate that prior internalization of S. sanguinis EMVs protects gingival epithelial cells from exposure to pathobiont Porphyromonas gingivalis outer membrane vesicles (OMVs), preventing dissociation and cell death. Our overall findings suggest that S. sanguinis EMVs trigger an immune response on gingival epithelial cells; however, this response suggests inhibition of some immune signaling pathways. Our results highlight an important role in commensalism, in which a microbe induces an immune response but avoids damage to host cells, thus discouraging infection by pathobionts.
Collapse
Affiliation(s)
- Emily Helliwell
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University, Portland, Oregon, USA
| | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Tim Nice
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Justin Merritt
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University, Portland, Oregon, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| | - Jens Kreth
- Division of Biomaterial and Biomedical Sciences, School of Dentistry, Oregon Health & Science University, Portland, Oregon, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
18
|
Wang Y, Li T, Dong Z, Zhang Q, Mi J, Wang Q, Lin G, Ma Q, Jia R, Huang S. Extracellular Vesicles From Lactobacillus fermentum Enhance Intestinal Barrier Integrity and Restore Gut Microbial Homeostasis in Experimental Murine Colitis. J Nutr 2025:S0022-3166(25)00151-8. [PMID: 40058701 DOI: 10.1016/j.tjnut.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/19/2025] [Accepted: 03/02/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Lactobacillus fermentum has been shown to improve intestinal health and treat colitis; however, its precise efficacy and mechanisms in inflammatory bowel disease remain unclear. OBJECTIVES This study aimed to evaluate whether L fermentum and its metabolites, extracellular vesicles, and other components could modulate intestinal barrier function and gut microbiota to alleviate dextran sulfate sodium (DSS)-induced colitis in mice. METHODS Forty-eight mice were randomly assigned to 6 groups: control, DSS, L fermentum+DSS group (LF+DSS), heat-inactivated L fermentum+DSS group (LHF+DSS), L fermentum supernatant solution+DSS group (LSF+DSS), and L fermentum extracellular vesicles+DSS group (LEV+DSS). After a 1-wk acclimation, mice were gavaged daily for 3 wk. Fresh cultures, including live (LF+DSS), heat-inactivated (LHF+DSS), supernatant (LSF+DSS), and extracellular vesicles (LEV+DSS), were prepared daily. During the final 7 d, the control group received normal water, and the other groups received 3% DSS. Data were collected daily, followed by sample collection from the mice. RESULTS In this study, significant reductions (P < 0.05) in body weight changes, disease activity index, intestinal damage, and histology scores were observed in the treatment groups, especially LEV+DSS and LF+DSS. Additionally, compared with the DSS group, colonic mucus secretion, as well as claudin-1 and occludin expression, increased significantly (P < 0.05) in the LEV+DSS and LF+DSS groups, whereas proinflammatory cytokines IL-1β and TNF-α decreased (P < 0.05) and IL-10 increased (P < 0.05) in the LEV+DSS group. L fermentum and its components significantly regulated gut microbiota α-diversity and β-diversity, affecting overall composition. Linear discriminant analysis effect size analysis revealed an enrichment of beneficial bacteria including Prevotellaceae_UCG-001, Romboutsia, and Ruminococcus species in the LF+DSS group and Akkermansia, Odoribacter, and Marvinbryantia species in the LEV+DSS group. Both L fermentum and its extracellular vesicles significantly downregulated the gene expression of TNF-α and IL-1β, whereas the expression of IL-10 was upregulated, thereby contributing to the alleviation of colitis symptoms. CONCLUSIONS This study reveals that L fermentum alleviates colitis through modulation of the gut microbiota and reinforcement of the intestinal mucosal barrier, with its extracellular vesicles potentially playing a key role in this regulatory process.
Collapse
Affiliation(s)
- Yanwei Wang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; School of Life Science, Shanxi University, Taiyuan, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Tiantian Li
- Academy of National Food and Strategic Reserves Administration, Beijing, China
| | - Zhuo Dong
- Hubei International Travel Healthcare Center, Hubei, China
| | - Qiyue Zhang
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jingqiu Mi
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Qingfeng Wang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Gang Lin
- Institute of Quality Standards and Testing Technology for Agricultural Products, Chinese Academy of Agricultural Science, Beijing, China
| | - Qiugang Ma
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China
| | - Ru Jia
- School of Life Science, Shanxi University, Taiyuan, China.
| | - Shimeng Huang
- National Key Laboratory of Livestock and Poultry Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, China; Laboratory of Feed grain Safety and Healthy Poultry Farming, Beijing Jingwa Agricultural Science and Technology Innovation Center, Beijing, China.
| |
Collapse
|
19
|
Puagsopa J, Tongviseskul N, Jaroentomeechai T, Meksiriporn B. Recent Progress in Developing Extracellular Vesicles as Nanovehicles to Deliver Carbohydrate-Based Therapeutics and Vaccines. Vaccines (Basel) 2025; 13:285. [PMID: 40266147 DOI: 10.3390/vaccines13030285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 04/24/2025] Open
Abstract
Cell-derived, nanoscale extracellular vesicles (EVs) have emerged as promising tools in diagnostic, therapeutic, and vaccine applications. Their unique properties including the capability to encapsulate diverse molecular cargo as well as the versatility in surface functionalization make them ideal candidates for safe and effective vehicles to deliver a range of biomolecules including gene editing cassettes, therapeutic proteins, glycans, and glycoconjugate vaccines. In this review, we discuss recent advances in the development of EVs derived from mammalian and bacterial cells for use in a delivery of carbohydrate-based protein therapeutics and vaccines. We highlight key innovations in EVs' molecular design, characterization, and deployment for treating diseases including Alzheimer's disease, infectious diseases, and cancers. We discuss challenges for their clinical translation and provide perspectives for future development of EVs within biopharmaceutical research and the clinical translation landscape.
Collapse
Affiliation(s)
- Japigorn Puagsopa
- Department of Physiology and Aging, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Niksa Tongviseskul
- Department of Biology, School of Science, King Mongkut's Institute of Technology Ladkrabang, Bangkok 10520, Thailand
| | - Thapakorn Jaroentomeechai
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen, Denmark
| | - Bunyarit Meksiriporn
- Department of Biology, School of Science, King Mongkut's Institute of Technology Ladkrabang, Bangkok 10520, Thailand
| |
Collapse
|
20
|
Zavan L, Hor L, Johnston EL, Paxman J, Heras B, Kaparakis-Liaskos M. Antigen 43 associated with Escherichia coli membrane vesicles contributes to bacterial cell association and biofilm formation. Microbiol Spectr 2025; 13:e0189024. [PMID: 39840972 DOI: 10.1128/spectrum.01890-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/23/2024] [Indexed: 01/23/2025] Open
Abstract
Bacterial membrane vesicles (MVs) are produced by all bacteria and contribute to numerous bacterial functions due to their ability to package and transfer bacterial cargo. In doing so, MVs have been shown to facilitate horizontal gene transfer, mediate antimicrobial activity, and promote biofilm formation. Uropathogenic Escherichia coli is a pathogenic Gram-negative organism that persists in the urinary tract of its host due to its ability to form persistent, antibiotic-resistant biofilms. The formation of these biofilms is dependent upon proteins such as Antigen 43 (Ag43), which belongs to the widespread Autotransporter group of bacterial surface proteins. In E. coli, the autotransporter Ag43 has been shown to contribute to bacterial cell aggregation and biofilm formation via self-association of Ag43 between neighboring Ag43-expressing bacteria. As MVs package bacterial proteins, we investigated whether MVs produced by E. coli contained Ag43, and the ability of Ag43-expressing MVs to facilitate cell aggregation and biofilm formation. We showed that Ag43 expressing E. coli produced MVs that contained Ag43 on their surface and had an enhanced ability to bind to E. coli bacteria. Furthermore, we demonstrated that the addition of Ag43-containing MVs to Ag43-expressing E. coli significantly enhanced biofilm formation. These findings reveal the contribution of MVs harboring autotransporters in promoting bacterial aggregation and enhancing biofilm formation, highlighting the impact of MVs and their specific composition to bacterial adaptation and pathogenesis.IMPORTANCEAutotransporter proteins are the largest family of outer membrane and secreted proteins in Gram-negative bacteria which contribute to pathogenesis by promoting aggregation, biofilm formation, persistence, and cytotoxicity. Although the roles of bacterial autotransporters are well known, the ability of bacterial membrane vesicles (MVs) naturally released from the surface of bacteria to contain autotransporters and their role in promoting virulence remains less investigated. Our findings reveal that MVs produced by E. coli contain the autotransporter protein Ag43. Furthermore, we show that Ag43-containing MVs function to enhance bacterial cell interactions and biofilm formation. By demonstrating the ability of MVs to carry functional autotransporter adhesins, this work highlights the importance of MVs in disseminating autotransporters beyond the bacterial cell membrane to ultimately promote cellular interactions and enhance biofilm development. Overall, these findings have significant implications in furthering our understanding of the numerous ways in which MVs can facilitate bacterial persistence and pathogenesis.
Collapse
Affiliation(s)
- Lauren Zavan
- Department of Microbiology, Anatomy, Physiology, and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Lilian Hor
- Department of Microbiology, Anatomy, Physiology, and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
| | - Ella L Johnston
- Department of Microbiology, Anatomy, Physiology, and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
| | - Jason Paxman
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Begoña Heras
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Maria Kaparakis-Liaskos
- Department of Microbiology, Anatomy, Physiology, and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Yadav N, Misra SK. Nitroaromatic Compounds Dictate Electrochemical Properties of Escherichia coli by Manipulating the Cellular Membrane. Mol Pharm 2025; 22:1707-1724. [PMID: 39932503 DOI: 10.1021/acs.molpharmaceut.4c01537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Nitroaromatic compounds (NACs) are generally used as starting materials and/or generated as byproducts during the manufacturing of dyes, fertilizers, and therapeutic agents. Though NACs are beneficial when used appropriately, inadequate management, disposal, and application methods have led to their introduction to bacterial ecosystems where NACs act as mutagenic agents and may even contribute to antimicrobial resistance. Many of these bacterial systems are known to have different pathways to adapt to the presence of NACs such as altering the lipid composition of cellular membranes and intracellular degradation of NACs. In general, these processes require sophisticated techniques and skilled human resources to detect the changes by conventional characterization techniques. Hence, alternative methods are needed to investigate the short-term effects of NACs on bacterial cells with better precision. Herein, we report that bacterial cells adapt to the presence of NACs initially by incorporation in the cellular membrane, which can be predicted by further altered electrical and electrochemical properties of the cells. It was observed that the whole cell bacteria were negatively charged entities that could generate varying levels of surface charges on being incubated with model NACs of biomedical importance viz. niclosamide and p-nitrophenol. Such variations were also reflected in dye entrapment assays performed by using lipidic membranes collected from NAC-treated bacterial cells after the cells. Further studies with gel electrophoresis and differential pulse voltammetry revealed the significant alterations in electrochemical properties of NAC-incubated bacterial cells. Overall, results indicate that bacterial adaptation to NACs was found to be closely linked to variations in the electrochemical properties of the bacterial cells. These outcomes advance our understanding of influences imparted by NACs during bacterial infections and might facilitate the way for developing therapies to combat antibacterial resistance in the near future.
Collapse
Affiliation(s)
- Neha Yadav
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, U.P. 208016, India
| | - Santosh K Misra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, U.P. 208016, India
- The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, U.P. 208016, India
- Centre of Excellence in Point-of-Care Diagnosis, Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Kalyanpur, Kanpur, U.P. 208016, India
| |
Collapse
|
22
|
Biller SJ, Ryan MG, Li J, Burger A, Eppley JM, Hackl T, DeLong EF. Distinct horizontal gene transfer potential of extracellular vesicles versus viral-like particles in marine habitats. Nat Commun 2025; 16:2126. [PMID: 40032822 DOI: 10.1038/s41467-025-57276-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 02/13/2025] [Indexed: 03/05/2025] Open
Abstract
Horizontal gene transfer (HGT) is enabled in part through the movement of DNA within two broad groups of small (<0.2 µm), diffusible nanoparticles: extracellular vesicles (EVs) and virus-like particles (VLPs; including viruses, gene transfer agents, and phage satellites). The information enclosed within these structures represents a substantial portion of the HGT potential available in planktonic ecosystems, but whether some genes might be preferentially transported through one type of nanoparticle versus another is unknown. Here we use long-read sequencing to compare the genetic content of EVs and VLPs from the oligotrophic North Pacific. Fractionated EV-enriched and VLP-enriched subpopulations contain diverse DNA from the surrounding microbial community, but differ in their capacity and encoded functions. The sequences carried by both particle types are enriched in mobile genetic elements (MGEs) as compared with other cellular chromosomal regions, and we highlight how this property enables novel MGE discovery. Examining the Pelagibacter mobilome reveals >7200 distinct chromosomal fragments and MGEs, many differentially partitioned between EVs and VLPs. Together these results suggest that distinctions in nanoparticle contents contribute to the mode and trajectory of microbial HGT networks and evolutionary dynamics in natural habitats.
Collapse
Affiliation(s)
- Steven J Biller
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA.
| | - M Gray Ryan
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Jasmine Li
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Andrew Burger
- Department of Oceanography, Daniel K. Inouye Center for Microbial Oceanography: Research and Education (C-MORE), University of Hawai'i at Manoa, Honolulu, HI, USA
| | - John M Eppley
- Department of Oceanography, Daniel K. Inouye Center for Microbial Oceanography: Research and Education (C-MORE), University of Hawai'i at Manoa, Honolulu, HI, USA
| | - Thomas Hackl
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Edward F DeLong
- Department of Oceanography, Daniel K. Inouye Center for Microbial Oceanography: Research and Education (C-MORE), University of Hawai'i at Manoa, Honolulu, HI, USA
| |
Collapse
|
23
|
Liu C, Udawatte NS, Liaw A, Staples R, Salomon C, Seneviratne CJ, Ivanovski S, Han P. Microbial DNA Profiles of Bacterial Extracellular Vesicles from 3D Salivary Polymicrobial Biofilms - A Pilot Study. Adv Healthc Mater 2025; 14:e2403300. [PMID: 39748613 DOI: 10.1002/adhm.202403300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/19/2024] [Indexed: 01/04/2025]
Abstract
With the advent of multi-layered and 3D scaffolds, the understanding of microbiome composition and pathogenic mechanisms within polymicrobial biofilms is continuously evolving. A fundamental component in mediating the microenvironment and bacterial-host communication within the biofilm are bilayered nanoparticles secreted by bacteria, known as bacterial extracellular vesicles (BEVs), which transport key biomolecules including proteins, nucleic acids, and metabolites. Their characteristics and microbiome profiles are yet to be explored in the context of in vitro salivary polymicrobial biofilm. This pilot study aimed to compare the profiles of BEVs from salivary biofilm cultured on a 2D tissue culture plate and 3D melt electrowritten medical-grade polycaprolactone (MEW mPCL) scaffold. BEVs derived from MEW mPCL biofilm exhibited enhanced purity and yield without altered EV morphology and lipopolysaccharide (LPS) content, with enriched BEVs-associated DNA from Capnocytophaga, porphyromonas, and veillonella genus. Moreover, compared to saliva controls, MEW mPCL BEVs showed comparable DNA expression of Tannerella forsythia, and Treponema denticola and significantly higher expression in Porphyromonas gingivalis, Eikenella corrodens and Lactobacillus acidophilus. Together, these findings highlight a more detailed microbial profile with BEVs derived from salivary biofilms cultured on 3D MEW PCL scaffolds, which facilitates an effective in vitro model with a greater resemblance to naturally occurring biofilms.
Collapse
Affiliation(s)
- Chun Liu
- School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, The University of Queensland, Brisbane, QLD, 4006, Australia
| | - Nadeeka S Udawatte
- School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, The University of Queensland, Brisbane, QLD, 4006, Australia
| | - Andrew Liaw
- School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, The University of Queensland, Brisbane, QLD, 4006, Australia
| | - Reuben Staples
- School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, The University of Queensland, Brisbane, QLD, 4006, Australia
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, The University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia
- UQ Centre for Extracellular Vesicle Nanomedicine, The University of Queensland, Brisbane, 4029, Australia
| | - Chaminda Jayampath Seneviratne
- School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, The University of Queensland, Brisbane, QLD, 4006, Australia
| | - Sašo Ivanovski
- School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, The University of Queensland, Brisbane, QLD, 4006, Australia
- UQ Centre for Extracellular Vesicle Nanomedicine, The University of Queensland, Brisbane, 4029, Australia
| | - Pingping Han
- School of Dentistry, Center for Oral-facial Regeneration, Rehabilitation and Reconstruction (COR3), Epigenetics nanodiagnostic and therapeutic group, The University of Queensland, Brisbane, QLD, 4006, Australia
- UQ Centre for Extracellular Vesicle Nanomedicine, The University of Queensland, Brisbane, 4029, Australia
| |
Collapse
|
24
|
Liu J, Chen S, Zhao J. The role and mechanisms of Helicobacter pylori outer membrane vesicles in the pathogenesis of extra-gastrointestinal diseases. Microb Pathog 2025; 200:107312. [PMID: 39855489 DOI: 10.1016/j.micpath.2025.107312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 12/20/2024] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
Helicobacter pylori (H. pylori) infection have been closely associated with several extra-gastrointestinal disorders. Outer membrane vesicles (OMVs), as lipid-membrane-bounded nanoparticles, are usually shed from Gram-negative both in vitro and in vivo. H. pylori is also capable of producing OMVs, which can enter the systemic circulation and be delivered to various cells, tissues or organs, eliciting a range of inflammatory and immune modulation responses. In this current review, we summarize the biogenesis and functions of H. pylori OMVs, describe the contribution of H. pylori OMVs to the generation and progression of extra-gastrointestinal diseases, such as neuronal damage, Alzheimer disease, hepatic fibrosis and atherosclerosis. We also explored the effect of H. pylori OMVs in inflammatory and immune modulation of diverse immune cells, including macrophages, mononuclear cells and dendritic cells. By elucidating the molecular mechanism of H. pylori OMVs-mediated extra-gastrointestinal diseases and immunomodulatory effect, it may promote the development of efficient treatments and vaccinations against H. pylori.
Collapse
Affiliation(s)
- Jin Liu
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, People's Republic of China
| | - Sheqing Chen
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, People's Republic of China
| | - Jingjing Zhao
- Department of Pharmaceutical Engineering, School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
25
|
Zhou H, Huang W, Li J, Chen P, Shen L, Huang W, Mai K, Zou H, Shi X, Weng Y, Liu Y, Yang Z, Ou C. Oral probiotic extracellular vesicle therapy mitigates Influenza A Virus infection via blunting IL-17 signaling. Bioact Mater 2025; 45:401-416. [PMID: 39697241 PMCID: PMC11652895 DOI: 10.1016/j.bioactmat.2024.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/12/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
The influenza A virus (IAV) damages intestinal mucosal tissues beyond the respiratory tract. Probiotics play a crucial role in maintaining the balance and stability of the intestinal microecosystem. Extracellular vesicles (EVs) derived from probiotics have emerged as potential mediators of host immune response and anti-inflammatory effect. However, the specific anti-inflammatory effects and underlying mechanisms of probiotics-derived EVs on IAV remain unclear. In the present study, we investigated the therapeutic efficacy of Lactobacillus reuteri EHA2-derived EVs (LrEVs) in a mouse model of IAV infection. Oral LrEVs were distributed in the liver, lungs, and gastrointestinal tract. In mice infected with IAV, oral LrEVs administration alleviated IAV-induced damages in the lungs and intestines, modified the microbiota compositions, and increased the levels of short-chain fatty acids in those organs. Mechanistically, LrEVs exerted their protective effects against IAV infection by blunting the pro-inflammatory IL-17 signaling. Furthermore, FISH analysis detected miR-4239, one of the most abundant miRNAs in LrEVs, in both lung and intestinal tissues. We confirmed that miR-4239 directly targets IL-17a. Our findings paved the ground for future application of LrEVs in influenza treatment and offered new mechanistic insights regarding the anti-inflammatory role of miR-4239.
Collapse
Affiliation(s)
- Hongxia Zhou
- Dongguan Institute of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Wenbo Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Jieting Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Peier Chen
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Lihan Shen
- Dongguan Institute of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Wenjing Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Kailin Mai
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Heyan Zou
- Dongguan Institute of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Xueqin Shi
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Yunceng Weng
- Becton Dickinson Medical Devices (Shanghai) Co., Ltd., Guangzhou, 510180, China
| | - Yuhua Liu
- Department of General Practice, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Zifeng Yang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
- Guangzhou National Laboratory, Guangzhou, 510000, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau SAR, 519020, China
| | - Caiwen Ou
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| |
Collapse
|
26
|
Tao S, Fan J, Li J, Wu Z, Yao Y, Wang Z, Wu Y, Liu X, Xiao Y, Wei H. Extracellular vesicles derived from Lactobacillus johnsonii promote gut barrier homeostasis by enhancing M2 macrophage polarization. J Adv Res 2025; 69:545-563. [PMID: 38508446 PMCID: PMC11954842 DOI: 10.1016/j.jare.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/19/2024] [Accepted: 03/16/2024] [Indexed: 03/22/2024] Open
Abstract
INTRODUCTION Diarrheic disease is a common intestinal health problem worldwide, causing great suffering to humans and animals. Precise manipulation strategies based on probiotics to combat diarrheic diseases have not been fully developed. OBJECTIVES The aim of this study was to investigate the molecular mechanisms by which probiotics manipulate macrophage against diarrheic disease. METHODS Metagenome reveals gut microbiome profiles of healthy and diarrheic piglets. Fecal microbial transplantation (FMT) was employed to explore the causal relationship between gut microbes and diarrhea. The protective role of probiotics and their derived extracellular vesicles (EVs) was investigated in ETEC K88-infected mice. Macrophage depletion was performed to assess the role of macrophages in EVs against diarrhea. Execution of in vitro cell co-culture and transcriptome analyses elucidated the molecular mechanisms by which EVs modulate the macrophage and intestinal epithelial barrier. RESULTS Escherichia coli was enriched in weaned diarrheic piglets, while Lactobacillus johnsonii (L. john) showed a negative correlation with Escherichia coli. The transmission of diarrheic illness symptoms was achieved by transferring fecal microbiota, but not metabolites, from diarrheic pigs to germ-free (GF) mice. L. john's intervention prevented the transmission of disease phenotypes from diarrheic piglets to GF mice. L. john also reduces the gut inflammation induced by ETEC K88. The EVs secreted by L. john demonstrated enhanced efficacy in mitigating the adverse impacts induced by ETEC K88 through the modulation of macrophage phenotype. In vitro experiments have revealed that EVs activate M2 macrophages in a manner that shuts down ERK, thereby inhibiting NLRP3 activation in intestinal epithelial cells. CONCLUSION Our results reveal that intestinal microbiota drives the onset of diarrheic disease and that probiotic-derived EVs ameliorate diarrheic disease symptoms by modulating macrophage phenotypes. These findings can enhance the advancement of innovative therapeutic approaches for diarrheic conditions based on probiotic-derived EVs.
Collapse
Affiliation(s)
- Shiyu Tao
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jinping Fan
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jingjing Li
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhifeng Wu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Yong Yao
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhenyu Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Yujun Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing 100193, China
| | - Xiangdong Liu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China.
| | - Hong Wei
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
27
|
Ou Q, Lu L, Zhai L, Sang S, Guan Y, Xiong Y, Liu C, Wang H, Hu Q, Wang Y. Exploration of a GMMA-Based Bivalent Vaccine Against Klebsiella pneumoniae. Vaccines (Basel) 2025; 13:226. [PMID: 40266066 DOI: 10.3390/vaccines13030226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND An emerging trend of mutual convergence between drug-resistant and highly virulent strains of K. pneumoniae has been identified, highlighting the urgent need for the development of novel vaccines. METHODS To delete the target genes and eliminate the plasmids carrying antibiotic resistance genes, CRISPR-Cas9 technology was employed to perform genome editing on a clinically isolated O2 serotype of K. pneumoniae. Subsequently, this strain was utilized as a host to express genes associated with the synthesis of O1 serotype LPSs to construct the recombinant strain capable of simultaneously expressing LPSs of both O1 and O2 serotypes. This recombinant strain was then used as the production strain for the preparation of vaccines based on GMMAs (Generalized Modules for Membrane Antigens), and its biological characteristics were characterized. Finally, the safety and immunogenicity of the vaccine were evaluated using mice as the model animals. RESULT a GMMA vaccine characterized by a high yield and low toxicity was gained. Importantly, the lipopolysaccharides (LPSs) of both O1 and O2 serotypes of K. pneumoniae were successfully expressed on the surface of the outer membrane vesicles. Following immunization with the GMMA vaccine, mice were capable of producing antibodies against the GMMA and demonstrated resistance to the invasion of both serotypes of clinically isolated K. pneumoniae. CONCLUSIONS The GMMA vaccine showed significant promise as a bivalent vaccine against K. pneumoniae.
Collapse
Affiliation(s)
- Qikun Ou
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, China
| | - Lu Lu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongda Street, Beijing 100071, China
| | - Lina Zhai
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongda Street, Beijing 100071, China
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Shuli Sang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongda Street, Beijing 100071, China
| | - Yiyan Guan
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongda Street, Beijing 100071, China
| | - Yuling Xiong
- Department of Clinical Laboratory, The Fourth Medical Centre, Chinese PLA General Hospital, No. 51 Fucheng Road, Beijing 100037, China
| | - Chunjie Liu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongda Street, Beijing 100071, China
| | - Haibin Wang
- Department of Clinical Laboratory, The Fourth Medical Centre, Chinese PLA General Hospital, No. 51 Fucheng Road, Beijing 100037, China
| | - Qiping Hu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, China
| | - Yanchun Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, 20 Dongda Street, Beijing 100071, China
| |
Collapse
|
28
|
Cuinat C, Pan J, Comelli EM. Host-dependent alteration of the gut microbiota: the role of luminal microRNAs. MICROBIOME RESEARCH REPORTS 2025; 4:15. [PMID: 40207285 PMCID: PMC11977366 DOI: 10.20517/mrr.2024.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 01/22/2025] [Accepted: 02/10/2025] [Indexed: 04/11/2025]
Abstract
MicroRNAs (miRNAs) are short, non-coding RNAs that play gene expression regulatory roles in eukaryotes. MiRNAs are also released in body fluids, and in the intestine, they are found in the lumen and feces. Here, together with exogenous dietary-derived miRNAs, they constitute the fecal miRNome. Several miRNAs were identified in the feces of healthy adults, including, as shown here, core miRNAs hsa-miR-21-5p and hsa-miR-1246. These miRNAs are important for intestinal homeostasis. Recent evidence suggests that miRNAs may interact with gut bacteria. This represents a new avenue to understand host-bacteria crosstalk in the gut and its role in health and disease. This review provides a comprehensive overview of current knowledge on fecal miRNAs, their representation across individuals, and their effects on the gut microbiota. It also discusses existing evidence on potential mechanisms of uptake and interaction with bacterial genomes, drawing from knowledge of prokaryotic small RNAs (sRNAs) regulation of gene expression. Finally, we review in silico and experimental approaches for profiling miRNA-mRNA interactions in bacterial species, highlighting challenges in target validation. This work emphasizes the need for further research into host miRNA-bacterial interactions to better understand their regulatory roles in the gut ecosystem and support their exploitation for disease prevention and treatment.
Collapse
Affiliation(s)
- Céline Cuinat
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto M5S 1A8, Canada
- Authors contributed equally
| | - Jiali Pan
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto M5S 1A8, Canada
- Authors contributed equally
| | - Elena M. Comelli
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto M5S 1A8, Canada
- Joannah and Brian Lawson Centre for Child Nutrition, Faculty of Medicine, University of Toronto, Toronto M5S 1A8, Canada
| |
Collapse
|
29
|
Dai F, Liang W, Liu J, Guo M, Li C. Eeukaryotic-like Sppsk1 from Vibrio splendidus AJ01 mediates phagosome escape via inhibiting phagosome acidification and maturation. Cell Mol Life Sci 2025; 82:88. [PMID: 39985586 PMCID: PMC11846826 DOI: 10.1007/s00018-025-05610-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/16/2024] [Accepted: 01/30/2025] [Indexed: 02/24/2025]
Abstract
The intracellular pathogen has evolved sophisticated mechanisms to evade host immune defenses by secreting different virulence factors. In our previous study, the eukaryotic factor STPKLRR was identified from the intracellular pathogen Vibrio splendidus AJ01 and shown to facilitate promote AJ01 internalization by mediating actin-dependent coelomocytes phagocytosis. However, the molecular mechanisms underlying AJ01'escaped from the phagosome remained largely unclear. In this study, a novel eukaryotic-like factor was identified, containing both the Serine/Threonine/Tyrosine (STYKc) domain and protein phosphatase 2 C (PP2C) domain (denoted as Sppsk1), which was essential for AJ01 phagosome escape. Deletion of Sppsk1 significantly increased phagolysosome maturation and reduced the intracellular AJ01 levels compared to the wild AJ01. Mechanistic analysis showed that the STYKc domain of Sppsk1 directly phosphorylated phagosome H+ transport complex subunit ATP6V1C at Serine-356, resulting in the inhibition of phagosome acidification in coelomocytes and promoting AJ01 phagosome survival. Moreover, the PP2C domain of Sppsk1 dephosphorylated phosphatidylinositol-3-bisphosphate [PtdIns(3)P], converting it to PtdIns(3)P to phosphatidylinositol (PtdIns). Reduction of PtdIns(3)P on phagosomes hindered early endosome antigen 1 (EEA1) recruitment, thereby inhibiting phagosome maturation. These findings demonstrated that Sppsk1 in AJ01 could achieve phagosome escape by two strategies including inhibiting host coelomocytes' phagosome acidification and maturation, which advanced our knowledge of the general biology of pathogen-host interactions.
Collapse
Affiliation(s)
- Fa Dai
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Weikang Liang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Jiqing Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China
| | - Ming Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China.
| | - Chenghua Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, PR China.
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266071, PR China.
| |
Collapse
|
30
|
Gu W, Huang Z, Fan Y, Li T, Yu X, Chen Z, Hu Y, Li A, Zhang F, Fu Y. Peripheral blood microbiome signature and Mycobacterium tuberculosis-derived rsRNA as diagnostic biomarkers for tuberculosis in human. J Transl Med 2025; 23:204. [PMID: 39972378 PMCID: PMC11837313 DOI: 10.1186/s12967-025-06190-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/29/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Tuberculosis (TB) is a major global health issue. Early diagnosis of TB is still a challenge. Studies are seeking non-sputum biomarker-based TB test. Emerging evidence indicates potential significance of blood microbiome signatures for diseases. However, blood microbiome RNA profiles are unknown in TB. We aimed to characterize the blood microbiome of TB patients and identify Mycobacterium tuberculosis (Mtb) genome-derived small RNA molecules to serve as diagnostic biomarkers for TB. METHODS RNA sequencing data of the blood from TB patients and healthy controls were retrieved from the NCBI-SRA database for analyzing the blood microbiome and identifying rRNA-derived small RNA (rsRNA) of Mtb. Small RNA-seq was performed on plasma exosomes from TB patients and healthy controls. The levels of the candidate Mtb rsRNAs were determined by real-time quantitative reverse transcription PCR (RT-qPCR) on plasma from a separate cohort of 73 TB patients and 62 healthy controls. RESULTS The blood microbiome of TB patients consisted of RNA signals from bacteria, fungi, archaea, and viruses, with bacteria accounting for more than 97% of the total. Reduced blood microbial diversity and abundance of 6 Mycobacterium-associated bacterial genera, including Mycobacterium, Priestia, Nocardioides, Agrobacterium, Bradyrhizobium, and Escherichia, were significantly altered in the blood of TB patients. A diagnostic model for TB based on the 6 genera achieved an area under the curve (AUC) of 0.8945. rsRNAs mapped to the Mtb genome were identified from blood and plasma exosomes of TB patients. RT-qPCR results showed that 2 Mtb-derived rsRNAs, 16 S-L1 and 16 S-L2, could be used as diagnostic biomarkers to differentiate TB patients from healthy controls, with a high co-diagnostic efficacy (AUC = 0.7197). CONCLUSIONS A panel of blood microbiome signatures and Mtb-derived rsRNAs can serve as blood biomarkers for TB diagnosis.
Collapse
Affiliation(s)
- Wei Gu
- Department of Microbiology, School of Basic Medical Sciences, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Zhigang Huang
- Department of Microbiology, School of Basic Medical Sciences, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Yunfan Fan
- Department of Microbiology, School of Basic Medical Sciences, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
- Department of Clinical Laboratory, Chongqing Public Health Medical Center, Chongqing, China
| | - Ting Li
- Department of Microbiology, School of Basic Medical Sciences, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
- Department of Clinical Laboratory, Chongqing Public Health Medical Center, Chongqing, China
| | - Xinyuan Yu
- Department of Microbiology, School of Basic Medical Sciences, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Zhiyuan Chen
- Department of Microbiology, School of Basic Medical Sciences, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Yan Hu
- Department of Microbiology, School of Basic Medical Sciences, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
| | - Aimei Li
- Department of Microbiology, School of Basic Medical Sciences, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China
| | - Fengmin Zhang
- Department of Microbiology, School of Basic Medical Sciences, WU Lien-Teh Institute, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China
| | - Yingmei Fu
- Department of Microbiology, School of Basic Medical Sciences, WU Lien-Teh Institute, Harbin Medical University, Harbin, China.
- Heilongjiang Provincial Key Laboratory of Infection and Immunity, Harbin Medical University, Harbin, China.
| |
Collapse
|
31
|
Long L, Xu XL, Duan YF, Long L, Chen JY, Yin YH, Zhu YG, Huang Q. Extracellular Vesicles Are Prevalent and Effective Carriers of Environmental Allergens in Indoor Dust. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:1969-1983. [PMID: 39711517 PMCID: PMC11800389 DOI: 10.1021/acs.est.4c10056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
The global incidence of allergic diseases is rising and poses a substantial threat to human health. Allergenic proteins released by various allergenic species play a critical role in the pathogenesis of allergic diseases and have been widely detected in the environmental matrix. However, the release, presence and interaction of environmental allergens with human body remain to be elucidated. In this study, we reported the widespread of allergen-harboring extracellular vesicles (EVs) in indoor dust from 75 households across five provinces in China. Particle size and abundance of EVs were correlated with specific environmental factors. EVs showed long persistence and high resistance to environmental stress. Metagenomics and metaproteomics data revealed that most indoor allergenic species released allergens within the EVs into dust. A higher abundance of allergenic species and their derived EVs was observed in urban areas compared to rural areas. ELISA confirmed the allergenic activity of the EV-associated allergens. Allergens are common components and even markers of EVs, as evidenced by the data compilation of various allergenic species. The proportion of EV-associated allergens varied across species. EVs facilitated allergen entry into epithelial cells. Intranasally administered EVs can be rapidly transported to the lungs and gastrointestinal tract. EV-associated allergens exhibited higher allergenicity compared with non-EV allergens. Our findings elucidate a vesicle pathway through which environmental allergens are released, persist, and trigger allergic responses within EVs.
Collapse
Affiliation(s)
- Lu Long
- Xiamen
Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment
and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue-Li Xu
- Xiamen
Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment
and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
- College
of Resources and Environment, University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi-Fang Duan
- Xiamen
Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment
and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Li Long
- Health
Management Center, The First Affiliated
Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Jing-Yu Chen
- Xiamen
Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment
and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Yu-Han Yin
- Xiamen
Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment
and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Yong-Guan Zhu
- Xiamen
Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment
and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| | - Qiansheng Huang
- Xiamen
Key Laboratory of Indoor Air and Health, Key Lab of Urban Environment
and Health, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen 361021, China
| |
Collapse
|
32
|
Gao S, Li X, Han B. Bacterial and bacterial derivatives-based drug delivery systems: a novel approach for treating central nervous system disorders. Expert Opin Drug Deliv 2025; 22:163-180. [PMID: 39688950 DOI: 10.1080/17425247.2024.2444364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/19/2024]
Abstract
INTRODUCTION Bacteria and their derivatives show great potential as drug delivery systems due to their unique chemotaxis, biocompatibility, and targeting abilities. In CNS disease treatment, bacterial carriers can cross the blood-brain barrier (BBB) and deliver drugs precisely, overcoming limitations of traditional methods. Advances in genetic engineering, synthetic biology, and nanotechnology have transformed these systems into multifunctional platforms for personalized CNS treatment. AREAS COVERED This review examines the latest research on bacterial carriers for treating ischemic brain injury, neurodegenerative diseases, and gliomas. Bacteria efficiently cross the blood-brain barrier via active targeting, endocytosis, paracellular transport, and the nose-to-brain route for precise drug delivery. Various bacterial drug delivery systems, such as OMVs and bacterial ghosts, are explored for their design and application. Databases were searched in Google Scholar for the period up to December 2024. EXPERT OPINION Future developments in bacterial drug delivery will rely on AI-driven design and high-throughput engineering, enhancing treatment precision. Personalized medicine will further optimize bacterial carriers for individual patients, but challenges such as biosafety, immune rejection, and scalability must be addressed. As multimodal diagnostic and therapeutic strategies advance, bacterial carriers are expected to play a central role in CNS disease treatment, offering novel precision medicine solutions.
Collapse
Affiliation(s)
- Shizhu Gao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| | - Xin Li
- Orthopedic Medical Center, 2nd hospital of Jilin University, Changchun, PR China
| | - Bing Han
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, Changchun, PR China
| |
Collapse
|
33
|
Li H, Chen X, Rao S, Zhou M, Lu J, Liang D, Zhu B, Meng L, Lin J, Ding X, Zhang Q, Hu D. Recent development of micro-nano carriers for oral antineoplastic drug delivery. Mater Today Bio 2025; 30:101445. [PMID: 39866789 PMCID: PMC11762190 DOI: 10.1016/j.mtbio.2025.101445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 12/17/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Chemotherapy is widely recognized as a highly efficacious modality for cancer treatment, involving the administration of chemotherapeutic agents to target and eradicate tumor cells. Currently, oral administration stands as the prevailing and widely utilized method of delivering chemotherapy drugs. However, the majority of anti-tumor medications exhibit limited solubility and permeability, and poor stability in harsh gastrointestinal environments, thereby impeding their therapeutic efficacy for chemotherapy. Therefore, more and more micro-nano drug delivery carriers have been developed and used to effectively deliver anti-cancer drugs, which can overcome physiological barriers, facilitate oral administration, and ultimately improve drug efficacy. In this paper, we first discuss the effects of various biological barriers on micro-nano drug carriers and oral administration approach. Then, the development of micro-nano drug carriers based on various biomedical components, such as micelles, dendrimers, hydrogels, liposomes, inorganic nanoparticles, etc. were introduced. Finally, the current dilemma and the potential of oral drug delivery for clinical treatment were discussed. The primary objective of this review is to introduce various oral delivery methods and serve as a point of reference for the advancement of novel oral delivery carriers, with the ultimate goal of informing the development of future clinical applications.
Collapse
Affiliation(s)
- Hongzheng Li
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Xiang Chen
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Shangrui Rao
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Minyu Zhou
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jianhua Lu
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Danna Liang
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, 20520, Finland
| | - Bingzi Zhu
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Letian Meng
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Ji Lin
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaoya Ding
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Qingfei Zhang
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Danhong Hu
- Department of Neurological Rehabilitation, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| |
Collapse
|
34
|
Qin J, Liu J, Wei Z, Li X, Chen Z, Li J, Zheng W, Liu H, Xu S, Yong T, Zhao B, Gou S, Ju S, Teng GJ, Yang X, Gan L. Targeted intervention in nerve-cancer crosstalk enhances pancreatic cancer chemotherapy. NATURE NANOTECHNOLOGY 2025; 20:311-324. [PMID: 39496914 DOI: 10.1038/s41565-024-01803-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 09/04/2024] [Indexed: 11/06/2024]
Abstract
Nerve-cancer crosstalk has gained substantial attention owing to its impact on tumour growth, metastasis and therapy resistance. Effective therapeutic strategies targeting tumour-associated nerves within the intricate tumour microenvironment remain a major challenge in pancreatic cancer. Here we develop Escherichia coli Nissle 1917-derived outer membrane vesicles conjugated with nerve-binding peptide NP41, loaded with the tropomyosin receptor kinase (Trk) inhibitor larotrectinib (Lar@NP-OMVs) for tumour-associated nerve targeting. Lar@NP-OMVs achieve efficient nerve intervention to diminish neurite growth by disrupting the neurotrophin/Trk signalling pathway. Moreover, OMV-mediated repolarization of M2-like tumour-associated macrophages to an M1-like phenotype results in nerve injury, further accentuating Lar@NP-OMV-induced nerve intervention to inhibit nerve-triggered proliferation and migration of pancreatic cancer cells and angiogenesis. Leveraging this strategy, Lar@NP-OMVs significantly reduce nerve infiltration and neurite growth promoted by gemcitabine within the tumour microenvironment, leading to augmented chemotherapy efficacy in pancreatic cancer. This study sheds light on a potential avenue for nerve-targeted therapeutic intervention for enhancing pancreatic cancer therapy.
Collapse
Affiliation(s)
- Jiaqi Qin
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjie Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaohan Wei
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaoxia Chen
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jianye Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Wenxia Zheng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Haojie Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Shiyi Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, China
| | - Ben Zhao
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Shanmiao Gou
- Department of Pancreatic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenghong Ju
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China
| | - Gao-Jun Teng
- Cultivation and Construction Site of the State Key Laboratory of Intelligent Imaging and Interventional Medicine, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, China.
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
35
|
Wang M, Yao Y, Yang Y, Zhu D, Wang M, Jia R, Chen S, Zhao X, Yang Q, Wu Y, Zhang S, Huang J, Ou X, Tian B, Sun D, Zhang L, Yu Y, He Y, Wu Z, Cheng A, Liu M. The characterization of outer membrane vesicles (OMVs) and their role in mediating antibiotic-resistance gene transfer through natural transformation in Riemerella anatipestifer. Poult Sci 2025; 104:104730. [PMID: 39729729 PMCID: PMC11742308 DOI: 10.1016/j.psj.2024.104730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 12/09/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024] Open
Abstract
Riemerella anatipestifer (R. anatipestifer, RA) is the etiological agent of duck serositis, an acute multisystemic disease in ducks that is globally distributed and causes serious economic losses in the duck industry. Despite exhibiting multidrug resistance, the transmission mechanism of its antibiotic resistance genes (ARGs) remains incompletely identified. To contribute to addressing this gap, in this study, outer membrane vesicles (OMVs) from the RA strain CH-1 were isolated and characterized to investigate their involvement in ARG transfer in RA. Sequencing and data analysis revealed that RA CH-1 OMVs had ∼2.04 Mb genomic size, representing 88.3 % of the RA CH-1 genomic length. Proteomic analysis showed that OMVs contained 577 proteins, representing 27.2 % of the bacterial proteins. Subsequent investigations demonstrated that OMVs from antibiotic-resistant strains transferred ARG fragments and plasmids to the sensitive strain RA ATCC11845, relying on the natural transformation system, and the transformants exhibited corresponding resistance. Overall, OMV-mediated horizontal transfer of ARGs serving as a significant mechanism for acquiring multiple resistance genes in R. anatipestifer.
Collapse
Affiliation(s)
- Mengying Wang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yizhou Yao
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuhao Yang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Dekang Zhu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingshu Wang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Renyong Jia
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Shun Chen
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xinxin Zhao
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Qiao Yang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ying Wu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Shaqiu Zhang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Juan Huang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xumin Ou
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Bin Tian
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Di Sun
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Ling Zhang
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yanling Yu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yu He
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhen Wu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Anchun Cheng
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| | - Mafeng Liu
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu 611130, China; Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu 611130, China; International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu 611130, China; Research Center of Avian Disease, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
36
|
Wang Y, Huang X, Hu Z, Peng H, Yang Y, Chen J, Dou J, Xiao C, Shang W, Rao X. Oxacillin promotes membrane vesicle secretion in Staphylococcus aureus via a SarA-Sle1 regulatory cascade. NANOSCALE 2025; 17:2488-2497. [PMID: 39589411 DOI: 10.1039/d4nr04321a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Membrane vesicles (MVs) are nanoscale particles secreted by living bacteria in vitro and in vivo. Bacterial MVs encapsulate various proteins, making them promising candidates for developing vaccines, drug carriers, and cancer immunotherapy agents. However, the mechanisms underlying MV secretion in Gram-positive bacteria remain unclear. Here, we showed that the subinhibitory concentration of oxacillin (OXA) stimulated MV production in Staphylococcus aureus with diverse genetic backgrounds. OXA treatment remarkably increased the expression of sle1, which encodes a main peptidoglycan hydrolase for adjusting peptidoglycan cross-linking. Deletion of sle1 decreased the OXA-mediated MV yield, whereas overexpression of sle1 considerably increased MV production. The accessory regulator SarA increased in response to OXA treatment, and SarA inactivation substantially attenuated OXA-stimulated MV production. We also demonstrated that SarA controlled sle1 expression by directly binding to its promoter region. Thus, the SarA-Sle1 regulatory axis was formed to mediate OXA-induced MV production in S. aureus. MVs derived from OXA-treated S. aureus RN4220 (MVs/OXA) exhibited a smaller particle size compared with those purified from wild-type RN4220; however, proteomic analysis revealed a comparable protein profile between MVs and MVs/OXA. Overall, our research reveals a mechanism underlying OXA-promoted S. aureus MV secretion and highlights the potential application of OXA-induced MVs.
Collapse
Affiliation(s)
- Yuting Wang
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China.
| | - Xiaonan Huang
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China.
| | - Zhen Hu
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China.
| | - Huagang Peng
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China.
| | - Yi Yang
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China.
| | - Juan Chen
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China.
| | - Jianxiong Dou
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China.
| | - Chuan Xiao
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China.
| | - Weilong Shang
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China.
| | - Xiancai Rao
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Under the Educational Committee in Chongqing, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
37
|
Wood NA, Gopinath T, Shin K, Marassi FM. In situ NMR reveals a pH sensor motif in an outer membrane protein that drives bacterial vesicle production. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634179. [PMID: 39896486 PMCID: PMC11785132 DOI: 10.1101/2025.01.21.634179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The outer membrane vesicles (OMVs) produced by diderm bacteria have important roles in cell envelope homeostasis, secretion, interbacterial communication, and pathogenesis. The facultative intracellular pathogen Salmonella enterica Typhimurium (STm) activates OMV biogenesis inside the acidic vacuoles of host cells by upregulating the expression of the outer membrane (OM) protein PagC, one of the most robustly activated genes in a host environment. Here, we used solid-state nuclear magnetic resonance (NMR) and electron microscopy (EM), with native bacterial OMVs, to demonstrate that three histidines, essential for the OMV biogenic function of PagC, constitute a key pH-sensing motif. The NMR spectra of PagC in OMVs show that they become protonated around pH 6, and His protonation is associated with specific perturbations of select regions of PagC. The use of bacterial OMVs is an essential aspect of this work enabling NMR structural studies in the context of the physiological environment. PagC expression upregulates OMV production in E. coli, replicating its function in STm. Moreover, the presence of PagC drives a striking aggregation of OMVs and increases bacterial cell pellicle formation at acidic pH, pointing to a potential role as an adhesin active in biofilm formation. The data provide experimental evidence for a pH-dependent mechanism of OMV biogenesis and aggregation driven by an outer membrane protein.
Collapse
Affiliation(s)
- Nicholas A Wood
- Department of Biophysics, Medical College of Wisconsin 8701 Watertown Plank Road, Milwaukee, WI 53226-3548 USA
| | - Tata Gopinath
- Department of Biophysics, Medical College of Wisconsin 8701 Watertown Plank Road, Milwaukee, WI 53226-3548 USA
| | - Kyungsoo Shin
- Department of Biophysics, Medical College of Wisconsin 8701 Watertown Plank Road, Milwaukee, WI 53226-3548 USA
| | - Francesca M. Marassi
- Department of Biophysics, Medical College of Wisconsin 8701 Watertown Plank Road, Milwaukee, WI 53226-3548 USA
| |
Collapse
|
38
|
Zhao X, Wei Y, Bu Y, Ren X, Dong Z. Review on bacterial outer membrane vesicles: structure, vesicle formation, separation and biotechnological applications. Microb Cell Fact 2025; 24:27. [PMID: 39833809 PMCID: PMC11749425 DOI: 10.1186/s12934-025-02653-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Outer membrane vesicles (OMVs), shed by Gram-negative bacteria, are spherical nanostructures that play a pivotal role in bacterial communication and host-pathogen interactions. Comprising an outer membrane envelope and encapsulating a variety of bioactive molecules from their progenitor bacteria, OMVs facilitate material and informational exchange. This review delves into the recent advancements in OMV research, providing a comprehensive overview of their structure, biogenesis, and mechanisms of vesicle formation. It also explores their role in pathogenicity and the techniques for their enrichment and isolation. Furthermore, the review highlights the burgeoning applications of OMVs in the field of biomedicine, emphasizing their potential as diagnostic tools, vaccine candidates, and drug delivery vectors.
Collapse
Affiliation(s)
- Xiaofei Zhao
- Graduate School, Hebei Medical University, Shijiazhuang, China
- Department of Pharmacy, Hebei Key Laboratory of Clinical Pharmacy, Hebei General Hospital, Shijiazhuang, China
| | - Yusen Wei
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Yuqing Bu
- Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Xiaokai Ren
- Graduate School, Hebei Medical University, Shijiazhuang, China
- Department of Pharmacy, Hebei Key Laboratory of Clinical Pharmacy, Hebei General Hospital, Shijiazhuang, China
| | - Zhanjun Dong
- Graduate School, Hebei Medical University, Shijiazhuang, China.
- Department of Pharmacy, Hebei Key Laboratory of Clinical Pharmacy, Hebei General Hospital, Shijiazhuang, China.
| |
Collapse
|
39
|
Chen J, Liu M, Chen S, Chou CP, Liu H, Wu D, Liu Y. Engineered Therapeutic Bacteria with High-Yield Membrane Vesicle Production Inspired by Eukaryotic Membrane Curvature for Treating Inflammatory Bowel Disease. ACS NANO 2025; 19:2405-2418. [PMID: 39772458 DOI: 10.1021/acsnano.4c13069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Bacterial membrane vesicles (BMVs) are emerging as powerful natural nanoparticles with transformative potential in medicine and industry. Despite their promise, scaling up BMV production and ensuring stable isolation and storage remain formidable challenges that limit their broader application. Inspired by eukaryotic mechanisms of membrane curvature, we engineered Escherichia coli DH5α to serve as a high-efficiency BMV factory. By fusing the ethanolamine utilization microcompartment shell protein EutS with the outer membrane via the ompA signal peptide, we induced dramatic membrane curvatures that drove enhanced vesiculation. Simultaneously, overexpression of fatty acyl reductase led to the production of amphiphilic fatty alcohols, further amplifying the BMV yield. Dynamic modulation of peptidoglycan hydrolase (PGase) expression facilitated efficient BMV release, resulting in a striking 149.11-fold increase in vesicle production. Notably, the high-yield BMVs from our engineered strain, without the need for purification, significantly bolstered innate immune responses and demonstrated therapeutic efficacy in treating inflammatory bowel disease (IBD). This study presents a strategy to overcome BMV production barriers, showcasing the therapeutic potential of engineered bacteria and BMVs for IBD treatment, while highlighting their potential applications in diverse biomedical fields.
Collapse
Affiliation(s)
- Jinjin Chen
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Mingkang Liu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Shiyi Chen
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - C Perry Chou
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Hongmei Liu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Decheng Wu
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yilan Liu
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| |
Collapse
|
40
|
Zhao Y, Chen J, Tian Y, Huang H, Zhao F, Deng X. Treponema denticola major surface protein (Msp): a key player in periodontal pathogenicity and immune evasion. Arch Microbiol 2025; 207:36. [PMID: 39825920 DOI: 10.1007/s00203-024-04223-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/08/2024] [Accepted: 12/19/2024] [Indexed: 01/30/2025]
Abstract
Treponema denticola, a bacterium that forms a "red complex" with Porphyromonas gingivalis and Tannerella forsythia, is associated with periodontitis, pulpitis, and other oral infections. The major surface protein (Msp) is a surface glycoprotein with a relatively well-established overall domain structure (N-terminal, central and C-terminal regions) and a controversial tertiary structure. As one of the key virulence factors of T. denticola, Msp is associated with adherence, immune response, and pore formation by the microorganism. It also mediates several pathological changes in histocytes, such as cytoskeleton disruption, neutrophil phagocytosis, and phosphoinositide balance interruption. In addition, the Msp of T. denticola is also an ortholog of the Treponema pallidum repeat (Tpr) proteins and Msp or Msp-like proteins that have been detected in other oral treponeme species. This review will discuss the structure, pathogenicity and homologs of Msp produced by T. denticola, illuminate the controversy regarding the structure and membrane topology of native Msp, explore the potential roles of Msp in the mechanism of T. denticola immune escape and provide an overview of the cytotoxicity and adherence ability of Msp. Further understanding of the structure and functions of Msp will offer new insights that will help promote further investigations of the pathogenic mechanisms of T. denticola and other treponemes, leading to more effective prophylactic or therapeutic treatments for relevant diseases.
Collapse
Affiliation(s)
- Yue Zhao
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Jiaxin Chen
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Yifei Tian
- Department of Clinical Medicine, Hengyang Medical College, University of South China, Hengyang, 421001, People's Republic of China
| | - Hong Huang
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Feijun Zhao
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Xuan Deng
- Department of Stomatology, The Second Affiliated Hospital, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
41
|
He J, Li J, Li M, He Z, Ye Y, Li J, Rao J, Zhao X, Li M, He Q. Platelet backpacking nanoparticles based on bacterial outer membrane vesicles enhanced photothermal-immune anti-tumor therapy. NANOSCALE 2025; 17:1510-1523. [PMID: 39621096 DOI: 10.1039/d4nr02757d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Bacterial outer membrane vesicles (OMVs), produced by Gram-negative bacteria, retain the immunostimulatory capacity of parental bacteria. OMVs have been recognized as potent natural immune adjuvants and drug delivery vehicles. Photothermal therapy that triggers immunogenic cell death further stimulates the immune system by releasing damage-associated molecular patterns. This therapeutic effect can be synergized with OMVs to achieve enhanced anti-tumor outcomes. We also observed that tumors can recruit platelets. Leveraging the phenomenon, we have innovatively employed platelets as "couriers" to boost the tumor-targeting delivery efficiency of both OMVs and photothermal agents. In detail, based on OMVs, we meticulously engineered nanoparticles (IR780-SLN@O-P) with platelet-binding capacity. These "courier" platelets carry "cargo" IR780-SLN@O-P NPs to tumor sites via P-selectin, ensuring targeted delivery. Under laser irradiation, the photothermal agents generate significant photothermal effects, which combined with the immune-stimulating properties of OMVs, creating a robust anti-tumor immune response. For "cold" tumors such as triple-negative breast cancer (TNBC), our IR780-SLN@O-P NPs not only prolonged the survival of mice bearing 4T1 orthotopic tumors, but also significantly suppressed tumor growth. Moreover, they facilitated dendritic cell maturation and the infiltration of CD8+ T cells to ameliorate the immunosuppressive tumor environment. Our research aims to highlight the unique advantages of OMVs and explore the potential of the tumor-targeting strategy that synergizes photothermal therapy with immunotherapy. We hope that our findings can offer insights into TNBC clinical treatments.
Collapse
Affiliation(s)
- Jiao He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Jiayu Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Min Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Zhidi He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Yunxia Ye
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Jiaxin Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Jingdong Rao
- Department of Applied Biology and Chemical Technology, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Xin Zhao
- Department of Applied Biology and Chemical Technology, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
42
|
Plavec TV, Žagar Soderžnik K, Della Pelle G, Zupančič Š, Vidmar R, Berlec A. Incorporation of recombinant proteins into extracellular vesicles by Lactococcus cremoris. Sci Rep 2025; 15:1768. [PMID: 39815011 PMCID: PMC11736121 DOI: 10.1038/s41598-025-86492-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/10/2025] [Indexed: 01/18/2025] Open
Abstract
Extracellular vesicles (EVs) are nanosized lipid bilayer particles released by various cellular organisms that carry an array of bioactive molecules. EVs have diagnostic potential, as they play a role in intercellular interspecies communication, and could be applied in drug delivery. In contrast to mammalian cell-derived EVs, the study of EVs from bacteria, particularly Gram-positive bacteria, received less research attention. This study aimed to investigate the production of EVs by lactic acid bacterium Lactococcus cremoris NZ9000 and to examine the impact of recombinant protein expression on their formation and protein content. Four different recombinant proteins were expressed in L. cremoris NZ9000, in different forms of expression and combinations, and the produced EVs were isolated using the standard ultracentrifugation method. The presence of vesicular structures (50-200 nm) in the samples was confirmed by transmission electron microscopy and by flow cytometry using membrane-specific stain. Mass spectrometry analyses confirmed the presence of recombinant proteins in the EVs fraction, with amounts ranging from 13.17 to 100%, highlighting their significant incorporation into the vesicles, together with intrinsic L. cremoris NZ9000 proteins that were either more abundant in the cytoplasm (ribosomal proteins, metabolic enzymes) or present in the membrane. The presence of the most abundant lactococcal proteins in EVs fraction suggests that protein cargo-loading of EVs in L. cremoris NZ9000 is not regulated. However, our data suggests that L. cremoris NZ9000 genetically engineered to express recombinant proteins can produce EVs containing these proteins in scalable manner. As L. cremoris NZ9000 is considered safe bacterium, EVs from L. cremoris NZ9000 could have several advantages over EVs from other bacteria, implying possible biotechnological applications, e.g. in therapeutic protein delivery.
Collapse
Affiliation(s)
- Tina Vida Plavec
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | | | - Giulia Della Pelle
- Department for Nanostructured Materials, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Špela Zupančič
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Robert Vidmar
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Aleš Berlec
- Department of Biotechnology, Jožef Stefan Institute, Ljubljana, Slovenia.
- Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
43
|
Li X, Cui J, Ding Z, Tian Z, Kong Y, Li L, Liu Y, Zhao W, Chen X, Guo H, Cui Z, Li X, Yuan J, Zhang H. Klebsiella pneumoniae-derived extracellular vesicles impair endothelial function by inhibiting SIRT1. Cell Commun Signal 2025; 23:21. [PMID: 39800699 PMCID: PMC11726972 DOI: 10.1186/s12964-024-02002-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The potential role of Klebsiella pneumoniae (K.pn) in hypertension development has been emphasized, although the specific mechanisms have not been well understood. Bacterial extracellular vesicles (BEVs) released by Gram-negative bacteria modulate host cell functions by delivering bacterial components to host cells. Endothelial dysfunction is an important early event in the pathogenesis of hypertension, yet the impact of K.pn-secreted EVs (K.pn EVs) on endothelial function remains unclear. This study aimed to investigate the effects of K.pn EVs on endothelial function and to elucidate the underlying mechanisms. METHODS K.pn EVs were purified from the bacterial suspension using ultracentrifugation and characterized by transmission electron microscopy nanoparticle tracking analysis, and EV marker expression. Endothelium-dependent relaxation was measured using a wire myograph after in vivo or ex vivo treatment with K.pn EVs. Superoxide anion production was measured by confocal microscopy and HUVEC senescence was assessed by SA-β-gal activity. SIRT1 overexpression or activator was utilized to investigate the underlying mechanisms. RESULTS Our data showed that K.pn significantly impaired acetylcholine-induced endothelium-dependent relaxation and increased superoxide anion production in endothelial cells in vivo. Similarly, in vivo and ex vivo studies showed that K.pn EVs caused significant endothelial dysfunction, endothelial provocation, and increased blood pressure. Further examination revealed that K.pn EVs reduced the levels of SIRT1 and p-eNOS and increased the levels of NOX2, COX-2, ET-1, and p53 in endothelial cells. Notably, overexpression or activation of SIRT1 attenuated the adverse effects and protein changes induced by K.pn EVs on endothelial cells. CONCLUSION This study reveals a novel role of K.pn EVs in endothelial dysfunction and dissects the relevant mechanism involved in this process, which will help to establish a comprehensive understanding of K.pn EVs in endothelial dysfunction and hypertension from a new scope.
Collapse
Affiliation(s)
- Xinxin Li
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Jinghua Cui
- Microbiology Department, Capital Institute of Pediatrics, China No.2 Yabao Road, Chaoyang District, Beijing, 100020, China
| | - Zanbo Ding
- Microbiology Department, Capital Institute of Pediatrics, China No.2 Yabao Road, Chaoyang District, Beijing, 100020, China
| | - Ziyan Tian
- Microbiology Department, Capital Institute of Pediatrics, China No.2 Yabao Road, Chaoyang District, Beijing, 100020, China
| | - Yiming Kong
- Microbiology Department, Capital Institute of Pediatrics, China No.2 Yabao Road, Chaoyang District, Beijing, 100020, China
| | - Linghai Li
- Department of Anesthesiology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Yang Liu
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Wen Zhao
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Xueying Chen
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Han Guo
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Zhengshuo Cui
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Xinwei Li
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China
| | - Jing Yuan
- Microbiology Department, Capital Institute of Pediatrics, China No.2 Yabao Road, Chaoyang District, Beijing, 100020, China.
| | - Huina Zhang
- Beijing An Zhen Hospital, Capital Medical University, The Key Laboratory of Remodeling Cardiovascular Diseases, Ministry of Education; Collaborative Innovation Center for Cardiovascular Disorders, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing, 100029, China.
| |
Collapse
|
44
|
Su D, Li M, Xie Y, Xu Z, Lv G, Jiu Y, Lin J, Chang CJ, Chen H, Cheng F. Gut commensal bacteria Parabacteroides goldsteinii-derived outer membrane vesicles suppress skin inflammation in psoriasis. J Control Release 2025; 377:127-145. [PMID: 39532207 DOI: 10.1016/j.jconrel.2024.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/19/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Despite gut microbiota-derived extracellular vesicles (EVs) serving as pivotal mediators in bacteria-host cell interactions, their potential role in modulating skin inflammation remains poorly understood. Here, we developed strategies for mass production of Parabacteroides goldsteinii-derived outer membrane vesicles (Pg OMVs), commonly known as EVs. We found that orally administered Pg OMVs can reach the colon, traverse the intestinal barrier, and circulate to the inflamed skin of psoriasis-like mice, resulting in reduced epidermal hyperplasia, suppressed infiltration of inflammatory cells in the skin lesions, and effective amelioration of both skin and systemic inflammation. Additionally, subcutaneous injection of thermosensitive PF-127 hydrogel loaded with Pg OMVs exerts similar immunomodulatory effects, allowing sustained release of Pg OMVs into skin cells, effectively suppressing skin inflammation and ameliorating symptoms of psoriasis. This study unveils the importance of gut microbiota-derived OMVs, which can target inflamed skin via both the gut-skin axis and local skin administration, providing a promising alternative to live bacteria therapy for the treatment of skin inflammatory diseases.
Collapse
Affiliation(s)
- Dandan Su
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Manchun Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Yuedong Xie
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Zhanxue Xu
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Department of Pharmacy, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, China
| | - Guowen Lv
- College of Life Sciences, Northwest A&F University, Yangling 712100, China
| | - Yaming Jiu
- Unit of Cell Biology and Imaging Study of Pathogen Host Interaction, The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Yuquan Road No. 19(A), Shijingshan District, Beijing 100049, China
| | - Jingxiong Lin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Chih-Jung Chang
- Medical Research Center and Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen 361028, China; School of Medicine, Huaqiao University, Quanzhou 362021, China.
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
45
|
Li G, Pu S, You L, Gao Y, Zhong Y, Zhao H, Fan D, Lu X. Innovative Strategies in Oncology: Bacterial Membrane Vesicle-Based Drug Delivery Systems for Cancer Diagnosis and Therapy. Pharmaceutics 2025; 17:58. [PMID: 39861706 PMCID: PMC11768367 DOI: 10.3390/pharmaceutics17010058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/18/2024] [Accepted: 12/25/2024] [Indexed: 01/27/2025] Open
Abstract
Outer membrane vesicles (OMVs) are double-layered structures of nanoscale lipids released by gram-negative bacteria. They have the same membrane composition and characteristics as primitive cells, which enables them to penetrate cells and tissues efficiently. These OMVs exhibit excellent membrane stability, immunogenicity, safety, and permeability (which makes it easier for them to penetrate into tumour tissue), making them suitable for developing cancer vaccines and drug delivery systems. Recent studies have focused on engineering OMVs to enhance tumour-targeting capabilities, reduce toxicity, and extend circulation time in vivo. This article reviews the latest progress in OMV engineering for tumour treatment and discusses the challenges associated with the use of OMV-based antitumour therapy in clinical practice.
Collapse
Affiliation(s)
- Guodong Li
- College of Life Sciences, Northwest University, Xi’an 710069, China; (G.L.)
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Shuangpeng Pu
- College of Life Sciences, Northwest University, Xi’an 710069, China; (G.L.)
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Lisiyao You
- College of Life Sciences, Northwest University, Xi’an 710069, China; (G.L.)
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Yuan Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Biotechnology Center, School of Pharmacy, The Fourth Military Medical University, Xi’an 710032, China
| | - Yuexia Zhong
- Outpatient Department of the Second Affiliated Hospital of the Fourth Military Medical University, Xi’an 710032, China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an 710038, China;
| | - Dong Fan
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an 710038, China;
| | - Xiyan Lu
- Outpatient Department of the Second Affiliated Hospital of the Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
46
|
Ayala-García P, Herrero-Gómez I, Jiménez-Guerrero I, Otto V, Moreno-de Castro N, Müsken M, Jänsch L, van Ham M, Vinardell JM, López-Baena FJ, Ollero FJ, Pérez-Montaño F, Borrero-de Acuña JM. Extracellular Vesicle-Driven Crosstalk between Legume Plants and Rhizobia: The Peribacteroid Space of Symbiosomes as a Protein Trafficking Interface. J Proteome Res 2025; 24:94-110. [PMID: 39665174 PMCID: PMC11705226 DOI: 10.1021/acs.jproteome.4c00444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
Prokaryotes and eukaryotes secrete extracellular vesicles (EVs) into the surrounding milieu to preserve and transport elevated concentrations of biomolecules across long distances. EVs encapsulate metabolites, DNA, RNA, and proteins, whose abundance and composition fluctuate depending on environmental cues. EVs are involved in eukaryote-to-prokaryote communication owing to their ability to navigate different ecological niches and exchange molecular cargo between the two domains. Among the different bacterium-host relationships, rhizobium-legume symbiosis is one of the closest known to nature. A crucial developmental stage of symbiosis is the formation of N2-fixing root nodules by the plant. These nodules contain endocytosed rhizobia─called bacteroids─confined by plant-derived peribacteroid membranes. The unrestricted interface between the bacterial external membrane and the peribacteroid membrane is the peribacteroid space. Many molecular aspects of symbiosis have been studied, but the interbacterial and interdomain molecule trafficking by EVs in the peribacteroid space has not been questioned yet. Here, we unveil intensive EV trafficking within the symbiosome interface of several rhizobium-legume dual systems by developing a robust EV isolation procedure. We analyze the EV-encased proteomes from the peribacteroid space of each bacterium-host partnership, uncovering both conserved and differential traits of every symbiotic system. This study opens the gates for designing EV-based biotechnological tools for sustainable agriculture.
Collapse
Affiliation(s)
- Paula Ayala-García
- Department
of Microbiology, Faculty of Biology, Universidad
de Sevilla, Av. de la Reina Mercedes 6, 41012 Sevilla, Spain
| | - Irene Herrero-Gómez
- Department
of Microbiology, Faculty of Biology, Universidad
de Sevilla, Av. de la Reina Mercedes 6, 41012 Sevilla, Spain
| | - Irene Jiménez-Guerrero
- Department
of Microbiology, Faculty of Biology, Universidad
de Sevilla, Av. de la Reina Mercedes 6, 41012 Sevilla, Spain
| | - Viktoria Otto
- Institute
of Microbiology, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany
| | - Natalia Moreno-de Castro
- Department
of Microbiology, Faculty of Biology, Universidad
de Sevilla, Av. de la Reina Mercedes 6, 41012 Sevilla, Spain
| | - Mathias Müsken
- Central
Facility for Microscopy, Helmholtz Centre
for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Lothar Jänsch
- Cellular
Proteome Research, Helmholtz Centre for
Infection Research, Inhoffenstraße
7, 38124 Braunschweig, Germany
| | - Marco van Ham
- Cellular
Proteome Research, Helmholtz Centre for
Infection Research, Inhoffenstraße
7, 38124 Braunschweig, Germany
| | - José-María Vinardell
- Department
of Microbiology, Faculty of Biology, Universidad
de Sevilla, Av. de la Reina Mercedes 6, 41012 Sevilla, Spain
| | - Francisco Javier López-Baena
- Department
of Microbiology, Faculty of Biology, Universidad
de Sevilla, Av. de la Reina Mercedes 6, 41012 Sevilla, Spain
| | - Francisco Javier Ollero
- Department
of Microbiology, Faculty of Biology, Universidad
de Sevilla, Av. de la Reina Mercedes 6, 41012 Sevilla, Spain
| | - Francisco Pérez-Montaño
- Department
of Microbiology, Faculty of Biology, Universidad
de Sevilla, Av. de la Reina Mercedes 6, 41012 Sevilla, Spain
| | - José Manuel Borrero-de Acuña
- Department
of Microbiology, Faculty of Biology, Universidad
de Sevilla, Av. de la Reina Mercedes 6, 41012 Sevilla, Spain
| |
Collapse
|
47
|
Wang J, Zhang Y, Wang S, Wang X, Jing Y, Su J. Bone aging and extracellular vesicles. Sci Bull (Beijing) 2024; 69:3978-3999. [PMID: 39455324 DOI: 10.1016/j.scib.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/01/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Bone aging, a major global health concern, is the natural decline in bone mass and strength. Concurrently, extracellular vesicles (EVs), tiny membrane-bound particles produced by cells, have gained recognition for their roles in various physiological processes and age-related diseases. The interaction between EVs and bone aging is of growing interest, particularly their effects on bone metabolism, which become increasingly critical with advancing age. In this review, we explored the biology, types, and functions of EVs and emphasized their regulatory roles in bone aging. We examined the effects of EVs on bone metabolism and highlighted their potential as biomarkers for monitoring bone aging progression. Furthermore, we discussed the therapeutic applications of EVs, including targeted drug delivery and bone regeneration, and addressed the challenges associated with EV-based therapies, including the technical complexities and regulatory issues. We summarized the current research and clinical trials investigating the role of EVs in bone aging and suggested future research directions. These include the potential for personalized medicine using EVs and the integration of EV research with advanced technologies to enhance the management of age-related bone health. This analysis emphasized the transformative potential of EVs in understanding and managing bone aging, thereby marking a significant advancement in skeletal health research.
Collapse
Affiliation(s)
- Jian Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; School of Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China
| | - Yuanwei Zhang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China
| | - Sicheng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China
| | - Xinglong Wang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA.
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China.
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
48
|
Jia X, Yuan B, Wang W, Wang K, Ling D, Wei M, Hu Y, Guo W, Chen Z, Du L, Jin Y. Gene editing tool-loaded biomimetic cationic vesicles with highly efficient bacterial internalization for in vivo eradication of pathogens. J Nanobiotechnology 2024; 22:787. [PMID: 39710679 DOI: 10.1186/s12951-024-03065-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 12/05/2024] [Indexed: 12/24/2024] Open
Abstract
In the post-COVID-19 era, drug-resistant bacterial infections emerge as one of major death causes, where multidrug-resistant Acinetobacter baumannii (MRAB) and drug-resistant Pseudomonas aeruginosa (DRPA) represent primary pathogens. However, the classical antibiotic strategy currently faces the bottleneck of drug resistance. We develop an antimicrobial strategy that applies the selective delivery of CRISPR/Cas9 plasmids to pathogens with biomimetic cationic hybrid vesicles (BCVs), irrelevant to bacterial drug resistance. CRISPR/Cas9 plasmids were constructed, replicating in MRAB or DRPA and expressing ribonucleic proteins, leading to irreparable chromosomal lesions; however, delivering the negatively charged plasmids with extremely large molecular weight to the pathogens at the infection site became a huge challenge. We found that the BCVs integrating the bacterial out membrane vesicles and cationic lipids efficiently delivered the plasmids in vitro/in vivo to the pathogens followed by effective internalization. The BCVs were used by intratracheal or topical hydrogel application against MRAB pulmonary infection or DRPA wound infection, and both of the two pathogens were eradicated from the lung or the wound. CRISPR/Cas9 plasmid-loaded BCVs become a promising medication for drug-resistant bacteria infections.
Collapse
Affiliation(s)
- Xueli Jia
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
- Department of Pharmaceutical Science, School of Pharmacy, Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China
| | - Bochuan Yuan
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China.
| | - Wanmei Wang
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Ke Wang
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Dandan Ling
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Meng Wei
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Yadan Hu
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Wanting Guo
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Ziyuan Chen
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Lina Du
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Yiguang Jin
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China.
| |
Collapse
|
49
|
Zhang R, Li G, Wu Y, Wang X, Luan Q. Pathogenic mechanisms and potential applications of extracellular vesicles from periodontal pathogens in periodontitis. Front Immunol 2024; 15:1513983. [PMID: 39759521 PMCID: PMC11695242 DOI: 10.3389/fimmu.2024.1513983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
Periodontitis is a multifactorial disease characterized by chronic destruction of the periodontal supporting tissues and is closely associated with the dysbiosis of the plaque biofilm. It is the leading cause of tooth loss in adults. Bacterial extracellular vesicles (BEVs) are released from bacteria, which range in size from 20 to 400 nm. These vesicles contain various components derived from their parent bacteria, including nucleic acids, proteins, lipids, and other molecules, which facilitate functions such as molecular transfer, metabolic regulation, bacterial interactions, biofilm formation, and immune modulation. BEVs participated in the pathophysiological process of periodontitis. Recently emerging evidence also showed that the contents of EVs in saliva and gingival crevicular fluid (miRNAs, mRNAs, and proteins) could be used as potential biomarkers for periodontitis. While most current research focuses on human-derived components, much less is known about BEVs. Therefore, this review introduces the formation mechanisms and components of BEVs related to periodontitis. Then, this review summarizes the current information about the mechanism, the diagnostic and theraputic value of periodontal pathogen-derived extracellular vesicles in the development of periodontitis. Furthermore, the future challenges of exploring the role of BEVs in periodontitis are also discussed.
Collapse
Affiliation(s)
- Ruiqing Zhang
- Department of Periodontology, Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Guoliang Li
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yingtao Wu
- Department of Periodontology, Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, Shandong, China
| | - Xiaoxuan Wang
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Qingxian Luan
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Beijing, China
| |
Collapse
|
50
|
Han JM, Mwiti G, Yeom SJ, Lim J, Kim WS, Lim S, Lim ST, Byun EB. Radiation-Resistant Bacteria Deinococcus radiodurans-Derived Extracellular Vesicles as Potential Radioprotectors. Adv Healthc Mater 2024:e2403192. [PMID: 39676336 DOI: 10.1002/adhm.202403192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/26/2024] [Indexed: 12/17/2024]
Abstract
The increasing use of radiation presents a risk of radiation exposure, making the development of radioprotectors necessary. In the previous study, it is investigated that Deinococcus radiodurans (R1-EVs) exert the antioxidative properties. However, the radioprotective activity of R1-EVs remains unclear. In the present study, the protective effects of R1-EVs against total body irradiation (TBI)-induced acute radiation syndrome (ARS) are investigated. To assess R1-EVs' radioprotective efficacy, ARS is induced in mice with 8 Gy of TBI, and protection against hematopoietic (H)- and gastrointestinal (GI)-ARS is evaluated. The survival rate of irradiated mice group decreases substantially after irradiation. In contrast, pretreatment with R1-EVs increases the survival rates of the mice. The administration of R1-EVs provides effective protection against radiation-induced death of bone marrow cells and splenocytes by scavenging reactive oxygen species (ROS). Additionally, R1-EVs protect both intestinal stem and epithelial cells from radiation-induced apoptosis. R1-EVs stimulate the production of short-chain fatty acids in the gastrointestinal tract, suppress proinflammatory cytokines, and increase regulatory T cells in pretreated mice versus the irradiation-only group. Proteomic analysis shows that the R1-EV proteome is significantly enriched with proteins involved in oxidative stress response. These findings highlight R1-EVs as potent radioprotectors with applications against radiation damage and ROS-mediated diseases.
Collapse
Affiliation(s)
- Jeong Moo Han
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, 03080, South Korea
- Institute for Data Innovation in Science, Seoul National University, Seoul, 08826, South Korea
| | - Godfrey Mwiti
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, 3000, Australia
- Department of Food and Nutrition, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Seo-Joon Yeom
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jaeyoon Lim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Food and Nutrition, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Woo Sik Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| | - Sangyong Lim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Radiation Science, University of Science and Technology, Daejeon, 01812, Republic of Korea
| | - Seung-Taik Lim
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Eui-Baek Byun
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| |
Collapse
|