1
|
Xie J, Van Hoecke L, Van Wonterghem E, Van Imschoot G, Andries V, Vereecke L, Vandenbroucke RE. The gut-brain axis in Alzheimer's disease is shaped by commensal gut microbiota derived extracellular vesicles. Gut Microbes 2025; 17:2501193. [PMID: 40355382 PMCID: PMC12077474 DOI: 10.1080/19490976.2025.2501193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/31/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025] Open
Abstract
Emerging clinical and experimental evidence highlight the involvement of gut microbiota in the onset and progression of neurodegenerative diseases such as Alzheimer's disease (AD) via neuroinflammatory processes along the gut-brain axis. Despite this, the precise mechanisms governing gut microbial involvement in AD remain elusive. In this study, we observed that AppNL-G-F AD mice raised under germ-free (GF) conditions, display a reduced amyloid-β (Aβ) pathology, accompanied by a shift in microglial cells toward a less inflammatory state and increased phagocytotic efficiency. In addition, we demonstrate that gut microbiota depletion can protect against synaptic deficits in AD mice. Notably, administering bacterial extracellular vesicles (bEVs), i.e. nano-sized particles packed with bacterial components, derived from fecal slurry from specific pathogen-free housed AppNL-G-F AD mice, reversed the effects of GF conditions on both microglial activation and Aβ plaque accumulation. These findings reveal for the first time that commensal gut microbiota-derived bEVs have a major impact on AD pathology progression.
Collapse
Affiliation(s)
- Junhua Xie
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Elien Van Wonterghem
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Griet Van Imschoot
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Vanessa Andries
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Ghent Gut Inflammation Group (GGIG), Ghent University, Ghent, Belgium
| | - Lars Vereecke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Ghent Gut Inflammation Group (GGIG), Ghent University, Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Ghent Gut Inflammation Group (GGIG), Ghent University, Ghent, Belgium
| |
Collapse
|
2
|
Vuong N, Alomia M, Byne A, Gade P, Philipson TR, Alhammad RI, Skislak CJ, Alruwaili I, Alsaab FM, Zhou W, Howard M, Brothers A, Still AH, Araujo RP, Van Hoek M, Birkaya B, Espina V, Hoefer RA, Liotta L, Luchini A. Lactobacillus rhamnosus-derived extracellular vesicles influence calcium deposition in a model of breast cancer intraductal calcium stress. iScience 2025; 28:112538. [PMID: 40491474 PMCID: PMC12148604 DOI: 10.1016/j.isci.2025.112538] [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: 10/22/2024] [Revised: 02/21/2025] [Accepted: 03/24/2025] [Indexed: 06/11/2025] Open
Abstract
Extracellular calcium export by the breast ductal epithelium is crucial during lactation and plays a significant role in breast cancer progression. Intraductal calcium deposition is a hallmark of aggressive premalignant lesions. This study tested the hypothesis that microbiome-derived extracellular vesicles (EVs) influence calcium modulation in premalignant breast cancer lesions. Based on the analysis of plasma, serum, saliva, and tissue collected from breast cancer patients and controls (N = 150), Lactobacillus rhamnosus (Lr) was chosen as the model microbiota. In a BT-474 human breast cancer cell line monolayer culture under acute calcium stress, Lr EVs enhanced intracellular calcium intake. In a BT474 3D spheroid model under chronic calcium stress, Lr EVs increased extracellular calcium deposition and mRNA expression of calcium export channel plasma membrane calcium-transporting ATPase 2 (PMCA2) and stromal interaction molecule 1 (STIM1) in a dose-dependent manner. We propose that Lr EVs influence calcium regulation and mineral deposition, thereby affecting premalignant breast cancer progression.
Collapse
Affiliation(s)
- Ngoc Vuong
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Melany Alomia
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Ahana Byne
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Purva Gade
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Thomas Raymond Philipson
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Rayan Ibrahim Alhammad
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
- Clinical Laboratory Department, College of Applied Medical Science, King Saud bin Abdulaziz University for Health Sciences, Al Ahsa, Saudi Arabia
- King Abdullah International Medical Research Center, Al Ahsa, Saudi Arabia
| | - Cade J. Skislak
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Intisar Alruwaili
- School of Systems Biology, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Fahad M. Alsaab
- School of Systems Biology, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
- College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Al Ahsa, Saudi Arabia
| | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Marissa Howard
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Andrea Brothers
- College Dean’s Office, American University, 4400 Massachusetts Avenue, Washington, DC 20016, USA
| | - Amanda Haymond Still
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Robyn P. Araujo
- The University of Melbourne, Grattan Street, Parkville, VIC 3010, Australia
| | - Monique Van Hoek
- School of Systems Biology, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Barbara Birkaya
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Virginia Espina
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Richard A. Hoefer
- Dorothy G. Hoefer Comprehensive Breast Center, 11803 Jefferson Avenue, Suite 130, Newport News, VA 23606, USA
| | - Lance Liotta
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| | - Alessandra Luchini
- Center for Applied Proteomics and Molecular Medicine, George Mason University, 10920 George Mason Circle, MSN 1A9, Manassas, VA 20110, USA
| |
Collapse
|
3
|
Zhang J, Yang R, Liang J, Huang Q, Huang AM, Qiu W, Li F, Wang L, Liang Y, Liu Z. Bacillus Coagulans Secretes Extracellular Vesicles and Modulates the Proliferation of Conjunctival Epithelial Cells via the P53/CDKN1A Signaling Pathway. Curr Eye Res 2025:1-12. [PMID: 40491050 DOI: 10.1080/02713683.2025.2513556] [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: 05/07/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025]
Abstract
PURPOSE To investigate the microbial strains in pterygium patients' tear samples via 16S rRNA sequencing and evaluate their impact on human conjunctival epithelial cells (HConEpics) and pterygium fibroblasts (HPFs) proliferation. METHODS Tear samples were cultured aerobically and anaerobically on blood agar plates. Bacterial colonies were characterized by 16S rRNA sequencing. Proliferation of HConEpics and HPFs was assessed using CCK-8 and EDU assays. The role of bacterial extracellular vesicles (EVs) was explored using the exosome inhibitor GW4869. EVs were isolated and characterized by transmission electron microscopy and nanoparticle tracking analysis (NTA), with DIL staining confirming their internalization by host cells. Transcriptomic sequencing and the SIRT2-IN-11 inhibitor were used to elucidate the molecular mechanisms and regulatory pathways. RESULTS 16S rRNA analysis revealed a significant reduction in the concentration of Bacillus coagulans (BC) in pterygium patients. BC significantly promoted HConEpics proliferation while inhibiting HPFs proliferation. GW4869 significantly reduced the stimulatory effect of BC culture supernatant on HConEpics proliferation, confirming EVs-medicated regulation. BC-derived EVs, isolated by ultracentrifugation, were internalized by HConEpics, promoting proliferation and inducing G1 phase cell cycle accumulation. These EVs also inhibited TGF-β-induced damage to HConEpics. Transcriptomic sequencing identified the p53 pathway as a key regulatory pathway, further clarified by SIRT2-IN-11. CONCLUSIONS This study offers novel insights into pterygium pathogenesis and identifies potential therapeutic targets.
Collapse
Affiliation(s)
- Jingyu Zhang
- Ophthalmic Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ruiming Yang
- Ophthalmic Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiamian Liang
- Ophthalmic Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qiudan Huang
- Ophthalmic Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Amy Michelle Huang
- Department of Ophthalmology, University of Colorado, Aurora, CO, United States
| | - Wangjian Qiu
- Ophthalmic Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Shenzhen Songgang People's Hospital of Bao 'an District, Shenzhen, Guangdong
| | - Fang Li
- Ophthalmic Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Li Wang
- Ophthalmic Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yue Liang
- Ophthalmic Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Zhiping Liu
- Ophthalmic Center, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
4
|
Qin LN, Yu YF, Ma L, Yu R. Intestinal bacteria-derived extracellular vesicles in metabolic dysfunction-associated steatotic liver disease: From mechanisms to therapeutics. Mol Cells 2025; 48:100216. [PMID: 40239896 DOI: 10.1016/j.mocell.2025.100216] [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: 01/14/2025] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a progressive disease that affects the health of approximately one-third of the world's population. It is the primary cause of end-stage liver disease, liver malignancy, and liver transplantation, resulting in a great medical burden. No medications have yet been approved by the US Food and Drug Administration for treating MASLD without liver inflammation or scarring. Therefore, the development of specific drugs to treat MASLD remains a key task in the ongoing research objective. Extracellular vesicles (EVs) play an important role in the communication between organs, tissues, and cells. Recent studies have found that intestinal microbiota are closely related to the pathogenesis and progression of MASLD. EVs produced by bacteria (BEVs) play an indispensable role in this process. Thus, this study provides a new direction for MASLD treatment. However, the mechanism by which BEVs affect MASLD remains unclear. Therefore, this study investigated the influence and function of intestinal microbiota in MASLD. Additionally, we focus on the research progress of BEVs in recent years and explain the relationship between BEVs and MASLD from the perspectives of glucose and lipid metabolism, immune responses, and intestinal homeostasis. Finally, we summarized the potential therapeutic value of BEVs and EVs from other sources, such as adipocytes, immunocytes, stem cells, and plants.
Collapse
Affiliation(s)
- Li-Na Qin
- Department of Endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yun-Feng Yu
- Department of Endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lie Ma
- Department of Reproductive Medicine, The Third Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Rong Yu
- Department of Endocrinology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China; College of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China.
| |
Collapse
|
5
|
Gal-Mandelbaum N, Carasso S, Kedem A, Ziv T, Keshet-David R, Abboud R, Zaatry R, Gefen T, Geva-Zatorsky N. Dietary carbohydrates alter immune-modulatory functionalities and DNA inversions in Bacteroides thetaiotaomicron. Nat Commun 2025; 16:4938. [PMID: 40436824 PMCID: PMC12120099 DOI: 10.1038/s41467-025-60202-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 05/15/2025] [Indexed: 06/01/2025] Open
Abstract
The gut bacteria environment is highly dynamic. Environmental conditions were shown to affect microbial composition. Yet, their influences on bacterial functionality (e.g., immune-modulation activity) are mostly overlooked. Distinct strains of the same species, and even the same bacterial strain, may have different effects on the immune system depending on their growth environment. Therefore, studying the functionality of strains under different conditions is crucial. We analyzed functional alterations in the gut symbiont Bacteroides thetaiotaomicron (B. theta) under different dietary components consumption in humans, upon white sugar consumption in mice, and in response to 190 different carbon sources in vitro. Dietary alterations affected the orientation of phase variable regions in B. theta in humans, in vivo, and in vitro, and altered B. theta's proteome and immune-modulatory functionality. Studying the effects of dietary components on the immune-modulatory functionalities of key members of the gut microbiota will allow for personalized dietary recommendations.
Collapse
Grants
- The Technion Institute of Technology, “Keren haNasi,” Cathedra, the Rappaport Technion Integrated Cancer Center, the Alon Fellowship for Outstanding Young Researchers, the Israeli Science Foundation (3165/20), the D. Dan and Betty Kahn Foundation’s gift to the University of Michigan, the Weizmann Institute, the Technion–Israel Institute of Technology Collaboration for Research, the Seerave Foundation, CIFAR (grant FL-000969/FL-001245/FL-001381), and the European Union (ERC, ExtractABact, 101078712).
Collapse
Affiliation(s)
- Noa Gal-Mandelbaum
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine and Research Institute, Rappaport Technion Integrated Cancer Center (RTICC), Technion-Israel Institute of Technology, Haifa, Israel
| | - Shaqed Carasso
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine and Research Institute, Rappaport Technion Integrated Cancer Center (RTICC), Technion-Israel Institute of Technology, Haifa, Israel
| | - Alon Kedem
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine and Research Institute, Rappaport Technion Integrated Cancer Center (RTICC), Technion-Israel Institute of Technology, Haifa, Israel
| | - Tamar Ziv
- Smoler Proteomics Center, Lokey Interdisciplinary Center for Life Sciences & Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Roni Keshet-David
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine and Research Institute, Rappaport Technion Integrated Cancer Center (RTICC), Technion-Israel Institute of Technology, Haifa, Israel
| | - Roberto Abboud
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine and Research Institute, Rappaport Technion Integrated Cancer Center (RTICC), Technion-Israel Institute of Technology, Haifa, Israel
| | - Rawan Zaatry
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine and Research Institute, Rappaport Technion Integrated Cancer Center (RTICC), Technion-Israel Institute of Technology, Haifa, Israel
| | - Tal Gefen
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine and Research Institute, Rappaport Technion Integrated Cancer Center (RTICC), Technion-Israel Institute of Technology, Haifa, Israel
| | - Naama Geva-Zatorsky
- Department of Cell Biology and Cancer Science, Rappaport Faculty of Medicine and Research Institute, Rappaport Technion Integrated Cancer Center (RTICC), Technion-Israel Institute of Technology, Haifa, Israel.
- CIFAR Humans & the Microbiome program, CIFAR, Toronto, Canada.
| |
Collapse
|
6
|
Zhu W, Yuan Y, Guan X, Xie Q, Sun X, Dong Z. Gut microbiota-derived extracellular vesicles: Exploring the pathogenesis and treatment of neuropsychiatric disorders. Life Sci 2025; 376:123750. [PMID: 40412609 DOI: 10.1016/j.lfs.2025.123750] [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: 03/09/2025] [Revised: 05/11/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
The gut microbiome is connected to the pathogenesis of various neuropsychiatric disorders, including cognitive impairment and depression. The gut microbiota can act on the brain through multiple pathways along the gut-brain axis. However, the specific mechanisms are unclear. Bacterial extracellular vesicles (BEVs), bacteria-derived membrane-bound vesicles that can carry a variety of bioactive substances and cross various barriers in the host, are used by bacteria to communicate and interact with the host. Recent studies have shown that BEVs from the gut microbiota are involved in gut-brain communication and may play a role in the pathogenesis and treatment of neuropsychiatric disorders. This review provides an overview of the biogenesis, structure, and function of BEVs and emphasizes their role in the development and treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Wenjie Zhu
- West China School of Medicine, Sichuan University, Chengdu 610041, PR China.
| | - Yanling Yuan
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xuan Guan
- Chengdu Medical College, Chengdu 610041, PR China
| | - Qinglian Xie
- Department of Outpatient, West China Hospital, Sichuan University, Chengdu 610041, PR China; Department of Outpatient, West China Xiamen Hospital of Sichuan University, Fujian 361022, PR China
| | - Xueli Sun
- Mental Health Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zaiquan Dong
- Mental Health Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
7
|
Moghaddam ZS, Dehghan A, Halimi S, Najafi F, Nokhostin A, Naeini AE, Akbarzadeh I, Ren Q. Bacterial Extracellular Vesicles: Bridging Pathogen Biology and Therapeutic Innovation. Acta Biomater 2025:S1742-7061(25)00352-6. [PMID: 40349898 DOI: 10.1016/j.actbio.2025.05.028] [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: 12/16/2024] [Revised: 04/14/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
The main role of bacterial extracellular vesicles (BEVs) has been associated with various processes such as intercellular communication and host-pathogen interactions. This comprehensive review explores the multifaceted functions of BEVs across different biological domains, emphasizing their dual nature as contributors to disease and potential vehicles for therapeutic intervention. We examine the intricate interactions of BEVs within bacterial communities and between bacteria and hosts, their involvement in disease development through cargo delivery mechanisms, and their beneficial impact on microbial ecology. The review also highlights BEVs' applications in biomedical field, where they are revolutionizing vaccine development, targeted drug delivery, and cancer therapy. By utilizing the inherent properties of BEVs for controlled drug release, targeted antigen delivery, and immune modulation, they offer a promising frontier in precision medicine. In addition, the diagnostic potential of BEVs is explored through their utility as biomarkers, providing valuable insights into disease states and treatment efficacy. Looking forward, this review underscores the challenges and opportunities in translating BEV research to clinical practice, promoting the need of standardized methods in BEV characterization and scaling up production. The diverse abilities of BEVs, ranging from contributing to pathogen virulence to driving therapeutic innovation, highlight their potential as a cornerstone in the future of biomedical advancements. STATEMENT OF SIGNIFICANCE: Bacterial extracellular vesicles (BEVs) are emerging as pivotal players in both pathogenesis and therapeutic innovation. This review explores their dual nature as agents of disease and as promising biomaterials for biomedical applications, and provides a comprehensive survey on their involvement in disease mechanisms and microbial ecology, and their potential in biomedical applications such as vaccine development, targeted drug delivery, cancer therapy, and diagnosis. It highlights the complex interactions of BEVs within bacterial communities and between bacteria and hosts. This review also addresses current advancements, challenges, and opportunities in translating BEV research into clinical practice. The insights presented here position BEVs as a cornerstone in the future of biomedical advancements, advocating for standardized methods in BEV characterization and scalable production techniques.
Collapse
Affiliation(s)
| | - Ashkan Dehghan
- W Booth School of Engineering Practice and Technology Faculty of Engineering, McMaster University Hamilton, ON, Canada, L8S 0A3
| | - Saba Halimi
- Department of Microbial Biotechnology, School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Fatemeh Najafi
- Department of Chemical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802-1503, United States
| | - Ali Nokhostin
- Medical Sciences & Technologies Faculty, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | | | - Iman Akbarzadeh
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia.
| | - Qun Ren
- Laboratory for Biointerfaces, Empa Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland.
| |
Collapse
|
8
|
Li N, Xin H, Deng K. Separation and Characterization of Heterogeneity Among Various Sizes of Outer Membrane Vesicles Derived from the Probiotic Escherichia coli Nissle 1917. MEMBRANES 2025; 15:141. [PMID: 40422751 DOI: 10.3390/membranes15050141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/22/2025] [Accepted: 05/02/2025] [Indexed: 05/28/2025]
Abstract
Outer membrane vesicles (OMVs) are extracellular vesicles secreted by Gram-negative bacteria with diameters of 20-250 nm. OMVs contain various biologically active substances from their parent bacteria, such as proteins, lipids, and nucleic acids. Escherichia coli Nissle 1917 (EcN) is a Gram-negative probiotic that resides in the human intestine. EcN-derived OMVs are pivotal in modulating intestinal immune responses. However, few studies have addressed the heterogeneity of EcN-derived OMVs in terms of size, significantly limiting the research on their clinical applications. Currently, there are a lack of feasible methods for obtaining EcN-derived OMVs of different sizes. To address this knowledge gap, we developed a membrane filtration method to isolate EcN-derived OMVs of varying sizes. In this study, we first used gradient filtration to isolate high-purity EcN-derived OMVs and conducted a proteomic analysis. Subsequently, we used membrane filtration to separate the EcN-derived OMVs by size. We successfully obtained EcN-derived OMVs of three specific sizes: <50 nm, 50-100 nm, and 100-300 nm. We then performed proteomic analyses of these EcN-derived OMVs and compared their protein profiles. Finally, we compared the ability of each EcN-derived OMV type to induce RAW264.7 macrophages to secrete the pro-inflammatory factor interleukin (IL)-1β and the anti-inflammatory factor IL-10. The EcN-derived OMVs contained 646 different proteins overall; those of different sizes contained different protein types. Among them, the EcN-derived OMVs in the <50 nm group contained significantly fewer proteins (262 different types in total) than those in the 50-100 nm (1603 types) and 100-300 nm (1568 types) groups. Furthermore, the <50 nm group had fewer membrane proteins (40) than the 50-100 nm (215) and 100-300 nm (209) groups. We also found that RAW264.7 macrophages secreted different concentrations of IL-1β and IL-10 following co-incubation with the three EcN-derived OMV types. The 50-100 nm EcN-derived OMV group showed a stronger effect in terms of inducing inflammatory cytokine secretion compared to the other two groups. This study provides direct experimental evidence that EcN-derived OMVs of different sizes exhibit heterogeneous properties.
Collapse
Affiliation(s)
- Ning Li
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Province Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Hongbo Xin
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Province Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Keyu Deng
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Province Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| |
Collapse
|
9
|
Li H, He B, Ma N, Liu C, Cai K, Zhang X, Ma X. Quorum sensing of Bifidobacteria: Research and progress. Microbiol Res 2025; 294:128102. [PMID: 39965277 DOI: 10.1016/j.micres.2025.128102] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 01/17/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025]
Abstract
Quorum sensing (QS) is a common method of communication among bacteria. While previous studies have discovered the mechanisms of QS in a variety of pathogenic bacteria, relatively little research has focused on probiotics, such as Bifidobacteria. Recent studies have detected QS signalling molecules in Bifidobacteria, but it remains unclear whether the probiotic properties of Bifidobacteria are mediated by QS. This review aims to provide an overview of the QS system in Bifidobacteria and its role in promoting the secretion of metabolites such as extracellular vesicles and biofilms. The review further examines the inhibition of virulence gene expression by Bifidobacteria QS through the luxS/AI-2 system, as well as its role in promoting host-microbial interactions. Understanding the QS mechanisms of Bifidobacteria can reveal beneficial interactions with hosts, which may facilitate the control of bacterial infections, including therapeutic strategies for intestinal diseases. This knowledge can also help improve gut health, thereby addressing the opportunities and challenges of enhancing the body's nutritional status.
Collapse
Affiliation(s)
- Huahui Li
- College of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Bin He
- College of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Ning Ma
- College of Animal Science and Technology, China Agricultural University, Haidian, Beijing 100193, China
| | - Chunchen Liu
- College of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China
| | - Kun Cai
- College of Animal Science and Technology, China Agricultural University, Haidian, Beijing 100193, China
| | - Xiujun Zhang
- College of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China.
| | - Xi Ma
- College of Public Health, North China University of Science and Technology, Tangshan, Hebei 063210, China; College of Animal Science and Technology, China Agricultural University, Haidian, Beijing 100193, China.
| |
Collapse
|
10
|
Oh SY, Kim DY, Lee KY, Ha DL, Kim TL, Kwon TG, Kim JW, Lee HJ, Choi SY, Hong SH. Streptococcus mutans-derived extracellular vesicles promote skin wound healing via tRNA cargo. J Nanobiotechnology 2025; 23:322. [PMID: 40296033 PMCID: PMC12036164 DOI: 10.1186/s12951-025-03410-1] [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: 01/08/2025] [Accepted: 04/19/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND The human oral cavity harbors a diverse microbiota, including Streptococcus species. Oral mucosal wounds heal rapidly, although the exact cause remains unclear. This study investigates the impact of Streptococcus mutans-derived extracellular vesicles (Sm EVs) on wound healing in both oral mucosal organoids and mouse skin. To explore whether microbial EV RNA cargo influences wound healing, RNA sequences from Sm EVs were identified, and the most abundant sequences were synthesized into oligomers and encapsulated in E. coli EVs (Ec EVs) for further in vivo testing. We assessed the role of Toll-like receptor 3 (TLR3) in the wound healing mechanism in TLR3 knockout (KO) mice. RESULTS Sm EVs significantly enhanced cell proliferation and migration in oral mucosa, with enhanced focal adhesion complex formation. Sm EVs improved wound healing in mouse dorsal skin compared to PBS controls. RNA sequencing revealed that bacterial tRNAs, particularly the tRNA-Met variant (Oligo 1), were the most abundant RNAs in Sm EVs. Ec EVs carrying Oligo 1 produced similar wound healing effects to Sm EVs in mucosal organoids and mouse dorsal skin. However, in TLR3 knockout mice, Oligo 1 did not improve wound healing. CONCLUSIONS This study highlights the role of Sm EVs, particularly their tRNA variants, in promoting skin wound healing through a TLR3-dependent mechanism. These findings suggest that EVs from oral commensal bacteria may offer therapeutic potential for chronic, non-healing skin wounds.
Collapse
Affiliation(s)
- Su Young Oh
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Dong Yeon Kim
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Kah Young Lee
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Dae-Lyong Ha
- Department of Dermatology, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Tae-Lyn Kim
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Tae-Geon Kwon
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Jin-Wook Kim
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Heon-Jin Lee
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea.
| | - So-Young Choi
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University, Daegu, South Korea.
| | - Su-Hyung Hong
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
11
|
Ciobanasu C. Bacterial Extracellular Vesicles and Antimicrobial Peptides: A Synergistic Approach to Overcome Antimicrobial Resistance. Antibiotics (Basel) 2025; 14:414. [PMID: 40298572 PMCID: PMC12024098 DOI: 10.3390/antibiotics14040414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Antimicrobial resistance is already a major global health threat, contributing to nearly 5 million deaths annually. The rise of multidrug-resistant pathogens has made many infections increasingly difficult to treat. This growing threat has driven the search for alternative therapeutic approaches. Among the most promising candidates are bacterial extracellular vesicles (BEVs) and antimicrobial peptides (AMPs), which offer unique mechanisms of action, potential synergistic effects, and the ability to bypass conventional resistance pathways. This review summarizes the current research on synergistic effects of BEVs and AMPs to overcome antimicrobial resistance.
Collapse
Affiliation(s)
- Corina Ciobanasu
- Department of Exact and Natural Sciences, Institute of Interdisciplinary Research, Alexandru I. Cuza University, Bulevardul Carol I, Nr. 11, 700506 Iasi, Romania
| |
Collapse
|
12
|
Dora D, Revisnyei P, Pasic A, Galffy G, Dulka E, Mihucz A, Roskó B, Szincsak S, Iliuk A, Weiss GJ, Lohinai Z. Host and bacterial urine proteomics might predict treatment outcomes for immunotherapy in advanced non-small cell lung cancer patients. Front Immunol 2025; 16:1543817. [PMID: 40297587 PMCID: PMC12035445 DOI: 10.3389/fimmu.2025.1543817] [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: 12/11/2024] [Accepted: 03/05/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Urine samples are non-invasive approaches to study potential circulating biomarkers from the host organism. Specific proteins cross the bloodstream through the intestinal barrier and may also derive from gut microbiota. In this study, we aimed to evaluate the predictive role of the host and bacterial urine extracellular vesicle (EV) proteomes in patients with non-small cell lung cancer (NSCLC) treated with anti-PD1 immunotherapy. Methods We analyzed the urine EV proteome of 33 advanced-stage NSCLC patients treated with anti-PD1 immunotherapy with LC-MS/MS, stratifying patients according to long (>6 months) and short (≤6 months) progression-free survival (PFS). Gut microbial communities on a subcohort of 23 patients were also analyzed with shotgun metagenomics. Internal validation was performed using the Random Forest (RF) machine learning (ML) algorithm. RF was validated with a non-linear Bayesian ML model. Gene enrichment, and pathway analysis of host urine proteins were analyzed using the Reactome and Gene Ontology databases. Results We identified human (n=3513), bacterial (n=2647), fungal (n=19), and viral (n=4) proteins. 186 human proteins showed differential abundance (p<0.05) according to PFS groups, 101 being significantly more abundant in patients with short PFS and n=85 in patients with long PFS. We found several pathways that were significantly enriched in patients with short PFS (vs long PFS). Multivariate Cox regression showed that human urine proteins MPP5, IGKV6-21, NT5E, and KRT27 were strongly associated with long PFS, and LMAN2, NUTF2, NID1, TNC, IGF1, BCR, GPHN, and PPBP showed the strongest association with short PFS. We revealed that an increased bacterial/host protein ratio in the urine is more frequent in patients with long PFS. Increased abundance of E. coli and E. faecalis proteins in the urine positively correlates with their gut metagenomic abundance. RF ML model supported the reliability in predicting PFS for critical human urine proteins (AUC=0.89), accuracy (95%) and Bacterial proteins (AUC=0.74). Conclusion To our knowledge, this is the first study to depict the predictive role of the host and bacterial urine proteome in anti-PD1-treated advanced NSCLC.
Collapse
Affiliation(s)
- David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Peter Revisnyei
- Department of Telecommunications and Media Informatics, Budapest University of Technology and Economics, Budapest, Hungary
- HUN-REN-BME Information Systems Research Group, Budapest, Hungary
| | - Alija Pasic
- Department of Telecommunications and Media Informatics, Budapest University of Technology and Economics, Budapest, Hungary
| | | | - Edit Dulka
- County Hospital of Torokbalint, Torokbalint, Hungary
| | - Anna Mihucz
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Brigitta Roskó
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Sara Szincsak
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| | - Anton Iliuk
- Tymora Analytical Operations, West Lafayette, IN, United States
| | - Glen J. Weiss
- Department of Medicine, UMass Chan Medical School, Worcester, MA, United States
| | - Zoltan Lohinai
- County Hospital of Torokbalint, Torokbalint, Hungary
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| |
Collapse
|
13
|
Preet R, Islam MA, Shim J, Rajendran G, Mitra A, Vishwakarma V, Kutz C, Choudhury S, Pathak H, Dai Q, Sun W, Madan R, Zhong C, Markiewicz MA, Zhang J. Gut commensal Bifidobacterium-derived extracellular vesicles modulate the therapeutic effects of anti-PD-1 in lung cancer. Nat Commun 2025; 16:3500. [PMID: 40221398 PMCID: PMC11993705 DOI: 10.1038/s41467-025-58553-4] [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: 10/22/2023] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Although immunotherapy such as anti-programmed death-1 and its ligand 1 (PD-1/L1) is a standard treatment for advanced non-small cell lung cancer (NSCLC), many patients do not derive benefit directly. Several studies have elucidated new strategies to improve the antitumor immune response through gut microbiota modulation. However, it remains largely debatable regarding how gut microbiota remotely affect lung cancer microenvironment and subsequently modulate immunotherapy response. Here we show that commensal Bifidobacterium-derived extracellular vesicles (Bif.BEVs) can modulate the therapeutic effect of anti-PD-1 therapy in NSCLC. These Bif.BEVs are up-taken by lung cancer cells predominantly via dynamin-dependent endocytosis and upregulate PD-L1 expression through TLR4-NF-κB pathway. They also efficiently penetrate murine intestinal and patient-derived lung cancer organoids. Oral gavage of these Bif.BEVs result in their accumulation in tumors in mice. Using a syngeneic mouse model, Bif.BEVs are found to synergize the anti-tumor effect of anti-PD-1 via modulation of key cytokines, immune response and oncogenic pathways, and increase in tumor-infiltrating CD8+ T cells. Our study therefore identifies a link between Bif.BEVs and the tumor microenvironment, providing an alternative mechanism to explain how gut microbiota can influence immunotherapy response, particularly in tumors located anatomically distant from the gut.
Collapse
Affiliation(s)
- Ranjan Preet
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Md Atiqul Islam
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jiyoung Shim
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Ganeshkumar Rajendran
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Amrita Mitra
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Vikalp Vishwakarma
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Caleb Kutz
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Sonali Choudhury
- Department of Cancer Biology, University of Kansas Comprehensive Cancer Center, Kansas City, KS, 66160, USA
| | - Harsh Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Qun Dai
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Weijing Sun
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Rashna Madan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Cuncong Zhong
- Department of Electrical Engineering and Computer Science, University of Kansas, Lawrence, KS, 66045, USA
| | - Mary A Markiewicz
- Department of Microbiology, Molecular Genetics & Immunology, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Department of Cancer Biology, University of Kansas Comprehensive Cancer Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
14
|
Ahmadishoar S, Mones Saeed S, Salih Mahdi M, Mohammed Taher W, Alwan M, Jasem Jawad M, Khdyair Hamad A, Gandomkar H. The potential use of bacteria and their derivatives as delivery systems for nanoparticles in the treatment of cancer. J Drug Target 2025:1-34. [PMID: 40186857 DOI: 10.1080/1061186x.2025.2489979] [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: 02/04/2025] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Cancer is a leading cause of mortality and morbidity worldwide. Nanomaterials, unique optical, magnetic, and electrical properties at the nanoscale (1-100 nm), have been engineered to improve drug capacity, bioavailability, and specificity in cancer treatment. These advancements address toxicity and lack of selectivity in conventional therapies, enabling precise targeting of cancer cells, the tumour microenvironment, and the immune system. Among emerging approaches, bacterial treatment shows promise due to its natural ability to target cancer and its diverse therapeutic mechanisms, which nanotechnology can further enhance. Bacteria-based drug delivery systems leverage bacteria's adaptability and survival strategies within the human body. Bacterial derivatives, such as bacterial ghosts (BGs), bacterial extracellular vesicles (BEVs), and dietary toxins, are recognised as effective biological nanomaterials capable of carrying nanoparticles (NPs). These systems have attracted increasing attention for their potential in targeted NP delivery for cancer treatment. This study explores the use of various bacteria and their byproducts as NP delivery vehicles, highlighting their potential in treating different types of cancer. By combining the strengths of nanotechnology and bacterial therapy, these innovative approaches aim to revolutionise cancer treatment with improved precision and efficacy.
Collapse
Affiliation(s)
- Shiva Ahmadishoar
- Department of Microbiology, Male.C., Islamic Azad University, Malekan, Iran
| | - Samaa Mones Saeed
- Dental Prosthetics Techniques Department, Health and Medical Techniques College/AlNoor University, Mosul, Iraq
| | | | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | | - Hossein Gandomkar
- Department of Surgical Oncology, Tehran University of Medical Medicine, Tehran, Iran
| |
Collapse
|
15
|
Xing J, Niu T, Yu T, Zou B, Shi C, Wang Y, Fan S, Li M, Bao M, Sun Y, Gao K, Qiu J, Zhang D, Wang N, Jiang Y, Huang H, Cao X, Zeng Y, Wang J, Zhang S, Hu J, Zhang D, Sun W, Yang G, Yang W, Wang C. Faecalibacterium prausnitzii-derived outer membrane vesicles reprogram gut microbiota metabolism to alleviate Porcine Epidemic Diarrhea Virus infection. MICROBIOME 2025; 13:90. [PMID: 40176190 PMCID: PMC11963522 DOI: 10.1186/s40168-025-02078-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/04/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND The Porcine Epidemic Diarrhea Virus (PEDV) is one of the major challenges facing the global pig farming industry, and vaccines and treatments have proven difficult in controlling its spread. Faecalibacterium prausnitzii (F.prausnitzii), a key commensal bacterium in the gut, has been recognized as a promising candidate for next-generation probiotics due to its potential wide-ranging health benefits. A decrease in F.prausnitzii abundance has been associated with certain viral infections, suggesting its potential application in preventing intestinal viral infections. In this study, we utilized a piglet model to examine the potential role of F.prausnitzii in PEDV infections. RESULTS A piglet model of PEDV infection was established and supplemented with F.prausnitzii, revealing that F.prausnitzii mitigated PEDV infection. Further studies found that outer membrane vesicles (OMVs) are the main functional components of F.prausnitzii, and proteomics, untargeted metabolomics, and small RNA-seq were used to analyze the composition of OMVs. Exhaustion of the gut microbiota demonstrated that the function of Fp. OMVs relies on the presence of the gut microbiota. Additionally, metagenomic analysis indicated that Fp. OMVs altered the gut microbiota composition, enhancing the abundance of Faecalibacterium prausnitzii, Prevotellamassilia timonensis, and Limosilactobacillus reuteri. Untargeted metabolomics analysis showed that Fp. OMVs increased phosphatidylcholine (PC) levels, with PC identified as a key metabolite in alleviating PEDV infection. Single-cell sequencing revealed that PC altered the relative abundance of intestinal cells, increased the number of intestinal epithelial cells, and reduced necroptosis in target cells. PC treatment in infected IPEC-J2 and Vero cells alleviated necroptosis and reduced the activation of the RIPK1-RIPK3-MLKL signaling axis, thereby improving PEDV infection. CONCLUSION F.prausnitzii and its OMVs play a critical role in mitigating PEDV infections. These findings provide a promising strategy to ameliorate PEDV infection in piglets. Video Abstract.
Collapse
Affiliation(s)
- JunHong Xing
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - TianMing Niu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Tong Yu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - BoShi Zou
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - ChunWei Shi
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - YingJie Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - ShuHui Fan
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - MingHan Li
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - MeiYing Bao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Yu Sun
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - KuiPeng Gao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - JingJing Qiu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - DongXing Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Nan Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - YanLong Jiang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - HaiBin Huang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Xin Cao
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Yan Zeng
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - JianZhong Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - ShuMin Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - JingTao Hu
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - Di Zhang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - WuSheng Sun
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China
| | - GuiLian Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| | - WenTao Yang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| | - ChunFeng Wang
- College of Veterinary Medicine, Jilin Provincial Engineering Research Center of Animal Probiotics, Jilin Provincial Key Laboratory of Animal Microecology and Healthy Breeding, Engineering Research Center of Microecological Vaccines (Drugs) for Major Animal Diseases, Ministry of Education, Jilin Agricultural University, 2888 Xincheng Street, Changchun, 130118, China.
| |
Collapse
|
16
|
Yun H, Seo JH, Kim YK, Yang J. Examining the bacterial diversity including extracellular vesicles in air and soil: implications for human health. PLoS One 2025; 20:e0320916. [PMID: 40168325 PMCID: PMC11960916 DOI: 10.1371/journal.pone.0320916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
As the significance of human health continues to rise, the microbiome has shifted its focus from microbial composition to the functional roles it plays. In parallel, interest in ultrafine particles associated with clinically important impact has been increasing. Bacterial extracellular vesicles (BEVs), involved in systemic microbiome activity, are nano-sized spherical vesicles (20 - 100 nm in diameter) containing DNA, RNA, proteins, and lipids. They are known to be absorbed into the body potentially through air and soil, circulate in the blood, and directly impact diseases by affecting organs. Therefore, the aim of this study is to examine the biodiversity of bacteria and BEVs and predicted functional pathways. We sampled air and soil samples in Seoul, Korea and analyzed metagenomics based on 16S rRNA sequencing. At the phylum levels, Firmicutes in BEVs from soil and air were significantly higher than in bacteria, and Acidobacteria in both bacteria and BEVs from soil were significantly higher than from air (p < 0.05). The most dominant genera were Pseudomonas in bacteria from air and soil; and Escherichia-Shigella in BEVs from air and soil. In addition, Two-component system (ko02020) and ATP-binding cassette transporters (ko02010) were dominant functional pathways in both air and soil. The most functional pathways and orthologous groups were significantly different between air and soil (p < 0.05). In conclusion, human health can be affected differently depending on type of environment. Future study is necessary to have a better understanding of human health effects from environmental microbiota.
Collapse
Affiliation(s)
- Hyunjun Yun
- The AI Convergence Appliances Research Center, Korea Electronics Technology Institute, Gwangju, Republic of Korea
| | - Ji Hoon Seo
- Department of Environmental Health, Korea University, Seoul, Republic of Korea
| | | | - Jinho Yang
- Department of Occupational Health and Safety, Semyung University, Jecheon-si, Chungcheongbuk-do, Republic of Korea
| |
Collapse
|
17
|
Daghrery A, Araújo IJDS, Marques JF, Alipour M, Ünsal RBK, Chathoth BM, Sivaramakrishnan G, Delgadillo-Barrera S, Chaurasia A. Role of exosomes in dental and craniofacial regeneration - A review. Tissue Cell 2025; 93:102684. [PMID: 39740273 DOI: 10.1016/j.tice.2024.102684] [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: 08/16/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND The treatment of congenital deformities, traumatic injuries, infectious diseases, and tumors in the craniomaxillofacial (CMF) region is complex due to the intricate nature of the tissues involved. Conventional treatments such as bone grafts and cell transplantation face limitations, including the need for multiple surgeries, complications, and safety concerns. OBJECTIVE This paper aims to provide a comprehensive analysis of the role of exosomes (EXOs) in CMF and dental tissue regeneration and to explore their potential applications in regenerative dental medicine. METHODS An extensive review of advancements in tissue engineering, materials sciences, and nanotechnology was conducted to evaluate the development of delivery systems for EXOs-based therapies. The analysis included how EXOs, as nanovesicles released by cells, can be modified to target specific cells or loaded with functional molecules for drug or gene delivery. RESULTS EXOs have emerged as a promising alternative to cell transplant therapy, offering a safer method for cell communication and epigenetic control. EXOs transport important proteins and genetic materials, facilitating intercellular communication and delivering therapeutics effectively. The potential of EXOs in personalized medicine, particularly in diagnosing, customizing treatment, and predicting patient responses, is highlighted. CONCLUSION EXO-mediated therapy holds significant potential for advancing tissue regeneration, offering targeted, personalized treatment options with reduced side effects. However, challenges in purification, production, and standardized protocols need to be addressed before its clinical application can be fully realized.
Collapse
Affiliation(s)
- Arwa Daghrery
- Department of Restorative Dental Sciences, School of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia.
| | | | - Joana Faria Marques
- Faculdade de Medicina Dentária, Universidade de Lisboa, Cidade Universitária, Lisboa 1600-277, Portugal.
| | - Mahdieh Alipour
- Dental and Periodontal Research Center, Faculty of Dentistry, Tabriz University of Medical Sciences, Iran; Departments of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, USA.
| | - Revan Birke Koca Ünsal
- Department of Periodontology, University of Kyrenia, Faculty of Dentistry, Kyrenia, Cyprus.
| | | | | | - Sara Delgadillo-Barrera
- Grupo de Investigacion Básica y Aplicada en Odontología - IBAPO, Facultad de Odontologia, Universidad Nacional de Colombia, Bogotá, Colombia.
| | - Akhilanand Chaurasia
- Department of Oral Medicine and Radiology, Faculty of Dental Sciences. King George's Medical University, Lucknow, India.
| |
Collapse
|
18
|
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] [Download PDF] [Figures] [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
|
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 PMCID: PMC11946770 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
|
Motaung TE, Ratsoma FM, Kunene S, Santana QC, Steenkamp ET, Wingfield BD. Harnessing exogenous membrane vesicles for studying Fusarium circinatum and its biofilm communities. Microb Pathog 2025; 200:107368. [PMID: 39947354 DOI: 10.1016/j.micpath.2025.107368] [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: 01/26/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Extracellular vesicles (EVs) are tiny messengers that convey bioactive molecules from donor to recipient cells, leading to changes in their physiology and function. We investigated the role of EVs in shaping growth and the biofilm biology of the tree pathogen Fusarium circinatum and its interaction with the susceptible host, Pinus patula. Vesicles were collected from fungal planktonic and biofilm cultures and from pine seedling needles and roots. The physical properties of these vesicles were analysed using nanoparticle tracking analysis and transmission electron microscopy, which revealed a diverse range of sizes and shapes, respectively. Furthermore, uptake of vesicles by conidia was conducted. The results demonstrated that F. circinatum EVs significantly but variably affected spore viability during the early phase (2-4 h) although they enhanced fungal biofilm integrity. In contrast, P. patula EVs greatly inhibited hyphal formation and biofilm biomass, but failed to inhibit matrix production in the fungal biofilm. Our results therefore show that conidial germination is essential for late fungal development including hyphal and biofilm formation while matrix production is a counter measure against harsh environmental conditions including the effects of plant-derived EVs.
Collapse
Affiliation(s)
- Thabiso E Motaung
- Department of Biochemistry, Genetics, and Microbiology (BGM), University of Pretoria, Private Bag X20, Hatfield, 0028, South Africa; Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Hatfield, 0083, Pretoria, South Africa.
| | - Francinah M Ratsoma
- Department of Biochemistry, Genetics, and Microbiology (BGM), University of Pretoria, Private Bag X20, Hatfield, 0028, South Africa; Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Hatfield, 0083, Pretoria, South Africa
| | - Sithembile Kunene
- Department of Plant and Soil Sciences (DePSS), University of Pretoria, Hatfield, 0083, Pretoria, South Africa; Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Hatfield, 0083, Pretoria, South Africa
| | - Quentin C Santana
- Department of Biochemistry, Genetics, and Microbiology (BGM), University of Pretoria, Private Bag X20, Hatfield, 0028, South Africa; Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Hatfield, 0083, Pretoria, South Africa; Agricultural Research Council (ARC), Biotechnology Platform, Private Bag X5 Onderstepoort, Pretoria, South Africa
| | - Emma T Steenkamp
- Department of Biochemistry, Genetics, and Microbiology (BGM), University of Pretoria, Private Bag X20, Hatfield, 0028, South Africa; Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Hatfield, 0083, Pretoria, South Africa
| | - Brenda D Wingfield
- Department of Biochemistry, Genetics, and Microbiology (BGM), University of Pretoria, Private Bag X20, Hatfield, 0028, South Africa; Forestry and Agricultural Biotechnology Institute (FABI), University of Pretoria, Hatfield, 0083, Pretoria, South Africa
| |
Collapse
|
21
|
Angeles Flores G, Cusumano G, Venanzoni R, Angelini P. Advancements in Antibacterial Therapy: Feature Papers. Microorganisms 2025; 13:557. [PMID: 40142450 PMCID: PMC11945154 DOI: 10.3390/microorganisms13030557] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 03/28/2025] Open
Abstract
Antimicrobial resistance (AMR) is a growing global health crisis that threatens the efficacy of antibiotics and modern medical interventions. The emergence of multidrug-resistant (MDR) pathogens, exacerbated by the misuse of antibiotics in healthcare and agriculture, underscores the urgent need for innovative solutions. (1) Background: AMR arises from complex interactions between human, animal, and environmental health, further aggravated by the overuse and inadequate regulation of antibiotics. Conventional treatments are increasingly ineffective, necessitating alternative strategies. Emerging approaches, including bacteriophage therapy, antimicrobial peptides (AMPs), nanotechnology, microbial extracellular vesicles (EVs), and CRISPR-based antimicrobials, provide novel mechanisms that complement traditional antibiotics in combating resistant pathogens. (2) Methods: This review critically analyzes advanced antibacterial strategies in conjunction with systemic reforms such as antimicrobial stewardship programs, the One Health framework, and advanced surveillance tools. These methods can enhance resistance detection, guide interventions, and promote sustainable practices. Additionally, economic, logistical, and regulatory challenges impeding their implementation are evaluated. (3) Results: Emerging technologies, such as CRISPR and nanotechnology, exhibit promising potential in targeting resistance mechanisms. However, disparities in resource distribution and regulatory barriers hinder widespread adoption. Public-private partnerships and sustainable agriculture practices are critical to overcoming these obstacles. (4) Conclusions: A holistic and integrated approach is essential for mitigating the impact of AMR. By aligning innovative therapeutic strategies with global health policies, fostering interdisciplinary collaboration, and ensuring equitable resource distribution, we can develop a sustainable response to this 21st-century challenge.
Collapse
Affiliation(s)
- Giancarlo Angeles Flores
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06122 Perugia, Italy; (G.A.F.); (G.C.); (R.V.)
- Centro di Ricerca per l’Innovazione, Digitalizzazione, Valorizzazione e Fruizione del Patrimonio Culturale e Ambientale (CE.D.I.PA.), Piazza San Gabriele dell’Addolorata, 4, 06049 Spoleto, Italy
| | - Gaia Cusumano
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06122 Perugia, Italy; (G.A.F.); (G.C.); (R.V.)
| | - Roberto Venanzoni
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06122 Perugia, Italy; (G.A.F.); (G.C.); (R.V.)
| | - Paola Angelini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06122 Perugia, Italy; (G.A.F.); (G.C.); (R.V.)
- Centro di Ricerca per l’Innovazione, Digitalizzazione, Valorizzazione e Fruizione del Patrimonio Culturale e Ambientale (CE.D.I.PA.), Piazza San Gabriele dell’Addolorata, 4, 06049 Spoleto, Italy
| |
Collapse
|
22
|
Margolis LB, Sadovsky Y. When Extracellular Vesicles Go Viral: A Bird's Eye View. Pathog Immun 2025; 10:140-158. [PMID: 40017586 PMCID: PMC11867185 DOI: 10.20411/pai.v10i1.787] [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: 12/19/2024] [Accepted: 01/22/2025] [Indexed: 03/01/2025] Open
Abstract
The science of extracellular vesicles (EVs) is a rapidly growing field that spans multiple aspects of normal physiology and pathophysiology. EVs play a critical role in most basic biological processes of cell-cell communications under normal conditions and in disease. EVs have "gone viral" not only in terms of research popularity, but also in our realization that they exhibit an elaborate crosstalk with viruses, particularly with the enveloped ones, which are also extracellular vesicles that are released by cells as a part of their virulence cycle yet are replicative. Here, we highlight some of the complexities underlying EV-virus crosstalk and pathways and provide our insights on key challenges from the viewpoint of EV biology.
Collapse
Affiliation(s)
- Leonid B. Margolis
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of OBGYN and Reproductive Sciences, Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
23
|
Ortiz Camargo AR, van Mastrigt O, Gouw JW, Liu Y, Bongers RS, van Bergenhenegouwen J, Knol J, Abee T, Smid EJ. Characterization of Extracellular Vesicles from Streptococcus thermophilus 065 and Their Potential to Modulate the Immune Response. Probiotics Antimicrob Proteins 2025:10.1007/s12602-024-10422-0. [PMID: 39891859 DOI: 10.1007/s12602-024-10422-0] [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] [Accepted: 12/03/2024] [Indexed: 02/03/2025]
Abstract
Bacteria can release membrane-derived nanoparticles made of lipid bilayers, so-called extracellular vesicles (EVs), which can carry diverse cargo and are important for microbe-microbe and microbe-host interactions. Here, we studied the production of EVs by Streptococcus thermophilus 065, the protein composition of the EVs, and how the produced EVs impact the immune response in vitro. Cultures of S. thermophilus grown for 6 h at 40 °C in M17 broth with 2% lactose reached high biomass yields and a high level of EVs quantified by lipophilic fluorescent dye staining. Proteome analysis of the isolated EVs revealed a high abundance of membrane-associated binding proteins of ABC transporters, ribosomal proteins, and glycolytic enzymes. In addition, phage proteins were found to be present in the EVs, which suggests a low-level expression of prophage genes during growth most likely supporting the release of EVs without causing cell lysis. The role of prophage activation was confirmed in an experiment with the addition of mitomycin C resulting in the expression of phage proteins including holin and endolysin causing a drop in culture OD and concomitant EV release. Subsequent in vitro immune assays using non-activated and activated human peripheral blood mononuclear cells (PBMCs) showed immune regulation in both cases upon exposure to S. thermophilus EVs and producer cells. This study shows the capacity of S. thermophilus EVs to act as immune modulators and opens the possibility for their use as postbiotics.
Collapse
Affiliation(s)
| | - Oscar van Mastrigt
- Food Microbiology, Wageningen University & Research, PO Box 17, 6700 AA, Wageningen, The Netherlands
| | - Joost W Gouw
- Danone Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands
| | - Yue Liu
- Food Microbiology, Wageningen University & Research, PO Box 17, 6700 AA, Wageningen, The Netherlands
| | - Roger S Bongers
- Danone Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands
| | | | - Jan Knol
- Danone Research, Uppsalalaan 12, 3584 CT, Utrecht, The Netherlands
- Laboratory of Microbiology, Wageningen University & Research, Wageningen, The Netherlands
| | - Tjakko Abee
- Food Microbiology, Wageningen University & Research, PO Box 17, 6700 AA, Wageningen, The Netherlands
| | - Eddy J Smid
- Food Microbiology, Wageningen University & Research, PO Box 17, 6700 AA, Wageningen, The Netherlands.
| |
Collapse
|
24
|
Watabe N, Subsomwong P, Yamane K, Asano K, Nakane A. Polygonum tinctorium extract suppresses the virulence of methicillin-resistant Staphylococcus aureus by disrupting its extracellular vesicles. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118933. [PMID: 39396717 DOI: 10.1016/j.jep.2024.118933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Methicillin-resistant S. aureus (MRSA) is a significant global health concern, causing both hospital- and community-acquired infections. The extracellular vesicles released by S. aureus (SaEVs) contain essential factors related to the bacterial survival and pathogenicity. Polygonum tinctorium is traditionally used as a natural dye (indigo) and for treating various infectious diseases caused by microorganisms. However, the effect of P. tinctorium extract (Indigo Ex) and its mechanism on SaEVs is unknown. AIM OF THE STUDY We investigated the effect and mechanism of Indigo Ex on SaEVs, which could be used in controlling S. aureus, especially MRSA infection. MATERIALS AND METHODS Indigo Ex was prepared from pesticide-free P. tinctorium, which was dried, powdered, and extracted with d-limonene. SaEVs were isolated and purified from MRSA culture supernatant by step-gradient ultracentrifugation. The effect of Indigo Ex on SaEVs morphology was observed by both transmission and scanning electron microscopy after incubating the Indigo Ex and SaEVs under shaking conditions. The cytotoxicity of Indigo Ex was performed using mouse macrophage cell line, RAW 264.7. In addition, the ability of Indigo Ex-treated SaEVs to stimulate the immune response and cytotoxicity in RAW 264.7 cells were evaluated by ELISA and WST-1 assay, respectively. RESULTS SaEV particles were disrupted when treated with undiluted Indigo Ex in a time-dependent manner. For the cytotoxicity of Indigo Ex on RAW 264.7 cells, over 50% of the cell viability decreased when diluted Indigo Ex 1000-fold and no cytotoxic effect was observed at a 25,000-fold dilution of Indigo Ex. Interestingly, the Indigo Ex-treated SaEVs showed less cytotoxic effect than SaEVs alone. Similarly, SaEVs treated with Indigo Ex reduced stimulation of pro-inflammatory cytokines (TNF-α and IL-6) and anti-inflammatory cytokine (IL-10) in RAW 264.7 cells compared to untreated SaEVs. Our results indicate that Indigo Ex disrupted SaEV particles, resulting in reduced virulence and stimulation of immune response. CONCLUSIONS This study reveals that the low concentration of Indigo Ex can suppresses the virulence of SaEVs without causing cytotoxicity to the host cells. Therefore, Indigo Ex may have the potential to be used to control S. aureus infection.
Collapse
Affiliation(s)
- Naoko Watabe
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.
| | - Phawinee Subsomwong
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan.
| | | | - Krisana Asano
- Department of Microbiology and Immunology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan; Department of Biopolymer and Health Science, Hirosaki University Graduate School of. Medicine, Hirosaki, Aomori, Japan.
| | - Akio Nakane
- Department of Biopolymer and Health Science, Hirosaki University Graduate School of. Medicine, Hirosaki, Aomori, Japan.
| |
Collapse
|
25
|
Almousa S, Kim S, Kumar A, Su Y, Singh S, Mishra S, Fonseca MM, Rather HA, Romero-Sandoval EA, Hsu FC, Singh R, Yadav H, Mishra S, Deep G. Bacterial Nanovesicles as Interkingdom Signaling Moieties Mediating Pain Hypersensitivity. ACS NANO 2025; 19:3210-3225. [PMID: 39818729 DOI: 10.1021/acsnano.4c10529] [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] [Indexed: 01/18/2025]
Abstract
Gut dysbiosis contributes to multiple pathologies, yet the mechanisms of the gut microbiota-mediated influence on systemic and distant responses remain largely elusive. This study aimed to identify the role of nanosized bacterial extracellular vesicles (bEVs) in mediating allodynia, i.e., pain hypersensitivity, in a diet-induced obesity (DIO) gut dysbiosis model. bEVs were enriched from the feces of lean (bEVLean) and DIO (bEVDIO) mice by an approach combining ultracentrifugation and immunoprecipitation and then extensively analyzed for purity and bacterial characteristics. Next, bEVs were injected, either intraplantarly or intravenously, in mice to assess pain sensitivity. Fluorescence-labeled bEVs were injected in mice by enema to assess biodistribution. The effect of bEV on immune cells and inflammation was analyzed by array, immunophenotyping, microscopy, NF-κB activation, and cellular uptake assays. Results showed that bEVDIO administration in wild-type mice replicated the allodynia phenotype observed in DIO mice for both mechanical and thermal stimuli. Importantly, this effect was compromised in TRPA1/TRPV1 double-knockout mice. Biodistribution analyses showed bEV entry into systemic circulation with subsequent localization at distant sites. Multiple analyses revealed that bEVDIO exposure incited systemic inflammation, primarily through modulating the innate immune system. This inflammatory mechanism involved LPS on the bEV surface, activating TLR2- and TLR4-related pathways, as confirmed using TLR2 and TLR4 inhibitors and shaving bEV surface proteins. Interestingly, the enhanced cellular uptake of bEVDIO was contingent on interactions involving LPS and proteins on bEVs and TLR2/TLR4 on monocytes. These findings illuminate the hitherto unexplored role of bEV as pivotal mediators of allodynia and inflammation linked to gut dysbiosis.
Collapse
Affiliation(s)
- Sameh Almousa
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Susy Kim
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Ashish Kumar
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Yixin Su
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Sangeeta Singh
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Shalini Mishra
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Miriam M Fonseca
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Hilal A Rather
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - E Alfonso Romero-Sandoval
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Fang-Chi Hsu
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| | - Rakesh Singh
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332, United States
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, Florida 33612, United States
| | - Santosh Mishra
- Comparative Pain Research and Education Center, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina 27607, United States
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
| | - Gagan Deep
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
- Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, United States
| |
Collapse
|
26
|
Dai K, Liao B, Huang X, Liu Q. Consistency in bacterial extracellular vesicle production: key to their application in human health. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2025; 6:1-20. [PMID: 40206807 PMCID: PMC11977363 DOI: 10.20517/evcna.2024.76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 04/11/2025]
Abstract
Bacterial extracellular vesicles (BEVs) are naturally occurring functional structures that play critical roles in bacterial life processes. These vesicles, commonly known as outer membrane vesicles (OMVs), were first found to be released by Gram-negative bacteria; however, it has since been confirmed that Gram-positive bacteria also secrete BEVs. As research advances, BEVs are increasingly utilized in diverse applications, including vaccine development and drug delivery. Nevertheless, the effective employment of BEVs in these contexts requires the acquisition of vesicles with consistent properties and functions through appropriate culture, isolation, and purification methods. This review examines the advantages and disadvantages of various purification techniques alongside the heterogeneity they may introduce. We utilize the heterogeneity of BEVs as a framework to critically analyze the barriers to their application and the factors influencing their characteristics. Additionally, we constructively propose solutions to enhance the consistency of BEVs, thereby facilitating their further development and application.
Collapse
Affiliation(s)
- Ke Dai
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- First Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- Authors contributed equally
| | - Bo Liao
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- First Clinical Medical College, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
- Authors contributed equally
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Qiong Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi, China
| |
Collapse
|
27
|
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
|
28
|
Dias MKHM, Jayathilaka ET, Jayasinghe JNC, Tennakoon N, Nikapitiya C, Whang I, De Zoysa M. Exploring the Proteomic Landscape and Immunomodulatory Functions of Edwardsiella piscicida Derived Extracellular Vesicles. J Microbiol Biotechnol 2024; 35:e2410001. [PMID: 39849936 PMCID: PMC11813346 DOI: 10.4014/jmb.2410.10001] [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: 10/02/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 01/25/2025]
Abstract
Extracellular vesicles (EVs) have garnered attention in research for their potential as biochemical transporters and immune modulators, crucial for regulating the host immune system. The present study was conducted to isolate and characterize EVs from Gram negative bacteria Edwardsiella piscicida (EpEVs) and investigate their proteomic profile and immune responses. Isolation of EpEVs was carried out using ultracentrifugation method. Transmission electron microscopy results confirmed the spherical shape of EpEVs. The average size and zeta potential were 85.3 ± 1.8 nm and -8.28 ± 0.41 mV, respectively. EpEVs consisted of 1,487 distinct proteins. Subcellular localization analysis revealed that "cell" and "cell part" were the most predominant areas for protein localization. Proteins associated with virulence, along with several chaperones that facilitate protein folding and stability, were also present. No toxicity was detected when EpEVs were treated to fathead minnow (FHM) cells up to 100 μg/ml. Fluorescent-labeled EpEVs showed cellular internalization in FHM cells at 24 h post treatment (hpt). In-vitro gene expression in Raw 264.7 cells showed upregulation of interleukin (Il)6, Il1β, and interferon (Ifn)β with simultaneous upregulation of anti-inflammatory Il10. In vivo, gene expression revealed that except for heat shock protein (hsp)70, all other genes were upregulated suggesting that EpEVs induced the expression of immune-related genes. Western blot analysis showed increased protein levels of tumor necrosis factor (Tnf)α in EpEVs-treated spleen tissue of zebrafish. Our results confirm that EpEVs can be successfully isolated using the ultracentrifugation method. Furthermore, exploring immunomodulatory mechanism of EpEVs is essential for their potential use as novel therapeutics in fish medicine.
Collapse
Affiliation(s)
| | - E.H.T. Thulshan Jayathilaka
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | | | - Nipuna Tennakoon
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Chamilani Nikapitiya
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Ilson Whang
- National Marine Biodiversity Institute of Korea (MABIK), Janghang-eup, Seocheon 33662, Republic of Korea
| | - Mahanama De Zoysa
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea
| |
Collapse
|
29
|
Sharifpour MF, Sikder S, Wong Y, Koifman N, Thomas T, Courtney R, Seymour J, Loukas A. Characterization of Spirulina-derived extracellular vesicles and their potential as a vaccine adjuvant. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70025. [PMID: 39676887 PMCID: PMC11635480 DOI: 10.1002/jex2.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/07/2024] [Accepted: 11/21/2024] [Indexed: 12/17/2024]
Abstract
Spirulina is an edible cyanobacterium that increasingly gaining recognition for it untapped potential in the biomanufacturing of pharmaceuticals. Despite the rapidly accumulating information on extracellular vesicles (EVs) from most other bacteria, nothing is known about Spirulina extracellular vesicles (SPEVs). This study reports the successful isolation, characterization and visualization of SPEVs for the first time and it further investigates the potential therapeutic benefits of SPEVs using a mouse model. SPEVs were isolated using ultracentrifugation and size-exclusion-chromatography. Cryo-Transmission Electron Microscopy revealed pleomorphic outer-membrane-vesicles and outer-inner-membrane-vesicles displaying diverse shapes, sizes and corona densities. To assess short- and long-term immune responses, mice were injected intraperitoneally with SPEVs, which demonstrated a significant increase in neutrophils and M1 macrophages at the injection site, indicating a pro-inflammatory effect induced by SPEVs without clinical signs of toxicity or hypersensitivity. Furthermore, SPEVs demonstrated potent adjuvanticity by enhancing antigen-specific IgG responses in mice by over 100-fold compared to an unadjuvanted model vaccine antigen. Mass-spectrometry identified 54 proteins within SPEVs, including three protein superfamily members linked to the observed pro-inflammatory effects. Our findings highlight the potential of SPEVs as a new class of vaccine adjuvant and warrant additional studies to further characterize the nature of the immune response.
Collapse
Affiliation(s)
| | - Suchandan Sikder
- Australian Institute of Tropical Health and MedicineJames Cook UniversitySmithfieldQueenslandAustralia
| | - Yide Wong
- Australian Institute of Tropical Health and MedicineJames Cook UniversitySmithfieldQueenslandAustralia
| | - Na'ama Koifman
- Centre for Microscopy and MicroanalysisThe University of QueenslandSt LuciaQueenslandAustralia
| | - Tamara Thomas
- Australian Institute of Tropical Health and MedicineJames Cook UniversitySmithfieldQueenslandAustralia
| | - Robert Courtney
- Australian Institute of Tropical Health and MedicineJames Cook UniversitySmithfieldQueenslandAustralia
| | - Jamie Seymour
- Australian Institute of Tropical Health and MedicineJames Cook UniversitySmithfieldQueenslandAustralia
| | - Alex Loukas
- Australian Institute of Tropical Health and MedicineJames Cook UniversitySmithfieldQueenslandAustralia
| |
Collapse
|
30
|
He M, Yin S, Huang X, Li Y, Li B, Gong T, Liu Q. Insights into the regulatory role of bacterial sncRNA and its extracellular delivery via OMVs. Appl Microbiol Biotechnol 2024; 108:29. [PMID: 38159117 DOI: 10.1007/s00253-023-12855-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: 05/23/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 01/03/2024]
Abstract
Small noncoding RNAs (sncRNAs) play important regulatory roles in bacterial physiological processes and host-pathogen interactions. Meanwhile, bacterial outer membrane vesicles (OMVs), as naturally secreted outer membrane structures, play a vital role in the interaction between bacteria and their living environment, including the host environment. However, most current studies focus on the biological functions of sncRNAs in bacteria or hosts, while neglecting the roles and regulatory mechanisms of the OMVs that encapsulate these sncRNAs. Therefore, this review aims to summarize the intracellular regulatory roles of bacterial sncRNAs in promoting pathogen survival by regulating virulence, modulating bacterial drug resistance, and regulating iron metabolism, and their extracellular regulatory function for influencing host immunity through host-pathogen interactions. Additionally, we introduce the key role played by OMVs, which serve as important cargoes in bacterial sncRNA-host interactions. We propose emerging pathways of sncRNA action to further discuss the mode of host-pathogen interactions, highlighting that the inhibition of sncRNA delivery by OMVs may prevent the occurrence of infection to some extent. Hence, this review lays the foundation for future prophylactic treatments against bacterial infections and strategies for addressing bacterial drug resistance. KEY POINTS: •sncRNAs have intracellular and extracellular regulatory functions in bacterial physiological processes and host-pathogen interactions. •OMVs are potential mediators between bacterial sncRNAs and host cells. •OMVs encapsulating sncRNAs have more potential biological functions.
Collapse
Affiliation(s)
- Mengdan He
- Center for Molecular Diagnosis and Precision Medicine, The Department of Clinical Laboratory, Jiangxi Provincial Center for Advanced Diagnostic Technology and Precision Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, 330006, China
| | - Shuanshuan Yin
- Center for Molecular Diagnosis and Precision Medicine, The Department of Clinical Laboratory, Jiangxi Provincial Center for Advanced Diagnostic Technology and Precision Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, 330006, China
| | - Xinlei Huang
- Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Yi Li
- Center for Molecular Diagnosis and Precision Medicine, The Department of Clinical Laboratory, Jiangxi Provincial Center for Advanced Diagnostic Technology and Precision Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, 330006, China
| | - Biaoxian Li
- Center for Molecular Diagnosis and Precision Medicine, The Department of Clinical Laboratory, Jiangxi Provincial Center for Advanced Diagnostic Technology and Precision Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Tian Gong
- Center for Molecular Diagnosis and Precision Medicine, The Department of Clinical Laboratory, Jiangxi Provincial Center for Advanced Diagnostic Technology and Precision Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Qiong Liu
- Center for Molecular Diagnosis and Precision Medicine, The Department of Clinical Laboratory, Jiangxi Provincial Center for Advanced Diagnostic Technology and Precision Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
- Department of Medical Microbiology, School of Medicine, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
31
|
Yang J. Insight into the potential of algorithms using AI technology as in vitro diagnostics utilizing microbial extracellular vesicles. Mol Cell Probes 2024; 78:101992. [PMID: 39580006 DOI: 10.1016/j.mcp.2024.101992] [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: 08/08/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
Recently, the microbiome has been gaining significant attention in the healthcare sector as a next-generation factor. However, there remains a substantial gap in our understanding of the fundamental mechanisms of microbes, particularly regarding the effector microbial products exchanged between the microbiota and the host. Consequently, research on microbial extracellular vesicles (MEVs) has increased. MEVs, which are nano-sized, can circulate throughout the body and penetrate the bloodstream, carrying diverse information. Consequently, they are increasingly being utilized in medical applications. Additionally, AI technologies are being utilized in medicine. The combination of MEVs and AI technology is being explored for the development of algorithm-based in vitro diagnostics (IVD). Therefore, this study aims to review the integration of MEVs and AI technology as diagnostic tools for personalized medicine. This paper reviewed the MEV-based algorithms developed by a variety of human samples and AI technology. Additionally, most of MEV-based diagnostic models showed higher clinical performance. Several important factors are crucial for accurate diagnosis. First, optimizing sample types according to specific diseases is essential. Second, AI technology with higher diagnostic power yields more accurate results. Finally, incorporating additional markers can enhance diagnostic power. However, applying this tool in situ faces several limitations, including method standardization, sample size, and analysis techniques. In the future, we anticipate that research on MEVs will advance our understanding of their role in disease and establish the foundation for precision medicine strategies.
Collapse
Affiliation(s)
- Jinho Yang
- Department of Occupational Health and Safety, Semyung University, Jecheon, Chungcheongbuk-do, Republic of Korea.
| |
Collapse
|
32
|
Wang K, Cunha E Rocha K, Qin H, Zeng Z, Ying W. Host metabolic inflammation fueled by bacterial DNA. Trends Endocrinol Metab 2024:S1043-2760(24)00294-7. [PMID: 39609222 DOI: 10.1016/j.tem.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/29/2024] [Accepted: 11/06/2024] [Indexed: 11/30/2024]
Abstract
Metabolic diseases, characterized by chronic low-grade inflammation, exhibit a compromised gut barrier allowing the translocation of bacteria-derived products to bloodstream and distant metabolic organs. Bacterial DNA can be detected in metabolic tissues during the onset of these diseases, highlighting its role in the development of metabolic diseases. Extracellular vesicles (EVs) are involved in the delivery of bacterial DNA to the local tissues, and its sensing by the host triggers local and system inflammation. Understanding bacterial DNA translocation and its induced inflammation is crucial in deciphering metabolic disease pathways. Here, we delve into the mechanisms dictating the interaction between host physiology and bacterial DNA, focusing on its origin and delivery, host immune responses against it, and its roles in metabolic disorders.
Collapse
Affiliation(s)
- Ke Wang
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Karina Cunha E Rocha
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Houji Qin
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Zixuan Zeng
- School of Biological Sciences, University of California, San Diego, La Jolla, CA, USA
| | - Wei Ying
- Division of Endocrinology and Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
33
|
Tan W, Nian H, Tran LSP, Jin J, Lian T. Small peptides: novel targets for modulating plant-rhizosphere microbe interactions. Trends Microbiol 2024; 32:1072-1083. [PMID: 38670883 DOI: 10.1016/j.tim.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: 12/17/2023] [Revised: 03/24/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024]
Abstract
The crucial role of rhizosphere microbes in plant growth and their resilience to environmental stresses underscores the intricate communication between microbes and plants. Plants are equipped with a diverse set of signaling molecules that facilitate communication across different biological kingdoms, although our comprehension of these mechanisms is still evolving. Small peptides produced by plants (SPPs) and microbes (SPMs) play a pivotal role in intracellular signaling and are essential in orchestrating various plant development stages. In this review, we posit that SPPs and SPMs serve as crucial signaling agents for the bidirectional cross-kingdom communication between plants and rhizosphere microbes. We explore several potential mechanistic pathways through which this communication occurs. Additionally, we propose that leveraging small peptides, inspired by plant-rhizosphere microbe interactions, represents an innovative approach in the field of holobiont engineering.
Collapse
Affiliation(s)
- Weiyi Tan
- The State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, China
| | - Hai Nian
- The State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, China
| | - Lam-Son Phan Tran
- Institute of Genomics for Crop Abiotic Stress Tolerance, Department of Plant and Soil Science, Texas Tech University, Lubbock, TX, USA.
| | - Jing Jin
- The State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, China.
| | - Tengxiang Lian
- The State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, South China Agricultural University, Guangzhou, Guangdong, China.
| |
Collapse
|
34
|
Jayathilaka EHTT, Hasitha Madhawa Dias MK, Tennakoon MSBWTMNS, Chulhong O, Nikapitiya C, Shin HJ, De Zoysa M. Mapping the proteomic landscape and anti-inflammatory role of Streptococcus parauberis extracellular vesicles. FISH & SHELLFISH IMMUNOLOGY 2024; 154:109945. [PMID: 39378979 DOI: 10.1016/j.fsi.2024.109945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/28/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024]
Abstract
Bacterial extracellular vesicles (BEVs) are nanoscale membrane-bound structures involved in intercellular communication and transport of bioactive molecules. In this study, we described the proteomic insight and anti-inflammatory activity of Streptococcus parauberis BEVs (SpEVs). Proteomics analysis of SpEVs identified 6209 distinct peptides and 1039 proteins. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis demonstrated enrichment in pathways related to the biosynthesis of aminoacyl tRNA, amino acids, and secondary metabolites. Based on the predicted protein-protein interactions, we discovered key immunological proteins such as IL12A, IL12B, IL8, CD28, and NF-κB between SpEVs and human proteins. Functionally, SpEVs exhibit strong anti-inflammatory activity in LPS-stimulated Raw 264.7 cells by reducing the production of key inflammatory mediators. These include nitric oxide (NO), reactive oxygen species (ROS), inflammatory cytokines such as TNFα and IL6, as well as inflammation-related proteins like inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). qRT-PCR and immunoblotting results clearly indicate that SpEVs modulate the NF-κB and MAPK pathways to induce anti-inflammatory activity. Furthermore, in vivo experiments with zebrafish larvae demonstrated that SpEVs treatment reduced the NO and ROS production with minimal cell mortality. Finally, we validated the anti-inflammatory activity of SpEVs in vivo by systematically assessing the inhibition of NO production, reduction in ROS generation, prevention of cell death, and modulation of NF-κB and MAPK signaling pathways. In conclusion, SpEVs contain rich in unique proteins that play crucial roles in mediating anti-inflammatory effects.
Collapse
Affiliation(s)
- E H T Thulshan Jayathilaka
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea
| | | | - M S B W T M Nipuna Sudaraka Tennakoon
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Oh Chulhong
- Jeju Bio Research Center, Korea Institute of Ocean Science and Technology, Gujwa-eup, Jeju 2670, Republic of Korea
| | - Chamilani Nikapitiya
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Hyun-Jin Shin
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Mahanama De Zoysa
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon 34134, Republic of Korea.
| |
Collapse
|
35
|
Du X, Zhang M, Wang R, Zeng Z, Zhao W, Fang B, Lan H, Hung W, Gao H. Bifidobacterium lactis-Derived Vesicles Attenuate Hippocampal Neuroinflammation by Targeting IL-33 to Regulate FoxO6/P53 Signaling. Nutrients 2024; 16:3586. [PMID: 39519420 PMCID: PMC11547434 DOI: 10.3390/nu16213586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/01/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Hippocampal Neuroinflammation (HNF) is a critical driver of cognitive impairment. The lipopolysaccharide (LPS) accumulate amyloid beta (Aβ) and lead to HNF. The Bifidobacterium lactis (BL) 99 have anti-inflammatory ability. However, whether BL99-derived microbiota-derived vesicles (MV) could alleviate LPS-induced HNF remains unclear. METHODS To investigate, we used ultrafiltration with ultracentrifuge to extract BL99-derived-MV (BL99-MV). We used hippocampal neuronal HT22 cells (HT22) to establish the LPS-induced HNF model, and explored whether BL99-MV alleviate LPS-induced HNF. RESULTS The confocal microscopy showed that BL99-MV were taken up by HT22 and reduced the oxidative stress (ROS) level. The PCR showed that BL99-MV up-regulate IL-10 level, and down-regulate TNF-α, IL-1β, and IL-6. Transcriptomic analysis revealed 4127 differentially expressed genes, with 2549 genes upregulated and 1578 genes downregulated in the BL99-MV group compared to the LPS group. Compared to the LPS group, BL99-MV decreased FoxO6, IL-33, P53, and NFκB expression, but increased FoxO1 and Bcl2 expression. The WB showed that BL99-MV modulated NFκB, FoxO6, P53, Caspase9, and Caspase3 protein expression by reducing IL-33 expression in HT22. The findings demonstrated IL-33 as a regulator for FoxO6/P53 signaling. CONCLUSIONS Here, we hypothesized that BL99-MV alleviated LPS-induced HNF to promote HT22 survival and synaptic development by regulating FoxO6/P53 signaling by targeting IL-33.
Collapse
Affiliation(s)
- Xiaoyu Du
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (X.D.); (M.Z.)
| | - Ming Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (X.D.); (M.Z.)
| | - Ran Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.W.); (W.Z.); (B.F.)
| | - Zhaozhong Zeng
- National Center of Technology Innovation for Dairy, Hohhot 010110, China; (Z.Z.); (H.L.)
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010100, China
| | - Wen Zhao
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.W.); (W.Z.); (B.F.)
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (R.W.); (W.Z.); (B.F.)
| | - Hanglian Lan
- National Center of Technology Innovation for Dairy, Hohhot 010110, China; (Z.Z.); (H.L.)
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010100, China
| | - Weilian Hung
- National Center of Technology Innovation for Dairy, Hohhot 010110, China; (Z.Z.); (H.L.)
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot 010100, China
| | - Haina Gao
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China; (X.D.); (M.Z.)
| |
Collapse
|
36
|
Cong Z, Li Y, Xie L, Chen Q, Tang M, Thongpon P, Jiao Y, Wu S. Engineered Microrobots for Targeted Delivery of Bacterial Outer Membrane Vesicles (OMV) in Thrombus Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400847. [PMID: 38801399 DOI: 10.1002/smll.202400847] [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: 02/01/2024] [Revised: 05/06/2024] [Indexed: 05/29/2024]
Abstract
In the realm of thrombosis treatment, bioengineered outer membrane vesicles (OMVs) offer a novel and promising approach, as they have rich content of bacterial-derived components. This study centers on OMVs derived from Escherichia coli BL21 cells, innovatively engineered to encapsulate the staphylokinase-hirudin fusion protein (SFH). SFH synergizes the properties of staphylokinase (SAK) and hirudin (HV) to enhance thrombolytic efficiency while reducing the risks associated with re-embolization and bleeding. Building on this foundation, this study introduces two cutting-edge microrobotic platforms: SFH-OMV@H for venous thromboembolism (VTE) treatment, and SFH-OMV@MΦ, designed specifically for cerebral venous sinus thrombosis (CVST) therapy. These platforms have demonstrated significant efficacy in dissolving thrombi, with SFH-OMV@H showcasing precise vascular navigation and SFH-OMV@MΦ effectively targeting cerebral thrombi. The study shows that the integration of these bioengineered OMVs and microrobotic systems marks a significant advancement in thrombosis treatment, underlining their potential to revolutionize personalized medical approaches to complex health conditions.
Collapse
Affiliation(s)
- Zhaoqing Cong
- The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu People's Hospital, Shenzhen, 518000, P. R. China
- South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, 95817, USA
| | - Yangyang Li
- The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu People's Hospital, Shenzhen, 518000, P. R. China
| | - Leiming Xie
- The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu People's Hospital, Shenzhen, 518000, P. R. China
| | - Qiwei Chen
- The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu People's Hospital, Shenzhen, 518000, P. R. China
| | - Menghuan Tang
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, 95817, USA
| | - Phonpilas Thongpon
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, 95817, USA
| | - Yanxiao Jiao
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Sacramento, 95817, USA
| | - Song Wu
- The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu People's Hospital, Shenzhen, 518000, P. R. China
- South China Hospital, Medical School, Shenzhen University, Shenzhen, 518116, P. R. China
| |
Collapse
|
37
|
Alvarado-Ocampo J, Abrahams-Sandí E, Retana-Moreira L. Overview of extracellular vesicles in pathogens with special focus on human extracellular protozoan parasites. Mem Inst Oswaldo Cruz 2024; 119:e240073. [PMID: 39319874 PMCID: PMC11421424 DOI: 10.1590/0074-02760240073] [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: 04/01/2024] [Accepted: 07/09/2024] [Indexed: 09/26/2024] Open
Abstract
Extracellular vesicles (EVs) are lipid-bilayered membrane-delimited particles secreted by almost any cell type, involved in different functions according to the cell of origin and its state. From these, cell to cell communication, pathogen-host interactions and modulation of the immune response have been widely studied. Moreover, these vesicles could be employed for diagnostic and therapeutic purposes, including infections produced by pathogens of diverse types; regarding parasites, the secretion, characterisation, and roles of EVs have been studied in particular cases. Moreover, the heterogeneity of EVs presents challenges at every stage of studies, which motivates research in this area. In this review, we summarise some aspects related to the secretion and roles of EVs from several groups of pathogens, with special focus on the most recent research regarding EVs secreted by extracellular protozoan parasites.
Collapse
Affiliation(s)
- Johan Alvarado-Ocampo
- Universidad de Costa Rica, Facultad de Microbiología, Centro de Investigación en Enfermedades Tropicales, San José, Costa Rica
| | - Elizabeth Abrahams-Sandí
- Universidad de Costa Rica, Facultad de Microbiología, Centro de Investigación en Enfermedades Tropicales, San José, Costa Rica
- Universidad de Costa Rica, Facultad de Microbiología, Departamento de Parasitología, San José, Costa Rica
| | - Lissette Retana-Moreira
- Universidad de Costa Rica, Facultad de Microbiología, Centro de Investigación en Enfermedades Tropicales, San José, Costa Rica
- Universidad de Costa Rica, Facultad de Microbiología, Departamento de Parasitología, San José, Costa Rica
| |
Collapse
|
38
|
Kang M, Kim MJ, Jeong D, Lim HJ, Go GE, Jeong U, Moon E, Kweon HS, Kang NG, Hwang SJ, Youn SH, Hwang BK, Kim D. A nanoscale visual exploration of the pathogenic effects of bacterial extracellular vesicles on host cells. J Nanobiotechnology 2024; 22:548. [PMID: 39238028 PMCID: PMC11378492 DOI: 10.1186/s12951-024-02817-6] [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] [Accepted: 08/30/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Bacterial extracellular vesicles (EVs) are pivotal mediators of intercellular communication and influence host cell biology, thereby contributing to the pathogenesis of infections. Despite their significance, the precise effects of bacterial EVs on the host cells remain poorly understood. This study aimed to elucidate ultrastructural changes in host cells upon infection with EVs derived from a pathogenic bacterium, Staphylococcus aureus (S. aureus). RESULTS Using super-resolution fluorescence microscopy and high-voltage electron microscopy, we investigated the nanoscale alterations in mitochondria, endoplasmic reticulum (ER), Golgi apparatus, lysosomes, and microtubules of skin cells infected with bacterial EVs. Our results revealed significant mitochondrial fission, loss of cristae, transformation of the ER from tubular to sheet-like structures, and fragmentation of the Golgi apparatus in cells infected with S. aureus EVs, in contrast to the negligible effects observed following S. epidermidis EV infection, probably due to the pathogenic factors in S. aureus EV, including protein A and enterotoxin. These findings indicate that bacterial EVs, particularly those from pathogenic strains, induce profound ultrastructural changes of host cells that can disrupt cellular homeostasis and contribute to infection pathogenesis. CONCLUSIONS This study advances the understanding of bacterial EV-host cell interactions and contributes to the development of new diagnostic and therapeutic strategies for bacterial infections.
Collapse
Affiliation(s)
- Minjae Kang
- Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea
| | - Min Jeong Kim
- Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea
| | - Dokyung Jeong
- Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea
| | - Hyung-Jun Lim
- Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea
| | - Ga-Eun Go
- Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea
| | - Uidon Jeong
- Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea
| | - Eunyoung Moon
- Electron Microscopy Research Center, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Hee-Seok Kweon
- Electron Microscopy Research Center, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Nae-Gyu Kang
- R&D Center, LG H&H Co., Ltd, Seoul, 07795, Republic of Korea
| | - Seung Jin Hwang
- R&D Center, LG H&H Co., Ltd, Seoul, 07795, Republic of Korea
| | - Sung Hun Youn
- R&D Center, LG H&H Co., Ltd, Seoul, 07795, Republic of Korea
| | - Bo Kyoung Hwang
- R&D Center, LG H&H Co., Ltd, Seoul, 07795, Republic of Korea
| | - Doory Kim
- Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea.
- Research Institute for Convergence of Basic Science, Hanyang University, Seoul, 04763, Republic of Korea.
- Institute of Nano Science and Technology, Hanyang University, Seoul, 04763, Republic of Korea.
- Research Institute for Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
39
|
Mohammed S, Çon AH. Postbiotic nanoparticles (postbiotics-NPs): a novel strategy for providing probiotics' health advantages through food consumption. Food Sci Biotechnol 2024; 33:2729-2736. [PMID: 39184983 PMCID: PMC11339192 DOI: 10.1007/s10068-024-01629-6] [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: 02/08/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 08/27/2024] Open
Abstract
In recent years, the term "postbiotics" has become increasingly popular in food microbiology, food science, and commercial products. This importance has been raised due to the safety issues associated with live bacterial cells. Postbiotics are defined as bioactive substances of probiotics. It confers unique health-promoting functions with its chemical structure, safe profile, and long shelf life. Meanwhile, postbiotics nanoparticles (postbiotics-NPs) can be considered novel postbiotic delivery systems to deliver bioactive components with health benefits and therapeutic aims, promote the shelf-life of food products, and develop novel functional foods. The present scientific literature shows that nanotechnology approaches are not yet mature enough to be used in postbiotic delivery systems. For all of that, the potential applications of postbiotics-NPs in the food industry and biomedical fields will be a new trend in the future.
Collapse
Affiliation(s)
- Sarhan Mohammed
- Department of Food Engineering, Faculty of Engineering, Ondokuz Mayis University, 55139 Atakum, Samsun Turkey
| | - Ahmet Hilmi Çon
- Department of Food Engineering, Faculty of Engineering, Ondokuz Mayis University, 55139 Atakum, Samsun Turkey
| |
Collapse
|
40
|
Dias MKHM, Jayathilaka EHTT, De Zoysa M. Isolation, characterization, and immunomodulatory effects of extracellular vesicles isolated from fish pathogenic Aeromonas hydrophila. FISH & SHELLFISH IMMUNOLOGY 2024; 152:109787. [PMID: 39047924 DOI: 10.1016/j.fsi.2024.109787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/09/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Bacterial extracellular vesicles (BEVs) are natural nanocarriers that have shown great potential for biomedical applications such as biomarkers, cancer therapy, immunomodulators, vaccines, wound healing, tissue engineering, and drug carriers. In the present study, BEVs were isolated from the gram-negative bacterium, Aeromonas hydrophila using the ultracentrifugation method and denoted as AhEVs. Using transmission electron microscopy imaging, we confirmed the ultrastructure and spherical shape morphology of AhEVs. Nanoparticle-tracking analysis results showed a mean particle size of 105.5 ± 2.0 nm for AhEVs. Moreover, the particle concentration of AhEVs was 2.34 ± 0.12 × 1011 particles/mL of bacterial supernatant. AhEV-treated fathead minnow (FHM) cells did not show cytotoxicity effects up to 50 μg/mL with no significant decrease in cells. Moreover, no mortality was observed in larval zebrafish up to 50 μg/mL which indicates that the AhEVs are biocompatible at this concentration. Furthermore, fluorescent-labeled AhEVs were internalized into FHM cells. Results of qRT-PCR analysis in FHM cells revealed that cellular pro-inflammatory cytokines such as nuclear factor (NF)-κB, interferon (Ifn), Irf7, interleukin (Il) 8, and Il11 were upregulated while downregulating the expression of anti-inflammatory Il10 in a concentration-dependent manner. AhEV-treated adult zebrafish (5 μg/fish) induced toll-like receptor (tlr) 2 and tlr4; tumor necrosis factor-alpha (tnfα); heat shock protein (hsp) 70; and il10, il6, and il1β in kidney. Protein expression of NF-κB p65 and Tnfα presented amplified levels in the spleen of AhEVs-treated zebrafish. Based on the collective findings, we conclude that AhEVs exhibited morphological and physicochemical characteristics to known EVs of gram (-)ve bacteria. At biocompatible concentrations, the immunomodulatory activity of AhEVs was demonstrated by inducing different immune response genes in FHM cells and zebrafish. Hence, we suggest that AhEVs could be a novel vaccine candidate in fish medicine due to their ability to elicit strong immune responses.
Collapse
Affiliation(s)
| | - E H T Thulshan Jayathilaka
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Mahanama De Zoysa
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
41
|
Sandanusova M, Turkova K, Pechackova E, Kotoucek J, Roudnicky P, Sindelar M, Kubala L, Ambrozova G. Growth phase matters: Boosting immunity via Lacticasebacillus-derived membrane vesicles and their interactions with TLR2 pathways. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e169. [PMID: 39185335 PMCID: PMC11341917 DOI: 10.1002/jex2.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/10/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024]
Abstract
Lipid bi-layered particles known as membrane vesicles (MVs), produced by Gram-positive bacteria are a communication tool throughout the entire bacterial growth. However, the MVs characteristics may vary across all stages of maternal culture growth, leading to inconsistencies in MVs research. This, in turn, hinders their employment as nanocarriers, vaccines and other medical applications. In this study, we aimed to comprehensively characterize MVs derived from Lacticaseibacillus rhamnosus CCM7091 isolated at different growth stages: early exponential (6 h, MV6), late exponential (12 h, MV12) and late stationary phase (48 h, MV48). We observed significant differences in protein content between MV6 and MV48 (data are available via ProteomeXchange with identifier PXD041580), likely contributing to their different immunomodulatory capacities. In vitro analysis demonstrated that MV48 uptake rate by epithelial Caco-2 cells is significantly higher and they stimulate an immune response in murine macrophages RAW 264.7 (elevated production of TNFα, IL-6, IL-10, NO). This correlated with increased expression of lipoteichoic acid (LTA) and enhanced TLR2 signalling in MV48, suggesting that LTA contributes to the immunomodulation. In conclusion, we showed that Lacticaseibacillus rhamnosus CCM7091-derived MVs from the late stationary phase boost the immune response the most effectively, which pre-destines them for therapeutical application as nanocarriers.
Collapse
Affiliation(s)
- Miriam Sandanusova
- Faculty of Science, Department of Experimental BiologyMasaryk UniversityBrnoCzech Republic
- Department of Biophysics of Immune SystemInstitute of Biophysics of the Czech Academy of SciencesBrnoCzech Republic
| | - Kristyna Turkova
- Department of Biophysics of Immune SystemInstitute of Biophysics of the Czech Academy of SciencesBrnoCzech Republic
| | - Eva Pechackova
- Faculty of Science, Department of BiochemistryMasaryk UniversityBrnoCzech Republic
| | - Jan Kotoucek
- Department of Pharmacology and ToxicologyVeterinary Research InstituteBrnoCzech Republic
| | - Pavel Roudnicky
- Central European Institute of Technology (CEITEC)Masaryk UniversityBrnoCzech Republic
| | - Martin Sindelar
- Faculty of Science, Department of Experimental BiologyMasaryk UniversityBrnoCzech Republic
| | - Lukas Kubala
- Faculty of Science, Department of Experimental BiologyMasaryk UniversityBrnoCzech Republic
- Department of Biophysics of Immune SystemInstitute of Biophysics of the Czech Academy of SciencesBrnoCzech Republic
| | - Gabriela Ambrozova
- Department of Biophysics of Immune SystemInstitute of Biophysics of the Czech Academy of SciencesBrnoCzech Republic
| |
Collapse
|
42
|
Liu J, Nordin JZ, McLachlan AJ, Chrzanowski W. Extracellular vesicles as the next-generation modulators of pharmacokinetics and pharmacodynamics of medications and their potential as adjuvant therapeutics. Clin Transl Med 2024; 14:e70002. [PMID: 39167024 PMCID: PMC11337541 DOI: 10.1002/ctm2.70002] [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: 05/20/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND AND MAIN BODY Pharmacokinetics (PK) and pharmacodynamics (PD) are central concepts to guide the dosage and administration of drug therapies and are essential to consider for both healthcare professionals and researchers in therapeutic planning and drug discovery. PK/PD properties of a drug significantly influence variability in response to treatment, including therapeutic failure or excessive medication-related harm. Furthermore, suboptimal PK properties constitute a significant barrier to further development for some candidate treatments in drug discovery. This article describes how extracellular vesicles (EVs) affect different aspects of PK and PD of medications and their potential to modulate PK and PD properties to address problematic PK/PD profiles of drugs. We reviewed EVs' intrinsic effects on cell behaviours and medication responses. We also described how surface and cargo modifications can enhance EV functionalities and enable them as adjuvants to optimise the PK/PD profile of conventional medications. Furthermore, we demonstrated that various bioengineering strategies can be used to modify the properties of EVs, hence enhancing their potential to modulate PK and PD profile of medications. CONCLUSION This review uncovers the critical role of EVs in PK and PD modulation and motivates further research and the development of assays to unfold EVs' full potential in solving PK and PD-related problems. However, while we have shown that EVs play a vital role in modulating PK and PD properties of medications, we postulated that it is essential to define the context of use when designing and utilising EVs in pharmaceutical and medical applications. HIGHLIGHTS Existing solutions for pharmacokinetics and pharmacodynamics modulation are limited. Extracellular vesicles can optimise pharmacokinetics as a drug delivery vehicle. Biogenesis and administration of extracellular vesicles can signal cell response. The pharmaceutical potential of extracellular vesicles can be enhanced by surface and cargo bioengineering. When using extracellular vesicles as modulators of pharmacokinetics and pharmacodynamics, the 'context of use' must be considered.
Collapse
Affiliation(s)
- Jiaqi Liu
- Sydney Pharmacy SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
| | - Joel Z. Nordin
- Division of Biomolecular and Cellular MedicineDivision of Clinical ImmunologyDepartment of Laboratory MedicineKarolinska InstituteHuddingeSweden
| | - Andrew J. McLachlan
- Sydney Pharmacy SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
| | - Wojciech Chrzanowski
- Sydney Pharmacy SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
- Division of Biomolecular and Cellular MedicineDivision of Clinical ImmunologyDepartment of Laboratory MedicineKarolinska InstituteHuddingeSweden
- Division of Biomedical EngineeringDepartment of Materials Science and EngineeringUppsala UniversityUppsalaAustralia
| |
Collapse
|
43
|
Han HS, Hwang S, Choi SY, Hitayezu E, Humphrey MA, Enkhbayar A, Song D, Kim M, Park J, Park Y, Park J, Cha KH, Choi KY. Roseburia intestinalis-derived extracellular vesicles ameliorate colitis by modulating intestinal barrier, microbiome, and inflammatory responses. J Extracell Vesicles 2024; 13:e12487. [PMID: 39166405 PMCID: PMC11336657 DOI: 10.1002/jev2.12487] [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: 01/27/2024] [Revised: 05/16/2024] [Accepted: 06/29/2024] [Indexed: 08/22/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disorder characterized by recurrent gastrointestinal inflammation, lacking a precise aetiology and definitive cure. The gut microbiome is vital in preventing and treating IBD due to its various physiological functions. In the interplay between the gut microbiome and human health, extracellular vesicles secreted by gut bacteria (BEVs) are key mediators. Herein, we explore the role of Roseburia intestinalis (R)-derived EVs (R-EVs) as potent anti-inflammatory mediators in treating dextran sulfate sodium-induced colitis. R was selected as an optimal BEV producer for IBD treatment through ANCOM analysis. R-EVs with a 76 nm diameter were isolated from R using a tangential flow filtration system. Orally administered R-EVs effectively accumulated in inflamed colonic tissues and increased the abundance of Bifidobacterium on microbial changes, inhibiting colonic inflammation and prompting intestinal recovery. Due to the presence of Ile-Pro-Ile in the vesicular structure, R-EVs reduced the DPP4 activity in inflamed colonic tissue and increased the active GLP-1, thereby downregulating the NFκB and STAT3 via the PI3K pathway. Our results shed light on the impact of BEVs on intestinal recovery and gut microbiome alteration in treating IBD.
Collapse
Affiliation(s)
- Hwa Seung Han
- Department of Marine Bio‐Food ScienceGangneung‐Wonju National UniversityGangneungRepublic of Korea
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Soonjae Hwang
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | | | - Emmanuel Hitayezu
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Mabwi A. Humphrey
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Altai Enkhbayar
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Dae‐Geun Song
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Myungsuk Kim
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | | | - Young‐Tae Park
- Natural Product Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Jin‐Soo Park
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Ki Young Choi
- Department of Marine Bio‐Food ScienceGangneung‐Wonju National UniversityGangneungRepublic of Korea
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
- NVience Inc.SeoulRepublic of Korea
| |
Collapse
|
44
|
Meidaninikjeh S, Mohammadi P, Elikaei A. Bacteriophages and bacterial extracellular vesicles, threat or opportunity? Life Sci 2024; 350:122749. [PMID: 38821215 DOI: 10.1016/j.lfs.2024.122749] [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: 12/04/2023] [Revised: 03/25/2024] [Accepted: 05/23/2024] [Indexed: 06/02/2024]
Abstract
Emergence of antimicrobial-resistant bacteria (AMR) is one of the health major problems worldwide. The scientists are looking for a novel method to treat infectious diseases. Phage therapy is considered a suitable approach for treating infectious diseases. However, there are different challenges in this way. Some biological aspects can probably influence on therapeutic results and further investigations are necessary to reach a successful phage therapy. Bacteriophage activity can influence by bacterial defense system. Bacterial extracellular vesicles (BEVs) are one of the bacterial defense mechanisms which can modify the results of bacteriophage activity. BEVs have the significant roles in the gene transferring, invasion, escape, and spreading of bacteriophages. In this review, the defense mechanisms of bacteria against bacteriophages, especially BEVs secretion, the hidden linkage of BEVs and bacteriophages, and its possible consequences on the bacteriophage activity as well phage therapy will be discussed.
Collapse
Affiliation(s)
- Sepideh Meidaninikjeh
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran.
| | - Parisa Mohammadi
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran; Research Center for Applied Microbiology and Microbial Biotechnology, Alzahra University, Tehran, Iran.
| | - Ameneh Elikaei
- Department of Microbiology, Faculty of Biological Sciences, Alzahra University, Tehran, Iran; Research Center for Applied Microbiology and Microbial Biotechnology, Alzahra University, Tehran, Iran.
| |
Collapse
|
45
|
Santacroce L, Charitos IA, Colella M, Palmirotta R, Jirillo E. Blood Microbiota and Its Products: Mechanisms of Interference with Host Cells and Clinical Outcomes. Hematol Rep 2024; 16:440-453. [PMID: 39051416 PMCID: PMC11270377 DOI: 10.3390/hematolrep16030043] [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: 04/11/2024] [Revised: 06/01/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
In healthy conditions, blood was considered a sterile environment until the development of new analytical approaches that allowed for the detection of circulating bacterial ribosomal DNA. Currently, debate exists on the origin of the blood microbiota. According to advanced research using dark field microscopy, fluorescent in situ hybridization, flow cytometry, and electron microscopy, so-called microbiota have been detected in the blood. Conversely, others have reported no evidence of a common blood microbiota. Then, it was hypothesized that blood microbiota may derive from distant sites, e.g., the gut or external contamination of blood samples. Alteration of the blood microbiota's equilibrium may lead to dysbiosis and, in certain cases, disease. Cardiovascular, respiratory, hepatic, kidney, neoplastic, and immune diseases have been associated with the presence of Gram-positive and Gram-negative bacteria and/or their products in the blood. For instance, lipopolysaccharides (LPSs) and endotoxins may contribute to tissue damage, fueling chronic inflammation. Blood bacteria can interact with immune cells, especially with monocytes that engulf microorganisms and T lymphocytes via spontaneous binding to their membranes. Moreover, LPSs, extracellular vesicles, and outer membrane vesicles interact with red blood cells and immune cells, reaching distant organs. This review aims to describe the composition of blood microbiota in healthy individuals and those with disease conditions. Furthermore, special emphasis is placed on the interaction of blood microbiota with host cells to better understand disease mechanisms.
Collapse
Affiliation(s)
- Luigi Santacroce
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy (R.P.); (E.J.)
| | - Ioannis Alexandros Charitos
- Istituti Clinici Scientifici Maugeri IRCCS, Pneumology and Respiratory Rehabilitation Unit, Institute of Bari, 70124 Bari, Italy;
| | - Marica Colella
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy (R.P.); (E.J.)
- Doctoral School, eCampus University, 22060 Novedrate, Italy
| | - Raffaele Palmirotta
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy (R.P.); (E.J.)
| | - Emilio Jirillo
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari ‘Aldo Moro’, 70124 Bari, Italy (R.P.); (E.J.)
| |
Collapse
|
46
|
Amabebe E, Kumar A, Tatiparthy M, Kammala AK, Taylor BD, Menon R. Cargo exchange between human and bacterial extracellular vesicles in gestational tissues: a new paradigm in communication and immune development. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:297-328. [PMID: 39698538 PMCID: PMC11648491 DOI: 10.20517/evcna.2024.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 05/25/2024] [Accepted: 06/05/2024] [Indexed: 12/20/2024]
Abstract
Host-bacteria and bacteria-bacteria interactions can be facilitated by extracellular vesicles (EVs) secreted by both human and bacterial cells. Human and bacterial EVs (BEVs) propagate and transfer immunogenic cargos that may elicit immune responses in nearby or distant recipient cells/tissues. Hence, direct colonization of tissues by bacterial cells is not required for immunogenic stimulation. This phenomenon is important in the feto-maternal interface, where optimum tolerance between the mother and fetus is required for a successful pregnancy. Though the intrauterine cavity is widely considered sterile, BEVs from diverse sources have been identified in the placenta and amniotic cavity. These BEVs can be internalized by human cells, which may help them evade host immune surveillance. Though it appears logical, whether bacterial cells internalize human EVs or human EV cargo is yet to be determined. However, the presence of BEVs in placental tissues or amniotic cavity is believed to trigger a low-grade immune response that primes the fetal immune system for ex-utero survival, but is insufficient to disrupt the progression of pregnancy or cause immune intolerance required for adverse pregnancy events. Nevertheless, the exchange of bioactive cargos between human and BEVs, and the mechanical underpinnings and health implications of such interactions, especially during pregnancy, are still understudied. Therefore, while focusing on the feto-maternal interface, we discussed how human cells take up BEVs and whether bacterial cells take up human EVs or their cargo, the exchange of cargos between human and BEVs, host cell (feto-maternal) inflammatory responses to BEV immunogenic stimulation, and associations of these interactions with fetal immune priming and adverse reproductive outcomes such as preeclampsia and preterm birth.
Collapse
Affiliation(s)
| | | | | | | | | | - Ramkumar Menon
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| |
Collapse
|
47
|
Li N, Wu M, Wang L, Tang M, Xin H, Deng K. Efficient Isolation of Outer Membrane Vesicles (OMVs) Secreted by Gram-Negative Bacteria via a Novel Gradient Filtration Method. MEMBRANES 2024; 14:135. [PMID: 38921502 PMCID: PMC11205348 DOI: 10.3390/membranes14060135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024]
Abstract
Bacterial extracellular vesicles (bEVs) secreted by Gram-negative bacteria are referred to as outer membrane vesicles (OMVs) because they originate in the outer membrane. OMVs are membrane-coated vesicles 20-250 nm in size. They contain lipopolysaccharide (LPS), peptidoglycan, proteins, lipids, nucleic acids, and other substances derived from their parent bacteria and participate in the transmission of information to host cells. OMVs have broad prospects in terms of potential application in the fields of adjuvants, vaccines, and drug delivery vehicles. Currently, there remains a lack of efficient and convenient methods to isolate OMVs, which greatly limits OMV-related research. In this study, we developed a fast, convenient, and low-cost gradient filtration method to separate OMVs that can achieve industrial-scale production while maintaining the biological activity of the isolated OMVs. We compared the gradient filtration method with traditional ultracentrifugation to isolate OMVs from probiotic Escherichia coli Nissle 1917 (EcN) bacteria. Then, we used RAW264.7 macrophages as an in vitro model to study the influence on the immune function of EcN-derived OMVs obtained through the gradient filtration method. Our results indicated that EcN-derived OMVs were efficiently isolated using our gradient filtration method. The level of OMV enrichment obtained via our gradient filtration method was about twice as efficient as that achieved through traditional ultracentrifugation. The EcN-derived OMVs enriched through the gradient filtration method were successfully taken up by RAW264.7 macrophages and induced them to secrete pro-inflammatory cytokines such as tumor necrosis factor α (TNF-α) and interleukins (ILs) 6 and 1β, as well as anti-inflammatory cytokine IL-10. Furthermore, EcN-derived OMVs induced more anti-inflammatory response (i.e., IL-10) than pro-inflammatory response (i.e., TNF-α, IL-6, and IL-1β). These results were consistent with those reported in the literature. The related literature reported that EcN-derived OMVs obtained through ultracentrifugation could induce stronger anti-inflammatory responses than pro-inflammatory responses in RAW264.7 macrophages. Our simple and novel separation method may therefore have promising prospects in terms of applications involving the study of OMVs.
Collapse
Affiliation(s)
- Ning Li
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (M.W.); (L.W.); (M.T.); (H.X.)
| | | | | | | | | | - Keyu Deng
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Nanchang University, Nanchang 330031, China; (M.W.); (L.W.); (M.T.); (H.X.)
| |
Collapse
|
48
|
Peregrino ES, Castañeda-Casimiro J, Vázquez-Flores L, Estrada-Parra S, Wong-Baeza C, Serafín-López J, Wong-Baeza I. The Role of Bacterial Extracellular Vesicles in the Immune Response to Pathogens, and Therapeutic Opportunities. Int J Mol Sci 2024; 25:6210. [PMID: 38892397 PMCID: PMC11172497 DOI: 10.3390/ijms25116210] [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: 05/01/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Pathogenic bacteria have several mechanisms to evade the host's immune response and achieve an efficient infection. Bacterial extracellular vesicles (EVs) are a relevant cellular communication mechanism, since they can interact with other bacterial cells and with host cells. In this review, we focus on the EVs produced by some World Health Organization (WHO) priority Gram-negative and Gram-positive pathogenic bacteria; by spore-producing bacteria; by Mycobacterium tuberculosis (a bacteria with a complex cell wall); and by Treponema pallidum (a bacteria without lipopolysaccharide). We describe the classification and the general properties of bacterial EVs, their role during bacterial infections and their effects on the host immune response. Bacterial EVs contain pathogen-associated molecular patterns that activate innate immune receptors, which leads to cytokine production and inflammation, but they also contain antigens that induce the activation of B and T cell responses. Understanding the many effects of bacterial EVs on the host's immune response can yield new insights on the pathogenesis of clinically important infections, but it can also lead to the development of EV-based diagnostic and therapeutic strategies. In addition, since EVs are efficient activators of both the innate and the adaptive immune responses, they constitute a promising platform for vaccine development.
Collapse
Affiliation(s)
- Eliud S. Peregrino
- Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (E.S.P.); (J.C.-C.)
| | - Jessica Castañeda-Casimiro
- Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (E.S.P.); (J.C.-C.)
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (S.E.-P.); (J.S.-L.)
| | - Luis Vázquez-Flores
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (L.V.-F.); (C.W.-B.)
| | - Sergio Estrada-Parra
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (S.E.-P.); (J.S.-L.)
| | - Carlos Wong-Baeza
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (L.V.-F.); (C.W.-B.)
| | - Jeanet Serafín-López
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (S.E.-P.); (J.S.-L.)
| | - Isabel Wong-Baeza
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas (ENCB), Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico; (S.E.-P.); (J.S.-L.)
| |
Collapse
|
49
|
Li Y, Sun J, Wang Q, Su C, Chen X, Ma C, Yang X, Feng C, Shi C. Lysis-Free Isolation and Direct Amplification of Pathogenic Bacterial DNA Using Diatom Frustules. Anal Chem 2024; 96:9113-9121. [PMID: 38771353 DOI: 10.1021/acs.analchem.4c00671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
DNA has been implicated as an important biomarker for the diagnosis of bacterial infections. Herein, we developed a streamlined methodology that uses diatom frustules (DFs) to liberate and capture bacterial DNA and allows direct downstream amplification tests without any lysis, washing, or elution steps. Unlike most conventional DNA isolation methods that rely on cell lysis to release bacterial DNA, DFs can trigger the oxidative stress response of bacterial cells to promote bacterial membrane vesicle formation and DNA release by generating reactive oxygen species in aqueous solutions. Due to the hierarchical porous structure, DFs provided high DNA capture efficiency exceeding 80% over a wide range of DNA amounts from 10 pg to 10 ng, making only 10 μg DFs sufficient for each test. Since laborious liquid handling steps are not required, the entire DNA sample preparation process using DFs can be completed within 3 min. The diagnostic use of this DF-based methodology was illustrated, which showed that the DNA of the pathogenic bacteria in serum samples was isolated by DFs and directly detected using polymerase chain reaction (PCR) at concentrations as low as 102 CFU/mL, outperforming the most used approaches based on solid-phase DNA extraction. Furthermore, most of the bacterial cells were still alive after DNA isolation using DFs, providing the possibility of recycling samples for storage and further diagnosis. The proposed DF-based methodology is anticipated to simplify bacterial infection diagnosis and be broadly applied to various medical diagnoses and biological research.
Collapse
Affiliation(s)
- Yang Li
- Qingdao Nucleic Acid Rapid Testing International Science and Technology Cooperation Base, College of Life Sciences, Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, P. R. China
| | - Jiachen Sun
- Qingdao Nucleic Acid Rapid Testing International Science and Technology Cooperation Base, College of Life Sciences, Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, P. R. China
| | - Qing Wang
- Qingdao Nucleic Acid Rapid Testing International Science and Technology Cooperation Base, College of Life Sciences, Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, P. R. China
| | - Chang Su
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, P. R. China
| | - Xiguang Chen
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, P. R. China
- Sanya Oceanographic Institute, Ocean University of China, Floor 7, Building 1, Yonyou Industrial Park, Yazhou Bay Science & Technology City, Sanya 572025, P. R. China
| | - Cuiping Ma
- Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao Nucleic Acid Rapid Detection Engineering Research Center, Qingdao Key Laboratory of Nucleic Acid Rapid Detection, Sino-UAE International Cooperative Joint Laboratory of Pathogenic Microorganism Rapid Detection, College of Biological Engineering, Qingdao University of Science and Technology, Qingdao 266042, P. R. China
| | - Xuecheng Yang
- Qingdao Nucleic Acid Rapid Testing International Science and Technology Cooperation Base, College of Life Sciences, Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, P. R. China
| | - Chao Feng
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, P. R. China
- Sanya Oceanographic Institute, Ocean University of China, Floor 7, Building 1, Yonyou Industrial Park, Yazhou Bay Science & Technology City, Sanya 572025, P. R. China
| | - Chao Shi
- Qingdao Nucleic Acid Rapid Testing International Science and Technology Cooperation Base, College of Life Sciences, Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, P. R. China
- Qingdao JianMa Gene Technology Co., Ltd., Qingdao 266114, P. R. China
| |
Collapse
|
50
|
Metsäniitty M, Hasnat S, Öhman C, Salo T, Eklund KK, Oscarsson J, Salem A. Extracellular vesicles from Aggregatibacter actinomycetemcomitans exhibit potential antitumorigenic effects in oral cancer: a comparative in vitro study. Arch Microbiol 2024; 206:244. [PMID: 38702412 PMCID: PMC11068833 DOI: 10.1007/s00203-024-03976-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/24/2024] [Indexed: 05/06/2024]
Abstract
Aggregatibacter actinomycetemcomitans is an opportunistic Gram-negative periodontopathogen strongly associated with periodontitis and infective endocarditis. Recent evidence suggests that periodontopathogens can influence the initiation and progression of oral squamous cell carcinoma (OSCC). Herein we aimed to investigate the effect of A. actinomycetemcomitans-derived extracellular vesicles (EVs) on OSCC cell behavior compared with EVs from periodontopathogens known to associate with carcinogenesis. EVs were isolated from: A. actinomycetemcomitans and its mutant strains lacking the cytolethal distending toxin (CDT) or lipopolysaccharide (LPS) O-antigen; Porphyromonas gingivalis; Fusobacterium nucleatum; and Parvimonas micra. The effect of EVs on primary and metastatic OSCC cells was assessed using cell proliferation, apoptosis, migration, invasion, and tubulogenesis assays. A. actinomycetemcomitans-derived EVs reduced the metastatic cancer cell proliferation, invasion, tubulogenesis, and increased apoptosis, mostly in CDT- and LPS O-antigen-dependent manner. EVs from F. nucleatum impaired the metastatic cancer cell proliferation and induced the apoptosis rates in all OSCC cell lines. EVs enhanced cancer cell migration regardless of bacterial species. In sum, this is the first study demonstrating the influence of A. actinomycetemcomitans-derived EVs on oral cancer in comparison with other periodontopathogens. Our findings revealed a potential antitumorigenic effect of these EVs on metastatic OSCC cells, which warrants further in vivo investigations.
Collapse
Affiliation(s)
- Marjut Metsäniitty
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
| | - Shrabon Hasnat
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
| | - Carina Öhman
- Oral Microbiology, Department of Odontology, Umeå University, Umeå, 90187, Sweden
| | - Tuula Salo
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
| | - Kari K Eklund
- Department of Rheumatology, University of Helsinki and Helsinki University Hospital, Helsinki, 00014, Finland
- Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
| | - Jan Oscarsson
- Oral Microbiology, Department of Odontology, Umeå University, Umeå, 90187, Sweden
| | - Abdelhakim Salem
- Department of Oral and Maxillofacial Diseases, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland.
- Translational Immunology Research Program (TRIMM), Research Program Unit (RPU), Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland.
| |
Collapse
|