151
|
Chen J, Hu S, Liu J, Jiang H, Wang S, Yang Z. Exosomes: a double-edged sword in cancer immunotherapy. MedComm (Beijing) 2025; 6:e70095. [PMID: 39968497 PMCID: PMC11831209 DOI: 10.1002/mco2.70095] [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: 08/02/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 02/20/2025] Open
Abstract
Over the past few decades, immunotherapy has emerged as a powerful strategy to overcome the limitations of conventional cancer treatments. The use of extracellular vesicles, particularly exosomes, which carry cargoes capable of modulating the immune response, has been extensively explored as a potential therapeutic approach in cancer immunotherapy. Exosomes can deliver their cargo to target cells, thereby influencing their phenotype and immunomodulatory functions. They exhibit either immunosuppressive or immune-activating characteristics, depending on their internal contents. These exosomes originate from diverse cell sources, and their internal contents can vary, suggesting that there may be a delicate balance between immune suppression and stimulation when utilizing them for immunotherapy. Therefore, a thorough understanding of the molecular mechanisms underlying the role of exosomes in cancer progression is essential. This review focuses on the molecular mechanisms driving exosome function and their impact on the tumor microenvironment (TME), highlighting the intricate balance between immune suppression and activation that must be navigated in exosome-based therapies. Additionally, it underscores the challenges and ongoing efforts to optimize exosome-based immunotherapies, thereby making a significant contribution to the advancement of cancer immunotherapy research.
Collapse
Affiliation(s)
- Jiayi Chen
- School of Life SciencesJilin UniversityChangchunChina
| | - Siyuan Hu
- School of Life SciencesJilin UniversityChangchunChina
| | - Jiayi Liu
- School of Life SciencesJilin UniversityChangchunChina
| | - Hao Jiang
- School of Life SciencesJilin UniversityChangchunChina
| | - Simiao Wang
- School of Life SciencesJilin UniversityChangchunChina
| | - Zhaogang Yang
- School of Life SciencesJilin UniversityChangchunChina
| |
Collapse
|
152
|
Xu CX, Huang W, Shi XJ, Du Y, Liang JQ, Fang X, Chen HY, Cheng Y. Dysregulation of Serum Exosomal Lipid Metabolism in Schizophrenia: A Biomarker Perspective. Mol Neurobiol 2025; 62:3556-3567. [PMID: 39312067 DOI: 10.1007/s12035-024-04477-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 09/01/2024] [Indexed: 02/04/2025]
Abstract
Exosomes, crucial extracellular vesicles, have emerged as potential biomarkers for neurological conditions, including schizophrenia (SCZ). However, the exploration of exosomal lipids in the context of SCZ remains scarce, necessitating in-depth investigation. Leveraging ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS), this study aimed to characterize the lipidomic profile of serum exosomes from SCZ patients, assessing their potential as novel biomarkers for SCZ diagnosis through absolute quantitative lipidomics. Our comprehensive lipidomic analysis unveiled 39 serum exosomal lipids that were differentially expressed between SCZ patients (n = 20) and healthy controls (HC, n = 20). These findings revealed a profound dysregulation in lipid metabolism pathways, notably in sphingolipid metabolism, glycerophospholipid metabolism, and linoleic acid metabolism. Among these, seven exosomal lipids stood out for their diagnostic potential, exhibiting remarkable ability to differentiate SCZ patients from HCs with an unparalleled classification performance, evidenced by an area under the curve (AUC) of 0.94 (95% CI, 0.82-1.00). These lipids included specific ceramides and phosphoethanolamines, pointing to a distinct lipid metabolic fingerprint associated with SCZ. Furthermore, bioinformatic analyses reinforced the pivotal involvement of these lipids in SCZ-related lipid metabolic processes, suggesting their integral role in the disorder's pathophysiology. This study significantly advances our understanding of SCZ by pinpointing dysregulated exosomal lipid metabolism as a key factor in its pathology. The identified serum exosome-derived lipids emerge as compelling biomarkers for SCZ diagnosis, offering a promising avenue towards the development of objective and reliable diagnostic tools.
Collapse
Affiliation(s)
- Chen-Xi Xu
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, No. 27, South Street of Zhongguancun, Haidian District, Beijing, 100081, China
| | - Wei Huang
- The Third People's Hospital of Foshan, Foshan, Guangdong, China
| | - Xiao-Jie Shi
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China
| | - Yang Du
- Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jia-Quan Liang
- The Third People's Hospital of Foshan, Foshan, Guangdong, China
| | - Xuan Fang
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, No. 27, South Street of Zhongguancun, Haidian District, Beijing, 100081, China
| | - He-Yuan Chen
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, No. 27, South Street of Zhongguancun, Haidian District, Beijing, 100081, China
| | - Yong Cheng
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, No. 27, South Street of Zhongguancun, Haidian District, Beijing, 100081, China.
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China.
- Institute of National Security, Minzu University of China, Beijing, China.
| |
Collapse
|
153
|
Qian L, Chen P, Zhang S, Wang Z, Guo Y, Koutouratsas V, Fleishman JS, Huang C, Zhang S. The uptake of extracellular vesicles: Research progress in cancer drug resistance and beyond. Drug Resist Updat 2025; 79:101209. [PMID: 39893749 DOI: 10.1016/j.drup.2025.101209] [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: 12/11/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Extracellular vesicles (EVs) are heterogeneous vesicles released by donor cells that can be taken up by recipient cells, thus inducing cellular phenotype changes. Since their discovery decades ago, roles of EVs in modulating initiation, growth, survival and metastasis of cancer have been revealed. Recent studies from multifaceted perspectives have further detailed the contribution of EVs to cancer drug resistance; however, the role of EV uptake in conferring drug resistance seems to be overlooked. In this comprehensive review, we update the EV subtypes and approaches for determining EV uptake. The biological basis of EV uptake is systematically summarized. Moreover, we focus on the diverse uptake mechanisms by which EVs carry out the intracellular delivery of functional molecules and drug resistance signaling. Furthermore, we highlight how EV uptake confers drug resistance and identify potential strategies for targeting EV uptake to overcome drug resistance. Finally, we discuss the research gap on the role of EV uptake in promoting drug resistance. This updated knowledge provides a new avenue to overcome cancer drug resistance by targeting EV uptake.
Collapse
Affiliation(s)
- Luomeng Qian
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Pangzhou Chen
- Department of Breast Surgery, Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan 528200, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin 300121, China
| | - Zhenglu Wang
- Department of Pathology, Tianjin Key Laboratory for Organ Transplantation, Tianjin First Centre Hospital, Tianjin 300192, China
| | - Yuan Guo
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Vasili Koutouratsas
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Chuanqiang Huang
- Department of Breast Surgery, Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan 528200, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
154
|
Justa HCD, Baldissera AB, Machado MI, Souza SH, Polli NLC, Boia-Ferreira M, Schluga PHDC, Donatti L, Wille ACM, Minozzo JC, Gremski LH, Veiga SS. Induction of ectosome formation by binding of phospholipases D from Loxosceles venoms to endothelial cell surface: Mechanism of interaction. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159579. [PMID: 39547302 DOI: 10.1016/j.bbalip.2024.159579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/01/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Members of the phospholipase D (PLD) superfamily found in Loxosceles spider venoms are potent toxins with inflammatory and necrotizing activities. They degrade phospholipids in cell membranes, generating bioactive molecules that activate skin cells. These skin cells, in turn, activate leukocytes involved in dermonecrosis, characterized by aseptic coagulative necrosis. Although the literature has advanced in understanding the structure-function relationship, the cell biology resulting from the interactions of these molecules with cells remains poorly understood. In this study, we show that different cells exposed to recombinant PLDs bind these molecules to their plasma membrane, leading to the subsequent organization of extracellular microvesicles/ectosomes. The binding occurs as quickly as five minutes or less after exposure, increases over time, and eventually, the PLDs are expelled from the cell surface without generating cytotoxicity. PLDs are not endocytosed, nor do they spatially colocalize with acidic organelles in the intracellular environment. At least two regions of PLDs - the domain involved in magnesium ion coordination and the choline binding site - appear to play a role in cell surface binding and ectosome organization. However, the amino acids involved in catalysis do not participate in these events. The binding of these PLDs to the cell membrane, independent of catalytic activity, is sufficient to trigger intracellular signaling and enhance the expression of the pro-inflammatory IL-8 gene. These results are supported by the observation that isoforms of PLDs lacking catalytic activity induce an inflammatory response in vivo when injected into the skin of rabbits, without causing dermonecrosis. Our data indicate that these PLDs bind to the surface of target cells, promoting the organization of extracellular vesicles/ectosomes. Subsequently, these events activate pro-inflammatory genes and induce an inflammatory response in vivo. The binding to cells is not dependent on amino acids involved in catalysis but rather on amino acids involved in magnesium coordination. The binding of PLDs to the cell surface, formation of ectosomes, and activation of cells appear to initiate signals involved in inflammatory responses that can lead to dermonecrosis in accidents. This correlation is supported by experimental observations indicating that the events of toxin binding to cells, formation of microvesicles, and inflammatory responses observed both in vitro and in vivo are interconnected.
Collapse
Affiliation(s)
- Hanna Câmara da Justa
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | | | | | - Samira Hajjar Souza
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | | | - Marianna Boia-Ferreira
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | | | - Lucelia Donatti
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | - Ana Carolina M Wille
- Department of Structural, Molecular Biology and Genetics, State University of Ponta Grossa (UEPG), Ponta Grossa 84030-900, Brazil
| | - João Carlos Minozzo
- Production and Research Center of Immunobiological Products (CPPI), State Department of Health, Piraquara 83302-200, Brazil
| | - Luiza Helena Gremski
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil
| | - Silvio S Veiga
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil.
| |
Collapse
|
155
|
Zheng Z, Zhai Y, Yan X, Wang Z, Zhang H, Xu R, Liu X, Cai J, Zhang Z, Shang Y, Zhang J, Yin J. Functions and Clinical Applications of Exosomes in Gastric Cancer. Int J Biol Sci 2025; 21:2330-2345. [PMID: 40083701 PMCID: PMC11900809 DOI: 10.7150/ijbs.98087] [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: 05/04/2024] [Accepted: 02/04/2025] [Indexed: 03/16/2025] Open
Abstract
Gastric cancer is a common and highly invasive type of malignant tumor, the pathogenesis of which remains unclarified. However, exosomes are now known to play important roles in gastric cancer development and treatment. Cells use exosomes for the packaging and transportation of a variety of bioactive molecules, such as proteins, double-stranded DNA, and micro-ribonucleic acids, to other sites. Exosome-specific membrane structures and exosomal contents are widely involved in processes that facilitate material exchange and intercellular communication between gastric cancer cells. They help in forming a pre-metastatic microenvironment, promoting the proliferation and apoptosis of gastric cancer cells, and driving invasion, metastasis, and resistance to anti-tumor drugs. In this review, we aimed to summarize the findings of research articles indexed in the PubMed, Web of Science, and Embase databases and published up to May 31, 2024, on the role of exosomes in the pathogenesis of gastric cancer and their potential clinical applications in its treatment. Thus, research on exosomes may lead to breakthroughs in the early diagnosis of gastric cancer and identification of novel treatments.
Collapse
Affiliation(s)
- Zhi Zheng
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Institute of Clinical Medicine, Beijing, China
| | - Yuhao Zhai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Institute of Clinical Medicine, Beijing, China
| | - Xiaosheng Yan
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Institute of Clinical Medicine, Beijing, China
| | - Zimeng Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Institute of Clinical Medicine, Beijing, China
| | - Haiqiao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Institute of Clinical Medicine, Beijing, China
| | - Rui Xu
- Department of Pathology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaoye Liu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Institute of Clinical Medicine, Beijing, China
| | - Jun Cai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Institute of Clinical Medicine, Beijing, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Institute of Clinical Medicine, Beijing, China
| | - Yuxi Shang
- Department of Hematology, Fuxing Hospital, Eighth Clinical Medical College, Capital Medical University, Beijing, China
| | - Jun Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Institute of Clinical Medicine, Beijing, China
| | - Jie Yin
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Beijing, China
- National Clinical Research Center for Digestive Diseases, Beijing, China
- Beijing Institute of Clinical Medicine, Beijing, China
| |
Collapse
|
156
|
Godbole N, Lai A, Carrion F, Scholz-Romero K, Ravichandran A, Kalita-de Croft P, McCart Reed AE, Joshi V, Lakhani SR, Masud MK, Yamauchi Y, Perrin L, Hooper J, Bray L, Guanzon D, Salomon C. Extracellular vesicle miRNAs from three-dimensional ovarian cancer in vitro models and their implication in overall cancer survival. Heliyon 2025; 11:e42188. [PMID: 40034306 PMCID: PMC11872480 DOI: 10.1016/j.heliyon.2025.e42188] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 12/02/2024] [Accepted: 01/21/2025] [Indexed: 03/05/2025] Open
Abstract
Ovarian cancer is the most common gynaecological malignancy and the seventh most diagnosed cancer in females worldwide. Currently, it is the sixth leading cause of cancer related mortality among patients globally. The heterogenous origin of the disease and unambiguous nature of the clinical symptoms leading to delayed detection has been one of the key reasons for increasing mortality. Hence new approaches are required to understand the biology of ovarian cancer, where the use of cell culture models that mimic the physiology of the disease is fundamental. Cell culture serves as a crucial in vitro tool, contributing to our comprehension of various aspects of cell biology, tissue morphology, disease mechanisms, drug responses, protein production, and tissue engineering. A significant portion of in vitro studies rely on two-dimensional (2D) cell cultures, however, these cultures present notable limitations, for example disruptions in cellular and extracellular interactions, alterations in cell morphology, polarity, and division mechanisms. Recently, extracellular vesicles have been identified as crucial players in cell biology as part of the communication system that cancer cells use to metastasize. We optimized and compared three-dimensional (3D) culture of ovarian cancer cells lines (SKOV-3 and OVCAR-3) with two-dimensional models based on their protein and miRNA content. We further investigated whether extracellular vesicles from these models reflect changes in cancer cells, and aid in the identification of overall survival in women with ovarian cancer.
Collapse
Affiliation(s)
- Nihar Godbole
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, Centre for Clinical Diagnostics, UQ Centre for Clinical Research (UQCCR), Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
- UQ Centre for Extracellular Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
| | - Andrew Lai
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, Centre for Clinical Diagnostics, UQ Centre for Clinical Research (UQCCR), Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
- UQ Centre for Extracellular Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
| | - Flavio Carrion
- Departamento de Investigación, Postgrado y Educación Continua (DIPEC), Facultad de Ciencias de la Salud, Universidad del Alba, Santiago 8320000, Chile
| | - Katherin Scholz-Romero
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, Centre for Clinical Diagnostics, UQ Centre for Clinical Research (UQCCR), Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
- UQ Centre for Extracellular Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
| | - Akhilandeshwari Ravichandran
- Centre for Biomedical Technologies, Queensland University of Technology, 60 Musk Ave., Kelvin Grove, QLD 4059, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology, Kelvin Grove, QLD 5059, Australia
| | - Priyakshi Kalita-de Croft
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, Centre for Clinical Diagnostics, UQ Centre for Clinical Research (UQCCR), Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
- UQ Centre for Extracellular Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
| | - Amy E. McCart Reed
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
| | - Vaibhavi Joshi
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
| | - Sunil R. Lakhani
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
- Pathology Queensland, The Royal Brisbane and Women's Hospital, Herston, QLD 4029, Australia
| | - Mostafa Kamal Masud
- UQ Centre for Extracellular Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD 4072, Australia
| | - Yusuke Yamauchi
- Australian Institute for Bioengineering and Nanotechnology (AIBN), The University of Queensland, St Lucia, QLD 4072, Australia
| | - Lewis Perrin
- UQ Centre for Extracellular Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
- Mater Research Institute, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - John Hooper
- UQ Centre for Extracellular Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
- Mater Research Institute, Translational Research Institute, Woolloongabba, QLD 4102, Australia
| | - Laura Bray
- Centre for Biomedical Technologies, Queensland University of Technology, 60 Musk Ave., Kelvin Grove, QLD 4059, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane City, QLD 4000, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology, Kelvin Grove, QLD 5059, Australia
| | - Dominic Guanzon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, Centre for Clinical Diagnostics, UQ Centre for Clinical Research (UQCCR), Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
- UQ Centre for Extracellular Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
| | - Carlos Salomon
- Translational Extracellular Vesicles in Obstetrics and Gynae-Oncology Group, Centre for Clinical Diagnostics, UQ Centre for Clinical Research (UQCCR), Royal Brisbane and Women's Hospital, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
- UQ Centre for Extracellular Nanomedicine, Faculty of Medicine, The University of Queensland, Herston, QLD 4029, Australia
| |
Collapse
|
157
|
Xia W, Tan Y, Liu Y, Xie N, Zhu H. Prospect of extracellular vesicles in tumor immunotherapy. Front Immunol 2025; 16:1525052. [PMID: 40078996 PMCID: PMC11897508 DOI: 10.3389/fimmu.2025.1525052] [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: 11/08/2024] [Accepted: 01/28/2025] [Indexed: 03/14/2025] Open
Abstract
Extracellular vesicles (EVs), as cell-derived small vesicles, facilitate intercellular communication within the tumor microenvironment (TME) by transporting biomolecules. EVs from different sources have varied contents, demonstrating differentiated functions that can either promote or inhibit cancer progression. Thus, regulating the formation, secretion, and intake of EVs becomes a new strategy for cancer intervention. Advancements in EV isolation techniques have spurred interest in EV-based therapies, particularly for tumor immunotherapy. This review explores the multifaceted functions of EVs from various sources in tumor immunotherapy, highlighting their potential in cancer vaccines and adoptive cell therapy. Furthermore, we explore the potential of EVs as nanoparticle delivery systems in tumor immunotherapy. Finally, we discuss the current state of EVs in clinical settings and future directions, aiming to provide crucial information to advance the development and clinical application of EVs for cancer treatment.
Collapse
Affiliation(s)
- Wenbo Xia
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yunhan Tan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yongen Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Huili Zhu
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
158
|
Park C, Ryu T, Mohamed-Hinds R, Kim K, Kim JH, Zou L, Williams B, Na CH, Chao W. Proteomic profiling of plasma extracellular vesicles identifies signatures of innate immunity, coagulation, and endothelial activation in septic patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.21.25322420. [PMID: 40061316 PMCID: PMC11888535 DOI: 10.1101/2025.02.21.25322420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Plasma extracellular vesicles (EVs) are cell-derived lipid particles and reportedly play a role in sepsis pathogenesis. This study aimed to identify EV cargo proteins in septic patients and explore their association with key sepsis pathophysiology. Plasma EVs were subjected to Tandem Mass Tag (TMT)-based quantitative proteomic analysis. We identified 522 differentially expressed (DE) EV proteins in septic patients (n=15) compared to the healthy controls (n=10). The KEGG analysis of the DE proteins revealed multiple functional pathways linked to sepsis, e.g., complement/coagulation, platelet activation, phagosome, inflammation, and neutrophil extracellular trap formation. Weighted Gene Coexpression Network Analysis of 1,642 EV proteins identified nine unique protein modules, some of which were highly correlated with the sepsis diagnosis and diverse plasma markers, including organ injury, inflammation, coagulopathy, and endothelial activation. Cell type-specific enrichment analysis revealed the cellular origins of EVs, including immune and epithelial cells, neurons, and glial cells. Thus, the current study discovered complex proteomic signatures in plasma EVs that are closely associated with key pathophysiological responses in sepsis. These findings support the importance of EV cargo proteins in the patients' immune responses, coagulation, and endothelial activation and lay the foundation for future mechanistic study of plasma EVs in sepsis pathogenesis.
Collapse
Affiliation(s)
- Chanhee Park
- Translational Research Program, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Taekyung Ryu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rashida Mohamed-Hinds
- Translational Research Program, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kyungdo Kim
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jin Hyeok Kim
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lin Zou
- Translational Research Program, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brittney Williams
- Translational Research Program, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Chan Hyun Na
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Wei Chao
- Translational Research Program, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Center for Shock, Trauma and Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
159
|
Prado-Yupanqui JW, Ramírez-Orrego L, Cortez D, Vera-Ponce VJ, Chenet SM, Tejedo JR, Tapia-Limonchi R. The Hidden Power of the Secretome: Therapeutic Potential on Wound Healing and Cell-Free Regenerative Medicine-A Systematic Review. Int J Mol Sci 2025; 26:1926. [PMID: 40076553 PMCID: PMC11899803 DOI: 10.3390/ijms26051926] [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: 02/01/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Various types of wounds represent a persistent healthcare burden that demands innovative and effective therapeutic solutions. Innovative approaches have emerged that focus on skin regeneration with minimal side effects. One such method is cell-free therapy that utilizes the secretome of human mesenchymal stem cells (hMSCs) as a promising alternative to traditional cell-based therapies, leveraging a complex mixture of bioactive molecules, including growth factors, cytokines, and extracellular vesicles, to accelerate tissue regeneration. This systematic review synthesizes the findings of 35 studies evaluating the impact of hMSC-derived secretomes on wound healing, with a focus on their regenerative, immunomodulatory, and angiogenic effects. The influence of MSC sources (adipose tissue, bone marrow, umbilical cord) and culture conditions on secretome composition and efficacy in the cutaneous wound healing process is examined, highlighting their therapeutic potential in regenerative medicine. This review also explores emerging preclinical and clinical applications, highlighting promising results, such as enhanced fibroblast proliferation, reduced inflammation, and improved extracellular matrix remodeling. In addition, advances in secretome-based biomaterials, including hydrogels and scaffolds, which optimize therapeutic delivery and efficacy are discussed. Despite the growing body of evidence supporting the safety and efficacy of secretomes, challenges remain regarding standardization, large-scale production, and clinical validation. This review highlights the potential of MSC-derived secretomes as a next-generation cell-free approach for wound healing and regenerative medicine.
Collapse
Affiliation(s)
- Jhon W. Prado-Yupanqui
- Instituto de Investigación de Enfermedades Tropicales, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas 01001, Peru; (J.W.P.-Y.); (L.R.-O.); (D.C.); (V.J.V.-P.); (S.M.C.); (J.R.T.)
| | - Lourdes Ramírez-Orrego
- Instituto de Investigación de Enfermedades Tropicales, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas 01001, Peru; (J.W.P.-Y.); (L.R.-O.); (D.C.); (V.J.V.-P.); (S.M.C.); (J.R.T.)
| | - Denny Cortez
- Instituto de Investigación de Enfermedades Tropicales, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas 01001, Peru; (J.W.P.-Y.); (L.R.-O.); (D.C.); (V.J.V.-P.); (S.M.C.); (J.R.T.)
| | - Victor Juan Vera-Ponce
- Instituto de Investigación de Enfermedades Tropicales, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas 01001, Peru; (J.W.P.-Y.); (L.R.-O.); (D.C.); (V.J.V.-P.); (S.M.C.); (J.R.T.)
- Facultad de Medicina, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas 01001, Peru
| | - Stella M. Chenet
- Instituto de Investigación de Enfermedades Tropicales, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas 01001, Peru; (J.W.P.-Y.); (L.R.-O.); (D.C.); (V.J.V.-P.); (S.M.C.); (J.R.T.)
- Facultad de Medicina, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas 01001, Peru
| | - Juan R. Tejedo
- Instituto de Investigación de Enfermedades Tropicales, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas 01001, Peru; (J.W.P.-Y.); (L.R.-O.); (D.C.); (V.J.V.-P.); (S.M.C.); (J.R.T.)
- Departamento de Biología Molecular e Ingeniería Bioquímica, Universidad Pablo de Olavide (UPO), 41013 Seville, Spain
- Biomedical Research Network for Diabetes and Related Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rafael Tapia-Limonchi
- Instituto de Investigación de Enfermedades Tropicales, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas 01001, Peru; (J.W.P.-Y.); (L.R.-O.); (D.C.); (V.J.V.-P.); (S.M.C.); (J.R.T.)
- Facultad de Medicina, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas 01001, Peru
| |
Collapse
|
160
|
Cerveró-Varona A, Prencipe G, Peserico A, Canciello A, House AH, Santos HA, Perugini M, Sulcanese L, Takano C, Miki T, Iannetta A, Russo V, Mattioli M, Barboni B. Amniotic epithelial Cell microvesicles uptake inhibits PBMCs and Jurkat cells activation by inducing mitochondria-dependent apoptosis. iScience 2025; 28:111830. [PMID: 39967871 PMCID: PMC11834128 DOI: 10.1016/j.isci.2025.111830] [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: 08/01/2024] [Revised: 10/15/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Amniotic epithelial cells (AECs) exhibit significant immunomodulatory and pro-regenerative properties, largely due to their intrinsic paracrine functions that are currently harnessed through the collection of their secretomes. While there is increasing evidence of the role of bioactive components freely secreted or carried by exosomes, the bioactive cargo of AEC microvesicles (MVs) and their crosstalk with the immune cells remains to be fully explored. We showed that under intrinsic conditions or in response to LPS, AEC-derived MV carries components such as lipid-mediated signaling molecules, ER, and mitochondria. They foster the intra/interspecific mitochondrial transfer into immune cells (PBMCs and Jurkat cells) in vitro and in vivo on the zebrafish larvae model of injury. The internalization of MV cargoes through macropinocytosis induces hyperpolarization of PBMC mitochondrial membranes and triggers MV-mediated apoptosis. This powerful immune suppressive mechanism triggered by AEC-MV cargo delivery paves the way for controlled and targeted cell-free therapeutic approaches.
Collapse
Affiliation(s)
- Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Alessia Peserico
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Angelo Canciello
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Andrew H. House
- Helsinki University Lipidomics Unit, Helsinki Institute for Life Science (HiLIFE), Biocenter 3, Viikinkaari 1, 00790 Helsinki, Finland
| | - Hélder A. Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Monia Perugini
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Ludovica Sulcanese
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Chika Takano
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Toshio Miki
- Department of Physiology, Nihon University School of Medicine, Tokyo, Japan
| | - Annamaria Iannetta
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Valentina Russo
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Mauro Mattioli
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
161
|
Thongsit A, Oontawee S, Siriarchavatana P, Rodprasert W, Somparn P, Na Nan D, Osathanon T, Egusa H, Sawangmake C. Scalable production of anti-inflammatory exosomes from three-dimensional cultures of canine adipose-derived mesenchymal stem cells: production, stability, bioactivity, and safety assessment. BMC Vet Res 2025; 21:81. [PMID: 39979916 PMCID: PMC11841348 DOI: 10.1186/s12917-025-04517-1] [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: 11/21/2024] [Accepted: 01/24/2025] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND The therapeutic potential of exosomes derived from mesenchymal stem cells (MSCs) is increasingly recognized in veterinary medicine. This study explored the feasibility of a microcarrier-based three-dimensional (3D) culture system for producing the exosomes (cEXO). Investigations were conducted to enhance production efficiency, ensure stability, and evaluate the therapeutic potential of cEXO for anti-inflammatory applications while assessing their safety profile. RESULTS The microcarrier-based 3D culture system improved efficient production of cEXO, yielding exosomes with acceptable profiles, including a size of approximately 81.22 nm, negative surface charge, and high particle concentration (1.32 × 109 particles/mL). Confocal imaging proved dynamic changes in cell viability across culture phases, highlighting the challenges of maintaining cell viability during repeated exosome collection cycles. Characterization via transmission electron microscopy, nanoparticle tracking analysis, and zeta-potential measurements confirmed the stability and functionality of cEXO, particularly when stored at -20 °C. Functional assays showed that cEXO exerted significant anti-inflammatory activity in RAW264.7 macrophages in an inverse dose-dependent manner, with no observed cytotoxicity to fibroblasts or macrophages. Acute toxicity testing in rats revealed no adverse effects on clinical parameters, organ health, or body weight, supporting the safety of cEXO for therapeutic use. CONCLUSIONS This study highlights the potential of a microcarrier-based 3D culture system for scalable cEXO production with robust anti-inflammatory activity, stability, and safety profiles. These findings advance the development of cEXO-based therapies and support their application in veterinary regenerative medicine.
Collapse
Affiliation(s)
- Anatcha Thongsit
- Department of Social and Administrative Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology, Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Saranyou Oontawee
- Second Century Fund (C2F), Chulalongkorn University for Post-doctoral Fellowship, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology, Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Parkpoom Siriarchavatana
- Second Century Fund (C2F), Chulalongkorn University for Post-doctoral Fellowship, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology, Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Watchareewan Rodprasert
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology, Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Poorichaya Somparn
- Center of Excellence in Systems Biology, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Daneeya Na Nan
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Dental Stem Cell Biology Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Regenerative Dentistry (CERD), Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Hiroshi Egusa
- Center for Advanced Stem Cell and Regenerative Research, Division of Molecular and Regenerative Prosthodontics, Tohoku University Graduate School of Dentistry, Sendai, 980-8575, Japan
| | - Chenphop Sawangmake
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology, Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
- Department of Pharmacology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand.
- Center of Excellence in Regenerative Dentistry (CERD), Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
162
|
Marquez-Paradas E, Torrecillas-Lopez M, Barrera-Chamorro L, del Rio-Vazquez JL, Gonzalez-de la Rosa T, Montserrat-de la Paz S. Microbiota-derived extracellular vesicles: current knowledge, gaps, and challenges in precision nutrition. Front Immunol 2025; 16:1514726. [PMID: 40051622 PMCID: PMC11882860 DOI: 10.3389/fimmu.2025.1514726] [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: 10/21/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
The gut microbiota has co-evolved with its host, profoundly shaping the development and functioning of the immune system. This co-evolution has led to a dynamic relationship where microbial metabolites and molecular signals influence immune maturation, tolerance, and defense mechanisms, highlighting its essential role in maintaining host health. Recently, bacterial extracellular vesicles (BEVs), membrane nanoparticles produced by bacteria, have emerged as important players in gut balance and as potent immune modulators. These vesicles reflect the characteristics of the bacterial membrane and contain nucleic acids, proteins, lipids, and metabolites. They can regulate immune processes and are involved in neurological and metabolic diseases due to their ability to distribute both locally in the gut and systemically, affecting immune responses at both levels. This review provides a comprehensive overview of the characteristics and functional profile of BEVs, detailing how nutrition influences the production and function of these vesicles, how antibiotics can disrupt or alter their composition, and how these factors collectively impact immunity and disease development. It also highlights the potential of BEVs in the development of precision nutritional strategies through dietary modulation, such as incorporating prebiotic fibers to enhance beneficial BEV production, reducing intake of processed foods that may promote harmful BEVs, and tailoring probiotic interventions to influence specific microbial communities and their vesicular outputs.
Collapse
Affiliation(s)
- Elvira Marquez-Paradas
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| | - Maria Torrecillas-Lopez
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| | - Luna Barrera-Chamorro
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| | - Jose L. del Rio-Vazquez
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
| | - Teresa Gonzalez-de la Rosa
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| | - Sergio Montserrat-de la Paz
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Seville, Spain
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocio/CSIC /Universidad de Sevilla, Seville, Spain
| |
Collapse
|
163
|
Iwamoto Y, Salmon B, Yoshioka Y, Kojima R, Krull A, Ota S. High throughput analysis of rare nanoparticles with deep-enhanced sensitivity via unsupervised denoising. Nat Commun 2025; 16:1728. [PMID: 39979247 PMCID: PMC11842628 DOI: 10.1038/s41467-025-56812-y] [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: 04/23/2024] [Accepted: 01/31/2025] [Indexed: 02/22/2025] Open
Abstract
The large-scale multiparametric analysis of individual nanoparticles is increasingly vital in the diverse fields of biology, medicine, and materials science. However, the current methods struggle with the tradeoff between measurement scalability and sensitivity, especially when identifying rare nanoparticles in heterogeneous mixtures. By developing and combining an unsupervised deep learning-based denoising method and an optofluidic device tuned for nanoparticle detection, we realize a nanoparticle analyzer that simultaneously achieves high scalability, throughput, and sensitivity levels; we name this approach "Deep Nanometry" (DNM). DNM detects polystyrene beads with a detection of limit of 30 nm at a throughput of over 100,000 events/second. The sensitive and scalable DNM directly detects rare target extracellular vesicles (EVs) in non-purified serum, making up as little as 0.002% of the 1,214,392 total particles. Moreover, DNM accurately and sufficiently counts diagnostic marker EVs present in only 0.93% and 0.17% of particle detections in sera of colorectal cancer patients and healthy controls, demonstrating its potential application to the early detection of colorectal cancer.
Collapse
Affiliation(s)
- Yuichiro Iwamoto
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro 4-6-1, Shibuya, Tokyo, Japan
| | - Benjamin Salmon
- School of Computer Science, University of Birmingham, Birmingham, UK
| | - Yusuke Yoshioka
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Nishishinjuku 6-7-1, Shinjuku, Tokyo, Japan
| | - Ryosuke Kojima
- Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo, Tokyo, Japan
| | - Alexander Krull
- School of Computer Science, University of Birmingham, Birmingham, UK.
| | - Sadao Ota
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro 4-6-1, Shibuya, Tokyo, Japan.
| |
Collapse
|
164
|
Chitteti R, Zuniga-Hertz JP, Masso-Silva JA, Shin J, Niesman I, Bojanowski CM, Kumar AJ, Hepokoski M, Crotty Alexander LE, Patel HH, Roth DM. E-cigarette-induced changes in cell stress and mitochondrial function. Free Radic Biol Med 2025; 228:329-338. [PMID: 39756490 DOI: 10.1016/j.freeradbiomed.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/22/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Inhaling aerosols from electronic nicotine delivery systems, such as e-cigarettes (e-cigs), may pose health risks beyond those caused by nicotine intake. Exposure to e-cig aerosols can lead to the release of exosomes and metabolites into the bloodstream, potentially affecting mitochondrial physiology across the body, leading to chronic inflammatory diseases. In this study we assessed the effects of e-cig use by young healthy human subjects on the circulating exosome profile and markers of cell stress, and also defined the effects of e-cig user plasma on mitochondrial function in endothelial cells (EA. Hy 926) and epithelial cells (A549) via adoptive transfer. E-cig users had altered plasma exosome profiles, with significantly increased levels of cell free mitochondrial DNA (mtDNA), protein carbonyls, and 4-HNE relative to non-users. Plasma from e-cig users decreased maximal mitochondrial respiration and spare capacity of cells, while also increasing metabolic stress, as evidenced by changes in mitochondrial phenotype from basal to stressed in both endothelial and epithelial cells, which was corroborated by electron microscopy demonstrating structural changes in mitochondria. Mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) levels significantly increased in e-cig plasma-subjected cells. Overall, we identified alterations in plasma exosome profiles and increased markers of mitochondrial stress in e-cig users and evidence that circulating factors within plasma from e-cig users drives metabolic stress in endothelial and epithelial cells. Our results imply that e-cig use adversely affects mitochondrial function, leading to stress and potentially chronic inflammation across the body.
Collapse
Affiliation(s)
- Ramamurthy Chitteti
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Anesthesiology, School of Medicine, University of California San Diego, USA.
| | - Juan Pablo Zuniga-Hertz
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Anesthesiology, School of Medicine, University of California San Diego, USA
| | - Jorge A Masso-Silva
- VA San Diego Healthcare System, San Diego, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - John Shin
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - Ingrid Niesman
- San Diego State University, Electron Microscope Facility, 5500 Campanile Dr, San Diego, CA, 92182, USA
| | - Christine M Bojanowski
- Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA; Division of Pulmonary and Critical Care, Tulane University, New Orleans, LA, USA
| | - Avnee J Kumar
- VA San Diego Healthcare System, San Diego, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - Mark Hepokoski
- VA San Diego Healthcare System, San Diego, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - Laura E Crotty Alexander
- VA San Diego Healthcare System, San Diego, CA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, University of California San Diego, USA
| | - Hemal H Patel
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Anesthesiology, School of Medicine, University of California San Diego, USA
| | - David M Roth
- VA San Diego Healthcare System, San Diego, CA, USA; Department of Anesthesiology, School of Medicine, University of California San Diego, USA
| |
Collapse
|
165
|
Bara Z, Gozar H, Nagy N, Gurzu S, Derzsi Z, Forró T, Kovács E, Jung I. Fetoscopic Endoluminal Tracheal Occlusion-Synergic Therapies in the Prenatal Treatment of Congenital Diaphragmatic Hernia. Int J Mol Sci 2025; 26:1639. [PMID: 40004103 PMCID: PMC11855672 DOI: 10.3390/ijms26041639] [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: 01/24/2025] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Congenital diaphragmatic hernia (CDH) is a relatively rare and severe developmental disease. Even with the most recent multidisciplinary therapies, the risk for neonatal mortality and morbidity remains high. Recent advancements in prenatal treatments, alongside experimental and clinical data, suggest that fetoscopic endoluminal tracheal occlusion (FETO) promotes lung development and offers a promising strategy against lung hypoplasia and pulmonary hypertension. It is the only existing direct mechanical therapy that intervenes in the regulation of pulmonary pressure. Its influence on lung development also interferes with tissue homeostasis and cell differentiation; it also enhances inflammation and apoptosis. Its physiopathology on cellular and molecular levels is still poorly understood. Unfortunately, the procedure also carries significant pregnancy-, maternal-, and fetus-related risks. Assessing a multifaceted intervention requires a collective view of all aspects. This scoping review uncovers potential materno-fetal procedure-related risks and highlights innovative solutions. Future research on lung development therapies in CDH may focus on the "dual hit" mechanism, combining molecular-targeting drugs and regenerative medicine with the mechanical nature of FETO for synergistic effects.
Collapse
Affiliation(s)
- Zsolt Bara
- Department of Pediatric Surgery and Orthopedics, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (Z.B.); (Z.D.)
- Clinic of Pediatric Surgery and Orthopedics, Targu Mures, County Emergency Clinical Hospital, 540136 Targu Mures, Romania;
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Horea Gozar
- Department of Pediatric Surgery and Orthopedics, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (Z.B.); (Z.D.)
- Clinic of Pediatric Surgery and Orthopedics, Targu Mures, County Emergency Clinical Hospital, 540136 Targu Mures, Romania;
| | - Nándor Nagy
- Department of Anatomy, Histology and Embryology Semmelweis University, Tűzoltó Street 58, H-1094 Budapest, Hungary;
| | - Simona Gurzu
- Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (S.G.); (I.J.)
- Romanian Academy of Medical Sciences, 030173 Bucharest, Romania
| | - Zoltán Derzsi
- Department of Pediatric Surgery and Orthopedics, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (Z.B.); (Z.D.)
- Clinic of Pediatric Surgery and Orthopedics, Targu Mures, County Emergency Clinical Hospital, 540136 Targu Mures, Romania;
| | - Timea Forró
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Evelyn Kovács
- Clinic of Pediatric Surgery and Orthopedics, Targu Mures, County Emergency Clinical Hospital, 540136 Targu Mures, Romania;
| | - Ioan Jung
- Department of Pathology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (S.G.); (I.J.)
- Romanian Academy of Medical Sciences, 030173 Bucharest, Romania
| |
Collapse
|
166
|
Pacilio S, Lombardi S, Costa R, Paris F, Petrocelli G, Marrazzo P, Cenacchi G, Alviano F. Role of Perinatal Stem Cell Secretome as Potential Therapy for Muscular Dystrophies. Biomedicines 2025; 13:458. [PMID: 40002871 PMCID: PMC11852414 DOI: 10.3390/biomedicines13020458] [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: 12/30/2024] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Inflammation mechanisms play a critical role in muscle homeostasis, and in Muscular Dystrophies (MDs), the myofiber damage triggers chronic inflammation which significantly controls the disease progression. Immunomodulatory strategies able to target inflammatory pathways and mitigate the immune-mediated damage in MDs may provide new therapeutic options. Owing to its capacity of influencing the immune response and enhancing tissue repair, stem cells' secretome has been proposed as an adjunct or standalone treatment for MDs. In this review study, we discuss the challenging points related to the inflammation condition characterizing MD pathology and provide a concise summary of the literature supporting the potential of perinatal stem cells in targeting and modulating the MD inflammation.
Collapse
Affiliation(s)
- Serafina Pacilio
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Sara Lombardi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Roberta Costa
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Francesca Paris
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Giovannamaria Petrocelli
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Pasquale Marrazzo
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Giovanna Cenacchi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Francesco Alviano
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| |
Collapse
|
167
|
Tang L, Zhang W, Qi T, Jiang Z, Tang D. Exosomes play a crucial role in remodeling the tumor microenvironment and in the treatment of gastric cancer. Cell Commun Signal 2025; 23:82. [PMID: 39948541 PMCID: PMC11827163 DOI: 10.1186/s12964-024-02009-7] [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: 09/03/2024] [Accepted: 12/21/2024] [Indexed: 02/16/2025] Open
Abstract
Gastric cancer (GC) is a common and frequent malignant cancer of the digestive system with a poor prognosis. In addition to common therapies such as surgical resection and chemotherapy, novel biological interventions are quite valuable for research. Exosomes are extracellular vesicles (EVs) that originate from various cell types and contain proteins, RNA, DNA, and other components that transmit biological signals and mediate intercellular communication. Numerous studies have shown that exosomes shape the tumor microenvironment (TME) by affecting hypoxia, inflammation, immunity, metabolism, and interstitial changes in the tumor, playing a crucial role in the development and metastasis of GC. This article reviews the important role of exosomes in the TME of GC and explores their potential clinical applications in GC treatment.
Collapse
Affiliation(s)
- Lingyun Tang
- Clinical Medical College, Yangzhou University, Yangzhou, 225000, China
| | - Wenjie Zhang
- School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Teng Qi
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China
| | - Zhengting Jiang
- Center for Liver Transplantation, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430022, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital, Yangzhou, 225000, China.
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Northern Jiangsu People's Hospital; The Yangzhou Clinical Medical College of Xuzhou Medical University; The Yangzhou School of Clinical Medicine of Dalian Medical University; The Yangzhou School of Clinical Medicine of Nanjing Medical University; Northern Jiangsu People's Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou, 225000, China.
| |
Collapse
|
168
|
Aggio JB, Vedam VV, Nisimura LM, da Silva RV, Lovo-Martins MI, Borges BS, Mörking PA, Batista M, Marchini FK, Yamada-Ogatta SF, Pinge-Filho P, Goldenberg S, Eger I, Wowk PF. Trypanosomatid Extracellular Vesicles as Potential Immunogens for Chagas Disease. Int J Mol Sci 2025; 26:1544. [PMID: 40004010 PMCID: PMC11855489 DOI: 10.3390/ijms26041544] [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: 12/30/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Chagas disease remains a significant public health concern, with limited treatment options and an urgent need for novel preventive strategies. Extracellular vesicles (EVs) from Trypanosoma cruzi have been shown to modulate host immune responses, often favoring parasite persistence. In this study, we characterized EVs derived from the non-pathogenic trypanosomatids Trypanosoma rangeli and Phytomonas serpens and evaluated their potential as immunogens capable of inducing cross-protection against T. cruzi infection. Isolated EVs were characterized by Nanoparticle Tracking Analysis (NTA) and electron microscopy. A comparative proteomic analysis of EVs was performed using Mass Spectrometry-Based Proteomic Analysis (LC-MS/MS). The effects of EVs on immunomodulation and T. cruzi infection were assessed through in vitro and in vivo assays, using peripheral blood mononuclear cells (PBMCs) and BALB/c mice. The proteomic analysis identified shared proteins between the EVs of T. rangeli, P. serpens, and T. cruzi, including immunogenic candidates such as calpain-like cysteine peptidase and elongation factor 2. In vitro, pre-stimulation with the T. rangeli EVs reduced infection rates of the host cells by T. cruzi. In vivo, immunization with the EVs from T. rangeli and P. serpens led to a significant reduction in parasitemia in the BALB/c mice challenged with T. cruzi, though this did not translate into improved survival compared to controls. Interestingly, the EVs from T. cruzi also reduced parasitemia but did not confer protection against mortality. These findings suggest that while non-pathogenic trypanosomatid EVs exhibit potential immunogenic properties and can reduce parasitic load, their efficacy in preventing disease progression remains limited. Further research is needed to explore the mechanisms underlying these effects and to optimize EV-based strategies for protective immunity against Chagas disease.
Collapse
Affiliation(s)
- Juliana Bernardi Aggio
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil; (J.B.A.); (V.V.V.); (L.M.N.); (R.V.d.S.); (M.I.L.-M.); (B.S.B.); (P.A.M.); (S.G.)
| | - Verônica Vitória Vedam
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil; (J.B.A.); (V.V.V.); (L.M.N.); (R.V.d.S.); (M.I.L.-M.); (B.S.B.); (P.A.M.); (S.G.)
- Laboratório de Biologia Celular e Protozoologia, Universidade Estadual de Ponta Grossa, Ponta Grossa 84030-900, Brazil
| | - Líndice Mitie Nisimura
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil; (J.B.A.); (V.V.V.); (L.M.N.); (R.V.d.S.); (M.I.L.-M.); (B.S.B.); (P.A.M.); (S.G.)
- Grupo de Imunologia Molecular, Celular e Inteligência Artificial, Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil
| | - Rosiane Valeriano da Silva
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil; (J.B.A.); (V.V.V.); (L.M.N.); (R.V.d.S.); (M.I.L.-M.); (B.S.B.); (P.A.M.); (S.G.)
- Laboratório de Imunopatologia Experimental, Departamento de Imunologia, Parasitologia e Patologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Brazil;
| | - Maria Izabel Lovo-Martins
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil; (J.B.A.); (V.V.V.); (L.M.N.); (R.V.d.S.); (M.I.L.-M.); (B.S.B.); (P.A.M.); (S.G.)
- Laboratório de Imunopatologia Experimental, Departamento de Imunologia, Parasitologia e Patologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Brazil;
| | - Beatriz Santana Borges
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil; (J.B.A.); (V.V.V.); (L.M.N.); (R.V.d.S.); (M.I.L.-M.); (B.S.B.); (P.A.M.); (S.G.)
| | - Patrícia Alves Mörking
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil; (J.B.A.); (V.V.V.); (L.M.N.); (R.V.d.S.); (M.I.L.-M.); (B.S.B.); (P.A.M.); (S.G.)
| | - Michel Batista
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil; (J.B.A.); (V.V.V.); (L.M.N.); (R.V.d.S.); (M.I.L.-M.); (B.S.B.); (P.A.M.); (S.G.)
| | - Fabricio Klerynton Marchini
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil; (J.B.A.); (V.V.V.); (L.M.N.); (R.V.d.S.); (M.I.L.-M.); (B.S.B.); (P.A.M.); (S.G.)
| | - Sueli Fumie Yamada-Ogatta
- Laboratório de Biologia Molecular de Microrganismos, Departamento de Microbiologia, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Brazil;
| | - Phileno Pinge-Filho
- Laboratório de Imunopatologia Experimental, Departamento de Imunologia, Parasitologia e Patologia Geral, Centro de Ciências Biológicas, Universidade Estadual de Londrina, Londrina 86057-970, Brazil;
| | - Samuel Goldenberg
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil; (J.B.A.); (V.V.V.); (L.M.N.); (R.V.d.S.); (M.I.L.-M.); (B.S.B.); (P.A.M.); (S.G.)
| | - Iriane Eger
- Laboratório de Biologia Celular e Protozoologia, Universidade Estadual de Ponta Grossa, Ponta Grossa 84030-900, Brazil
| | - Pryscilla Fanini Wowk
- Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil; (J.B.A.); (V.V.V.); (L.M.N.); (R.V.d.S.); (M.I.L.-M.); (B.S.B.); (P.A.M.); (S.G.)
- Grupo de Imunologia Molecular, Celular e Inteligência Artificial, Instituto Carlos Chagas, Fundação Oswaldo Cruz (FIOCRUZ-PR), Curitiba 81350-010, Brazil
| |
Collapse
|
169
|
Wang Z, Ou Y, Zhu X, Zhou Y, Zheng X, Zhang M, Li S, Yang SN, Juntti-Berggren L, Berggren PO, Zheng X. Differential Regulation of miRNA and Protein Profiles in Human Plasma-Derived Extracellular Vesicles via Continuous Aerobic and High-Intensity Interval Training. Int J Mol Sci 2025; 26:1383. [PMID: 39941151 PMCID: PMC11818269 DOI: 10.3390/ijms26031383] [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: 12/17/2024] [Revised: 01/25/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Both continuous aerobic training (CAT) and high-intensity interval training (HIIT) are recommended to promote health and prevent diseases. Exercise-induced circulating extracellular vesicles (EX-EVs) have been suggested to play essential roles in mediating organ crosstalk, but corresponding molecular mechanisms remain unclear. To assess and compare the systemic effects of CAT and HIIT, five healthy male volunteers were assigned to HIIT and CAT, with a 7-day interval between sessions. Plasma EVs were collected at rest or immediately after each training section, prior to proteomics and miRNA profile analysis. We found that the differentially expressed (DE) miRNAs in EX-EVs were largely involved in the regulation of transcriptional factors, while most of the DE proteins in EX-EVs were identified as non-secreted proteins. Both CAT and HIIT play common roles in neuronal signal transduction, autophagy, and cell fate regulation. Specifically, CAT showed distinct roles in cognitive function and substrate metabolism, while HIIT was more associated with organ growth, cardiac muscle function, and insulin signaling pathways. Interestingly, the miR-379 cluster within EX-EVs was specifically regulated by HIIT, involving several biological functions, including neuroactive ligand-receptor interaction. Furthermore, EX-EVs likely originate from various tissues, including metabolic tissues, the immune system, and the nervous system. Our study provides molecular insights into the effects of CAT and HIIT, shedding light on the roles of EX-EVs in mediating organ crosstalk and health promotion.
Collapse
Affiliation(s)
- Zhenghao Wang
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.W.); (Y.O.); (X.Z.); (Y.Z.); (X.Z.); (S.L.); (P.-O.B.)
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-17176 Stockholm, Sweden; (S.-N.Y.); (L.J.-B.)
| | - Yiran Ou
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.W.); (Y.O.); (X.Z.); (Y.Z.); (X.Z.); (S.L.); (P.-O.B.)
| | - Xinyue Zhu
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.W.); (Y.O.); (X.Z.); (Y.Z.); (X.Z.); (S.L.); (P.-O.B.)
| | - Ye Zhou
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.W.); (Y.O.); (X.Z.); (Y.Z.); (X.Z.); (S.L.); (P.-O.B.)
| | - Xiaowei Zheng
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.W.); (Y.O.); (X.Z.); (Y.Z.); (X.Z.); (S.L.); (P.-O.B.)
- Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Meixia Zhang
- Research Laboratory of Macular Disease, Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Sheyu Li
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.W.); (Y.O.); (X.Z.); (Y.Z.); (X.Z.); (S.L.); (P.-O.B.)
| | - Shao-Nian Yang
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-17176 Stockholm, Sweden; (S.-N.Y.); (L.J.-B.)
| | - Lisa Juntti-Berggren
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-17176 Stockholm, Sweden; (S.-N.Y.); (L.J.-B.)
| | - Per-Olof Berggren
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.W.); (Y.O.); (X.Z.); (Y.Z.); (X.Z.); (S.L.); (P.-O.B.)
- The Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-17176 Stockholm, Sweden; (S.-N.Y.); (L.J.-B.)
| | - Xiaofeng Zheng
- Department of Endocrinology and Metabolism, Research Center for Islet Transplantation, West China Hospital, Sichuan University, Chengdu 610041, China; (Z.W.); (Y.O.); (X.Z.); (Y.Z.); (X.Z.); (S.L.); (P.-O.B.)
| |
Collapse
|
170
|
Yarahmadi A, Dorri Giv M, Hosseininejad R, Rezaie A, Mohammadi N, Afkhami H, Farokhi A. Mesenchymal stem cells and their extracellular vesicle therapy for neurological disorders: traumatic brain injury and beyond. Front Neurol 2025; 16:1472679. [PMID: 39974358 PMCID: PMC11835705 DOI: 10.3389/fneur.2025.1472679] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/08/2025] [Indexed: 02/21/2025] Open
Abstract
Traumatic brain injury (TBI) is a complex condition involving mechanisms that lead to brain dysfunction and nerve damage, resulting in significant morbidity and mortality globally. Affecting ~50 million people annually, TBI's impact includes a high death rate, exceeding that of heart disease and cancer. Complications arising from TBI encompass concussion, cerebral hemorrhage, tumors, encephalitis, delayed apoptosis, and necrosis. Current treatment methods, such as pharmacotherapy with dihydropyridines, high-pressure oxygen therapy, behavioral therapy, and non-invasive brain stimulation, have shown limited efficacy. A comprehensive understanding of vascular components is essential for developing new treatments to improve blood vessel-related brain damage. Recently, mesenchymal stem cells (MSCs) have shown promising results in repairing and mitigating brain damage. Studies indicate that MSCs can promote neurogenesis and angiogenesis through various mechanisms, including releasing bioactive molecules and extracellular vesicles (EVs), which help reduce neuroinflammation. In research, the distinctive characteristics of MSCs have positioned them as highly desirable cell sources. Extensive investigations have been conducted on the regulatory properties of MSCs and their manipulation, tagging, and transportation techniques for brain-related applications. This review explores the progress and prospects of MSC therapy in TBI, focusing on mechanisms of action, therapeutic benefits, and the challenges and potential limitations of using MSCs in treating neurological disorders.
Collapse
Affiliation(s)
- Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Masoumeh Dorri Giv
- Nuclear Medicine Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Hosseininejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Azin Rezaie
- Department of Microbiology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Narges Mohammadi
- Department of Molecular Cell Biology and Microbiology, Faculty of Biological Sciences and Technologies, University of Isfahan, Isfahan, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Arastoo Farokhi
- Department of Anesthesiology, Kermanshah University of Medical Sciences, Imam Reza Hospital, Kermanshah, Iran
| |
Collapse
|
171
|
Wille ACM, Machado MI, Souza SH, da Justa HC, de Fraga-Ferreira ME, Mello EDS, Gremski LH, Veiga SS. Brown Spider Venom Phospholipases D: From Potent Molecules Involved in Pathogenesis of Brown Spider Bites to Molecular Tools for Studying Ectosomes, Ectocytosis, and Its Applications. Toxins (Basel) 2025; 17:70. [PMID: 39998087 PMCID: PMC11860474 DOI: 10.3390/toxins17020070] [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: 11/29/2024] [Revised: 01/24/2025] [Accepted: 01/31/2025] [Indexed: 02/26/2025] Open
Abstract
Accidents caused by Loxosceles spiders, commonly known as brown spiders, are frequent in warm and temperate regions worldwide, with a higher prevalence in South America and the southern United States. In the venoms of species clinically associated with accidents, phospholipases D (PLDs) are the most expressed toxins. This classification is based on the toxins' ability to cleave various phospholipids, with a preference for sphingomyelin. Studies using purified PLDs have demonstrated that these enzymes cleave phospholipids from cells, producing derivatives that can activate leukocytes. A dysregulated inflammatory response is the primary effect following envenomation, leading to dermonecrosis, which is histopathologically characterized by aseptic coagulative necrosis-a key feature of envenomation. Although advances in understanding the structure-function relationship of enzymes have been achieved through molecular biology, heterologous expression, site-directed mutations, crystallography, and bioinformatic analyses-describing PLDs in the venoms of various species and highlighting the conservation of amino acid residues involved in catalysis, substrate binding, and magnesium stabilization-little is known about the cellular biology of these PLDs. Studies have shown that the treatment of various cells with recombinant PLDs stimulates the formation of ectosomes and ectocytosis, events that initiate a cascade of intracellular signaling in PLD-binding cells and lead to the release of extracellular microvesicles. These microvesicles may act as signalosomes for other target cells, thereby triggering an inflammatory response and dermonecrosis. In this review, we will discuss the biochemical properties of PLDs, the target cells that bind to them, and the ectocytosis-dependent pathophysiology of envenoming.
Collapse
Affiliation(s)
- Ana Carolina Martins Wille
- Department of Structural, Molecular Biology and Genetics, State University of Ponta Grossa (UEPG), Ponta Grossa 84030-900, Brazil;
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil; (M.I.M.); (S.H.S.); (H.C.d.J.); (M.E.d.F.-F.); (E.d.S.M.); (L.H.G.)
| | - Mariana Izabele Machado
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil; (M.I.M.); (S.H.S.); (H.C.d.J.); (M.E.d.F.-F.); (E.d.S.M.); (L.H.G.)
| | - Samira Hajjar Souza
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil; (M.I.M.); (S.H.S.); (H.C.d.J.); (M.E.d.F.-F.); (E.d.S.M.); (L.H.G.)
| | - Hanna Câmara da Justa
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil; (M.I.M.); (S.H.S.); (H.C.d.J.); (M.E.d.F.-F.); (E.d.S.M.); (L.H.G.)
| | - Maria Eduarda de Fraga-Ferreira
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil; (M.I.M.); (S.H.S.); (H.C.d.J.); (M.E.d.F.-F.); (E.d.S.M.); (L.H.G.)
| | - Eloise de Souza Mello
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil; (M.I.M.); (S.H.S.); (H.C.d.J.); (M.E.d.F.-F.); (E.d.S.M.); (L.H.G.)
| | - Luiza Helena Gremski
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil; (M.I.M.); (S.H.S.); (H.C.d.J.); (M.E.d.F.-F.); (E.d.S.M.); (L.H.G.)
| | - Silvio Sanches Veiga
- Department of Cell Biology, Federal University of Paraná (UFPR), Curitiba 81530-900, Brazil; (M.I.M.); (S.H.S.); (H.C.d.J.); (M.E.d.F.-F.); (E.d.S.M.); (L.H.G.)
| |
Collapse
|
172
|
Wang Q, Sun J, Jiang H, Yu M. Emerging roles of extracellular vesicles in oral and maxillofacial areas. Int J Oral Sci 2025; 17:11. [PMID: 39900916 PMCID: PMC11791077 DOI: 10.1038/s41368-024-00341-9] [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: 05/03/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 02/05/2025] Open
Abstract
The oral and maxillofacial region is a highly complex area composed of multiple tissue types and bears various critical functions of the human body. Diseases in this region pose significant diagnostic and management challenges; therefore, exploring new strategies for early diagnosis, targeted treatment, and tissue reconstruction is key to improving patient prognosis and quality of life. Extracellular vesicles are a group of heterogeneous lipid-bilayer membrane structures secreted by most cell types, including exosomes, microvesicles, and apoptotic bodies. Present in various body fluids and tissues, they act as messengers via the transfer of nucleic acids, proteins, and metabolites to recipient cells. To date, studies have revealed the different roles of extracellular vesicles in physiological or pathological processes, as well as applications in disease diagnosis, prognosis, and treatment. The importance and tissue specificity of the dental and maxillofacial tissues indicate that extracellular vesicles derived from this region are promising for further research. This paper reviews the published data on extracellular vesicles derived from cells, body fluids, and tissues in oral and maxillofacial regions, summarizes the latest advances in extracellular vesicles from extensive sources, and concludes with a focus on the current research progress and application prospects of engineered exosomes in oral science.
Collapse
Affiliation(s)
- Qianting Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Jiayu Sun
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Haci Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Mengfei Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of the Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China.
| |
Collapse
|
173
|
Ma X, Chen N, Zeng P, He Y, Zhang T, Lu Y, Li Z, Xu J, You J, Zheng Y, Wang L, Luo M, Wu J. Hypericum Perforatum-Derived Exosomes-Like Nanovesicles: A Novel Natural Photosensitizer for Effective Tumor Photodynamic Therapy. Int J Nanomedicine 2025; 20:1529-1541. [PMID: 39925681 PMCID: PMC11806729 DOI: 10.2147/ijn.s510339] [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: 12/09/2024] [Accepted: 01/25/2025] [Indexed: 02/11/2025] Open
Abstract
Background Natural photosensitizers hold potential for photodynamic therapy (PDT) but are often limited by poor visible light absorption. Plant-derived exosome-like nanovesicles offer an innovative platform for enhancing photosensitizer performance. Methods Hypericum perforatum-derived nanovesicles (HPDENs) were characterized using electron microscopy, dynamic light scattering, and proteomic and miRNA sequencing. High-performance liquid chromatography confirmed hypericin content. PDT efficacy was assessed in vitro and in vivo. Results HPDENs exhibited robust photosensitizing properties, generating reactive oxygen species (ROS) through both Type I and Type II pathways upon light activation. In vitro, HPDENs showed light dose-dependent cytotoxicity against human melanoma cells, characterized by elevated ROS production and apoptosis induction. In vivo, HPDEN-mediated PDT significantly suppressed tumor growth and induced extensive tumor necrosis, with no observable toxicity to major organs. Conclusion HPDENs represent a novel plant-derived photosensitizer with dual ROS generation pathways and significant therapeutic efficacy, providing a promising platform for enhancing photodynamic therapy.
Collapse
Affiliation(s)
- Xiaoyu Ma
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Ni Chen
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Peiyuan Zeng
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Yuqian He
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Tao Zhang
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Yu Lu
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Ziyu Li
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Jin Xu
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Jingcan You
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Youkun Zheng
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Liqun Wang
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Mao Luo
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| | - Jianbo Wu
- Basic Medicine Research Innovation Center for Cardiometabolic Diseases, Ministry of Education; Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy; Luzhou Municipal Key Laboratory of Thrombosis and Vascular Biology Southwest Medical University, Luzhou, 646000, People’s Republic of China
| |
Collapse
|
174
|
Teng Y, Luo C, Qiu X, Mu J, Sriwastva MK, Xu Q, Liu M, Hu X, Xu F, Zhang L, Park JW, Hwang JY, Kong M, Liu Z, Zhang X, Xu R, Yan J, Merchant ML, McClain CJ, Zhang HG. Plant-nanoparticles enhance anti-PD-L1 efficacy by shaping human commensal microbiota metabolites. Nat Commun 2025; 16:1295. [PMID: 39900923 PMCID: PMC11790884 DOI: 10.1038/s41467-025-56498-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
Diet has emerged as a key impact factor for gut microbiota function. However, the complexity of dietary components makes it difficult to predict specific outcomes. Here we investigate the impact of plant-derived nanoparticles (PNP) on gut microbiota and metabolites in context of cancer immunotherapy with the humanized gnotobiotic mouse model. Specifically, we show that ginger-derived exosome-like nanoparticle (GELN) preferentially taken up by Lachnospiraceae and Lactobacillaceae mediated by digalactosyldiacylglycerol (DGDG) and glycine, respectively. We further demonstrate that GELN aly-miR159a-3p enhances anti-PD-L1 therapy in melanoma by inhibiting the expression of recipient bacterial phospholipase C (PLC) and increases the accumulation of docosahexaenoic acid (DHA). An increased level of circulating DHA inhibits PD-L1 expression in tumor cells by binding the PD-L1 promoter and subsequently prevents c-myc-initiated transcription of PD-L1. Colonization of germ-free male mice with gut bacteria from anti-PD-L1 non-responding patients supplemented with DHA enhances the efficacy of anti-PD-L1 therapy compared to controls. Our findings reveal a previously unknown mechanistic impact of PNP on human tumor immunotherapy by modulating gut bacterial metabolic pathways.
Collapse
Affiliation(s)
- Yun Teng
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.
| | - Chao Luo
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Central Laboratory, The affiliated Huai'an First People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xiaolan Qiu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Breast and Thyroid Surgery, The affiliated Huai'an First People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Jingyao Mu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Mukesh K Sriwastva
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Qingbo Xu
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Minmin Liu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Breast and Thyroid Surgery, The affiliated Huai'an First People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xin Hu
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fangyi Xu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Lifeng Zhang
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Juw Won Park
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Bioinformatics and Biostatistics, SPHIS, University of Louisville, Louisville, KY, USA
| | - Jae Yeon Hwang
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Maiying Kong
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
- Department of Bioinformatics and Biostatistics, SPHIS, University of Louisville, Louisville, KY, USA
| | - Zhanxu Liu
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Xiang Zhang
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Raobo Xu
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
| | - Jun Yan
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA
| | - Michael L Merchant
- Kidney Disease Program and Clinical Proteomics Center, University of Louisville, Louisville, KY, USA
| | - Craig J McClain
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
| | - Huang-Ge Zhang
- Brown Cancer Center, University of Louisville School of Medicine, Louisville, USA.
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA.
- Robley Rex Veterans Affairs Medical Center, Louisville, KY, USA.
| |
Collapse
|
175
|
Mulzer LM, Felger T, Muñoz LE, Engl G, Reutter H, Schiffer M, Jahromi LP, Boros FA, Zunke F, Arnold P, Hilger AC. Dynamic changes of extracellular vesicles during zebrafish organogenesis. Cell Commun Signal 2025; 23:60. [PMID: 39901276 PMCID: PMC11789338 DOI: 10.1186/s12964-025-02053-x] [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: 05/26/2024] [Accepted: 01/17/2025] [Indexed: 02/05/2025] Open
Abstract
Extracellular Vesicles (EVs) play a crucial role in cell differentiation. Despite its role as a well-established vertebrate model, little is known about EVs during zebrafish embryogenesis. This study investigates EVs during zebrafish embryogenesis, analysing size- and concentration-changes over time.Wild-type AB strain zebrafish larvae (zfl) were collected at 24, 48, 72, and 96 h post fertilization (hpf) and homogenized. EVs were isolated and characterized using flow cytometry, negative staining transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western Blot.Flow cytometry and TEM showed a high purity of the samples. Small EVs (sEVs) and large EVs (lEVs) were differentiated using NTA, showing that zfl use different types of EVs during embryogenesis. It was observed that the total EV number increased significantly over the first 72 hpf, but not proportionally to zfl growth in length. Additionally, sEV size also increased significantly, with a maximum diameter at 72 hpf. Since most organs are formed during the first 72 hpf and from then on mainly maturation and growth occur, the elevated number and larger size before 72 hpf suggests an important role of EVs during zebrafish organogenesis. Since EVs serve as cargo delivery platforms, the increase in size of sEVs may reflect the need for a higher transport capacity.
Collapse
Affiliation(s)
- Linda-Marie Mulzer
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology and Pediatric, Intensive Care University Hospital Erlangen, Erlangen, Germany.
| | - Tim Felger
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology and Pediatric, Intensive Care University Hospital Erlangen, Erlangen, Germany
| | - Luis E Muñoz
- Department of Internal Medicine 3 Rheumatology and Immunology, Friedrich-Alexander- University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen- Nürnberg (FAU), Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | - Gesa Engl
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology and Pediatric, Intensive Care University Hospital Erlangen, Erlangen, Germany
| | - Heiko Reutter
- Department of Pediatrics and Adolescent Medicine, Division of Neonatology and Pediatric, Intensive Care University Hospital Erlangen, Erlangen, Germany
| | - Mario Schiffer
- Department of Internal Medicine 4 Nephrology, Friedrich-Alexander-University Erlangen- Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, 91054, Germany
| | | | - Fanni Annamária Boros
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander- University Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander- University Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
| | - Philipp Arnold
- Institute of Anatomy, Functional and Clinical Anatomy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, 91054, Germany
| | - Alina C Hilger
- Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Erlangen, Germany
- Research Center on Rare Kidney Diseases (RECORD), University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
176
|
Peppicelli S, Calorini L, Bianchini F, Papucci L, Magnelli L, Andreucci E. Acidity and hypoxia of tumor microenvironment, a positive interplay in extracellular vesicle release by tumor cells. Cell Oncol (Dordr) 2025; 48:27-41. [PMID: 39023664 PMCID: PMC11850579 DOI: 10.1007/s13402-024-00969-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] [Accepted: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
The complex and continuously evolving features of the tumor microenvironment, varying between tumor histotypes, are characterized by the presence of host cells and tumor cells embedded in a milieu shaped by hypoxia and low pH, resulting from the frequent imbalance between vascularity and tumor cell proliferation. These microenvironmental metabolic stressors play a crucial role in remodeling host cells and tumor cells, contributing to the stimulation of cancer cell heterogeneity, clonal evolution, and multidrug resistance, ultimately leading to progression and metastasis. The extracellular vesicles (EVs), membrane-enclosed structures released into the extracellular milieu by tumor/host cells, are now recognized as critical drivers in the complex intercellular communication between tumor cells and the local cellular components in a hypoxic/acidic microenvironment. Understanding the intricate molecular mechanisms governing the interactions between tumor and host cells within a hypoxic and acidic microenvironment, triggered by the release of EVs, could pave the way for innovative strategies to disrupt the complex interplay of cancer cells with their microenvironment. This approach may contribute to the development of an efficient and safe therapeutic strategy to combat cancer progression. Therefore, we review the major findings on the release of EVs in a hypoxic/acidic tumor microenvironment to appreciate their role in tumor progression toward metastatic disease.
Collapse
Affiliation(s)
- Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy.
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| |
Collapse
|
177
|
Huang Z, Whitehead B, Nejsum P, Corredig M, Rasmussen MK. Tomato-derived extracellular vesicles increase intestinal zinc transportation by potentially down-regulating the expression of the metallothionein family. Food Res Int 2025; 203:115804. [PMID: 40022334 DOI: 10.1016/j.foodres.2025.115804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 03/03/2025]
Abstract
Extracellular vesicles (EVs) have the ability to regulate physiological and pathological processes across species and have been shown to be present in plants. Tomatoes are one of the most widespread vegetables on the market and exhibit a broad range of health-promoting effects, including antioxidant and anti-inflammatory properties. However, little is known about the bioactivity of tomato-derived EVs. Here, we isolated EVs from tomatoes and explored their gene regulatory potential using array-based transcriptomics. Interestingly, using a differentiated Caco-2 monolayer model, tomato-derived EVs were shown to upregulate the transportation of zinc, which may involve the down-regulation of metallothionein proteins (MTs). Differentiated Caco-2 cells internalized tomato-derived EVs. Post-EV treatment the relative expression levels of MT-related mRNAs within the cells decreased by approximately threefold, accompanied by an approximately twofold reduction in intracellular zinc concentration. Additionally, the amount of secreted zinc in the basolateral medium increased by approximately threefold. Moreover, tomato-derived EV regulation of MT gene expression occurred only in differentiated epithelial cells. This effect was observed in differentiated Caco-2 and HIEC-6 cells, whereas no impact was seen on the MT gene in undifferentiated cells. This mechanistic study uniquely demonstrates the bioactivity of tomato-derived EVs, and for the first time, reveals the ability of plant-derived EVs to modify zinc regulation across the intestinal epithelia. This further suggests the potential of plant-derived EVs as functional food supplements in the future.
Collapse
Affiliation(s)
- Ziyu Huang
- Department of Food Science Aarhus University Denmark
| | - Bradley Whitehead
- Department of Clinical Medicine Aarhus University Aarhus Denmark; Department of Infectious Diseases Aarhus University Hospital Aarhus Denmark
| | - Peter Nejsum
- Department of Clinical Medicine Aarhus University Aarhus Denmark; Department of Infectious Diseases Aarhus University Hospital Aarhus Denmark
| | | | | |
Collapse
|
178
|
Singh D, Prasad S. A Pioneer Review on Lactoferrin-Conjugated Extracellular Nanovesicles for Targeting Cellular Melanoma: Recent Advancements and Future Prospects. Assay Drug Dev Technol 2025; 23:55-69. [PMID: 39654517 DOI: 10.1089/adt.2024.045] [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: 03/21/2025] Open
Abstract
Melanoma, a highly aggressive form of skin cancer, presents a formidable challenge in terms of treatment due to its propensity for metastasis and resistance to conventional therapies. The development of innovative nanocarriers for targeted drug delivery has opened new avenues in cancer therapy. Lactoferrin-conjugated extracellular nanovesicles (LF-EVs) have emerged as a promising vehicle in the targeted treatment of cellular melanoma, owing to their natural biocompatibility, enhanced bioavailability, and ability to traverse biological barriers effectively. This review synthesizes recent advancements in the use of LF-EVs as a novel drug delivery system for melanoma, emphasizing their unique capacity to enhance cellular uptake through LF's receptor-mediated endocytosis pathways. Key studies demonstrate that LF conjugation significantly increases the specificity of extracellular nanovesicles for melanoma cells, minimizes off-target effects, and promotes efficient intracellular drug release. Furthermore, we explore how LF-EVs interact with the tumor microenvironment, potentially inhibiting melanoma progression and metastasis while supporting antitumor immune responses. Future prospects in this field include optimizing LF conjugation techniques, improving the scalability of LF-EV production, and integrating multifunctional payloads to target drug resistance mechanisms. This review highlights the potential of LF-EVs to transform melanoma treatment strategies, bridging current gaps in therapeutic delivery and paving the way for personalized and less invasive melanoma therapies.
Collapse
Affiliation(s)
- Dilpreet Singh
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
- University Centre for Research and Development, Chandigarh University, Mohali, Punjab, India
| | - Sonima Prasad
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| |
Collapse
|
179
|
Rao C, Cater DT, Roy S, Xu J, De Oliveira AG, Evans-Molina C, Piganelli JD, Eizirik DL, Mirmira RG, Sims EK. Beta cell extracellular vesicle PD-L1 as a novel regulator of CD8 + T cell activity and biomarker during the evolution of type 1 diabetes. Diabetologia 2025; 68:382-396. [PMID: 39508879 DOI: 10.1007/s00125-024-06313-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/16/2024] [Indexed: 11/15/2024]
Abstract
AIMS/HYPOTHESIS Surviving beta cells in type 1 diabetes respond to inflammation by upregulating programmed death-ligand 1 (PD-L1) to engage immune cell programmed death protein 1 (PD-1) and limit destruction by self-reactive immune cells. Extracellular vesicles (EVs) and their cargo can serve as biomarkers of beta cell health and contribute to islet intercellular communication. We hypothesised that the inflammatory milieu of type 1 diabetes increases PD-L1 in beta cell EV cargo and that EV PD-L1 may protect beta cells against immune-mediated cell death. METHODS Beta cell lines and human islets were treated with proinflammatory cytokines to model the proinflammatory type 1 diabetes microenvironment. EVs were isolated using ultracentrifugation or size exclusion chromatography and analysed via immunoblot, flow cytometry and ELISA. EV PD-L1 binding to PD-1 was assessed using a competitive binding assay and in vitro functional assays testing the ability of EV PD-L1 to inhibit NOD CD8+ T cells. Plasma EV and soluble PD-L1 were assayed in the plasma of islet autoantibody-positive (Ab+) individuals or individuals with recent-onset type 1 diabetes and compared with levels in non-diabetic control individuals. RESULTS PD-L1 protein co-localised with tetraspanin-associated proteins intracellularly and was detected on the surface of beta cell EVs. Treatment with IFN-α or IFN-γ for 24 h induced a twofold increase in EV PD-L1 cargo without a corresponding increase in the number of EVs. IFN exposure predominantly increased PD-L1 expression on the surface of beta cell EVs and beta cell EV PD-L1 showed a dose-dependent capacity to bind PD-1. Functional experiments demonstrated specific effects of beta cell EV PD-L1 to suppress proliferation and cytotoxicity of murine CD8+ T cells. Plasma EV PD-L1 levels were increased in Ab+individuals, particularly in those positive for a single autoantibody. Additionally, in Ab+ individuals or those who had type 1 diabetes, but not in control individuals, plasma EV PD-L1 positively correlated with circulating C-peptide, suggesting that higher EV PD-L1 could be protective for residual beta cell function. CONCLUSIONS/INTERPRETATION IFN exposure increases PD-L1 on the beta cell EV surface. Beta cell EV PD-L1 binds PD1 and inhibits CD8+ T cell proliferation and cytotoxicity. Circulating EV PD-L1 is higher in Ab+ individuals than in control individuals. Circulating EV PD-L1 levels correlate with residual C-peptide at different stages in type 1 diabetes progression. These findings suggest that EV PD-L1 could contribute to heterogeneity in type 1 diabetes progression and residual beta cell function and raise the possibility that EV PD-L1 could be exploited as a means to inhibit immune-mediated beta cell death.
Collapse
Affiliation(s)
- Chaitra Rao
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Daniel T Cater
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Saptarshi Roy
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jerry Xu
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andre G De Oliveira
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jon D Piganelli
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Raghavendra G Mirmira
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL, USA
| | - Emily K Sims
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA.
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
180
|
Bao Y, Chen Y, Deng X, Wang Y, Zhang Y, Xu L, Huang W, Cheng S, Zhang H, Xie M. Boron Clusters Escort Doxorubicin Squashing Into Exosomes and Overcome Drug Resistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412501. [PMID: 39721006 PMCID: PMC11831453 DOI: 10.1002/advs.202412501] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/09/2024] [Indexed: 12/28/2024]
Abstract
Exosome-based drug delivery holds significant promise for cancer chemotherapy. However, current methods for loading drugs into exosomes are inefficient and cost-prohibitive for practical application. In this study, boron clusters are mixed with doxorubicin (DOX) and exosomes, enabling the efficient encapsulation of DOX into exosomes through a superchaotropic effect. Exosomes loaded with DOX and boron clusters (EDB) exhibit superior permeability and the ability to deliver higher concentrations of DOX into DOX-resistant breast cancer cells. Mechanistic analysis reveals that boron clusters form a supramolecular complex with DOX, which facilitates sustained drug release and effectively inhibits P-glycoprotein-mediated DOX efflux. As a result, EDB significantly enhance apoptosis in DOX-resistant breast cancer cells and suppress tumor growth in cases where DOX alone is ineffective, thereby extending the survival of nude mice. In summary, boron clusters effectively facilitate the incorporation of DOX into exosomes and inhibit DOX efflux, offering a novel strategy to overcome DOX resistance.
Collapse
Affiliation(s)
- Yi‐Ru Bao
- College of Chemistry and Molecular SciencesWuhan University299 Bayi RoadWuhan430072P. R. China
| | - Yi‐Jing Chen
- College of Chemistry and Molecular SciencesWuhan University299 Bayi RoadWuhan430072P. R. China
| | - Xue‐Fan Deng
- College of Chemistry and Molecular SciencesEngineering Research Center of Organosilicon Compounds & MaterialsMinistry of Education and National Demonstration Center for Experimental ChemistryWuhan University299 Bayi RoadWuhan430072P. R. China
| | - Yi‐Ke Wang
- College of Chemistry and Molecular SciencesWuhan University299 Bayi RoadWuhan430072P. R. China
| | - Yu‐Xin Zhang
- College of Chemistry and Molecular SciencesWuhan University299 Bayi RoadWuhan430072P. R. China
| | - Li‐Li Xu
- College of Chemistry and Molecular SciencesWuhan University299 Bayi RoadWuhan430072P. R. China
| | - Wei‐Hua Huang
- College of Chemistry and Molecular SciencesWuhan University299 Bayi RoadWuhan430072P. R. China
- Department of Hepatobiliary and Pancreatic SurgeryZhongnan HospitalWuhan University169East Lake RoadWuhan430072P. R. China
| | - Shi‐Bo Cheng
- School of Laboratory MedicineHubei University of Chinese Medicine16 Huangjia Lake West RoadWuhan430065P. R. China
| | - Hai‐Bo Zhang
- College of Chemistry and Molecular SciencesEngineering Research Center of Organosilicon Compounds & MaterialsMinistry of Education and National Demonstration Center for Experimental ChemistryWuhan University299 Bayi RoadWuhan430072P. R. China
| | - Min Xie
- College of Chemistry and Molecular SciencesWuhan University299 Bayi RoadWuhan430072P. R. China
| |
Collapse
|
181
|
Figueroa‐Hall LK, Burrows K, Alarbi AM, Hannafon BN, Hladik C, Tan C, Ramesh R, Stewart JL, Risbrough VB, Paulus MP, Teague TK. Comparison of Methods for Isolation and Characterization of Total and Astrocyte-Enriched Extracellular Vesicles From Human Serum and Plasma. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70035. [PMID: 39958973 PMCID: PMC11826443 DOI: 10.1002/jex2.70035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 02/18/2025]
Abstract
Extracellular vesicles (EV) which play critical roles in intercellular communication, have garnered interest as biomarkers with researchers studying brain-related disease processes due to their ability to be isolated from various biofluids. Astrocytes, a type of glial cell, play a critical role in neuronal regulation and function. As such, EV enriched from astrocytes can be used to interrogate cargo and identify mechanisms by which astrocytes communicate with other cells of the central nervous system or shed light on pathophysiological conditions. This manuscript compared five EV isolation methods (differential ultracentrifugation [dUC], precipitation, precipitation + purification, silicon carbon resin and size exclusion chromatography [SEC]) using small volumes of human plasma and serum with a focus on immunocapture of astrocyte-enriched EV (AEEV), with the excitatory amino acid transporter 1, or GLAST. Methods were evaluated on yield, purity, recovery and downstream application to include immunoassays for tetraspanin, immune and astrocyte markers. Results revealed that whilst precipitation-based methods such as ExoQuick yielded higher EV concentrations, size exclusion (SmartSEC, qEV) provided greater purity, emphasizing a trade-off between yield and purity. This study provides a comprehensive resource for researchers in selecting EV isolation methods tailored to small biobanked clinical samples, with the goal of advancing biomarker discovery in Neuroscience.
Collapse
Affiliation(s)
- Leandra K. Figueroa‐Hall
- Laureate Institute for Brain ResearchTulsaOklahomaUSA
- Oxley College of Health and Natural SciencesThe University of TulsaTulsaOklahomaUSA
| | | | - Ahlam M. Alarbi
- Integrative Immunology CenterUniversity of Oklahoma (OU)TulsaOklahomaUSA
| | - Bethany N. Hannafon
- Department of Obstetrics and GynecologyOU Health Sciences Center (OUHSC)Oklahoma CityOklahomaUSA
- Department of Cell BiologyOUHSCOklahoma CityOklahomaUSA
- OU Health Stephenson Cancer CenterOklahoma CityOklahomaUSA
| | - Cole Hladik
- Department of Obstetrics and GynecologyOU Health Sciences Center (OUHSC)Oklahoma CityOklahomaUSA
- Department of Cell BiologyOUHSCOklahoma CityOklahomaUSA
| | - Chibing Tan
- Integrative Immunology CenterUniversity of Oklahoma (OU)TulsaOklahomaUSA
| | - Rajagopal Ramesh
- OU Health Stephenson Cancer CenterOklahoma CityOklahomaUSA
- Department of PathologyOUHSCOklahoma CityOklahomaUSA
| | - Jennifer L. Stewart
- Laureate Institute for Brain ResearchTulsaOklahomaUSA
- Oxley College of Health and Natural SciencesThe University of TulsaTulsaOklahomaUSA
| | - Victoria B. Risbrough
- VA Center of Excellence for Stress and Mental HealthLa JollaCaliforniaUSA
- Department of PsychiatryUniversity of CaliforniaSan Diego, La JollaCaliforniaUSA
| | - Martin P. Paulus
- Laureate Institute for Brain ResearchTulsaOklahomaUSA
- Oxley College of Health and Natural SciencesThe University of TulsaTulsaOklahomaUSA
| | - T. Kent Teague
- Integrative Immunology CenterUniversity of Oklahoma (OU)TulsaOklahomaUSA
- Department of Biochemistry and MicrobiologyThe Oklahoma State University Center for Health SciencesTulsaOklahomaUSA
- Department of Pharmaceutical SciencesOUHSCOklahoma CityOklahomaUSA
- Departments of Surgery and PsychiatrySchool of Community MedicineOU, TulsaOklahomaUSA
| |
Collapse
|
182
|
Branco H, Xavier CPR, Riganti C, Vasconcelos MH. Hypoxia as a critical player in extracellular vesicles-mediated intercellular communication between tumor cells and their surrounding microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189244. [PMID: 39672279 DOI: 10.1016/j.bbcan.2024.189244] [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: 06/18/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
In the past years, increasing attention has been paid to the role of extracellular vesicles (EVs) as mediators of intercellular communication in cancer. These small size particles mediate the intercellular transfer of important bioactive molecules involved in malignant initiation and progression. Hypoxia, or low partial pressure of oxygen, is recognized as a remarkable feature of solid tumors and has been demonstrated to exert a profound impact on tumor prognosis and therapeutic efficacy. Indeed, the high-pitched growth rate and chaotic neovascular architecture that embodies solid tumors results in a profound reduction in oxygen pressure within the tumor microenvironment (TME). In response to oxygen-deprived conditions, tumor cells and their surrounding milieu develop homeostatic adaptation mechanisms that contribute to the establishment of a pro-tumoral phenotype. Latest evidence suggests that the hypoxic microenvironment that surrounds the tumor bulk may be a clincher for the observed elevated levels of circulating EVs in cancer patients. Thus, it is proposed that EVs may play a role in mediating intercellular communication in response to hypoxic conditions. This review focuses on the EVs-mediated crosstalk that is established between tumor cells and their surrounding immune, endothelial, and stromal cell populations, within the hypoxic TME.
Collapse
Affiliation(s)
- Helena Branco
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Cristina P R Xavier
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, University Institute of Health Sciences - CESPU, 4585-116 Gandra, Portugal.
| | - Chiara Riganti
- Department of Oncology, University of Torino, 10126 Torino, Italy; Interdepartmental Research Center for Molecular Biotechnology "G. Tarone", University of Torino, 10126 Torino, Italy
| | - M Helena Vasconcelos
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, 4200-135 Porto, Portugal; Cancer Drug Resistance Group, IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; Department of Biological Sciences, FFUP - Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
183
|
Bebesi T, Pálmai M, Szigyártó IC, Gaál A, Wacha A, Bóta A, Varga Z, Mihály J. Surface-enhanced infrared spectroscopic study of extracellular vesicles using plasmonic gold nanoparticles. Colloids Surf B Biointerfaces 2025; 246:114366. [PMID: 39531836 DOI: 10.1016/j.colsurfb.2024.114366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Extracellular vesicles (EVs), sub-micrometer lipid-bound particles released by most cells, are considered a novel area in both biology and medicine. Among characterization methods, infrared (IR) spectroscopy, especially attenuated total reflection (ATR), is a rapidly emerging label-free tool for molecular characterization of EVs. The relatively low number of vesicles in biological fluids (∼1010 particle/mL), however, and the complex content of the EVs' milieu (protein aggregates, lipoproteins, buffer molecules) might result in poor signal-to-noise ratio in the IR analysis of EVs. Exploiting the increment of the electromagnetic field at the surface of plasmonic nanomaterials, surface-enhanced infrared spectroscopy (SEIRS) provides an amplification of characteristic IR signals of EV samples. Negatively charged citrate-capped and positively charged cysteamine-capped gold nanoparticles with around 10 nm diameter were synthesized and tested with blood-derived EVs. Both types of gold nanoparticles contributed to an enhancement of the EVs' IR spectroscopic signature. Joint evaluation of UV-Vis and IR spectroscopic results, supported by FF-TEM images, revealed that proper interaction of gold nanoparticles with EVs is crucial, and an aggregation or clustering of gold nanoparticles is necessary to obtain the SEIRS effect. Positively charged gold nanoparticles resulted in higher enhancement, probably due to electrostatic interaction with EVs, commonly negatively charged.
Collapse
Affiliation(s)
- Tímea Bebesi
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary; Hevesy György PhD School of Chemistry, Eötvös Lóránd University, Pázmány Péter sétány 1/A, Budapest 1117, Hungary
| | - Marcell Pálmai
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary
| | - Imola Csilla Szigyártó
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary
| | - Anikó Gaál
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary
| | - András Wacha
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary
| | - Attila Bóta
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary
| | - Zoltán Varga
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary; Department of Physical Chemistry and Materials Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, Budapest 1111, Hungary
| | - Judith Mihály
- Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences (RCNS), Magyar tudósok körútja 2, Budapest 1117, Hungary; Department of Chemistry, Eszterházy Károly Catholic University, Leányka u. 6, Eger 3300, Hungary.
| |
Collapse
|
184
|
Chen M, Huang B, Su X. Mesenchymal stem cell-derived extracellular vesicles in periodontal bone repair. J Mol Med (Berl) 2025; 103:137-156. [PMID: 39821702 DOI: 10.1007/s00109-025-02513-4] [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: 08/08/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
Periodontitis is a chronic inflammatory disease that destroys tooth-supporting structures and poses significant public health challenges due to its high prevalence and links to systemic health conditions. Traditional treatments are effective in reducing the inflammatory response and improving the clinical symptoms of periodontitis. However, these methods are challenging to achieve an ideal treatment effect in alveolar bone repair. Mesenchymal stem cells (MSCs) represent a potential alternative for the treatment of periodontal bone defects due to their self-renewal and differentiation capabilities. Recent research indicates that MSCs exert their effects primarily through paracrine mechanisms. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) serve as pivotal mediators in intercellular communication, transferring microRNAs (miRNAs), messenger RNAs (mRNAs), proteins, and cytokines to recipient cells, thereby emulating the therapeutic effects of MSCs. In periodontitis, MSC-EVs play a pivotal role in immunomodulation and bone remodeling, thereby facilitating periodontal bone repair. As a cell-free therapy, MSC-EVs demonstrate considerable clinical potential due to their specialized membrane structure, minimal immunogenicity, low toxicity, high biocompatibility, and nanoscale size. This review indicates that MSC-EVs represent a promising approach for periodontitis treatment, with the potential to overcome the limitations of traditional therapies and offer a more effective solution for bone repair in periodontal disease. In this review, we introduce MSC-EVs, emphasizing their mechanisms and clinical applications in periodontal bone repair. It synthesizes recent advances, existing challenges, and future prospects to present up-to-date information and novel techniques for periodontal regeneration and to guide the improvement of MSC-EV therapy in clinical practice.
Collapse
Affiliation(s)
- Mengbing Chen
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases &, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Bo Huang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases &, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiaoxia Su
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases &, Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
185
|
Li H, Wang L, Cheng H, Zhang Q, Wang S, Zhong W, He C, Wei Q. Unlocking the Potential of Extracellular Vesicles in Cardiovascular Disease. J Cell Mol Med 2025; 29:e70407. [PMID: 39910696 PMCID: PMC11798870 DOI: 10.1111/jcmm.70407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 12/02/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
Extracellular vesicles (EVs) are micro-nanoscale biological particles encapsulated by phospholipid bilayers, which regulate cell migration, angiogenesis and tumour cell growth by transmitting various biomolecules such as nucleic acids and proteins. EVs are composed of exosomes, microparticles and apoptotic bodies. Its benefits pass through biofilms and are not degraded by various enzymes, so it can be used as a biomarker in potential diseases and has attracted much attention from researchers. Current studies have found that EVs are involved in the development of various cardiovascular diseases (CVD), such as heart failure and myocardial ischemia-reperfusion injury. In addition, stem cell-derived EVs play an important role in the diagnosis and treatment of a variety of CVD. In this review, we present the biological features of EVs, the role of EVs in various CVD, and the challenges they encounter in the treatment of CVD.
Collapse
Affiliation(s)
- Hanbin Li
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Lu Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Hongxin Cheng
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Qing Zhang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Shiqi Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Wen Zhong
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
186
|
Sonallya T, Juhász T, Szigyártó IC, Ilyés K, Singh P, Khamari D, Buzás EI, Varga Z, Beke-Somfai T. Categorizing interaction modes of antimicrobial peptides with extracellular vesicles: Disruption, membrane trespassing, and clearance of the protein corona. J Colloid Interface Sci 2025; 679:496-509. [PMID: 39378685 DOI: 10.1016/j.jcis.2024.09.244] [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: 05/04/2024] [Revised: 08/21/2024] [Accepted: 09/29/2024] [Indexed: 10/10/2024]
Abstract
Host antimicrobial peptides (AMPs) and extracellular vesicles (EVs) are known to play important roles as part of the immune system, from antimicrobial actions to immune regulation. Recent results also demonstrate that EVs could serve as carriers for AMPs. Related, it was shown that some AMPs can remove the protein corona (PC), the externally adsorbed layer of proteins, from EVs which can be exploited for subtractive proteomics strategies. The interaction of these compounds is thus interesting for multiple reasons from better insight to natural processes to direct applications in EV-based bioengineering. However, we have only limited information on the various ways these species may interact with each other. To reach a broader overview, here we selected twenty-six AMPs, including cell-penetrating peptides (CPPs), and investigated their interactions with red blood cell-derived vesicles (REVs). For this, we employed a complex lipid biophysics including linearly polarized light spectroscopy, flow cytometry, nanoparticle tracking analysis, electron microscopy and also zeta-potential measurements. This enabled the categorization of these peptides into distinct groups. At specific low concentrations, peptides such as LL-37 and lasioglossin-III were effective in PC elimination with minimal disruption of the membrane. In contrast, AMPs like KLA, bradykinin, histatin-5, and most of the tested CPPs (e.g. octa-arginine, penetratin, and buforin II), demonstrate cell-penetrating mechanisms as they could sustain large peptide concentrations with minimal membrane damage. The systematic overview presented here shows the potential mechanism of how AMPs and EVs could interact in vivo, and also how certain peptides may be employed to manipulate EVs for specific applications.
Collapse
Affiliation(s)
- Tasvilla Sonallya
- Biomolecular Self-assembly Research Group, Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Budapest H-1117, Magyar Tudósok Körútja 2, Hungary; Hevesy György PhD School of Chemistry, ELTE Eötvös Loránd University, Budapest H-1117, Pázmány Péter Sétány 1/A, Hungary
| | - Tünde Juhász
- Biomolecular Self-assembly Research Group, Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Budapest H-1117, Magyar Tudósok Körútja 2, Hungary
| | - Imola Cs Szigyártó
- Biomolecular Self-assembly Research Group, Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Budapest H-1117, Magyar Tudósok Körútja 2, Hungary
| | - Kinga Ilyés
- Hevesy György PhD School of Chemistry, ELTE Eötvös Loránd University, Budapest H-1117, Pázmány Péter Sétány 1/A, Hungary; Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Budapest H-1117, Magyar Tudósok Körútja 2, Hungary
| | - Priyanka Singh
- Biomolecular Self-assembly Research Group, Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Budapest H-1117, Magyar Tudósok Körútja 2, Hungary
| | - Delaram Khamari
- Department of Genetics, Cell and Immunobiology, Semmelweis University, H-1089 Budapest, Nagyvárad tér 4, Hungary
| | - Edit I Buzás
- Department of Genetics, Cell and Immunobiology, Semmelweis University, H-1089 Budapest, Nagyvárad tér 4, Hungary; HCEMM Extracellular Vesicle Research Group, Semmelweis University, H-1089 Budapest, Nagyvárad tér 4, Hungary; HUN-REN-SU Translational Extracellular Vesicle Research Group, H-1089 Budapest, Nagyvárad tér 4, Hungary
| | - Zoltán Varga
- Biological Nanochemistry Research Group, Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Budapest H-1117, Magyar Tudósok Körútja 2, Hungary; Department of Physical Chemistry and Materials Science, Budapest University of Technology and Economics, Műegyetem rkp. 3, Budapest 1111, Hungary
| | - Tamás Beke-Somfai
- Biomolecular Self-assembly Research Group, Institute of Materials and Environmental Chemistry, HUN-REN Research Centre for Natural Sciences, Budapest H-1117, Magyar Tudósok Körútja 2, Hungary.
| |
Collapse
|
187
|
Wang C, Zhang Y, Wang J, Han Y, Wang Y, Sun M, Liang Y, Huang M, Yu Y, Hu H, Liu H, Han L. Single-Cell Isolation Chip Integrated with Multicolor Barcode Array for High-Throughput Single-Cell Exosome Profiling in Tissue Samples. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411259. [PMID: 39659120 DOI: 10.1002/adma.202411259] [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: 07/31/2024] [Revised: 10/28/2024] [Indexed: 12/12/2024]
Abstract
Exosomes, functional biomarkers involved in cancer progression, have gained widespread attention for promoting tumor formation, growth, and metastasis. Current bulk exosome detections in bodily fluids enable cancer functional analysis, but average secretion levels from cell populations, losing parent cell information and ignoring exosome heterogeneity from diverse cell subgroups, necessitating an effective platform for analyzing single-cell exosome functional heterogeneity. Here, a high-throughput platform is presented, capable of efficient single-cell isolation and multi-color exosome phenotype analysis, as well as quantifying trace exosomes secreted by single cells. Photothermal-driven single-cell chips achieve significant single-cell isolation efficiency (≈97%) within 5 min, facilitating the ultra-high throughput single-cell exosome analysis. By conducting mass spectrometry and protein interaction of breast cancer exosome phenotypic proteins, key exosome phenotypes are identified. Tens of thousands of single cells from breast cancer cell lines, and clinical tissues are analyzed, revealing various subgroup differences. The study finds more CD44 and EGFR co-expressing exosome subgroups in breast cancer cell lines, while immune-evasion PD-L1 high-phenotype exosome subgroups are primarily presented in complex tumor microenvironments, especially in HER2-positive tissues. This platform offers powerful single-cell isolation, exosome quantification, and phenotypic analysis capabilities, making it a powerful tool for advancing single-cell exosome analysis in cancer research.
Collapse
Affiliation(s)
- Chao Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Yu Zhang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
- Department of Integrated Circuits, Shandong University, Jinan, 250100, China
| | - Jianbo Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, Shandong University, Jinan, 250012, China
| | - Yunrui Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Yihe Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Mingyuan Sun
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Yanbo Liang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Miao Huang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Yang Yu
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Huili Hu
- School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
- Department of Integrated Circuits, Shandong University, Jinan, 250100, China
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China
| |
Collapse
|
188
|
Liu L, Liu W, Han Z, Shan Y, Xie Y, Wang J, Qi H, Xu Q. Extracellular Vesicles-in-Hydrogel (EViH) targeting pathophysiology for tissue repair. Bioact Mater 2025; 44:283-318. [PMID: 39507371 PMCID: PMC11539077 DOI: 10.1016/j.bioactmat.2024.10.017] [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: 04/06/2024] [Revised: 10/08/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
Regenerative medicine endeavors to restore damaged tissues and organs utilizing biological approaches. Utilizing biomaterials to target and regulate the pathophysiological processes of injured tissues stands as a crucial method in propelling this field forward. The Extracellular Vesicles-in-Hydrogel (EViH) system amalgamates the advantages of extracellular vesicles (EVs) and hydrogels, rendering it a prominent biomaterial in regenerative medicine with substantial potential for clinical translation. This review elucidates the development and benefits of the EViH system in tissue regeneration, emphasizing the interaction and impact of EVs and hydrogels. Furthermore, it succinctly outlines the pathophysiological characteristics of various types of tissue injuries such as wounds, bone and cartilage injuries, cardiovascular diseases, nerve injuries, as well as liver and kidney injuries, underscoring how EViH systems target these processes to address related tissue damage. Lastly, it explores the challenges and prospects in further advancing EViH-based tissue regeneration, aiming to impart a comprehensive understanding of EViH. The objective is to furnish a thorough overview of EViH in enhancing regenerative medicine applications and to inspire researchers to devise innovative tissue engineering materials for regenerative medicine.
Collapse
Affiliation(s)
- Lubin Liu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Wei Liu
- Department of Emergency Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266003, China
| | - Zeyu Han
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Yansheng Shan
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Yutong Xie
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Jialu Wang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| | - Hongzhao Qi
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Quanchen Xu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
- School of Stomatology, Qingdao University, Qingdao, 266023, China
| |
Collapse
|
189
|
Saneh H, Wanczyk H, Walker J, Finck C. Stem cell-derived extracellular vesicles: a potential intervention for Bronchopulmonary Dysplasia. Pediatr Res 2025; 97:497-509. [PMID: 39251881 PMCID: PMC12014501 DOI: 10.1038/s41390-024-03471-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 06/06/2024] [Accepted: 07/16/2024] [Indexed: 09/11/2024]
Abstract
Despite advances in neonatal care, the incidence of Bronchopulmonary Dysplasia (BPD) remains high among extreme preterm infants. The pathogenesis of BPD is multifactorial, with inflammation playing a central role. There is strong evidence that stem cell therapy reduces inflammatory changes and restores normal lung morphology in animal models of hyperoxia-induced lung injury. These therapeutic effects occur without significant engraftment of the stem cells in the host lung, suggesting more of a paracrine mechanism mediated by their secretome. In addition, there are multiple concerns with stem cell therapy which may be alleviated by administering only the effective vesicles instead of the cells themselves. Extracellular vesicles (EVs) are cell-derived components secreted by most eukaryotic cells. They can deliver their bioactive cargo (mRNAs, microRNAs, proteins, growth factors) to recipient cells, which makes them a potential therapeutic vehicle in many diseases, including BPD. The following review will highlight recent studies that investigate the effectiveness of EVs derived from stem cells in preventing or repairing injury in the preterm lung, and the potential mechanisms of action that have been proposed. Current limitations will also be discussed as well as suggestions for advancing the field and easing the transition towards clinical translation in evolving or established BPD. IMPACT: Extracellular vesicles (EVs) derived from stem cells are a potential intervention for neonatal lung diseases. Their use might alleviate the safety concerns associated with stem cell therapy. This review highlights recent studies that investigate the effectiveness of stem cell-derived EVs in preclinical models of bronchopulmonary dysplasia. It adds to the existing literature by elaborating on the challenges associated with EV research. It also provides suggestions to advance the field and ease the transition towards clinical applications. Optimizing EV research could ultimately improve the quality of life of extreme preterm infants born at vulnerable stages of lung development.
Collapse
Affiliation(s)
- Hala Saneh
- Department of Neonatal Medicine, Connecticut Children's Medical Center, Hartford, CT, USA.
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA.
| | - Heather Wanczyk
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
| | - Joanne Walker
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
| | - Christine Finck
- Department of Pediatrics, University of Connecticut Health Center, Farmington, CT, USA
- Department of Pediatric Surgery, Connecticut Children's Medical Center, Hartford, CT, USA
| |
Collapse
|
190
|
Doerfler R, Yerneni S, LoPresti S, Chaudhary N, Newby A, Melamed JR, Malaney A, Whitehead KA. Maternal milk cell components are uptaken by infant liver macrophages via extracellular vesicle mediated transport. FASEB J 2025; 39:e70340. [PMID: 39835705 PMCID: PMC11748825 DOI: 10.1096/fj.202402365r] [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: 10/10/2024] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Milk is a multifaceted biofluid that is essential for infant nutrition and development, yet its cellular and bioactive components, particularly maternal milk cells, remain understudied. Early research on milk cells indicated that they cross the infant's intestinal barrier and accumulate within systemic organs. However, due to the absence of modern analytical techniques, these studies were limited in scope and mechanistic analysis. To overcome this knowledge gap, we have investigated the transintestinal transport of milk cells and components in pups over a 21-day period. Studies employed a mT/mG foster nursing model in which milk cells express a membrane-bound fluorophore, tdTomato. Using flow cytometry, we tracked the transport of milk cell-derived components across local and systemic tissues, including the intestines, blood, thymus, mesenteric lymph nodes, and liver. These experiments identified milk-derived fluorescent signals in intestinal epithelial and immune cells as well as liver macrophages in 7-day-old pups. However, the minute numbers of macrophages in mouse milk suggest that maternal cells are not systemically accumulating in the infant; instead, pup macrophages are consuming milk cell membrane components, such as apoptotic bodies or extracellular vesicles (EVs). Ex vivo experiments using primary macrophages support this hypothesis, showing that immune cells preferentially consumed EVs over milk cells. Together, these data suggest a more complex interplay between milk cells and the infant's immune and digestive systems than previously recognized and highlight the need for future research on the role of milk cells in infant health.
Collapse
Affiliation(s)
- Rose Doerfler
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | | | - Samuel LoPresti
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Namit Chaudhary
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Alexandra Newby
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Jilian R. Melamed
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Angela Malaney
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Kathryn A. Whitehead
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| |
Collapse
|
191
|
Joung H, Jang GJ, Jeong JY, Lim G, Han SY. Evaluating the In Situ Effects of Whole Protein Coronas on the Biosensing of Antibody-Immobilized Nanoparticles Using Two-Color Fluorescence Nanoparticle Tracking Analysis. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:220. [PMID: 39940196 PMCID: PMC11820540 DOI: 10.3390/nano15030220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/14/2025]
Abstract
The formation of protein coronas around engineered nanoparticles (ENPs) in biological environments is critical in nanomedicine, as these coronas significantly influence the biological behavior of ENPs. Despite extensive research on protein coronas, understanding the in situ influence of whole (soft plus hard) protein coronas has remained challenging. In this study, we demonstrate a strategy to assess the in situ effects of whole coronas on the model biosensing of anti-IgG using IgG-conjugated gold nanoparticles (IgG-AuNPs) through fluorescence nanoparticle tracking analysis (F-NTA), which enables the selective tracking of fluorescent particles within complex media. In our approach, anti-IgG and IgG-AuNPs were labeled with distinct fluorescent dyes. The accordance in hydrodynamic diameter distributions observed at two different wavelengths verifies the successful capture of anti-IgG on the IgG-AuNPs. The counting of fluorescent anti-IgG within the size distribution allows for a quantitative assessment of biosensing efficiency. This method was applied to evaluate the effects of four protein coronas-human serum albumin, high-density lipoproteins, immunoglobulin G, and fibrinogen-as well as their mixture across varying incubation times and concentrations. The results suggest that the physical presence of whole protein coronas surrounding the IgG-AuNPs may assist the biosensing interaction in situ rather than screening it.
Collapse
Affiliation(s)
| | | | | | | | - Sang Yun Han
- Department of Chemistry, Gachon University, Seongnam 13120, Gyeonggi, Republic of Korea; (H.J.); (G.J.J.)
| |
Collapse
|
192
|
Karbanová J, Thamm K, Fargeas CA, Deniz IA, Lorico A, Corbeil D. Prominosomes - a particular class of extracellular vesicles containing prominin-1/CD133? J Nanobiotechnology 2025; 23:61. [PMID: 39881297 PMCID: PMC11776279 DOI: 10.1186/s12951-025-03102-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: 09/17/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025] Open
Abstract
Extracellular membrane vesicles (EVs) offer promising values in various medical fields, e.g., as biomarkers in liquid biopsies or as native (or bioengineered) biological nanocarriers in tissue engineering, regenerative medicine and cancer therapy. Based on their cellular origin EVs can vary considerably in composition and diameter. Cell biological studies on mammalian prominin-1, a cholesterol-binding membrane glycoprotein, have helped to reveal new donor membranes as sources of EVs. For instance, small EVs can originate from microvilli and primary cilia, while large EVs might be produced by transient structures such as retracting cellular extremities of cancer cells during the mitotic rounding process, and the midbody at the end of cytokinesis. Here, we will highlight the various subcellular origins of prominin-1+ EVs, also called prominosomes, and the potential mechanism(s) regulating their formation. We will further discuss the molecular and cellular characteristics of prominin-1, notably those that have a direct effect on the release of prominin-1+ EVs, a process that might be directly implicated in donor cell reprogramming of stem and cancer stem cells. Prominin-1+ EVs also mediate intercellular communication during embryonic development and adult homeostasis in healthy individuals, while disseminating biological information during diseases.
Collapse
Affiliation(s)
- Jana Karbanová
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany.
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
- Tissue Engineering Laboratories, Biotechnology Center, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany.
| | - Kristina Thamm
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
- denovoMATRIX GmbH, Tatzberg 47, 01307, Dresden, Germany
| | - Christine A Fargeas
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Ilker A Deniz
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Aurelio Lorico
- College of Osteopathic Medicine, Touro University Nevada, 874 American Pacific Drive, Henderson, NV, 89014, USA
| | - Denis Corbeil
- Biotechnology Center (BIOTEC) and Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany.
- Tissue Engineering Laboratories, Medizinische Fakultät der Technischen Universität Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
- Tissue Engineering Laboratories, Biotechnology Center, Technische Universität Dresden, Tatzberg 47-49, 01307, Dresden, Germany.
| |
Collapse
|
193
|
Orefice NS, Petrillo G, Pignataro C, Mascolo M, De Luca G, Verde S, Pentimalli F, Condorelli G, Quintavalle C. Extracellular vesicles and microRNAs in cancer progression. Adv Clin Chem 2025; 125:23-54. [PMID: 39988407 DOI: 10.1016/bs.acc.2024.11.004] [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: 02/25/2025]
Abstract
Extracellular vesicles (EVs) have emerged as critical mediators of intercellular communication in cancer. These membranous structures, secreted by normal and cancerous cells, carry a cargo of bioactive molecules including microRNAs (miRNAs) that modulate various cellular processes. miRNAs are small non-coding RNAs that play pivotal roles in post-transcriptional gene regulation and have been implicated in cancer initiation, progression, and metastasis. In cancer, tumor-derived EVs transport specific miRNAs to recipient cells, modulating tumorigenesis, growth, angiogenesis, and metastasis. Dysregulation of miRNA expression profiles within EVs contributes to the acquisition of cancer hallmarks that include increased proliferation, survival, and migration. EV miRNAs influence the tumor microenvironment, promoting immune evasion, remodeling the extracellular matrix, and establishing pre-metastatic niches. Understanding the complex interplay between EVs, miRNAs, and cancer holds significant promise for developing novel diagnostic and therapeutic strategies. This chapter provides insights into the role of EV-mediated miRNA signaling in cancer pathogenesis, highlighting its potential as a biomarker for cancer detection, prognosis, and treatment response assessment.
Collapse
Affiliation(s)
- Nicola S Orefice
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| | - Gianluca Petrillo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Claudia Pignataro
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Martina Mascolo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Giada De Luca
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) National Research Council (CNR), Naples, Italy
| | - Sara Verde
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy; Aka biotech S.r.l., Napoli, Italy
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University "Giuseppe DeGennaro", Bari, Italy
| | - Gerolama Condorelli
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy; Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) National Research Council (CNR), Naples, Italy.
| | - Cristina Quintavalle
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) National Research Council (CNR), Naples, Italy
| |
Collapse
|
194
|
Wang B, Wu X, Cheng J, Ye J, Zhu H, Liu X. Regulatory role of S1P and its receptors in sepsis-induced liver injury. Front Immunol 2025; 16:1489015. [PMID: 39935473 PMCID: PMC11811114 DOI: 10.3389/fimmu.2025.1489015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
As an immune and metabolic organ, the liver affects the progression and prognosis of sepsis. Despite the severe adverse effects of sepsis liver injury on the body, treatment options remain limited. Sphingosine-1-phosphate (S1P) is a widely distributed lipid signaling molecule that binds to five sphingosine-1-phosphate receptors (S1PR) to regulate downstream signaling pathways involved in the pathophysiological processes of sepsis, including endothelial permeability, cytokine release, and vascular tone. This review summarizes current research on the role of S1P in normal liver biology and describes the mechanisms by which changes in S1P/S1PR affect the development of liver-related diseases. At the same time, the pathological processes underlying liver injury, as evidenced by clinical manifestations during sepsis, were comprehensively reviewed. This paper focused on the mechanistic pathways through which S1P and its receptors modulate immunity, bile acid metabolism, and liver-intestinal circulation in septic liver injury. Finally, the relationships between S1P and its receptors with liver inflammation and metabolism and the use of related drugs for the treatment of liver injury were examined. By elucidating the role of S1P and its receptor in the pathogenesis of sepsis liver injury, this review established a molecular targeting framework, providing novel insights into clinical and drug development.
Collapse
Affiliation(s)
- Bin Wang
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiaoyu Wu
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jiangfeng Cheng
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Junming Ye
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
- Clinical College, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Hongquan Zhu
- Department of Critical Care Medicine, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
- Department of Critical Care Medicine, The First Affiliated hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xiaofeng Liu
- Clinical College, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Emergency, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
195
|
Avalos-de Leon CG, Thomson AW. Regulatory Immune Cell-derived Exosomes: Modes of Action and Therapeutic Potential in Transplantation. Transplantation 2025:00007890-990000000-00994. [PMID: 39865513 DOI: 10.1097/tp.0000000000005309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Reduced dependence on antirejection agents, improved long-term allograft survival, and induction of operational tolerance remain major unmet needs in organ transplantation due to the limitations of current immunosuppressive therapies. To address this challenge, investigators are exploring the therapeutic potential of adoptively transferred host- or donor-derived regulatory immune cells. Extracellular vesicles of endosomal origin (exosomes) secreted by these cells seem to be important contributors to their immunoregulatory properties. Twenty years ago, it was first reported that donor-derived exosomes could extend the survival of transplanted organs in rodents. Recent studies have revealed that regulatory immune cells, such as regulatory myeloid cells (dendritic cells, macrophages, or myeloid-derived suppressor cells), regulatory T cells, or mesenchymal stem/stromal cells can suppress graft rejection via exosomes that express a cargo of immunosuppressive molecules. These include cell surface molecules that interact with adaptive immune cell receptors, immunoregulatory enzymes, and micro- and long noncoding RNAs that can regulate inflammatory gene expression via posttranscriptional changes and promote tolerance through promotion of regulatory T cells. This overview analyzes the diverse molecules and mechanisms that enable regulatory immune cell-derived exosomes to modulate alloimmunity and promote experimental transplant tolerance. We also discuss the potential benefits and limitations of their application as therapeutic entities in organ transplantation.
Collapse
Affiliation(s)
- Cindy G Avalos-de Leon
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh PA
| | - Angus W Thomson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh PA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh PA
| |
Collapse
|
196
|
Galbiati M, Maiullari F, Ceraolo MG, Bousselmi S, Fratini N, Gega K, Recchia S, Ferretti AM, Scala G, Costantini M, Sciarra T, Rizzi R, Bearzi C. Bioactive Hydrogel Supplemented with Stromal Cell-Derived Extracellular Vesicles Enhance Wound Healing. Pharmaceutics 2025; 17:162. [PMID: 40006529 PMCID: PMC11859224 DOI: 10.3390/pharmaceutics17020162] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/17/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Skin regeneration is a rapidly advancing field with significant implications for regenerative medicine, particularly in treating wounds and burns. This study explores the potential of hydrogels functionalized with fibroblast-derived extracellular vesicles (EVs) to enhance skin regeneration in vivo. Being immunoprivileged, EVs minimize immune rejection, offering an attractive alternative to whole-cell therapies by replicating fibroblasts' key roles in tissue repair. Methods: To promote EVs' versatility and effective application across different conditions, a lyophilization method with lyoprotectants was optimized. Then, EVs were used to functionalize a hydrogel to perform experiments on murine cutaneous wound models. Results: Gelatin methacrylate (GelMA) was selected as the polymeric hydrogel due to its biocompatibility, tunable mechanical properties, and ability to support wound healing. Mechanical tests confirmed GelMA's strength and elasticity for this application. Fibroblast-derived EVs were characterized using Western blot, Transmission Electron Microscopy, and NanoSight analysis, proving their integrity, size distribution, and stability. miRNome profiling identified enriched biological pathways related to cell migration, differentiation, and angiogenesis, emphasizing the critical role of EV cargo in promoting wound repair. In a murine model, hydrogels loaded with fibroblast-derived EVs significantly accelerated wound healing compared to controls (mean wound area 0.62 mm2 and 4.4 mm2, respectively), with faster closure, enhanced epithelialization, increased vascularization, and reduced fibrosis. Notably, the lyoprotectants successfully preserved the EVs' structure and bioactivity during freeze-drying, reducing EVs loss by 35% compared to the control group and underscoring the feasibility of this approach for long-term storage and clinical application. Conclusions: This study introduces a novel scalable and adaptable strategy for regenerative medicine by combining fibroblast-derived EVs with GelMA, optimizing EVs' stability and functionality for enhanced wound healing in clinical settings, even in challenging contexts such as combat zones or large-scale natural disasters.
Collapse
Affiliation(s)
- Matteo Galbiati
- Institute for Biomedical Technologies, National Research Council, Via Fratelli Cervi, 93, Segrate, 20054 Milan, Italy; (M.G.); (F.M.); (K.G.)
| | - Fabio Maiullari
- Institute for Biomedical Technologies, National Research Council, Via Fratelli Cervi, 93, Segrate, 20054 Milan, Italy; (M.G.); (F.M.); (K.G.)
- Institute of Physical Chemistry, Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224 Warsaw, Poland;
| | - Maria Grazia Ceraolo
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.G.C.); (S.B.)
| | - Salma Bousselmi
- Neurology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (M.G.C.); (S.B.)
- Ph.D. Program in Cellular and Molecular Biology, Department of Biology, University of Rome “Tor Vergata”, Via della Ricerca Scientifica, 1, 00133 Rome, Italy
| | - Nicole Fratini
- Department of Molecular Medicine, Sapienza University, Viale Regina Elena, 324, 00161 Rome, Italy;
| | - Klajdi Gega
- Institute for Biomedical Technologies, National Research Council, Via Fratelli Cervi, 93, Segrate, 20054 Milan, Italy; (M.G.); (F.M.); (K.G.)
| | - Sandro Recchia
- Department of Science and High Technology, University of Insubria, 22100 Como, Italy;
| | - Anna Maria Ferretti
- CNR-SCITEC Istituto di Scienze e Tecnologie Chimiche “G. Natta”, Via G. Fantoli 16/15, 20138 Milan, Italy;
| | - Giovanni Scala
- Department of Biology, University Federico II, 80128 Naples, Italy;
| | - Marco Costantini
- Institute of Physical Chemistry, Polish Academy of Sciences, Marcina Kasprzaka 44/52, 01-224 Warsaw, Poland;
| | - Tommaso Sciarra
- Joint Veteran Center, Scientific Department, Army Medical Center, 00184 Rome, Italy;
| | - Roberto Rizzi
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University, Corso della Repubblica 79, 04100 Latina, Italy
| | - Claudia Bearzi
- Institute for Biomedical Technologies, National Research Council, Via Fratelli Cervi, 93, Segrate, 20054 Milan, Italy; (M.G.); (F.M.); (K.G.)
| |
Collapse
|
197
|
Ilvonen P, Susila S, Impola U, Pusa R, Helin T, Joutsi-Korhonen L, Laitinen S, Lauronen J, Ilmakunnas M. Extracellular vesicles in ageing cold-stored whole blood may not compensate for the decreasing haemostatic function in vitro. Transfus Med 2025. [PMID: 39865366 DOI: 10.1111/tme.13122] [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: 06/05/2024] [Revised: 11/08/2024] [Accepted: 01/12/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND Extracellular vesicles (EVs) have procoagulative properties. As EVs are known to accumulate in stored blood products, we compared the EV content and coagulation capacity of leukoreduced cold-stored whole blood (CSWB) with current prehospital and in-hospital component therapies to understand the role of EVs in the haemostatic capacity of ageing CSWB. MATERIALS AND METHODS Blood was obtained from 12 O RhD-positive male donors. CSWB was compared with in-hospital component therapy of red blood cells (RBCs), OctaplasLG and buffy-coat platelets and prehospital component therapy of RBC and lyophilized plasma. Samples were drawn on Days 1 and 14 of CSWB and RBC cold storage. Blood count, haemolysis markers, rotational thromboelastometry, sonorheometry and thrombin generation were analysed. EVs were analysed using nanoparticle tracking analysis and cellular origin was determined using imaging flow cytometry. RESULTS There was a trend towards increased production of both platelet and RBC-derived EVs during CSWB storage. Particle count increased during storage, whereas thrombin generation slowed down and in viscoelastic assays, clotting times prolonged, clot formation became impaired, and stiffness of the resulting clot decreased. CONCLUSION Both platelet and RBC-derived EVs increased in number in CSWB during storage. This did not appear to compensate for the in vitro decreasing haemostatic capacity of ageing CSWB, suggesting EVs produced during storage may not have active procoagulative effects, but rather reflect the ageing of blood cells.
Collapse
Affiliation(s)
- Petra Ilvonen
- Research and Development, Finnish Red Cross Blood Service, Vantaa, Finland
| | - Sanna Susila
- Research and Development, Finnish Red Cross Blood Service, Vantaa, Finland
- Emergency Medical Service and Emergency Department, Päijät-Häme Wellbeing Services County, Lahti, Finland
| | - Ulla Impola
- Research and Development, Finnish Red Cross Blood Service, Vantaa, Finland
| | - Reetta Pusa
- Research and Development, Finnish Red Cross Blood Service, Vantaa, Finland
| | - Tuukka Helin
- Department of Clinical Chemistry, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Lotta Joutsi-Korhonen
- Department of Clinical Chemistry, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Saara Laitinen
- Research and Development, Finnish Red Cross Blood Service, Vantaa, Finland
| | - Jouni Lauronen
- Research and Development, Finnish Red Cross Blood Service, Vantaa, Finland
| | - Minna Ilmakunnas
- Research and Development, Finnish Red Cross Blood Service, Vantaa, Finland
- Department of Anesthesiology and Intensive Care Medicine, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Meilahti Hospital Blood Bank, Department of Clinical Chemistry, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
- Department of Perioperative and Intensive Care, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
198
|
Chen J, Zheng Y, Wang Z, Gao Q, Hao K, Chen X, Ke N, Lv X, Weng J, Zhong Y, Huang Z, Fu M, Zhao L, Lin F, Mi H, Tang H, Yu C, Huang Y. Development a glycosylated extracellular vesicle-derived miRNA Signature for early detection of esophageal squamous cell carcinoma. BMC Med 2025; 23:39. [PMID: 39849483 PMCID: PMC11755925 DOI: 10.1186/s12916-025-03871-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/14/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is often diagnosed at an advanced stage due to the lack of non-invasive early detection tools, which significantly impacts patient prognosis. Given that glycosylation alterations especially high sialylation and fucosylation, frequently occur during cellular malignant transformation, but their roles are not elucidated. We examined alterations in disease-specific glycosylated extracellular vesicles (EVs)-derived miRNAs in the serum of ESCC patients, evaluating their utility as diagnostic biomarkers. METHODS A total of 371 ESCC and 303 healthy controls (HCs) were recruited in this multi-stage, multicentre case-control study. Fucosylated (Fuc-) and sialylated (Sia-) EVs were isolated utilizing Lentil lectin (LCA) and wheat germ lectin (WGA)-coated magnetic beads, respectively. The glycosylated EVs-derived miRNAs-based signature (RiskscoreFuc-&Sia-) was established through logistic regression in a training cohort and subsequently validated in an internal and an external multicentre cohort. RESULTS The RiskscoreFuc-&Sia- effectively identified ESCC across all stages, demonstrating high AUC values in training (0.980), internal validation (0.957), and external multicentre validation (0.973) cohorts, markedly higher than carcinoembryonic antigen (CEA) (AUC = 0.769, training cohort; AUC = 0.749, internal validation cohort; AUC = 0.765, external validation cohort). Notably, this score exhibited robust accuracy in detecting CEA (-) ESCC cases (CEA < 5 ng/ml) (AUC = 0.974, training & internal cohort; AUC = 0.973, external multicentre validation cohort). Additionally, it displayed strong efficacy in differentiating early-stage ESCC patients (AUC = 0.982, training cohort; AUC = 0.977, external multicentre validation cohort). CONCLUSIONS Our study illustrates the effectiveness of glycosylated EVs capture strategy for isolating tumour-specific EVs. The unique glycosylated EVs-derived miRNAs-based signature shows the optimal potential as a biomarker for early detection of ESCC.
Collapse
Affiliation(s)
- Jianlin Chen
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Yue Zheng
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Zhen Wang
- Department of Clinical Laboratory, Shishi Hospital, Fujian, 362700, Shishi, China
| | - Qi Gao
- Research and development center, Beijing Youngen Technology Co. Ltd, Beijing, 102600, People's Republic of China
| | - Kun Hao
- Research and development center, Beijing Hotgen Biotech Co., Ltd, Beijing, 102600, People's Republic of China
| | - Xiongfeng Chen
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Scientific Research, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China
| | - Nantian Ke
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Xiang Lv
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Jiamiao Weng
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Yuhong Zhong
- Department of Clinical Laboratory, the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, 310009, China
| | - Zhixin Huang
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China
- Integrated Chinese and Western Medicine College, Fujian University of Traditional Chinese Medicine, Fujian, Fuzhou, 350108, China
| | - Miao Fu
- Department of Clinical Laboratory, Jinhua Municipal Central Hospital, Zhejiang, 321000, Jinhua, China
| | - Lilan Zhao
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Thoracic Surgery, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China
| | - Fan Lin
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China
- Department of Geriatric Medicine, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China
- Fujian Provincial Centre for Geriatrics, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China
| | - Hui Mi
- Departments of Clinical Laboratory, Changzhi People's Hospital, Shanxi, Changzhi, 046000, China
| | - Haijun Tang
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China.
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China.
| | - Chundong Yu
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China.
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China.
- State Key Laboratory of Cellular Stress Biology, Innovation Centre for Cell Signalling Network, School of Life Sciences, Xiamen University, Fujian, Xiamen, 361102, China.
| | - Yi Huang
- Shengli Clinical Medical College, Fujian Medical University, Fujian, Fuzhou, 350001, China.
- Department of Clinical Laboratory, Fujian Provincial Hospital, Fuzhou, 350001, China.
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China.
- Central Laboratory, Fujian Provincial Hospital, Fujian, Fuzhou, 350001, China.
- Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Key Laboratory of Critical Care Medicine, Fujian, Fuzhou, 350001, China.
| |
Collapse
|
199
|
Kasiński D, Szeliski K, Drewa T, Pokrywczyńska M. Extracellular vesicles-a new player in the development of urinary bladder cancer. Ther Adv Med Oncol 2025; 17:17588359241297529. [PMID: 39850919 PMCID: PMC11755519 DOI: 10.1177/17588359241297529] [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: 05/06/2024] [Accepted: 10/18/2024] [Indexed: 01/25/2025] Open
Abstract
Bladder cancer was the 10th most commonly diagnosed cancer worldwide in 2020. Extracellular vesicles (EVs) are nano-sized membranous structures secreted by all types of cells into the extracellular space. EVs can transport proteins, lipids, or nucleic acids to specific target cells. What brings more attention and potential implications is the fact that cancer cells secrete more EVs than non-malignant cells. EVs are widely studied for their role in cancer development. This publication summarizes the impact of EVs secreted by urinary bladder cancer cells on urinary bladder cancer development and metastasis. EVs isolated from urinary bladder cancer cells affect other lower-grade cancer cells or normal cells by inducing different metabolic pathways (transforming growth factor β/Smads pathway; phosphoinositide 3-kinase/Akt pathway) that promote epithelial-mesenchymal transition. The cargo carried by EVs can also induce angiogenesis, another critical element in the development of bladder cancer, and modulate the immune system response in a tumor-beneficial manner. In summary, the transfer of substances produced by tumor cells via EVs to the environment influences many stages of tumor progression. An in-depth understanding of the role EVs play in the development of urinary bladder cancer is crucial for the development of future anticancer therapies.
Collapse
Affiliation(s)
- Damian Kasiński
- Chair of Urology and Andrology, Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, Jagiellońska 13/15, 85-067 Bydgoszcz, Poland
| | - Kamil Szeliski
- Chair of Urology and Andrology, Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Drewa
- Chair of Urology and Andrology, Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Marta Pokrywczyńska
- Chair of Urology and Andrology, Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
200
|
Birjandi AA, Sharpe P. The Secretome of the Inductive Tooth Germ Exhibits Signals Required for Tooth Development. Bioengineering (Basel) 2025; 12:96. [PMID: 40001617 PMCID: PMC11851894 DOI: 10.3390/bioengineering12020096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 02/27/2025] Open
Abstract
Teeth develop from reciprocal signaling between inductive and receptive cells. The inductive signals for tooth development are initially in the epithelium of the developing branchial arch, but later shift to the underlying mesenchyme of a developing tooth germ. The inductive signals that are needed for tooth development have not yet been fully identified. Our lab previously provided a basis for bioengineering new teeth by separating the tooth germ's epithelial and mesenchyme cells into a single cell population and recombing them. This approach, however, is not clinically applicable as the cells lose their inductive ability when expanded in vitro. In this study, we investigate whether the secretome and small extracellular vehicles (sEV) derived from inductive tooth germ mesenchyme can contribute to inductive signals required for tooth development. To address this, small extracellular vesicles and secretome were purified from inductive tooth germ mesenchyme and characterized. We investigated the proteome of sEV and proteome of inductive tooth germ mesenchyme and the impact of the culture condition and duration on the proteome. Additionally, we investigated the transcriptomic changes in tooth germ epithelium after treatment with sEV from inductive tooth germ mesenchyme. We show that culture duration of inductive tooth germ mesenchyme has an impact on the proteome of sEV purified from these cells. Similarly, culturing these cells in 2D and 3D environments results in different protein content. Proteome unique to sEV derived from inductive shows an association with multiple signaling pathways related to tooth development. Our RNASeq results show that treatment of tooth germ epithelial cells with small extracellular vesicles derived from inductive tooth germ mesenchyme results in an increased expression of some of the known odontogenic genes. Whilst further analysis is required to harvest the full potential of these sEV, our results suggests that extracellular vehicles contribute to signals required during tooth development, potentially through modulation of cellular metabolism and ECM organization.
Collapse
Affiliation(s)
| | - Paul Sharpe
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK
| |
Collapse
|