1
|
Ou J, Li K, Yuan H, Du S, Wang T, Deng Q, Wu H, Zeng W, Cheng K, Nandakumar KS. Staphylococcus aureus vesicles impair cutaneous wound healing through p38 MAPK-MerTK cleavage-mediated inhibition of macrophage efferocytosis. Cell Commun Signal 2025; 23:14. [PMID: 39780180 PMCID: PMC11708000 DOI: 10.1186/s12964-024-01994-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/12/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Staphylococcus aureus, a known contributor to non-healing wounds, releases vesicles (SAVs) that influence the delicate balance of host-pathogen interactions. Efferocytosis, a process by which macrophages clear apoptotic cells, plays a key role in successful wound healing. However, the precise impact of SAVs on wound repair and efferocytosis remains unknown. METHODS Filtration, ultracentrifugation, and iodixanol density gradient centrifugation were used to purify the bacterial vesicles. Transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blot (WB) were used to characterize the vesicles. Macrophage efferocytosis efficiency was assessed using flow cytometry and confocal microscopy, while efferocytosis at wound sites was analyzed through WB, FACS, and TUNEL staining. Hematoxylin and eosin (H&E) staining and wound size measurements were used to evaluate the wound healing process. Phosphorylation of signaling pathways was detected by WB, and efferocytosis receptor expression was measured using RNA sequencing, qPCR, and flow cytometry. siRNA and pathway inhibitors were used to investigate the roles of key receptors and signaling pathways in efferocytosis. RESULTS We identified SAVs at infected wound sites, linking them to delayed healing of wounds. SAVs inhibit efferocytosis by activating the TLR2-MyD88-p38 MAPK signaling pathway, which regulates efferocytosis receptor genes. This activation promoted cleavage and shedding of MerTK, a crucial receptor for macrophage-driven efferocytosis. Notably, selective inhibition of p38 MAPK prevented MerTK shedding, restored efferocytosis and accelerated wound healing significantly, offering a promising therapeutic approach for chronic, non-healing wounds. CONCLUSION These findings uncover a novel mechanism in S. aureus-infected wounds, highlighting how the disruption of efferocytosis via the TLR2-MyD88-p38 MAPK-MerTK axis becomes a key force behind impaired healing of wounds. Targeting this pathway could open up a new therapeutic avenue facilitating the treatment of chronic, non-healing skin injuries.
Collapse
Affiliation(s)
- Jiaxin Ou
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
- Center for Cancer Immunology, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Kangxin Li
- Henan International Joint Laboratory of Infection and Immunity, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450001, China.
- Department of Respiratory and Critical Care Medicine, the Tenth Affiliated Hospital (Dongguan Peoples Hospital), Southern Medical University, Dongguan, 523059, China.
- Department of Endocrinology, the Fifth Affiliated Hospital of Southern Medical University, Guangzhou, 510030, China.
| | - Hui Yuan
- Henan International Joint Laboratory of Infection and Immunity, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, 450001, China
| | - Shaohua Du
- Department of Musculoskeletal Oncology, the Third Affiliated Hospital of Southern Medical University, Guangzhou, 510642, China
| | - Tingting Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Qiannan Deng
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou, 510075, China
| | - Huimei Wu
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Weiyan Zeng
- Department of Pharmacy, Sun Yat-Sen University Cancer Center, Guangzhou, 510030, China
| | - Kui Cheng
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Kutty Selva Nandakumar
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
2
|
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
|
3
|
Hu W, Chen S, Zou X, Chen Y, Luo J, Zhong P, Ma D. Oral microbiome, periodontal disease and systemic bone-related diseases in the era of homeostatic medicine. J Adv Res 2024:S2090-1232(24)00362-X. [PMID: 39159722 DOI: 10.1016/j.jare.2024.08.019] [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: 03/18/2024] [Revised: 08/03/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND Homeostasis is a state of self-regulation and dynamic equilibrium, maintaining the good physiological functions of each system in living organisms. In the oral cavity, the interaction between the host and the oral microbiome forms oral microbial homeostasis. Physiological bone remodeling and renewal can occur under the maintenance of oral microbial homeostasis. The imbalance of bone homeostasis is a key mechanism leading to the occurrence of systemic bone-related diseases. Considering the importance of oral microbial homeostasis in the maintenance of bone homeostasis, it still lacks a complete understanding of the relationship between oral microbiome, periodontal disease and systemic bone-related diseases. AIM OF REVIEW This review focuses on the homeostatic changes, pathogenic routes and potential mechanisms in the oral microbiome in periodontal disease and systemic bone-related diseases such as rheumatoid arthritis, osteoarthritis, osteoporosis and osteomyelitis. Additionally, this review discusses oral microbiome-based diagnostic approaches and explores probiotics, mesenchymal stem cells, and oral microbiome transplantation as promising treatment strategies. KEY SCIENTIFIC CONCEPTS OF REVIEW This review highlights the association between oral microbial homeostasis imbalance and systemic bone-related diseases, and highlights the possibility of remodeling oral microbial homeostasis for the prevention and treatment of systemic bone-related diseases.
Collapse
Affiliation(s)
- Weiqi Hu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Shuoling Chen
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Xianghui Zou
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Yan Chen
- Department of Pediatric Dentistry, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Jiayu Luo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Peiliang Zhong
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China
| | - Dandan Ma
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, No 366 Jiangnan Avenue South, Guangzhou, Guangdong Province 510280, China.
| |
Collapse
|
4
|
Zhang Y, Chen S, Qin X, Guo A, Li K, Chen L, Yi W, Deng Z, Tay FR, Geng W, Miao L, Jiao Y, Tao B. A Versatile Chitosan-Based Hydrogel Accelerates Infected Wound Healing via Bacterial Elimination, Antioxidation, Immunoregulation, and Angiogenesis. Adv Healthc Mater 2024; 13:e2400318. [PMID: 38408212 DOI: 10.1002/adhm.202400318] [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: 01/26/2024] [Indexed: 02/28/2024]
Abstract
Drug-resistant bacterial infection of cutaneous wounds causes great harm to the human body. These infections are characterized by a microenvironment with recalcitrant bacterial infections, persistent oxidative stress, imbalance of immune regulation, and suboptimal angiogenesis. Treatment strategies available to date are incapable of handling the healing dynamics of infected wounds. A Schiff base and borate ester cross-linked hydrogel, based on phenylboronic acid-grafted chitosan (CS-PBA), dibenzaldehyde-grafted poly(ethylene glycol), and tannic acid (TA), is fabricated in the present study. Customized phenylboronic acid-modified zinc oxide nanoparticles (ZnO) are embedded in the hydrogel prior to gelation. The CPP@ZnO-P-TA hydrogel effectively eliminates methicillin-resistant Staphylococcus aureus (MRSA) due to the pH-responsive release of Zn2+ and TA. Killing is achieved via membrane damage, adenosine triphosphate reduction, leakage of intracellular components, and hydrolysis of bacterial o-nitrophenyl-β-d-galactopyranoside. The CPP@ZnO-P-TA hydrogel is capable of scavenging reactive oxygen and nitrogen species, alleviating oxidative stress, and stimulating M2 polarization of macrophages. The released Zn2+ and TA also induce neovascularization via the PI3K/Akt pathway. The CPP@ZnO-P-TA hydrogel improves tissue regeneration in vivo by alleviating inflammatory responses, stimulating angiogenesis, and facilitating collagen deposition. These findings suggest that this versatile hydrogel possesses therapeutic potential for the treatment of MRSA-infected cutaneous wounds.
Collapse
Affiliation(s)
- Ye Zhang
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Sinan Chen
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Xian Qin
- Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Ai Guo
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Kai Li
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Lixue Chen
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Weiwei Yi
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Zhongliang Deng
- Department of OrthopediCP, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA, 30912, USA
| | - Wenbo Geng
- Chongqing Key Laboratory of Ophthalmology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Li Miao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, P. R. China
| | - Yang Jiao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, P. R. China
| | - Bailong Tao
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| |
Collapse
|
5
|
Lin Y, Wang Z, Liu S, Liu J, Zhang Z, Ouyang Y, Su Z, Chen D, Guo L, Luo T. Roles of extracellular vesicles on macrophages in inflammatory bone diseases. Mol Cell Biochem 2024; 479:1401-1414. [PMID: 37436653 DOI: 10.1007/s11010-023-04809-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 07/02/2023] [Indexed: 07/13/2023]
Abstract
Inflammatory bone disease is a general term for a series of diseases caused by chronic inflammation, which leads to the destruction of bone homeostasis, that is, the osteolytic activity of osteoclasts increases, and the osteogenic activity of osteoblasts decreases, leading to osteolysis. Macrophages are innate immune cell with plasticity, and their polarization is related to inflammatory bone diseases. The dynamic balance of macrophages between the M1 phenotype and the M2 phenotype affects the occurrence and development of diseases. In recent years, an increasing number of studies have shown that extracellular vesicles existing in the extracellular environment can act on macrophages, affecting the progress of inflammatory diseases. This process is realized by influencing the physiological activity or functional activity of macrophages, inducing macrophages to secrete cytokines, and playing an anti-inflammatory or pro-inflammatory role. In addition, by modifying and editing extracellular vesicles, the potential of targeting macrophages can be used to provide new ideas for developing new drug carriers for inflammatory bone diseases.
Collapse
Affiliation(s)
- Yifan Lin
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ziyan Wang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shirong Liu
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiaohong Liu
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhiyi Zhang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuanting Ouyang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhikang Su
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ding Chen
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lvhua Guo
- Guangzhou Medical University, Guangzhou, Guangdong, China.
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Tao Luo
- Guangzhou Medical University, Guangzhou, Guangdong, China.
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
6
|
Dai C, Xu Q, Li L, Liu Y, Qu S. Milk Extracellular Vesicles: Natural Nanoparticles for Enhancing Oral Drug Delivery against Bacterial Infections. ACS Biomater Sci Eng 2024; 10:1988-2000. [PMID: 38529792 DOI: 10.1021/acsbiomaterials.3c01824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Oral drug delivery is typically preferred as a therapeutic intervention due to the complexities and expenses associated with intravenous administration. However, some drugs are poorly absorbed orally, requiring intravenous administration to bypass the gastrointestinal tract and deliver the drug directly into the bloodstream. Thus, there is an urgent need to develop novel drug delivery platforms to overcome the challenges of oral drug delivery with low solubility, low permeability, oral degradation, and low bioavailability. Advances in extracellular vesicles (EVs) as natural carriers have provided emerging approaches to improve potential therapeutic applications. Milk not only contains traditional nutrients but is also rich in EVs. In this Review, we focus mainly on the purification of milk EVs (mEVs), their safety, and the advantages of mEV-based drug carriers in combatting intestinal infections. Additionally, we summarize several advantages of mEVs over conventional synthetic carriers, such as low immunogenicity, high biocompatibility, and the ability to transfer bioactive molecules between cells. Considering the unmet gaps of mEVs in clinical translation, it is essential to review the cargo loading into mEVs and future perspectives for their use as natural drug carriers for oral delivery. This overview of mEV-based drug carriers for oral delivery sheds light on alternative approaches to treat clinical infections associated with intestinal pathogens and the development of novel oral delivery systems.
Collapse
Affiliation(s)
- Cunchun Dai
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Qingjun Xu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Lin Li
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Ying Liu
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing 100097, China
| | - Shaoqi Qu
- Animal-Derived Food Safety Innovation Team, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
7
|
Lu G, Zhao G, Wang S, Li H, Yu Q, Sun Q, Wang B, Wei L, Fu Z, Zhao Z, Yang L, Deng L, Zheng X, Cai M, Lu M. Injectable Nano-Micro Composites with Anti-bacterial and Osteogenic Capabilities for Minimally Invasive Treatment of Osteomyelitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306964. [PMID: 38234236 DOI: 10.1002/advs.202306964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/17/2023] [Indexed: 01/19/2024]
Abstract
The effective management of osteomyelitis remains extremely challenging due to the difficulty associated with treating bone defects, the high probability of recurrence, the requirement of secondary surgery or multiple surgeries, and the difficulty in eradicating infections caused by methicillin-resistant Staphylococcus aureus (MRSA). Hence, smart biodegradable biomaterials that provide effective and precise local anti-infection effects and can promote the repair of bone defects are actively being developed. Here, a novel nano-micro composite is fabricated by combining calcium phosphate (CaP) nanosheets with drug-loaded GelMA microspheres via microfluidic technology. The microspheres are covalently linked with vancomycin (Van) through an oligonucleotide (oligo) linker using an EDC/NHS carboxyl activator. Accordingly, a smart nano-micro composite called "CaP@MS-Oligo-Van" is synthesized. The porous CaP@MS-Oligo-Van composites can target and capture bacteria. They can also release Van in response to the presence of bacterial micrococcal nuclease and Ca2+, exerting additional antibacterial effects and inhibiting the inflammatory response. Finally, the released CaP nanosheets can promote bone tissue repair. Overall, the findings show that a rapid, targeted drug release system based on CaP@MS-Oligo-Van can effectively target bone tissue infections. Hence, this agent holds potential in the clinical treatment of osteomyelitis caused by MRSA.
Collapse
Affiliation(s)
- Guanghua Lu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Gang Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Shen Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Hanqing Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Qiang Yu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Qi Sun
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Bo Wang
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Li Wei
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Zi Fu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Zhenyu Zhao
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Linshan Yang
- Taikang Bybo Dental, Shanghai, 200001, P. R. China
| | - Lianfu Deng
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| | - Xianyou Zheng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ming Cai
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Min Lu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, P. R. China
| |
Collapse
|
8
|
Liu BD, Akbar R, Oliverio A, Thapa K, Wang X, Fan GC. BACTERIAL EXTRACELLULAR VESICLES IN THE REGULATION OF INFLAMMATORY RESPONSE AND HOST-MICROBE INTERACTIONS. Shock 2024; 61:175-188. [PMID: 37878470 PMCID: PMC10921997 DOI: 10.1097/shk.0000000000002252] [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] [Indexed: 10/27/2023]
Abstract
ABSTRACT Extracellular vesicles (EVs) are a new revelation in cross-kingdom communication, with increasing evidence showing the diverse roles of bacterial EVs (BEVs) in mammalian cells and host-microbe interactions. Bacterial EVs include outer membrane vesicles released by gram-negative bacteria and membrane vesicles generated from gram-positive bacteria. Recently, BEVs have drawn attention for their potential as biomarkers and therapeutic tools because they are nano-sized and can deliver bacterial cargo into host cells. Importantly, exposure to BEVs significantly affects various physiological and pathological responses in mammalian cells. Herein, we provide a comprehensive overview of the various effects of BEVs on host cells (i.e., immune cells, endothelial cells, and epithelial cells) and inflammatory/infectious diseases. First, the biogenesis and purification methods of BEVs are summarized. Next, the mechanisms and pathways identified by BEVs that stimulate either proinflammatory or anti-inflammatory responses are highlighted. In addition, we discuss the mechanisms by which BEVs regulate host-microbe interactions and their effects on the immune system. Finally, this review focuses on the contribution of BEVs to the pathogenesis of sepsis/septic shock and their therapeutic potential for the treatment of sepsis.
Collapse
Affiliation(s)
- Benjamin D. Liu
- Department of Chemistry and Biochemistry, The Ohio State University College of Arts and Sciences, Columbus, OH, 43210, USA
| | - Rubab Akbar
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Anna Oliverio
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Kajol Thapa
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Xiaohong Wang
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| |
Collapse
|
9
|
Xie J, Haesebrouck F, Van Hoecke L, Vandenbroucke RE. Bacterial extracellular vesicles: an emerging avenue to tackle diseases. Trends Microbiol 2023; 31:1206-1224. [PMID: 37330381 DOI: 10.1016/j.tim.2023.05.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 05/20/2023] [Accepted: 05/22/2023] [Indexed: 06/19/2023]
Abstract
A growing body of research, especially in recent years, has shown that bacterial extracellular vesicles (bEVs) are one of the key underlying mechanisms behind the pathogenesis of various diseases like pulmonary fibrosis, sepsis, systemic bone loss, and Alzheimer's disease. Given these new insights, bEVs are proposed as an emerging vehicle that can be used as a diagnostic tool or to tackle diseases when used as a therapeutic target. To further boost the understanding of bEVs in health and disease we thoroughly discuss the contribution of bEVs in disease pathogenesis and the underlying mechanisms. In addition, we speculate on their potential as novel diagnostic biomarkers and how bEV-related mechanisms can be exploited as therapeutic targets.
Collapse
Affiliation(s)
- Junhua Xie
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium; Department of Pathobiology, Pharmacology, and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Freddy Haesebrouck
- Department of Pathobiology, Pharmacology, and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, 9820 Merelbeke, Belgium
| | - Lien Van Hoecke
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Roosmarijn E Vandenbroucke
- VIB Center for Inflammation Research, VIB, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
10
|
Zhang S, Yang H, Wang M, Mantovani D, Yang K, Witte F, Tan L, Yue B, Qu X. Immunomodulatory biomaterials against bacterial infections: Progress, challenges, and future perspectives. Innovation (N Y) 2023; 4:100503. [PMID: 37732016 PMCID: PMC10507240 DOI: 10.1016/j.xinn.2023.100503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/24/2023] [Indexed: 09/22/2023] Open
Abstract
Bacterial infectious diseases are one of the leading causes of death worldwide. Even with the use of multiple antibiotic treatment strategies, 4.95 million people died from drug-resistant bacterial infections in 2019. By 2050, the number of deaths will reach 10 million annually. The increasing mortality may be partly due to bacterial heterogeneity in the infection microenvironment, such as drug-resistant bacteria, biofilms, persister cells, intracellular bacteria, and small colony variants. In addition, the complexity of the immune microenvironment at different stages of infection makes biomaterials with direct antimicrobial activity unsatisfactory for the long-term treatment of chronic bacterial infections. The increasing mortality may be partly attributed to the biomaterials failing to modulate the active antimicrobial action of immune cells. Therefore, there is an urgent need for effective alternatives to treat bacterial infections. Accordingly, the development of immunomodulatory antimicrobial biomaterials has recently received considerable interest; however, a comprehensive review of their research progress is lacking. In this review, we focus mainly on the research progress and future perspectives of immunomodulatory antimicrobial biomaterials used at different stages of infection. First, we describe the characteristics of the immune microenvironment in the acute and chronic phases of bacterial infections. Then, we highlight the immunomodulatory strategies for antimicrobial biomaterials at different stages of infection and their corresponding advantages and disadvantages. Moreover, we discuss biomaterial-mediated bacterial vaccines' potential applications and challenges for activating innate and adaptive immune memory. This review will serve as a reference for future studies to develop next-generation immunomodulatory biomaterials and accelerate their translation into clinical practice.
Collapse
Affiliation(s)
- Shutao Zhang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Hongtao Yang
- School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Minqi Wang
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Department of Min-Met-Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Ke Yang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Frank Witte
- Department of Prosthodontics, Geriatric Dentistry and Craniomandibular Disorders, Charite Medical University, Assmannshauser Strasse 4–6, 14197 Berlin, Germany
| | - Lili Tan
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Bing Yue
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| | - Xinhua Qu
- Department of Bone and Joint Surgery, Department of Orthopedics, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200001, China
| |
Collapse
|
11
|
Chen J, Lv Y, Shang W, Yang Y, Wang Y, Hu Z, Huang X, Zhang R, Yuan J, Huang J, Rao X. Loaded delta-hemolysin shapes the properties of Staphylococcus aureus membrane vesicles. Front Microbiol 2023; 14:1254367. [PMID: 37869662 PMCID: PMC10588482 DOI: 10.3389/fmicb.2023.1254367] [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: 07/07/2023] [Accepted: 09/05/2023] [Indexed: 10/24/2023] Open
Abstract
Background Membrane vesicles (MVs) are nanoscale vesicular structures produced by bacteria during their growth in vitro and in vivo. Some bacterial components can be loaded in bacterial MVs, but the roles of the loaded MV molecules are unclear. Methods MVs of Staphylococcus aureus RN4220 and its derivatives were prepared. Dynamic light scattering analysis was used to evaluate the size distribution, and 4D-label-free liquid chromatography-tandem mass spectrometry analysis was performed to detect protein composition in the MVs. The site-mutation S. aureus RN4220-Δhld and agrA deletion mutant RN4220-ΔagrA were generated via allelic replacement strategies. A hemolysis assay was performed with rabbit red blood cells. CCK-8 and lactate dehydrogenase release assays were used to determine the cytotoxicity of S. aureus MVs against RAW264.7 macrophages. The serum levels of inflammatory factors such as IL-6, IL-1β, and TNFα in mice treated with S. aureus MVs were detected with an enzyme-linked immunosorbent assay kit. Results Delta-hemolysin (Hld) was identified as a major loaded factor in S. aureus MVs. Further study showed that Hld could promote the production of staphylococcal MVs with smaller sizes. Loaded Hld affected the diversity of loaded proteins in MVs of S. aureus RN4220. Hld resulted in decreased protein diversity in MVs of S. aureus. Site-mutation (RN4220-Δhld) and agrA deletion (RN4220-ΔagrA) mutants produced MVs (ΔhldMVs and ΔagrAMVs) with a greater number of bacterial proteins than those derived from wild-type RN4220 (wtMVs). Moreover, Hld contributed to the hemolytic activity of wtMVs. Hld-loaded wtMVs were cytotoxic to macrophage RAW264.7 cells and could stimulate the production of inflammatory factor IL-6 in vivo. Conclusion This study presented that Hld was a major loaded factor in S. aureus MVs, and the loaded Hld played vital roles in the MV-property modification.
Collapse
Affiliation(s)
- Juan Chen
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Yuhuan Lv
- Department of Microbiology, College of Basic Medical Science, Army Medical University, Chongqing, China
- Department of Clinical Laboratory, The 971st Hospital of Chinese People's Liberation Army Navy, Qingdao, China
| | - Weilong Shang
- Department of Microbiology, College of Basic Medical Science, Army Medical University, Chongqing, China
| | - Yi Yang
- Department of Microbiology, College of Basic Medical Science, Army Medical University, Chongqing, China
| | - Yuting Wang
- Department of Microbiology, College of Basic Medical Science, Army Medical University, Chongqing, China
| | - Zhen Hu
- Department of Microbiology, College of Basic Medical Science, Army Medical University, Chongqing, China
| | - Xiaonan Huang
- Department of Microbiology, College of Basic Medical Science, Army Medical University, Chongqing, China
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jizhen Yuan
- Department of Clinical Laboratory, The 971st Hospital of Chinese People's Liberation Army Navy, Qingdao, China
| | - Jingbin Huang
- Department of Pharmacy, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Xiancai Rao
- Department of Microbiology, College of Basic Medical Science, Army Medical University, Chongqing, China
| |
Collapse
|
12
|
Wang X, Li H, Wang J, Xu H, Xue K, Liu X, Zhang Z, Liu J, Liu Y. Staphylococcus aureus extracellular vesicles induce apoptosis and restrain mitophagy-mediated degradation of damaged mitochondria. Microbiol Res 2023; 273:127421. [PMID: 37267814 DOI: 10.1016/j.micres.2023.127421] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/04/2023]
Abstract
Extracellular vesicles (EVs) are nano-sized bilayer EVs with various components. EV secretion in pathogenic Gram-positive bacteria is a universal feature that can cause disease and damage the targeted host. In this study, we isolated and purified Staphylococcus aureus (S. aureus) EVs, and liquid chromatography-tandem mass spectrometry (LC-MS/MS) analyzed Ev's protein composition. After that, the pathway of EVs internalized into MAC-T cells was evaluated. Moreover, the activation of mitogen-activated protein kinase (MAPK) and the nuclear factor κB (NF-κB) pathway was measured by Western blot. Meanwhile, Western blot and confocal microscopy detected mitochondrial damage, apoptosis, and Parkin-mediated mitophagy. Results showed that purified S. aureus EVs exhibited a typical cup-shaped structure and internalized into MAC-T cells by lipid raft-mediated endocytic pathway. S. aureus EVs caused mitochondrial damage and apoptosis in MAC-T cells. However, degradation of the damaged mitochondria was impeded due to the Parkin-mediated mitophagy pathway being restrained by the disruption of the acidic environment of lysosomes by S. aureus EVs. Hence, our study reveals the role of S. aureus EVs in immune stimulation, disruption of mitochondria, and lysosomal acidic environment in bovine mammary epithelial cells. These findings help us understand the role of EVs in the pathogenic mechanism of S. aureus.
Collapse
Affiliation(s)
- Xiaozhou Wang
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an 271018, China
| | - Hongyan Li
- The Affiliated Taian City Central Hospital of Qingdao University, Tai`an 271000, China
| | - Jie Wang
- Xinjiang Key Laboratory of Animal Infectious Diseases, Institute of Veterinary Medicine, Xinjiang Academy of Animal Science, Urumqi 830013, China
| | - Huiling Xu
- Research Center for Animal Disease Control Engineering, Shandong Agricultural University, Tai`an 271018, China
| | - Kun Xue
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an 271018, China
| | - Xiaoting Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an 271018, China
| | - Zhizhong Zhang
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an 271018, China
| | - Jianzhu Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an 271018, China; Research Center for Animal Disease Control Engineering, Shandong Agricultural University, Tai`an 271018, China.
| | - Yongxia Liu
- College of Veterinary Medicine, Shandong Agricultural University, Tai`an 271018, China.
| |
Collapse
|
13
|
Zhou Z, Xun J, Wu C, Ji C, Ji S, Shu F, Wang Y, Chen H, Zheng Y, Xiao S. Acceleration of burn wound healing by micronized amniotic membrane seeded with umbilical cord-derived mesenchymal stem cells. Mater Today Bio 2023; 20:100686. [PMID: 37334186 PMCID: PMC10276167 DOI: 10.1016/j.mtbio.2023.100686] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/11/2023] [Accepted: 05/29/2023] [Indexed: 06/20/2023] Open
Abstract
Umbilical cord-derived mesenchymal stem cells (UC-MSC) are promising candidates for wound healing. However, the low amplification efficiency of MSC in vitro and their low survival rates after transplantation have limited their medical application. In this study, we fabricated a micronized amniotic membrane (mAM) as a microcarrier to amplify MSC in vitro and used mAM and MSC (mAM-MSC) complexes to repair burn wounds. Results showed that MSC could live and proliferate on mAM in a 3D culture system, exhibiting higher cell activity than in 2D culture. Transcriptome sequencing of MSC showed that the expression of growth factor-related, angiogenesis-related, and wound healing-related genes was significantly upregulated in mAM-MSC compared to traditional 2D-cultured MSC, which was verified via RT-qPCR. Gene ontology (GO) analysis of differentially expressed genes (DEGs) showed significant enrichment of terms related to cell proliferation, angiogenesis, cytokine activity, and wound healing in mAM-MSC. In a burn wound model of C57BL/6J mice, topical application of mAM-MSC significantly accelerated wound healing compared to MSC injection alone and was accompanied by longer survival of MSC and greater neovascularization in the wound.
Collapse
Affiliation(s)
- Zixuan Zhou
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Jingnan Xun
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Chenghao Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, People's Republic of China
| | - Chao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shizhao Ji
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Futing Shu
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Yuxiang Wang
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Hao Chen
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Yongjun Zheng
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| | - Shichu Xiao
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, People's Republic of China
| |
Collapse
|
14
|
Kengmo Tchoupa A, Kretschmer D, Schittek B, Peschel A. The epidermal lipid barrier in microbiome-skin interaction. Trends Microbiol 2023:S0966-842X(23)00027-6. [PMID: 36822953 DOI: 10.1016/j.tim.2023.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 02/25/2023]
Abstract
The corneocyte layers forming the upper surface of mammalian skin are embedded in a lamellar-membrane matrix which repels harmful molecules while retaining solutes from subcutaneous tissues. Only certain bacterial and fungal taxa colonize skin surfaces. They have ways to use epidermal lipids as nutrients while resisting antimicrobial fatty acids. Skin microorganisms release lipophilic microbe-associated molecular pattern (MAMP) molecules which are largely retained by the epidermal lipid barrier. Skin barrier defects, as in atopic dermatitis, impair lamellar-membrane integrity, resulting in altered skin microbiomes, which then include the pathogen Staphylococcus aureus. The resulting increased penetration of MAMPs and toxins promotes skin inflammation. Elucidating how microorganisms manipulate the epidermal lipid barrier will be key for better ways of preventing inflammatory skin disorders.
Collapse
Affiliation(s)
- Arnaud Kengmo Tchoupa
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Infection Biology Section, University of Tübingen, Tübingen, Germany; Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany; German Center for Infection Research (DZIF), partner site Tübingen, Germany
| | - Dorothee Kretschmer
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Infection Biology Section, University of Tübingen, Tübingen, Germany; Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany; German Center for Infection Research (DZIF), partner site Tübingen, Germany
| | - Birgit Schittek
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany; Dermatology Department, University Hospital Tübingen, Tübingen, Germany
| | - Andreas Peschel
- Interfaculty Institute of Microbiology and Infection Medicine Tübingen, Infection Biology Section, University of Tübingen, Tübingen, Germany; Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, University of Tübingen, Tübingen, Germany; German Center for Infection Research (DZIF), partner site Tübingen, Germany.
| |
Collapse
|
15
|
Jeong D, Kim MJ, Park Y, Chung J, Kweon HS, Kang NG, Hwang SJ, Youn SH, Hwang BK, Kim D. Visualizing extracellular vesicle biogenesis in gram-positive bacteria using super-resolution microscopy. BMC Biol 2022; 20:270. [PMID: 36464676 PMCID: PMC9720944 DOI: 10.1186/s12915-022-01472-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/21/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Recently, bacterial extracellular vesicles (EVs) have been considered to play crucial roles in various biological processes and have great potential for developing cancer therapeutics and biomedicine. However, studies on bacterial EVs have mainly focused on outer membrane vesicles released from gram-negative bacteria since the outermost peptidoglycan layer in gram-positive bacteria is thought to preclude the release of EVs as a physical barrier. RESULTS Here, we examined the ultrastructural organization of the EV produced by gram-positive bacteria using super-resolution stochastic optical reconstruction microscopy (STORM) at the nanoscale, which has not been resolved using conventional microscopy. Based on the super-resolution images of EVs, we propose three major mechanisms of EV biogenesis, i.e., membrane blebbing (mechanisms 1 and 2) or explosive cell lysis (mechanism 3), which are different from the mechanisms in gram-negative bacteria, despite some similarities. CONCLUSIONS These findings highlight the significant role of cell wall degradation in regulating various mechanisms of EV biogenesis and call for a reassessment of previously unresolved EV biogenesis in gram-positive bacteria.
Collapse
Affiliation(s)
- Dokyung Jeong
- grid.49606.3d0000 0001 1364 9317Department of Chemistry, Hanyang University, Seoul, 04763 Republic of Korea
| | - Min Jeong Kim
- grid.49606.3d0000 0001 1364 9317Department of Chemistry, Hanyang University, Seoul, 04763 Republic of Korea
| | - Yejin Park
- grid.49606.3d0000 0001 1364 9317Department of Chemistry, Hanyang University, Seoul, 04763 Republic of Korea
| | - Jinkyoung Chung
- grid.49606.3d0000 0001 1364 9317Department of Chemistry, Hanyang University, Seoul, 04763 Republic of Korea
| | - Hee-Seok Kweon
- grid.410885.00000 0000 9149 5707Electron 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
| | | | - Sung Hun Youn
- R&D Center, LG H&H Co., Ltd, Seoul, 07795 Republic of Korea
| | | | - Doory Kim
- grid.49606.3d0000 0001 1364 9317Department of Chemistry, Hanyang University, Seoul, 04763 Republic of Korea ,grid.49606.3d0000 0001 1364 9317Research Institute for Convergence of Basic Science, Hanyang University, Seoul, 04763 Republic of Korea ,grid.49606.3d0000 0001 1364 9317Institute of Nano Science and Technology, Hanyang University, Seoul, 04763 Republic of Korea ,grid.49606.3d0000 0001 1364 9317Research Institute for Natural Sciences, Hanyang University, Seoul, 04763 Republic of Korea
| |
Collapse
|
16
|
Hagberg K, Ghasemi Jahani SA, Omar O, Thomsen P. Osseointegrated prostheses for the rehabilitation of patients with transfemoral amputations: A prospective ten-year cohort study of patient-reported outcomes and complications. J Orthop Translat 2022; 38:56-64. [PMCID: PMC9588992 DOI: 10.1016/j.jot.2022.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 02/16/2023] Open
Abstract
Background Methods Results Conclusion
Collapse
Affiliation(s)
- Kerstin Hagberg
- Department of Orthopaedics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Department of Orthopaedics, Sahlgrenska University Hospital, Gothenburg, Sweden,Corresponding author. Department of Orthopaedics, Sahlgrenska University Hospital, Gothenburg, Sweden.
| | - Shadi Afarin Ghasemi Jahani
- Department of Orthopaedics, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Omar Omar
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Peter Thomsen
- Department of Biomaterials, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
17
|
Wei S, Jiao D, Xing W. A rapid method for isolation of bacterial extracellular vesicles from culture media using epsilon-poly-L–lysine that enables immunological function research. Front Immunol 2022; 13:930510. [PMID: 36032173 PMCID: PMC9411643 DOI: 10.3389/fimmu.2022.930510] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/21/2022] [Indexed: 11/24/2022] Open
Abstract
Both Gram-negative and Gram-positive bacteria can release vesicle-like structures referred to as bacterial extracellular vesicles (BEVs), which contain various bioactive compounds. BEVs play important roles in the microbial community interactions and host-microbe interactions. Markedly, BEVs can be delivered to host cells, thus modulating the development and function of the innate immune system. To clarify the compositions and biological functions of BEVs, we need to collect these vesicles with high purity and bioactivity. Here we propose an isolation strategy based on a broad-spectrum antimicrobial epsilon-poly-L-lysine (ϵ-PL) to precipitate BEVs at a relatively low centrifugal speed (10,000 × g). Compared to the standard ultracentrifugation strategy, our method can enrich BEVs from large volumes of media inexpensively and rapidly. The precipitated BEVs can be recovered by adjusting the pH and ionic strength of the media, followed by an ultrafiltration step to remove ϵ-PL and achieve buffer exchange. The morphology, size, and protein composition of the ϵ-PL-precipitated BEVs are comparable to those purified by ultracentrifugation. Moreover, ϵ-PL-precipitated BEVs retained the biological activity as observed by confocal microscopy studies. And THP-1 cells stimulated with these BEVs undergo marked reprogramming of their transcriptome. KEGG analysis of the differentially expressed genes showed that the signal pathways of cellular inflammatory response were significantly activated. Taken together, we provide a new method to rapidly enrich BEVs with high purity and bioactivity, which has the potential to be applied to BEVs-related immune response studies.
Collapse
Affiliation(s)
- Shujin Wei
- School of Medicine, Tsinghua University, Beijing, China
| | - Dian Jiao
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Wanli Xing
- School of Medicine, Tsinghua University, Beijing, China
- *Correspondence: Wanli Xing,
| |
Collapse
|
18
|
Yang Y, Li M, Zhou B, Jiang X, Zhang D, Luo H. Graphene oxide/gallium nanoderivative as a multifunctional modulator of osteoblastogenesis and osteoclastogenesis for the synergistic therapy of implant-related bone infection. Bioact Mater 2022; 25:594-614. [PMID: 37056253 PMCID: PMC10087081 DOI: 10.1016/j.bioactmat.2022.07.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/24/2022] [Accepted: 07/12/2022] [Indexed: 11/30/2022] Open
Abstract
Currently, implant-associated bacterial infections account for most hospital-acquired infections in patients suffering from bone fractures or defects. Poor osseointegration and aggravated osteolysis remain great challenges for the success of implants in infectious scenarios. Consequently, developing an effective surface modification strategy for implants is urgently needed. Here, a novel nanoplatform (GO/Ga) consisting of graphene oxide (GO) and gallium nanoparticles (GaNPs) was reported, followed by investigations of its in vitro antibacterial activity and potential bacterium inactivation mechanisms, cytocompatibility and regulatory actions on osteoblastogenesis and osteoclastogenesis. In addition, the possible molecular mechanisms underlying the regulatory effects of GO/Ga nanocomposites on osteoblast differentiation and osteoclast formation were clarified. Moreover, an in vivo infectious microenvironment was established in a rat model of implant-related femoral osteomyelitis to determine the therapeutic efficacy and biosafety of GO/Ga nanocomposites. Our results indicate that GO/Ga nanocomposites with excellent antibacterial potency have evident osteogenic potential and inhibitory effects on osteoclast differentiation by modulating the BMP/Smad, MAPK and NF-κB signaling pathways. The in vivo experiments revealed that the administration of GO/Ga nanocomposites significantly inhibited bone infections, reduced osteolysis, promoted osseointegration located in implant-bone interfaces, and resulted in satisfactory biocompatibility. In summary, this synergistic therapeutic system could accelerate the bone healing process in implant-associated infections and can significantly guide the future surface modification of implants used in bacteria-infected environments.
Collapse
Affiliation(s)
- Ying Yang
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
- Corresponding author. Department of Plastic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, PR China.
| | - Min Li
- Department of Oncology, Changsha Central Hospital, University of South China, Changsha, 410006, China
| | - Bixia Zhou
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Xulei Jiang
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Dou Zhang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
| | - Hang Luo
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, China
- Corresponding author. State Key Laboratory of Powder Metallurgy, Central South University, 932 South Lushan Road, Changsha, 410083, Hunan, China.
| |
Collapse
|
19
|
Saenz-de-Juano MD, Silvestrelli G, Weber A, Röhrig C, Schmelcher M, Ulbrich SE. Inflammatory Response of Primary Cultured Bovine Mammary Epithelial Cells to Staphylococcus aureus Extracellular Vesicles. BIOLOGY 2022; 11:biology11030415. [PMID: 35336789 PMCID: PMC8944978 DOI: 10.3390/biology11030415] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/28/2022] [Accepted: 03/04/2022] [Indexed: 11/22/2022]
Abstract
Simple Summary Mastitis, the inflammation of the mammary gland, is one of the most common and costly diseases worldwide, and Staphylococcus aureus (S. aureus) is among the most prevalent microorganisms that cause it. To obtain new insights into S. aureus mammary gland infections, we have isolated S. aureus extracellular vesicles to challenge in vitro primary bovine mammary epithelial cells. Despite the toxic content of the vesicles, we observed only a minor pro-inflammatory response. The latter can contribute to the explanation of how S. aureus evades mammary epithelial defence mechanisms and successfully colonizes the mammary gland. Abstract In dairy cows, Staphylococcus aureus (S. aureus) is among the most prevalent microorganisms worldwide, causing mastitis, an inflammation of the mammary gland. Production of extracellular vesicles (EVs) is a common feature of S. aureus strains, which contributes to its pathogenesis by delivering bacterial effector molecules to host cells. In the current study, we evaluated the differences between five S. aureus mastitis isolates regarding their EV production. We found that different mastitis-related S. aureus strains differ in their behaviour of shedding EVs, with M5512VL producing the largest amount of EVs containing alpha-haemolysin, a strong cytotoxic agent. We stimulated primary cultured bovine mammary epithelial cells (pbMECs) with EVs from the S. aureus strain M5512VL. After 24 h of incubation, we observed a moderate increase in gene expression of tumour necrosis factor-alpha (TNF-α) but, surprisingly, a lack of an associated pronounced pro-inflammatory response. Our results contribute to understanding the damaging nature of S. aureus in its capacity to effectively affect mammary epithelial cells.
Collapse
Affiliation(s)
- Mara D. Saenz-de-Juano
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, 8092 Zurich, Switzerland; (M.D.S.-d.-J.); (G.S.); (A.W.)
| | - Giulia Silvestrelli
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, 8092 Zurich, Switzerland; (M.D.S.-d.-J.); (G.S.); (A.W.)
| | - Andres Weber
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, 8092 Zurich, Switzerland; (M.D.S.-d.-J.); (G.S.); (A.W.)
| | - Christian Röhrig
- Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zurich, Switzerland; (C.R.); (M.S.)
| | - Mathias Schmelcher
- Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zurich, Switzerland; (C.R.); (M.S.)
| | - Susanne E. Ulbrich
- Animal Physiology, Institute of Agricultural Sciences, ETH Zurich, 8092 Zurich, Switzerland; (M.D.S.-d.-J.); (G.S.); (A.W.)
- Correspondence:
| |
Collapse
|