1
|
Lin Z, Luo X, Wickman JR, Reddy D, DaCunza JT, Pande R, Tian Y, Kasimoglu EE, Triana V, Lee J, Furdui CM, Pink D, Sacan A, Ajit SK. Inflammatory pain resolution by mouse serum-derived small extracellular vesicles. Brain Behav Immun 2025; 123:422-441. [PMID: 39349284 DOI: 10.1016/j.bbi.2024.09.032] [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: 03/15/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/02/2024] Open
Abstract
Current treatments for chronic pain have limited efficacy and significant side effects, warranting research on alternative strategies for pain management. One approach involves using small extracellular vesicles (sEVs), or exosomes, to transport beneficial biomolecular cargo to aid pain resolution. Exosomes are 30-150 nm sEVs that can be beneficial or harmful depending on their source and cargo composition. We report a comprehensive multi-modal analysis of different aspects of sEV characterization, miRNAs, and protein markers across sEV sources. To investigate the short- and long-term effects of mouse serum-derived sEVs in pain modulation, sEVs from naïve control or spared nerve injury (SNI) model male donor mice were injected intrathecally into naïve male recipient mice. These sEVs transiently increased basal mechanical thresholds, an effect mediated by opioid signaling as this outcome was blocked by naltrexone. Mass spectrometry of sEVs detected endogenous opioid peptide leu-enkephalin. sEVs from naïve female mice have higher levels of leu-enkephalin compared to male, matching the analgesic onset of leu-enkephalin in male recipient mice. In investigating the long-term effect of sEVs, we observed that a single prophylactic intrathecal injection of sEVs two weeks prior to induction of the pain model in recipient mice accelerated recovery from inflammatory pain after complete Freund's adjuvant (CFA) injection. Our exploratory studies examining immune cell populations in spinal cord and dorsal root ganglion using ChipCytometry suggested alterations in immune cell populations 14 days post-CFA. Flow cytometry confirmed increases in CD206+ macrophages in the spinal cord in sEV-treated mice. Collectively, these studies demonstrate multiple mechanisms by which sEVs can attenuate pain.
Collapse
Affiliation(s)
- Zhucheng Lin
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Mail Stop 488, Room 8223, Philadelphia, PA 19102, USA
| | - Xuan Luo
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Mail Stop 488, Room 8223, Philadelphia, PA 19102, USA
| | - Jason R Wickman
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Mail Stop 488, Room 8223, Philadelphia, PA 19102, USA
| | - Deepa Reddy
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Mail Stop 488, Room 8223, Philadelphia, PA 19102, USA
| | - Jason T DaCunza
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Mail Stop 488, Room 8223, Philadelphia, PA 19102, USA
| | - Richa Pande
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Mail Stop 488, Room 8223, Philadelphia, PA 19102, USA
| | - Yuzhen Tian
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Mail Stop 488, Room 8223, Philadelphia, PA 19102, USA
| | - Ezgi E Kasimoglu
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Mail Stop 488, Room 8223, Philadelphia, PA 19102, USA
| | | | - Jingyun Lee
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Cristina M Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Desmond Pink
- Nanostics Inc., Edmonton, Alberta T5J 4P6, Canada
| | - Ahmet Sacan
- School of Biomedical Engineering, Science & Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, USA
| | - Seena K Ajit
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Mail Stop 488, Room 8223, Philadelphia, PA 19102, USA.
| |
Collapse
|
2
|
Zheng M, Chavda VP, Vaghela DA, Bezbaruah R, Gogoi NR, Patel K, Kulkarni M, Shen B, Singla RK. Plant-derived exosomes in therapeutic nanomedicine, paving the path toward precision medicine. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156087. [PMID: 39388922 DOI: 10.1016/j.phymed.2024.156087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Plant-derived exosomes (PDEs), are nanoscale vesicles secreted by multivesicular bodies, play pivotal roles in critical biological processes, including gene regulation, cell communication, and immune defense against pathogens. Recognized for their potential health-promoting properties, PDEs are emerging as innovative components in functional nutrition, poised to enhance dietary health benefits. PURPOSE To describe the efficacy of PDEs in nanoform and their application as precision therapy in many disorders. STUDY DESIGN The design of this review was carried out in PICO format using randomized clinical trials and research articles based on in vivo and in vitro studies. METHODS All the relevant clinical and research studies conducted on plant-derived nanovesicle application and efficacy were included, as retrieved from PubMed and Cochrane, after using specific search terms. This review was performed to determine PDEs' efficacy as nanomedicine and precision therapy. Sub-group analysis and primary data were included to determine the relationship with PDEs. RESULT PDEs are extracted from plant materials using sophisticated techniques like precipitation, size exclusion, immunoaffinity capture, and ultracentrifugation, encapsulating vital molecules such as lipids, proteins, and predominantly microRNAs. Although their nutritional impact may be minimal in small quantities, the broader application of PDEs in biomedicine, particularly as vehicles for drug delivery, underscores their significance. They offer a promising strategy to improve the bioavailability and efficacy of therapeutic agents carrying nano-bioactive substances that exhibit anti-inflammatory, antioxidant, cardioprotective, and anti-cancer activities. CONCLUSION PDEs enhance the therapeutic potency of plant-derived phytochemicals, supporting their use in disease prevention and therapy. This comprehensive review explores the multifaceted aspects of PDEs, including their isolation methods, biochemical composition, health implications, and potential to advance medical and nutritional interventions.
Collapse
Affiliation(s)
- Min Zheng
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; West China Tianfu Hospital, Sichuan University, Chengdu, Sichuan, 610218, China
| | - Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M College of Pharmacy, Ahmedabad 380009, Gujrat, India.
| | - Dixa A Vaghela
- Pharmacy section, L.M College of Pharmacy Ahmedabad 380009, Gujrat, India
| | - Rajashri Bezbaruah
- Department of Pharmacology, Dibrugarh University, Dibrugarh 786004, Assam
| | - Niva Rani Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh 786004, Assam
| | - Kaushika Patel
- Department of Pharmaceutical Technology, L. J. Institute of Pharmacy, LJ University, Ahmedabad 382210, Gujarat, India
| | - Mangesh Kulkarni
- Department of Pharmaceutical Technology, L. J. Institute of Pharmacy, LJ University, Ahmedabad 382210, Gujarat, India; Department of Pharmaceutics, Gandhinagar Institute of Pharmacy, Gandhinagar University, Moti Bhoyan, Khatraj-Kalol Road 382721, Gujarat, India
| | - Bairong Shen
- Institutes for Systems Genetics, West China Tianfu Hospital, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Rajeev K Singla
- Department of Pharmacy and Institutes for Systems Genetics, Center for High Altitude Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| |
Collapse
|
3
|
Wang B, Zhang B, Wu M, Xu T. Unlocking therapeutic potential: Targeting lymphocyte activation Gene-3 (LAG-3) with fibrinogen-like protein 1 (FGL1) in systemic lupus erythematosus. J Transl Autoimmun 2024; 9:100249. [PMID: 39228513 PMCID: PMC11369448 DOI: 10.1016/j.jtauto.2024.100249] [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: 02/22/2024] [Revised: 07/21/2024] [Accepted: 07/26/2024] [Indexed: 09/05/2024] Open
Abstract
Systemic lupus erythematosus (SLE) represents an autoimmune disorder that affects multiple systems. In the treatment of this condition, the focus primarily revolves around inflammation suppression and immunosuppression. Consequently, targeted therapy has emerged as a prevailing approach. Currently, the quest for highly sensitive and specifically effective targets has gained significant momentum in the context of SLE treatment. Lymphocyte activation gene-3 (LAG-3) stands out as a crucial inhibitory receptor that binds to pMHC-II, thereby effectively dampening autoimmune responses. Fibrinogen-like protein 1 (FGL1) serves as the principal immunosuppressive ligand for LAG-3, and their combined action demonstrates a potent immunosuppressive effect. This intricate mechanism paves the way for potential SLE treatment by targeting LAG-3 with FGL1. This work provides a comprehensive summary of LAG-3's role in the pathogenesis of SLE and elucidates the feasibility of leveraging FGL1 as a therapeutic approach for SLE management. It introduces a novel therapeutic target and opens up new avenues of therapeutic consideration in the clinical context of SLE treatment.
Collapse
Affiliation(s)
- Bing Wang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Biqing Zhang
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Min Wu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| | - Ting Xu
- Department of Rheumatology and Immunology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu Province, China
| |
Collapse
|
4
|
Wang L, Wang C, Huang C, Zhou Z, Yang R, Huang Y, Chen Z, Zhang Y, Wang S, Feng K. Role of microRNAs in diabetic foot ulcers: Mechanisms and possible interventions. Diabetes Res Clin Pract 2024; 217:111858. [PMID: 39284457 DOI: 10.1016/j.diabres.2024.111858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/24/2024] [Accepted: 09/13/2024] [Indexed: 09/21/2024]
Abstract
Diabetic foot ulcer (DFU) is a common and serious complication among diabetic patients, and its incidence and difficulty in treatment have placed large burdens on patient health and quality of life. Diabetic foot tissue typically exhibits chronic wounds, ulcers, or necrosis that are difficult to heal, are prone to infection, and, in severe cases, may even lead to amputation. Recent studies have shown that microRNAs (miRNAs) play key roles in the development and healing of DFUs. miRNAs are a class of short noncoding RNA molecules that regulate gene expression to affect cellular functions and physiological processes. miRNAs may be involved in the development of DFUs by regulating cell growth, proliferation, differentiation and apoptosis. miRNAs can also participate in the healing and recovery of DFUs by regulating key steps, such as inflammation, angiogenesis, cell migration and proliferation, tissue repair and matrix remodeling. Therefore, altering the pathological processes of diabetic foot by modulating the expression of miRNAs could improve the recovery and treatment outcomes of patients. This review provides new insights and perspectives for the treatment of DFUs by summarizing the roles of miRNAs in the development and healing of DFUs and the mechanisms.
Collapse
Affiliation(s)
- Lin Wang
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118, Guangdong, China
| | - Cong Wang
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118, Guangdong, China
| | - Caiyan Huang
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118, Guangdong, China
| | - Zhongyu Zhou
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118, Guangdong, China
| | - Ruihong Yang
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118, Guangdong, China
| | - Ying Huang
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118, Guangdong, China
| | - Zhuangsen Chen
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118, Guangdong, China
| | - Yanrong Zhang
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118, Guangdong, China
| | - Shanshan Wang
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118, Guangdong, China
| | - Kun Feng
- Pingshan District People's Hospital of Shenzhen, Pingshan General Hospital of Southern Medical University, Shenzhen 518118, Guangdong, China.
| |
Collapse
|
5
|
Hou J, Chen KX, He C, Li XX, Huang M, Jiang YZ, Jiao YR, Xiao QN, He WZ, Liu L, Zou NY, Huang M, Wei J, Xiao Y, Yang M, Luo XH, Zeng C, Lei GH, Li CJ. Aged bone marrow macrophages drive systemic aging and age-related dysfunction via extracellular vesicle-mediated induction of paracrine senescence. NATURE AGING 2024; 4:1562-1581. [PMID: 39266768 PMCID: PMC11564114 DOI: 10.1038/s43587-024-00694-0] [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: 12/02/2023] [Accepted: 07/25/2024] [Indexed: 09/14/2024]
Abstract
The accumulation and systemic propagation of senescent cells contributes to physiological aging and age-related pathology. However, which cell types are most susceptible to the aged milieu and could be responsible for the propagation of senescence has remained unclear. Here we found that physiologically aged bone marrow monocytes/macrophages (BMMs) propagate senescence to multiple tissues, through extracellular vesicles (EVs), and drive age-associated dysfunction in mice. We identified peroxisome proliferator-activated receptor α (PPARα) as a target of microRNAs within aged BMM-EVs that regulates downstream effects on senescence and age-related dysfunction. Demonstrating therapeutic potential, we report that treatment with the PPARα agonist fenofibrate effectively restores tissue homeostasis in aged mice. Suggesting conservation to humans, in a cohort study of 7,986 participants, we found that fenofibrate use is associated with a reduced risk of age-related chronic disease and higher life expectancy. Together, our findings establish that BMMs can propagate senescence to distant tissues and cause age-related dysfunction, and they provide supportive evidence for fenofibrate to extend healthy lifespan.
Collapse
Affiliation(s)
- Jing Hou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Kai-Xuan Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Chen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Xiao-Xiao Li
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
| | - Mei Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Yang-Zi Jiang
- School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Special Administrative Region of China, Hong Kong, China
| | - Yu-Rui Jiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Qiao-Ni Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Wen-Zhen He
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Ling Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Nan-Yu Zou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Min Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Jie Wei
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, China
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
| | - Xiang-Hang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Zeng
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China.
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, China.
- Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Guang-Hua Lei
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha, China.
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.
- Department of Orthopaedics, Xiangya Hospital of Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Chang-Jun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, China.
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Laboratory Animal Center, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
6
|
Carrabs V, Guillén MI, Ferrándiz ML, Alcaraz MJ, Ferrini F, Agostini R, Guescini M, Fimognari C, Capparucci I, Barbieri E, Sestili P. Hyaluronic Acid Hampers the Inflammatory Response Elicited by Extracellular Vesicles from Activated Monocytes in Human Chondrocytes. Pharmaceutics 2024; 16:1386. [PMID: 39598510 PMCID: PMC11597363 DOI: 10.3390/pharmaceutics16111386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Osteoarthritis (OA) is the most common joint disease in the adult population. OA is the result of multiple mechanisms leading to inflammation and the degradation of the cartilage. A complex series of etiological actors have been identified so far, including extracellular vesicles (EVs). The EV content of the synovial fluid (SF) can release inflammatory mediators that enhance OA progression. An intra-articular viscosupplementation of high-MW hyaluronic acid (HyA) constitutes the first-line conservative treatment for OA. Although attractive for the potential pharmacological implications, the possibility that HyA may interact with EVs in the context of OA has not yet been specifically investigated; therefore, the present study aimed to fill this gap. Methods: We studied the effect of a HyA preparation (a blend of crosslinked and linear polymers, CLHyA) on the relevant inflammatory markers in chondrocytes (HC cells or primary chondrocytes isolated from patients with advanced OA) exposed to the EVs collected from IL-1β-stimulated THP-1 human monocytes (EVs+). Results: EVs+ caused specific inflammatory responses in chondrocytes that could be prevented by coincubation with CLHyA. This anti-inflammatory activity is likely dependent on the direct binding of CLHyA to CD44 receptors highly expressed in EVs+ and on the subsequent hindrance to EVs+ diffusion and docking to target cells. Conclusions: On the whole, the tight interactions identified herein between HMW HyA and EVs+ represent a novel, pharmacologically exploitable mechanism potentially relevant in the context of OA treatment.
Collapse
Affiliation(s)
- Vittoria Carrabs
- Dipartimento di Scienze Biomolecolari, University of Urbino Carlo Bo, 61029 Urbino, Italy; (V.C.); (R.A.); (M.G.); (I.C.); (E.B.)
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, 46010 Valencia, Spain; (M.I.G.); (M.L.F.); (M.J.A.)
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Universidad CEU Cardenal Herrera, 46115 Valencia, Spain
| | - Maria Isabel Guillén
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, 46010 Valencia, Spain; (M.I.G.); (M.L.F.); (M.J.A.)
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Universidad CEU Cardenal Herrera, 46115 Valencia, Spain
| | - María Luisa Ferrándiz
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, 46010 Valencia, Spain; (M.I.G.); (M.L.F.); (M.J.A.)
| | - María José Alcaraz
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, 46010 Valencia, Spain; (M.I.G.); (M.L.F.); (M.J.A.)
| | - Fabio Ferrini
- Dipartimento di Scienze Biomolecolari, University of Urbino Carlo Bo, 61029 Urbino, Italy; (V.C.); (R.A.); (M.G.); (I.C.); (E.B.)
| | - Rachele Agostini
- Dipartimento di Scienze Biomolecolari, University of Urbino Carlo Bo, 61029 Urbino, Italy; (V.C.); (R.A.); (M.G.); (I.C.); (E.B.)
| | - Michele Guescini
- Dipartimento di Scienze Biomolecolari, University of Urbino Carlo Bo, 61029 Urbino, Italy; (V.C.); (R.A.); (M.G.); (I.C.); (E.B.)
| | - Carmela Fimognari
- Dipartimento di Scienze per la Qualità della Vita, Università degli Studi di Bologna, C.so d’Augusto 237, 47921 Rimini, Italy;
| | - Italo Capparucci
- Dipartimento di Scienze Biomolecolari, University of Urbino Carlo Bo, 61029 Urbino, Italy; (V.C.); (R.A.); (M.G.); (I.C.); (E.B.)
| | - Elena Barbieri
- Dipartimento di Scienze Biomolecolari, University of Urbino Carlo Bo, 61029 Urbino, Italy; (V.C.); (R.A.); (M.G.); (I.C.); (E.B.)
| | - Piero Sestili
- Dipartimento di Scienze Biomolecolari, University of Urbino Carlo Bo, 61029 Urbino, Italy; (V.C.); (R.A.); (M.G.); (I.C.); (E.B.)
| |
Collapse
|
7
|
Gaurav I, Thakur A, Zhang K, Thakur S, Hu X, Xu Z, Kumar G, Jaganathan R, Iyaswamy A, Li M, Zhang G, Yang Z. Peptide-Conjugated Vascular Endothelial Extracellular Vesicles Encapsulating Vinorelbine for Lung Cancer Targeted Therapeutics. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1669. [PMID: 39453005 PMCID: PMC11510406 DOI: 10.3390/nano14201669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
Lung cancer is one of the major cancer types and poses challenges in its treatment, including lack of specificity and harm to healthy cells. Nanoparticle-based drug delivery systems (NDDSs) show promise in overcoming these challenges. While conventional NDDSs have drawbacks, such as immune response and capture by the reticuloendothelial system (RES), extracellular vesicles (EVs) present a potential solution. EVs, which are naturally released from cells, can evade the RES without surface modification and with minimal toxicity to healthy cells. This makes them a promising candidate for developing a lung-cancer-targeting drug delivery system. EVs isolated from vascular endothelial cells, such as human umbilical endothelial-cell-derived EVs (HUVEC-EVs), have shown anti-angiogenic activity in a lung cancer mouse model; therefore, in this study, HUVEC-EVs were chosen as a carrier for drug delivery. To achieve lung-cancer-specific targeting, HUVEC-EVs were engineered to be decorated with GE11 peptides (GE11-HUVEC-EVs) via a postinsertional technique to target the epidermal growth factor receptor (EGFR) that is overexpressed on the surface of lung cancer cells. The GE11-HUVEC-EVs were loaded with vinorelbine (GE11-HUVEC-EVs-Vin), and then characterized and evaluated in in vitro and in vivo lung cancer models. Further, we examined the binding affinity of ABCB1, encoding P-glycoprotein, which plays a crucial role in chemoresistance via the efflux of the drug. Our results indicate that GE11-HUVEC-EVs-Vin effectively showed tumoricidal effects against cell and mouse models of lung cancer.
Collapse
Affiliation(s)
- Isha Gaurav
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China; (I.G.)
| | - Abhimanyu Thakur
- Department of Pharmacology, Delhi Pharmaceutical Sciences & Research University (DPSRU), New Delhi 110017, India
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kui Zhang
- Ben May Department for Cancer Research, Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Sudha Thakur
- National Institute for Locomotor Disabilities (Divyangjan), Kolkata 700090, India
| | - Xin Hu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing 400715, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410017, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410017, China
| | - Gaurav Kumar
- Clinical Research Division, Department of Biosciences, School of Basic and Applied Sciences, Galgotias University, Greater Noida 203201, India
| | - Ravindran Jaganathan
- Preclinical Department, Universiti Kuala Lumpur, Royal College of Medicine Perak (UniKL-RCMP), Ipoh 30450, Malaysia
| | - Ashok Iyaswamy
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China; (I.G.)
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641021, India
| | - Min Li
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China; (I.G.)
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
- Institute of Precision Medicine and Innovative Drug Discovery, HKBU Institute for Research and Continuing Education, Shenzhen 518000, China
| | - Zhijun Yang
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China; (I.G.)
| |
Collapse
|
8
|
Cetinkaya PG, Abras IF, Evcili I, Yildirim T, Ceylan Y, Kara Eroglu F, Kayaoglu B, İpekoglu EM, Akarsu A, Yıldırım M, Kahraman T, Cengiz AB, Sahiner UM, Sekerel BE, Ozsurekci Y, Soyer O, Gursel I. Plasma Extracellular Vesicles Derived from Pediatric COVID-19 Patients Modulate Monocyte and T Cell Immune Responses Based on Disease Severity. Immunol Invest 2024; 53:1141-1175. [PMID: 39115924 DOI: 10.1080/08820139.2024.2385992] [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: 10/02/2024]
Abstract
BACKGROUND The COVID-19 pandemic has caused significant morbidity and mortality globally. The role of plasma-derived extracellular vesicles (EVs) in pediatric COVID-19 patients remains unclear. METHODS We isolated EVs from healthy controls (n = 13) and pediatric COVID-19 patients (n = 104) with varying severity during acute and convalescent phases using serial ultracentrifugation. EV effects on healthy PBMCs, naïve CD4+ T cells, and monocytes were assessed through in vitro assays, flow cytometry, and ELISA. RESULTS Our findings indicate that COVID-19 severity correlates with diverse immune responses. Severe acute cases exhibited increased cytokine levels, decreased IFNγ levels, and lower CD4+ T cell and monocyte counts, suggesting immunosuppression. EVs from severe acute patients stimulated healthy cells to express higher PDL1, increased Th2 and Treg cells, reduced IFNγ secretion, and altered Th1/Th17 ratios. Patient-derived EVs significantly reduced proinflammatory cytokine production by monocytes (p < .001 for mild, p = .0025 for severe cases) and decreased CD4+ T cell (p = .043) and monocyte (p = .033) populations in stimulated healthy PBMCs. CONCLUSION This study reveals the complex relationship between immunological responses and EV-mediated effects, emphasizing the impact of COVID-19 severity. We highlight the potential role of plasma-derived EVs in early-stage immunosuppression in severe COVID-19 patients.
Collapse
Affiliation(s)
- Pınar Gur Cetinkaya
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Irem Fatma Abras
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Irem Evcili
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Tugçe Yildirim
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- Basic and Translational Research Program, Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Yasemin Ceylan
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Fehime Kara Eroglu
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Başak Kayaoglu
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Emre Mert İpekoglu
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Aysegul Akarsu
- Division of Pediatric Allergy and Asthma Unit, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Muzaffer Yıldırım
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- Basic and Translational Research Program, Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Tamer Kahraman
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Ali Bülent Cengiz
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Umit Murat Sahiner
- Division of Pediatric Allergy and Asthma, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Bulent Enis Sekerel
- Division of Pediatric Allergy and Asthma, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Yasemin Ozsurekci
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ozge Soyer
- Division of Pediatric Allergy and Asthma, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Ihsan Gursel
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
- Basic and Translational Research Program, Izmir Biomedicine and Genome Center, Izmir, Turkey
| |
Collapse
|
9
|
Cho J, Tae N, Song Y, Kim CW, Lee SJ, Ahn JH, Lee KH, Lee BH, Kim BS, Chang SY, Kim DH, Ko HJ. The expression of PD-L1 on tumor-derived exosomes enhances infiltration and anti-tumor activity of αCD3 × αPD-L1 bispecific antibody-armed T cells. Cancer Immunol Immunother 2024; 73:196. [PMID: 39105814 PMCID: PMC11303351 DOI: 10.1007/s00262-024-03785-4] [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: 06/10/2024] [Accepted: 07/17/2024] [Indexed: 08/07/2024]
Abstract
Anti-cluster of differentiation (CD) 3 × α programmed death-ligand 1 (PD-L1) bispecific T-cell engager (BsTE)-bound T-cells (BsTE:T) are a promising new cancer treatment agent. However, the mechanisms of action of bispecific antibody-armed activated T-cells are poorly understood. Therefore, this study aimed to investigate the anti-tumor mechanism and efficacy of BsTE:T. The BsTE:T migration was assessed in vivo and in vitro using syngeneic and xenogeneic tumor models, flow cytometry, immunofluorescence staining, transwell migration assays, microfluidic chips, Exo View R100, western blotting, and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 technology. In murine B16 melanoma, MC38 colon cancer, and human multiple myeloma cells, BsTE:T exhibited superior tumor elimination relative to that of T-cells or BsTE alone. Moreover, BsTE:T migration into tumors was significantly enhanced owing to the presence of PD-L1 in tumor cells and secretion of PD-L1-containing exosomes. Furthermore, increased infiltration of CD44highCD62Llow effector memory CD8+ T-cells into tumors was closely associated with the anti-tumor effect of BsTE:T. Therefore, BsTE:T is an innovative potential anti-tumor therapy, and exosomal PD-L1 plays a crucial role both in vitro and in vivo in the anti-tumor activity of BsTE:T.
Collapse
Affiliation(s)
- Jaewon Cho
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Nara Tae
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Yujeong Song
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Chae-Won Kim
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Seung-Joo Lee
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jae-Hee Ahn
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Kwang-Ho Lee
- Department of Advanced Material Science and Engineering, College of Engineering, Kangwon National University, Chuncheon, 25561, Korea
| | - Byung-Hyun Lee
- Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Byung Soo Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Sun-Young Chang
- Laboratory of Microbiology, College of Pharmacy, and Research Institute of Pharmaceutical Science and Technology (RIPST), Ajou University, Suwon, 16499, Korea
| | - Dae Hee Kim
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Hyun-Jeong Ko
- Department of Pharmacy, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Innovative Drug Development Research Team for Intractable Diseases (BK21 plus), Kangwon National University, Chuncheon, 24341, Republic of Korea.
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
10
|
Firouzabadi SR, Mohammadi I, Ghafourian K, Kiani A, Hashemi SM. Mesenchymal Stem Cell-Derived Extracellular Vesicle Therapy for Asthma in Murine Models: A Systematic Review and Meta-analysis. Stem Cell Rev Rep 2024; 20:1162-1183. [PMID: 38492133 DOI: 10.1007/s12015-024-10704-8] [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] [Accepted: 02/24/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND Asthma is a common disease, and among the most predominant causes of the years lived with disability. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have emerged as a promising avenue for asthma management. The objective of this study is to perform a systematic review and meta-analysis of pre-clinical studies investigating the therapeutic use of MSC-EVs in murine models of asthma. METHODS A systematic search of electronic databases was performed. Meta-analyses were conducted on broncho-alveolar lavage fluid (BALF) cells and cytokines, as well as airway hyper-responsiveness Penh values and histological staining scores to determine the efficacy of MSC-EVs-based therapy, comparing treated rodents with untreated ones. BALF IL-4, BALF total cells, and BALF eosinophils were chosen as the primary outcomes, while airway hyper-responsiveness Penh values, BALF cytokines excluding IL-4, and histological staining scores were chosen as secondary outcomes. RESULTS A total of 19 eligible studies were included in the current systematic review, with 9 assessing BALF IL-4, 11 assessing BALF total cells, and 10 assessing BALF eosinophils. Pooled Hedges' g (p-value) for each outcome was - 4.407 (< 0.001), -4.976 (< 0.001), and - 4.071 (< 0.001), showing that MSC-EVs therapy inhibits asthma pathology. Changes in secondary outcomes also indicated a reduction in inflammation, goblet cell hyperplasia, and airway hyper-responsiveness. Subgroup analyses did not reveal significant disparities between the type of rodents and administration routes, and meta-regressions were only significant for MSC-EVs source and dose in the IL-4 meta-analysis, and for administration frequency and time from the last challenge to sacrifice in the BALF total cell meta-analysis. CONCLUSION This review highlights the current pre-clinical evidence of MSC-EVs therapy for asthma and finds its application ameliorates multiple aspects of asthma's pathology. We further underline the importance of MSC-EVs source, dose, administration frequency, and timing on the therapeutic effect and warrant further investigation and clinical translation to assess the best treatment regimen and to gauge the efficacy of EV therapy in human asthma cases.
Collapse
Affiliation(s)
- Shahryar Rajai Firouzabadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Koodakyar street, Daneshjoo Boulevard, Tehran, Iran.
- Tehran Lung Research and Developmental Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ida Mohammadi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Koodakyar street, Daneshjoo Boulevard, Tehran, Iran
- Tehran Lung Research and Developmental Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kiana Ghafourian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Koodakyar street, Daneshjoo Boulevard, Tehran, Iran
| | - Arda Kiani
- Tehran Lung Research and Developmental Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Cabrera-Pastor A. Extracellular Vesicles as Mediators of Neuroinflammation in Intercellular and Inter-Organ Crosstalk. Int J Mol Sci 2024; 25:7041. [PMID: 39000150 PMCID: PMC11241119 DOI: 10.3390/ijms25137041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Neuroinflammation, crucial in neurological disorders like Alzheimer's disease, multiple sclerosis, and hepatic encephalopathy, involves complex immune responses. Extracellular vesicles (EVs) play a pivotal role in intercellular and inter-organ communication, influencing disease progression. EVs serve as key mediators in the immune system, containing molecules capable of activating molecular pathways that exacerbate neuroinflammatory processes in neurological disorders. However, EVs from mesenchymal stem cells show promise in reducing neuroinflammation and cognitive deficits. EVs can cross CNS barriers, and peripheral immune signals can influence brain function via EV-mediated communication, impacting barrier function and neuroinflammatory responses. Understanding EV interactions within the brain and other organs could unveil novel therapeutic targets for neurological disorders.
Collapse
Affiliation(s)
- Andrea Cabrera-Pastor
- Departamento de Farmacología, Facultad de Medicina y Odontología, Universitat de València, 46010 Valencia, Spain; or
- Fundación de Investigación del Hospital Clínico Universitario de Valencia, INCLIVA, 46010 Valencia, Spain
| |
Collapse
|
12
|
Li J, He S, Yang H, Zhang L, Xiao J, Liang C, Liu S. The Main Mechanisms of Mesenchymal Stem Cell-Based Treatments against COVID-19. Tissue Eng Regen Med 2024; 21:545-556. [PMID: 38573476 PMCID: PMC11087407 DOI: 10.1007/s13770-024-00633-5] [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: 04/11/2023] [Revised: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) has a clinical manifestation of hypoxic respiratory failure and acute respiratory distress syndrome. However, COVID-19 still lacks of effective clinical treatments so far. As a promising potential treatment against COVID-19, stem cell therapy raised recently and had attracted much attention. Here we review the mechanisms of mesenchymal stem cell-based treatments against COVID-19, and provide potential cues for the effective control of COVID-19 in the future. METHODS Literature is obtained from databases PubMed and Web of Science. Key words were chosen for COVID- 19, acute respiratory syndrome coronavirus 2, mesenchymal stem cells, stem cell therapy, and therapeutic mechanism. Then we summarize and critically analyze the relevant articles retrieved. RESULTS Mesenchymal stem cell therapy is a potential effective treatment against COVID-19. Its therapeutic efficacy is mainly reflected in reducing severe pulmonary inflammation, reducing lung injury, improving pulmonary function, protecting and repairing lung tissue of the patients. Possible therapeutic mechanisms might include immunoregulation, anti-inflammatory effect, tissue regeneration, anti-apoptosis effect, antiviral, and antibacterial effect, MSC - EVs, and so on. CONCLUSION Mesenchymal stem cells can effectively treat COVID-19 through immunoregulation, anti-inflammatory, tissue regeneration, anti-apoptosis, anti-virus and antibacterial, MSC - EVs, and other ways. Systematically elucidating the mechanisms of mesenchymal stem cell-based treatments for COVID-19 will provide novel insights into the follow-up research and development of new therapeutic strategies in next step.
Collapse
Affiliation(s)
- Jinling Li
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Laboratory of Basic Medicine Center, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Shipei He
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Hang Yang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Lizeai Zhang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Jie Xiao
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Chaoyi Liang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Sijia Liu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed by the Province and Ministry, Guangxi Key Laboratory of Regenerative Medicine and Key Laboratory of Longevity and Aging-Related Diseases of Chinese Ministry of Education, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
- Guangxi Colleges and Universities Key Laboratory of Biological Molecular Medicine Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangxi Medical University, Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
13
|
Ma K, Luo C, Du M, Wei Q, Luo Q, Zheng L, Liao M. Advances in stem cells treatment of diabetic wounds: A bibliometric analysis via CiteSpace. Skin Res Technol 2024; 30:e13665. [PMID: 38558448 PMCID: PMC10982678 DOI: 10.1111/srt.13665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/09/2024] [Indexed: 04/04/2024]
Abstract
Diabetes is a chronic medical condition that may induce complications such as poor wound healing. Stem cell therapies have shown promise in treating diabetic wounds with pre-clinical and clinical studies. However, little bibliometric analysis has been carried out on stem cells in the treatment of diabetic wounds. In this study, we retrieved relevant papers published from January 1, 2003, to December 31, 2023, from Chinese and English databases. CiteSpace software was used to analyze the authors, institutions, and keywords by standard bibliometric indicators. Our analysis findings indicated that publications on stem cells in the treatment of diabetic wounds kept increasing. The most prolific author was Qian Cai (n = 7) and Mohammad Bayat (n = 16) in Chinese and English databases, respectively. Institutions distribution analysis showed that Chinese institutions conducted most publications, and the most prolific institution was the Chinese People's Liberation Army General Hospital (n = 9) and Shahid Beheshti University of Medical Sciences (n = 17) in Chinese and English databases, respectively. The highest centrality keyword in Chinese and English databases was "wound healing" (0.54) and "in vitro" (0.13), respectively. There were 8 and 11 efficient and convincing keyword clusters produced by a log-likelihood ratio in the Chinese and English databases, respectively. The strongest burst keyword was "exosome" (strength 3.57) and "endothelial progenitor cells" (strength 7.87) in the Chinese and English databases, respectively. These findings indicated a direction for future therapies and research on stem cells in the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Ke Ma
- Department of Plastic & Cosmetic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
- Pharmaceutical CollegeGuangxi Medical UniversityNanningChina
| | - Chao Luo
- Shanghai Mental Health CenterShanghai Jiao Tong University, School of MedicineShanghaiChina
| | - Mindong Du
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
- Department of Orthopaedics Trauma and Hand SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Qiang Wei
- Department of Plastic & Cosmetic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Qianxuan Luo
- Department of Plastic & Cosmetic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ RegenerationThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
- Pharmaceutical CollegeGuangxi Medical UniversityNanningChina
| | - Mingde Liao
- Department of Plastic & Cosmetic SurgeryThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
14
|
Berriel Pinho VH, Daher JPL, Kanaan S, Medeiros T. Extracellular vesicles in Alzheimer's disease. ARQUIVOS DE NEURO-PSIQUIATRIA 2024; 82:1-8. [PMID: 38467392 PMCID: PMC10927369 DOI: 10.1055/s-0044-1779296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 12/01/2023] [Indexed: 03/13/2024]
Abstract
Extracellular vesicles (EVs) are small vesicles released by cells that facilitate cell signaling. They are categorized based on their biogenesis and size. In the context of the central nervous system (CNS), EVs have been extensively studied for their role in both normal physiological functions and diseases like Alzheimer's disease (AD). AD is a neurodegenerative disorder characterized by cognitive decline and neuronal death. EVs have emerged as potential biomarkers for AD due to their involvement in disease progression. Specifically, EVs derived from neurons, astrocytes, and neuron precursor cells exhibit changes in quantity and composition in AD. Neuron-derived EVs have been found to contain key proteins associated with AD pathology, such as amyloid beta (Aß) and tau. Increased levels of Aß in neuron-derived EVs isolated from the plasma have been observed in individuals with AD and mild cognitive impairment, suggesting their potential as early biomarkers. However, the analysis of tau in neuron-derived EVs is still inconclusive. In addition to Aß and tau, neuron-derived EVs also carry other proteins linked to AD, including synaptic proteins. These findings indicate that EVs could serve as biomarkers for AD, particularly for early diagnosis and disease monitoring. However, further research is required to validate their use and explore potential therapeutic applications. To summarize, EVs are small vesicles involved in cell signaling within the CNS. They hold promise as biomarkers for AD, potentially enabling early diagnosis and monitoring of disease progression. Ongoing research aims to refine their use as biomarkers and uncover additional therapeutic applications.
Collapse
Affiliation(s)
| | - João Paulo Lima Daher
- Universidade Federal Fluminense, Faculdade de Medicina, Departamento de Patologia, NIterói RJ, Brazil.
| | - Salim Kanaan
- Universidade Federal Fluminense, Faculdade de Medicina, Departamento de Patologia, NIterói RJ, Brazil.
| | - Thalia Medeiros
- Universidade Federal Fluminense, Faculdade de Medicina, Departamento de Patologia, NIterói RJ, Brazil.
| |
Collapse
|
15
|
Lin Z, Luo X, Wickman JR, Reddy D, Pande R, Tian Y, Triana V, Lee J, Furdui CM, Pink D, Sacan A, Ajit SK. Inflammatory pain resolution by mouse serum-derived small extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.16.578759. [PMID: 38405813 PMCID: PMC10888877 DOI: 10.1101/2024.02.16.578759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Chronic pain is a significant public health issue. Current treatments have limited efficacy and significant side effects, warranting research on alternative strategies for pain management. One approach involves using small extracellular vesicles (sEVs) to transport beneficial biomolecular cargo to aid pain resolution. Exosomes are 30-150 nm sEVs that can carry RNAs, proteins, and lipid mediators to recipient cells via circulation. Exosomes can be beneficial or harmful depending on their source and contents. To investigate the short and long-term effects of mouse serum-derived sEVs in pain modulation, sEVs from naïve control or spared nerve injury (SNI) model donor mice were injected intrathecally into naïve recipient mice. Basal mechanical thresholds transiently increased in recipient mice. This effect was mediated by opioid signaling as this outcome was blocked by naltrexone. Mass Spectrometry of sEVs detected endogenous opioid peptide leu-enkephalin. A single prophylactic intrathecal injection of sEVs two weeks prior to induction of the pain model in recipient mice delayed mechanical allodynia in SNI model mice and accelerated recovery from inflammatory pain after complete Freund's adjuvant (CFA) injection. ChipCytometry of spinal cord and dorsal root ganglion (DRG) from sEV treated mice showed that prophylactic sEV treatment reduced the number of natural killer (NK) and NKT cells in spinal cord and increased CD206+ anti-inflammatory macrophages in (DRG) after CFA injection. Further characterization of sEVs showed the presence of immune markers suggesting that sEVs can exert immunomodulatory effects in recipient mice to promote the resolution of inflammatory pain. Collectively, these studies demonstrate multiple mechanisms by which sEVs can attenuate pain.
Collapse
Affiliation(s)
- Zhucheng Lin
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA
| | - Xuan Luo
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA
| | - Jason R. Wickman
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA
| | - Deepa Reddy
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA
| | - Richa Pande
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA
| | - Yuzhen Tian
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA
| | | | - Jingyun Lee
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Cristina M. Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Desmond Pink
- Nanostics Inc., Edmonton, Alberta, T5J 4P6, Canada
| | - Ahmet Sacan
- School of Biomedical Engineering, Science & Health Systems, Drexel University, 3141 Chestnut Street, Philadelphia, PA 19104, USA
| | - Seena K. Ajit
- Department of Pharmacology & Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA 19102, USA
| |
Collapse
|
16
|
Kumar MA, Baba SK, Sadida HQ, Marzooqi SA, Jerobin J, Altemani FH, Algehainy N, Alanazi MA, Abou-Samra AB, Kumar R, Al-Shabeeb Akil AS, Macha MA, Mir R, Bhat AA. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct Target Ther 2024; 9:27. [PMID: 38311623 PMCID: PMC10838959 DOI: 10.1038/s41392-024-01735-1] [Citation(s) in RCA: 102] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 12/20/2023] [Accepted: 12/24/2023] [Indexed: 02/06/2024] Open
Abstract
Extracellular vesicles (EVs) are nano-sized, membranous structures secreted into the extracellular space. They exhibit diverse sizes, contents, and surface markers and are ubiquitously released from cells under normal and pathological conditions. Human serum is a rich source of these EVs, though their isolation from serum proteins and non-EV lipid particles poses challenges. These vesicles transport various cellular components such as proteins, mRNAs, miRNAs, DNA, and lipids across distances, influencing numerous physiological and pathological events, including those within the tumor microenvironment (TME). Their pivotal roles in cellular communication make EVs promising candidates for therapeutic agents, drug delivery systems, and disease biomarkers. Especially in cancer diagnostics, EV detection can pave the way for early identification and offers potential as diagnostic biomarkers. Moreover, various EV subtypes are emerging as targeted drug delivery tools, highlighting their potential clinical significance. The need for non-invasive biomarkers to monitor biological processes for diagnostic and therapeutic purposes remains unfulfilled. Tapping into the unique composition of EVs could unlock advanced diagnostic and therapeutic avenues in the future. In this review, we discuss in detail the roles of EVs across various conditions, including cancers (encompassing head and neck, lung, gastric, breast, and hepatocellular carcinoma), neurodegenerative disorders, diabetes, viral infections, autoimmune and renal diseases, emphasizing the potential advancements in molecular diagnostics and drug delivery.
Collapse
Affiliation(s)
- Mudasir A Kumar
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Kashmir, 192122, India
| | - Sadaf K Baba
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Kashmir, 192122, India
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Sara Al Marzooqi
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Jayakumar Jerobin
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Faisal H Altemani
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Naseh Algehainy
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Mohammad A Alanazi
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Abdul-Badi Abou-Samra
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Rakesh Kumar
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, India
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Kashmir, 192122, India
| | - Rashid Mir
- Department of Medical Laboratory Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
17
|
Miron RJ, Estrin NE, Sculean A, Zhang Y. Understanding exosomes: Part 2-Emerging leaders in regenerative medicine. Periodontol 2000 2024; 94:257-414. [PMID: 38591622 DOI: 10.1111/prd.12561] [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: 02/04/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with the ability to communicate with other tissues and cell types over long distances. Their use in regenerative medicine has gained tremendous momentum recently due to their ability to be utilized as therapeutic options for a wide array of diseases/conditions. Over 5000 publications are currently being published yearly on this topic, and this number is only expected to dramatically increase as novel therapeutic strategies continue to be developed. Today exosomes have been applied in numerous contexts including neurodegenerative disorders (Alzheimer's disease, central nervous system, depression, multiple sclerosis, Parkinson's disease, post-traumatic stress disorders, traumatic brain injury, peripheral nerve injury), damaged organs (heart, kidney, liver, stroke, myocardial infarctions, myocardial infarctions, ovaries), degenerative processes (atherosclerosis, diabetes, hematology disorders, musculoskeletal degeneration, osteoradionecrosis, respiratory disease), infectious diseases (COVID-19, hepatitis), regenerative procedures (antiaging, bone regeneration, cartilage/joint regeneration, osteoarthritis, cutaneous wounds, dental regeneration, dermatology/skin regeneration, erectile dysfunction, hair regrowth, intervertebral disc repair, spinal cord injury, vascular regeneration), and cancer therapy (breast, colorectal, gastric cancer and osteosarcomas), immune function (allergy, autoimmune disorders, immune regulation, inflammatory diseases, lupus, rheumatoid arthritis). This scoping review is a first of its kind aimed at summarizing the extensive regenerative potential of exosomes over a broad range of diseases and disorders.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Nathan E Estrin
- Advanced PRF Education, Venice, Florida, USA
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Anton Sculean
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| |
Collapse
|
18
|
Caño-Carrillo S, Castillo-Casas JM, Franco D, Lozano-Velasco E. Unraveling the Signaling Dynamics of Small Extracellular Vesicles in Cardiac Diseases. Cells 2024; 13:265. [PMID: 38334657 PMCID: PMC10854837 DOI: 10.3390/cells13030265] [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: 12/29/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024] Open
Abstract
Effective intercellular communication is essential for cellular and tissue balance maintenance and response to challenges. Cellular communication methods involve direct cell contact or the release of biological molecules to cover short and long distances. However, a recent discovery in this communication network is the involvement of extracellular vesicles that host biological contents such as proteins, nucleic acids, and lipids, influencing neighboring cells. These extracellular vesicles are found in body fluids; thus, they are considered as potential disease biomarkers. Cardiovascular diseases are significant contributors to global morbidity and mortality, encompassing conditions such as ischemic heart disease, cardiomyopathies, electrical heart diseases, and heart failure. Recent studies reveal the release of extracellular vesicles by cardiovascular cells, influencing normal cardiac function and structure. However, under pathological conditions, extracellular vesicles composition changes, contributing to the development of cardiovascular diseases. Investigating the loading of molecular cargo in these extracellular vesicles is essential for understanding their role in disease development. This review consolidates the latest insights into the role of extracellular vesicles in diagnosis and prognosis of cardiovascular diseases, exploring the potential applications of extracellular vesicles in personalized therapies, shedding light on the evolving landscape of cardiovascular medicine.
Collapse
Affiliation(s)
| | | | | | - Estefanía Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (S.C.-C.); (J.M.C.-C.); (D.F.)
| |
Collapse
|
19
|
Zhang R, Li M, Li H, Ran X, Jin F, Tan Q, Chen Z. Immune Cell-Derived Exosomes in Inflammatory Disease and Inflammatory Tumor Microenvironment: A Review. J Inflamm Res 2024; 17:301-312. [PMID: 38250144 PMCID: PMC10800116 DOI: 10.2147/jir.s421649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 10/18/2023] [Indexed: 01/23/2024] Open
Abstract
Inflammation is a common feature of many inflammatory diseases and tumors, and plays a decisive role in their development. Exosomes are extracellular vesicles unleashed by assorted types of cells, and it is widely known that exosomes of different immune cell sources play different functions. Exosome production has recently been reported for immune cells comprising macrophages, T cells, B cells, and dendritic cells (DCs). Immune cell-derived exosomes are involved in a variety of inflammatory responses.Herein, we summarize and review the role of macrophages, T cells, B cells, and dendritic cells (DC) in inflammatory diseases, with a focus on the role of immune cell-derived exosomes in osteoarthritis, rheumatoid arthritis, and the inflammatory tumor microenvironment (TME).These findings are expected to be important for developing new treatments for inflammatory diseases and ameliorating tumor-related inflammation.
Collapse
Affiliation(s)
- Runmin Zhang
- Department of Orthopaedics, The First Affiliated Hospital of University of South China, Hengyang, People’s Republic of China
| | - Muzhe Li
- Department of Orthopaedics, The First Affiliated Hospital of University of South China, Hengyang, People’s Republic of China
| | - Huiyun Li
- Department of Orthopaedics, The First Affiliated Hospital of University of South China, Hengyang, People’s Republic of China
| | - Xun Ran
- Department of Orthopaedics, The First Affiliated Hospital of University of South China, Hengyang, People’s Republic of China
| | - Fengtian Jin
- Department of Orthopaedics, The First Affiliated Hospital of University of South China, Hengyang, People’s Republic of China
| | - Qingshan Tan
- Department of Orthopaedics, The First Affiliated Hospital of University of South China, Hengyang, People’s Republic of China
| | - Zhiwei Chen
- Department of Orthopaedics, The First Affiliated Hospital of University of South China, Hengyang, People’s Republic of China
| |
Collapse
|
20
|
Wang J, Tao Y, Zhao F, Liu T, Shen X, Zhou L. Expression of urinary exosomal miRNA-615-3p and miRNA-3147 in diabetic kidney disease and their association with inflammation and fibrosis. Ren Fail 2023; 45:2121929. [PMID: 36695327 PMCID: PMC9879181 DOI: 10.1080/0886022x.2022.2121929] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Diabetic kidney disease (DKD) is one of the most common chronic complications of type 2 diabetes mellitus (T2DM), and it is particularly important to identify a high-quality method for evaluating disease progression. Urinary exosomes contain microRNA that might promise early diagnostic and monitoring markers of DKD. The present study aimed to identify novel exosome-related markers associated with inflammation and fibrosis to assess the progression of DKD. METHOD Exosomes were extracted from the urine of 83 participants to determine the expression levels of miRNA-615-3p and miRNA-3147 in 20 healthy people, 21 patients with T2DM and 42 patients with DKD, as determined by RT-qPCR. The circulating expression level of TGF-β1 was detected by ELISA. Serum Cystatin C was measured by a latex-enhanced immunoturbidimetric method. The correlation analyses were performed for all clinical and laboratory parameters. RESULT The expression level of urinary exosomal miRNA-615-3p in DKD patients was significantly higher than that in the control group and the T2DM group by RT-qPCR. The expression of miRNA-3147 showed an upward trend in the three groups of subjects, but it was not statistically significant. The urinary exosomal miRNA-615-3p was positively correlated with serum Cystatin C, plasma TGF-β1, creatinine, BUN, PCR and 24-h urine protein, and negatively correlated with eGFR and albumin. The diagnostic efficacy of urinary exosomal miRNA-615-3p combined with the ACR was higher than that of ACR alone. CONCLUSIONS Urinary exosomal miRNA-615-3p may be used as a novel biomarker for evaluating the progression of DKD, and may be involved in the process of inflammation and fibrosis in DKD. The combined diagnosis of urinary exosomal miRNA-615-3p and ACR may be used as more stable and sensitive diagnostic criteria for DKD.
Collapse
Affiliation(s)
- Jiaxin Wang
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yiying Tao
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fan Zhao
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Tong Liu
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiahong Shen
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ling Zhou
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou, China,CONTACT Ling Zhou Department of Nephrology, The First Affiliated Hospital of Soochow University, 889 Pinghai Rd, Suzhou, 215000, People’s Republic of China
| |
Collapse
|
21
|
Jin X, Sun H, Yang L. How Extracellular Nano-Vesicles Can Play a Role in Sepsis? An Evidence-Based Review of the Literature. Int J Nanomedicine 2023; 18:5797-5814. [PMID: 37869065 PMCID: PMC10588718 DOI: 10.2147/ijn.s427116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/08/2023] [Indexed: 10/24/2023] Open
Abstract
Sepsis is a systemic inflammatory reaction caused by infection. Severe sepsis can lead to multiple organ dysfunction, with a high incidence rate and mortality. The molecular pathogenesis of sepsis is complex and diverse. In recent years, with further study of the role of extracellular vesicles (EVs) in inflammatory diseases, it has been found that EVs play a dual role in the imbalance of inflammatory response in sepsis. Due to the great advantages such as lower toxicity, lower immunogenicity compared with stem cells and better circulation stability, EVs are increasingly used for the diagnosis and treatment of sepsis. The roles of EVs in the pathogenesis, diagnosis and treatment of sepsis were summarized to guide further clinical studies.
Collapse
Affiliation(s)
- Xiaolin Jin
- Department of International Physical Examination Center, The First Hospital of China Medical University, Shengyang, People’s Republic of China
| | - Haiyan Sun
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, People’s Republic of China
| | - Lina Yang
- Department of International Physical Examination Center, The First Hospital of China Medical University, Shengyang, People’s Republic of China
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
22
|
Levy D, Abadchi SN, Shababi N, Ravari MR, Pirolli NH, Bergeron C, Obiorah A, Mokhtari‐Esbuie F, Gheshlaghi S, Abraham JM, Smith IM, Powsner EH, Solomon TJ, Harmon JW, Jay SM. Induced Pluripotent Stem Cell-Derived Extracellular Vesicles Promote Wound Repair in a Diabetic Mouse Model via an Anti-Inflammatory Immunomodulatory Mechanism. Adv Healthc Mater 2023; 12:e2300879. [PMID: 37335811 PMCID: PMC10592465 DOI: 10.1002/adhm.202300879] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 06/07/2023] [Indexed: 06/21/2023]
Abstract
Extracellular vesicles (EVs) derived from mesenchymal stem/stromal cells (MSCs) have recently been explored in clinical trials for treatment of diseases with complex pathophysiologies. However, production of MSC EVs is currently hampered by donor-specific characteristics and limited ex vivo expansion capabilities before decreased potency, thus restricting their potential as a scalable and reproducible therapeutic. Induced pluripotent stem cells (iPSCs) represent a self-renewing source for obtaining differentiated iPSC-derived MSCs (iMSCs), circumventing both scalability and donor variability concerns for therapeutic EV production. Thus, it is initially sought to evaluate the therapeutic potential of iMSC EVs. Interestingly, while utilizing undifferentiated iPSC EVs as a control, it is found that their vascularization bioactivity is similar and their anti-inflammatory bioactivity is superior to donor-matched iMSC EVs in cell-based assays. To supplement this initial in vitro bioactivity screen, a diabetic wound healing mouse model where both the pro-vascularization and anti-inflammatory activity of these EVs would be beneficial is employed. In this in vivo model, iPSC EVs more effectively mediate inflammation resolution within the wound bed. Combined with the lack of additional differentiation steps required for iMSC generation, these results support the use of undifferentiated iPSCs as a source for therapeutic EV production with respect to both scalability and efficacy.
Collapse
Affiliation(s)
- Daniel Levy
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | | | - Niloufar Shababi
- Department of SurgeryJohns Hopkins University School of MedicineBaltimoreMD21224USA
| | | | - Nicholas H. Pirolli
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Cade Bergeron
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Angel Obiorah
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | | | - Shayan Gheshlaghi
- Department of SurgeryJohns Hopkins University School of MedicineBaltimoreMD21224USA
| | - John M. Abraham
- Department of SurgeryJohns Hopkins University School of MedicineBaltimoreMD21224USA
| | - Ian M. Smith
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Emily H. Powsner
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - Talia J. Solomon
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
| | - John W. Harmon
- Department of SurgeryJohns Hopkins University School of MedicineBaltimoreMD21224USA
| | - Steven M. Jay
- Fischell Department of BioengineeringUniversity of MarylandCollege ParkMD20742USA
- Program in Molecular and Cell BiologyUniversity of MarylandCollege ParkMD20742USA
| |
Collapse
|
23
|
Liu WS, Wu LL, Chen CM, Zheng H, Gao J, Lu ZM, Li M. Lipid-hybrid cell-derived biomimetic functional materials: A state-of-the-art multifunctional weapon against tumors. Mater Today Bio 2023; 22:100751. [PMID: 37636983 PMCID: PMC10448342 DOI: 10.1016/j.mtbio.2023.100751] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Tumors are among the leading causes of death worldwide. Cell-derived biomimetic functional materials have shown great promise in the treatment of tumors. These materials are derived from cell membranes, extracellular vesicles and bacterial outer membrane vesicles and may evade immune recognition, improve drug targeting and activate antitumor immunity. However, their use is limited owing to their low drug-loading capacity and complex preparation methods. Liposomes are artificial bionic membranes that have high drug-loading capacity and can be prepared and modified easily. Although they can overcome the disadvantages of cell-derived biomimetic functional materials, they lack natural active targeting ability. Lipids can be hybridized with cell membranes, extracellular vesicles or bacterial outer membrane vesicles to form lipid-hybrid cell-derived biomimetic functional materials. These materials negate the disadvantages of both liposomes and cell-derived components and represent a promising delivery platform in the treatment of tumors. This review focuses on the design strategies, applications and mechanisms of action of lipid-hybrid cell-derived biomimetic functional materials and summarizes the prospects of their further development and the challenges associated with it.
Collapse
Affiliation(s)
- Wen-Shang Liu
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, China
| | - Li-Li Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Cui-Min Chen
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Hao Zheng
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zheng-Mao Lu
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Meng Li
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, China
| |
Collapse
|
24
|
Jorfi S, Ansa-Addo EA, Mariniello K, Warde P, Bin Senian AA, Stratton D, Bax BE, Levene M, Lange S, Inal JM. A Coxsackievirus B1-mediated nonlytic Extracellular Vesicle-to-cell mechanism of virus transmission and its possible control through modulation of EV release. J Gen Virol 2023; 104. [PMID: 37665326 DOI: 10.1099/jgv.0.001884] [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: 09/05/2023] Open
Abstract
Like most non-enveloped viruses, CVB1 mainly uses cell lysis to spread. Details of a nonlytic virus transmission remain unclear. Extracellular Vesicles (EVs) transfer biomolecules between cells. We show that CVB1 entry into HeLa cells results in apoptosis and release of CVB1-induced 'medium-sized' EVs (CVB1i-mEVs). These mEVs (100-300 nm) harbour CVB1 as shown by immunoblotting with anti-CVB1-antibody; viral capsids were detected by transmission electron microscopy and RT-PCR revealed CVB1 RNA. The percentage of mEVs released from CVB1-infected HeLa cells harbouring virus was estimated from TEM at 34 %. Inhibition of CVB1i-mEV production, with calpeptin or siRNA knockdown of CAPNS1 in HeLa cells limited spread of CVB1 suggesting these vesicles disseminate CVB1 virions to new host cells by a nonlytic EV-to-cell mechanism. This was confirmed by detecting CVB1 virions inside HeLa cells after co-culture with CVB1i-mEVs; EV release may also prevent apoptosis of infected cells whilst spreading apoptosis to secondary sites of infection.
Collapse
Affiliation(s)
- Samireh Jorfi
- Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, London N7 8DB, UK
| | - Ephraim Abrokwa Ansa-Addo
- Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, London N7 8DB, UK
- Present address: Pelotonia Institute for Immuno-Oncology, The James, Ohio State University, Columbus, OH 43210, USA
| | - Katia Mariniello
- Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, London N7 8DB, UK
- Present address: William Harvey Research Institute, Queen Mary, University of London, London, UK
| | - Purva Warde
- Biosciences Research Group, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9EU, UK
| | - Ahmad Asyraf Bin Senian
- Biosciences Research Group, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9EU, UK
- Present address: Clinical Research Centre, Sarawak General Hospital, Kuching, Malaysia
| | - Dan Stratton
- School of Life, Health & Chemical Sciences, The Open University, Milton Keynes MK7 6AE, UK
| | - Bridget E Bax
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, UK
| | - Michelle Levene
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, UK
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, School of Life Sciences, University of Westminster, 116, New Cavendish St., London, UK
- University College London School of Pharmacy, Brunswick Sq., London, UK
| | - Jameel Malhador Inal
- Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, London N7 8DB, UK
- Biosciences Research Group, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9EU, UK
| |
Collapse
|
25
|
Pelassa S, Raggi F, Rossi C, Bosco MC. MicroRNAs in Juvenile Idiopathic Arthritis: State of the Art and Future Perspectives. BIOLOGY 2023; 12:991. [PMID: 37508421 PMCID: PMC10376583 DOI: 10.3390/biology12070991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023]
Abstract
Juvenile Idiopathic Arthritis (JIA) represents the most common chronic pediatric arthritis in Western countries and a leading cause of disability in children. Despite recent clinical achievements, patient management is still hindered by a lack of diagnostic/prognostic biomarkers and targeted treatment protocols. MicroRNAs (miRNAs) are short non-coding RNAs playing a key role in gene regulation, and their involvement in many pathologies has been widely reported in the literature. In recent decades, miRNA's contribution to the regulation of the immune system and the pathogenesis of autoimmune diseases has been demonstrated. Furthermore, miRNAs isolated from patients' biological samples are currently under investigation for their potential as novel biomarkers. This review aims to provide an overview of the state of the art on miRNA investigation in JIA. The literature addressing the expression of miRNAs in different types of biological samples isolated from JIA patients was reviewed, focusing in particular on their potential application as diagnostic/prognostic biomarkers. The role of miRNAs in the regulation of immune responses in affected joints will also be discussed along with their potential utility as markers of patients' responses to therapeutic approaches. This information will be of value to investigators in the field of pediatric rheumatology, encouraging further research to increase our knowledge of miRNAs' potential for future clinical applications in JIA.
Collapse
Affiliation(s)
- Simone Pelassa
- UOC Rheumatology and Autoinflammatory Diseases, Department of Pediatric Sciences, Istituto Giannina Gaslini, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 16147 Genova, Italy
| | - Federica Raggi
- UOC Rheumatology and Autoinflammatory Diseases, Department of Pediatric Sciences, Istituto Giannina Gaslini, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 16147 Genova, Italy
| | - Chiara Rossi
- UOC Rheumatology and Autoinflammatory Diseases, Department of Pediatric Sciences, Istituto Giannina Gaslini, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 16147 Genova, Italy
| | - Maria Carla Bosco
- UOC Rheumatology and Autoinflammatory Diseases, Department of Pediatric Sciences, Istituto Giannina Gaslini, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), 16147 Genova, Italy
| |
Collapse
|
26
|
Salazar-Puerta AI, Rincon-Benavides MA, Cuellar-Gaviria TZ, Aldana J, Martinez GV, Ortega-Pineda L, Das D, Dodd D, Spencer CA, Deng B, McComb DW, Englert JA, Ghadiali S, Zepeda-Orozco D, Wold LE, Gallego-Perez D, Higuita-Castro N. Engineered Extracellular Vesicles Derived from Dermal Fibroblasts Attenuate Inflammation in a Murine Model of Acute Lung Injury. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210579. [PMID: 37119468 PMCID: PMC10573710 DOI: 10.1002/adma.202210579] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 03/29/2023] [Indexed: 06/06/2023]
Abstract
Acute respiratory distress syndrome (ARDS) represents a significant burden to the healthcare system, with ≈200 000 cases diagnosed annually in the USA. ARDS patients suffer from severe refractory hypoxemia, alveolar-capillary barrier dysfunction, impaired surfactant function, and abnormal upregulation of inflammatory pathways that lead to intensive care unit admission, prolonged hospitalization, and increased disability-adjusted life years. Currently, there is no cure or FDA-approved therapy for ARDS. This work describes the implementation of engineered extracellular vesicle (eEV)-based nanocarriers for targeted nonviral delivery of anti-inflammatory payloads to the inflamed/injured lung. The results show the ability of surfactant protein A (SPA)-functionalized IL-4- and IL-10-loaded eEVs to promote intrapulmonary retention and reduce inflammation, both in vitro and in vivo. Significant attenuation is observed in tissue damage, proinflammatory cytokine secretion, macrophage activation, influx of protein-rich fluid, and neutrophil infiltration into the alveolar space as early as 6 h post-eEVs treatment. Additionally, metabolomics analyses show that eEV treatment causes significant changes in the metabolic profile of inflamed lungs, driving the secretion of key anti-inflammatory metabolites. Altogether, these results establish the potential of eEVs derived from dermal fibroblasts to reduce inflammation, tissue damage, and the prevalence/progression of injury during ARDS via nonviral delivery of anti-inflammatory genes/transcripts.
Collapse
Affiliation(s)
- Ana I. Salazar-Puerta
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
| | - María A. Rincon-Benavides
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States
| | | | - Julian Aldana
- Biochemistry Program, The Ohio State University, Columbus, Ohio, United States
| | - Gabriela Vasquez Martinez
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, Ohio, United States
| | - Lilibeth Ortega-Pineda
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Devleena Das
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Daniel Dodd
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Biomedical Science Graduate Program, The Ohio State University, Columbus, Ohio, United States
| | - Charles A. Spencer
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University, Columbus, Ohio, United States
| | - Binbin Deng
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, Ohio, United States
| | - David W. McComb
- Center for Electron Microscopy and Analysis (CEMAS), The Ohio State University, Columbus, Ohio, United States
- Department of Materials Science and Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Joshua A. Englert
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Samir Ghadiali
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Division of Pulmonary, Critical Care, and Sleep Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Diana Zepeda-Orozco
- Kidney and Urinary Tract Research Center, The Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, Ohio, United States
- Department of Pediatrics, The Ohio State University, Columbus, Ohio, United States
- Division of Pediatric Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, Ohio, United States
| | - Loren E. Wold
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University, Columbus, Ohio, United States
| | - Daniel Gallego-Perez
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States
- Division of General Surgery, Department of Surgery, The Ohio State University, Columbus, Ohio, United States
| | - Natalia Higuita-Castro
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, United States
- Biophysics Program, The Ohio State University, Columbus, Ohio, United States
- Division of General Surgery, Department of Surgery, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
27
|
Patel S, Guo MK, Abdul Samad M, Howe KL. Extracellular vesicles as biomarkers and modulators of atherosclerosis pathogenesis. Front Cardiovasc Med 2023; 10:1202187. [PMID: 37304965 PMCID: PMC10250645 DOI: 10.3389/fcvm.2023.1202187] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 04/20/2023] [Indexed: 06/13/2023] Open
Abstract
Extracellular vesicles (EVs) are small, lipid bilayer-enclosed structures released by various cell types that play a critical role in intercellular communication. In atherosclerosis, EVs have been implicated in multiple pathophysiological processes, including endothelial dysfunction, inflammation, and thrombosis. This review provides an up-to-date overview of our current understanding of the roles of EVs in atherosclerosis, emphasizing their potential as diagnostic biomarkers and their roles in disease pathogenesis. We discuss the different types of EVs involved in atherosclerosis, the diverse cargoes they carry, their mechanisms of action, and the various methods employed for their isolation and analysis. Moreover, we underscore the importance of using relevant animal models and human samples to elucidate the role of EVs in disease pathogenesis. Overall, this review consolidates our current knowledge of EVs in atherosclerosis and highlights their potential as promising targets for disease diagnosis and therapy.
Collapse
Affiliation(s)
- Sarvatit Patel
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Mandy Kunze Guo
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Majed Abdul Samad
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
28
|
Levy D, Abadchi SN, Shababi N, Ravari MR, Pirolli NH, Bergeron C, Obiorah A, Mokhtari-Esbuie F, Gheshlaghi S, Abraham JM, Smith IM, Powsner E, Solomon T, Harmon JW, Jay SM. Induced pluripotent stem cell-derived extracellular vesicles promote wound repair in a diabetic mouse model via an anti-inflammatory immunomodulatory mechanism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.19.533334. [PMID: 36993554 PMCID: PMC10055496 DOI: 10.1101/2023.03.19.533334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Extracellular vesicles (EVs) derived from mesenchymal stem/stromal cells (MSCs) have recently been widely explored in clinical trials for treatment of diseases with complex pathophysiology. However, production of MSC EVs is currently hampered by donor-specific characteristics and limited ex vivo expansion capabilities before decreased potency, thus restricting their potential as a scalable and reproducible therapeutic. Induced pluripotent stem cells (iPSCs) represent a self-renewing source for obtaining differentiated iPSC-derived MSCs (iMSCs), circumventing both scalability and donor variability concerns for therapeutic EV production. Thus, we initially sought to evaluate the therapeutic potential of iMSC EVs. Interestingly, while utilizing undifferentiated iPSC EVs as a control, we found that their vascularization bioactivity was similar and their anti-inflammatory bioactivity was superior to donor-matched iMSC EVs in cell-based assays. To supplement this initial in vitro bioactivity screen, we employed a diabetic wound healing mouse model where both the pro-vascularization and anti-inflammatory activity of these EVs would be beneficial. In this in vivo model, iPSC EVs more effectively mediated inflammation resolution within the wound bed. Combined with the lack of additional differentiation steps required for iMSC generation, these results support the use of undifferentiated iPSCs as a source for therapeutic EV production with respect to both scalability and efficacy.
Collapse
Affiliation(s)
- Daniel Levy
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | | | - Niloufar Shababi
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Mohsen Rouhani Ravari
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Nicholas H. Pirolli
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Cade Bergeron
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Angel Obiorah
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Farzad Mokhtari-Esbuie
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Shayan Gheshlaghi
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - John M. Abraham
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Ian M. Smith
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Emily Powsner
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Talia Solomon
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - John W. Harmon
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Steven M. Jay
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
- Program in Molecular and Cell Biology, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
29
|
Phospholipid fatty acid remodeling and carbonylated protein increase in extracellular vesicles released by airway epithelial cells exposed to cigarette smoke extract. Eur J Cell Biol 2023; 102:151285. [PMID: 36584599 DOI: 10.1016/j.ejcb.2022.151285] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 12/14/2022] [Accepted: 12/17/2022] [Indexed: 12/27/2022] Open
Abstract
Cigarette smoke (CS) represents one of the most relevant environmental risk factors for several chronic pathologies. Tissue damage caused by CS exposure is mediated, at least in part, by oxidative stress induced by its toxic and pro-oxidant components. Evidence demonstrates that extracellular vesicles (EVs) released by various cell types exposed to CS extract (CSE) are characterized by altered biochemical cargo and gained pathological properties. In the present study, we evaluated the content of oxidized proteins and phospholipid fatty acid profiles of EVs released by human bronchial epithelial BEAS-2B cells treated with CSE. This specific molecular characterization has hitherto not been performed. After confirmation that CSE reduces viability of BEAS-2B cells and elevates intracellular ROS levels, in a dose-dependent manner, we demonstrated that 24 h exposure at 1% CSE, a concentration that only slight modifies cell viability but increases ROS levels, was able to increase carbonylated protein levels in cells and released EVs. The release of oxidatively modified proteins via EVs might represent a mechanism used by cells to remove toxic proteins in order to avoid their intracellular overloading. Moreover, 1% CSE induced only few changes in the fatty acid asset in BEAS-2B cell membrane phospholipids, whereas several rearrangements were observed in EVs released by CSE-treated cells. The impact of changes in acyl chain composition of CSE-EVs accounted for the increased saturation levels of phospholipids, a membrane parameter that might influence EV stability, uptake and, at least in part, EV-mediated biological effects. The present in vitro study adds new information concerning the biochemical composition of CSE-related EVs, useful to predict their biological effects on target cells. Furthermore, the information regarding the presence of oxidized proteins and the specific membrane features of CSE-related EVs can be useful to define the utilization of circulating EVs as marker for diagnosing of CS-induced lung damage and/or CS-related diseases.
Collapse
|
30
|
Alternative biological sources for extracellular vesicles production and purification strategies for process scale-up. Biotechnol Adv 2023; 63:108092. [PMID: 36608746 DOI: 10.1016/j.biotechadv.2022.108092] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/22/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023]
Abstract
Extracellular vesicles (EVs) are phospholipidic bi-layer enclosed nanoparticles secreted naturally by all cell types. They are attracting increasing attention in the fields of nanomedicine, nutraceutics and cosmetics as biocompatible carriers for drug delivery, with intrinsic properties beneficial to human health. Scientific work now focuses on developing techniques for isolating EVs that can translate into industrial-scale production and meet rigorous clinical requirements. The science of EVs is ongoing, and many pitfalls must be addressed, such as the requirement for standard, reproducible, inexpensive, and Good Manufacturing Practices (GMP) adherent EV processing techniques. Researchers are exploring the use of alternative sources to EVs derived from mammalian cultures, such as plant EVs, as well as the use of bacteria, algae and milk. Regarding the downstream processing of EVs, many alternative techniques to the ultracentrifugation (UC) protocols most commonly used in the laboratory are emerging. In the context of process scale-up, membrane-based processes for isolation and purification of EVs are the most promising, either as stand-alone processes or in combination with chromatographic techniques. This review discusses current trends on EVs source selection and EVs downstream processing techniques, with a focus on plant-derived EVs and membrane-based techniques for EVs enrichment.
Collapse
|
31
|
Stem Cell-derived Extracellular Vesicles: A Promising Nano Delivery Platform to the Brain? Stem Cell Rev Rep 2023; 19:285-308. [PMID: 36173500 DOI: 10.1007/s12015-022-10455-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2022] [Indexed: 02/07/2023]
Abstract
A very important cause of the frustration with drug therapy for central nervous system (CNS) diseases is the failure of drug delivery. The blood-brain barrier (BBB) prevents most therapeutic molecules from entering the brain while maintaining CNS homeostasis. Scientists are keen to develop new brain drug delivery systems to solve this dilemma. Extracellular vesicles (EVs), as a class of naturally derived nanoscale vesicles, have been extensively studied in drug delivery due to their superior properties. This review will briefly present current brain drug delivery strategies, including invasive and non-invasive techniques that target the brain, and the application of nanocarriers developed for brain drug delivery in recent years, especially EVs. The cellular origin of EVs affects the surface protein, size, yield, luminal composition, and other properties of EVs, which are also crucial in determining whether EVs are useful as drug carriers. Stem cell-derived EVs, which inherit the properties of parental cells and avoid the drawbacks of cell therapy, have always been favored by researchers. Thus, in this review, we will focus on the application of stem cell-derived EVs for drug delivery in the CNS. Various nucleic acids, proteins, and small-molecule drugs are loaded into EVs with or without modification and undergo targeted delivery to the brain to achieve their therapeutic effects. In addition, the challenges facing the clinical application of EVs as drug carriers will also be discussed. The directions of future efforts may be to improve drug loading efficiency and precise targeting.
Collapse
|
32
|
Karnas E, Dudek P, Zuba-Surma EK. Stem cell- derived extracellular vesicles as new tools in regenerative medicine - Immunomodulatory role and future perspectives. Front Immunol 2023; 14:1120175. [PMID: 36761725 PMCID: PMC9902918 DOI: 10.3389/fimmu.2023.1120175] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/11/2023] [Indexed: 01/25/2023] Open
Abstract
In the last few decades, the practical use of stem cells (SCs) in the clinic has attracted significant attention in the regenerative medicine due to the ability of these cells to proliferate and differentiate into other cell types. However, recent findings have demonstrated that the therapeutic capacity of SCs may also be mediated by their ability to secrete biologically active factors, including extracellular vesicles (EVs). Such submicron circular membrane-enveloped vesicles may be released from the cell surface and harbour bioactive cargo in the form of proteins, lipids, mRNA, miRNA, and other regulatory factors. Notably, growing evidence has indicated that EVs may transfer their bioactive content into recipient cells and greatly modulate their functional fate. Thus, they have been recently envisioned as a new class of paracrine factors in cell-to-cell communication. Importantly, EVs may modulate the activity of immune system, playing an important role in the regulation of inflammation, exhibiting broad spectrum of the immunomodulatory activity that promotes the transition from pro-inflammatory to pro-regenerative environment in the site of tissue injury. Consequently, growing interest is placed on attempts to utilize EVs in clinical applications of inflammatory-related dysfunctions as potential next-generation therapeutic factors, alternative to cell-based approaches. In this review we will discuss the current knowledge on the biological properties of SC-derived EVs, with special focus on their role in the regulation of inflammatory response. We will also address recent findings on the immunomodulatory and pro-regenerative activity of EVs in several disease models, including in vitro and in vivo preclinical, as well as clinical studies. Finally, we will highlight the current perspectives and future challenges of emerging EV-based therapeutic strategies of inflammation-related diseases treatment.
Collapse
|
33
|
The Role of Neutrophils in Lower Limb Peripheral Artery Disease: State of the Art and Future Perspectives. Int J Mol Sci 2023; 24:ijms24021169. [PMID: 36674682 PMCID: PMC9866688 DOI: 10.3390/ijms24021169] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/11/2023] Open
Abstract
In recent years, increasing attention has been paid to the role of neutrophils in cardiovascular (CV) disease (CVD) with evidence supporting their role in the initiation, progression, and rupture of atherosclerotic plaque. Although these cells have long been considered as terminally differentiated cells with a relatively limited spectrum of action, recent research has revealed intriguing novel cellular functions, including neutrophil extracellular trap (NET) generation and inflammasome activation, which have been linked to several human diseases, including CVD. While most research to date has focused on the role of neutrophils in coronary artery and cerebrovascular diseases, much less information is available on lower limb peripheral artery disease (PAD). PAD is a widespread condition associated with great morbidity and mortality, though physician and patient awareness of the disease remains low. To date, several studies have produced some evidence on the role of certain biomarkers of neutrophil activation in this clinical setting. However, the etiopathogenetic role of neutrophils, and in particular of some of the newly discovered mechanisms, has yet to be fully elucidated. In the future, complementary assessment of neutrophil activity should improve CV risk stratification and provide personalized treatments to patients with PAD. This review aims to summarize the basic principles and recent advances in the understanding of neutrophil biology, current knowledge about the role of neutrophils in atherosclerosis, as well as available evidence on their role of PAD.
Collapse
|
34
|
Sterner RC, Sterner RM. Immune response following traumatic spinal cord injury: Pathophysiology and therapies. Front Immunol 2023; 13:1084101. [PMID: 36685598 PMCID: PMC9853461 DOI: 10.3389/fimmu.2022.1084101] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating condition that is often associated with significant loss of function and/or permanent disability. The pathophysiology of SCI is complex and occurs in two phases. First, the mechanical damage from the trauma causes immediate acute cell dysfunction and cell death. Then, secondary mechanisms of injury further propagate the cell dysfunction and cell death over the course of days, weeks, or even months. Among the secondary injury mechanisms, inflammation has been shown to be a key determinant of the secondary injury severity and significantly worsens cell death and functional outcomes. Thus, in addition to surgical management of SCI, selectively targeting the immune response following SCI could substantially decrease the progression of secondary injury and improve patient outcomes. In order to develop such therapies, a detailed molecular understanding of the timing of the immune response following SCI is necessary. Recently, several studies have mapped the cytokine/chemokine and cell proliferation patterns following SCI. In this review, we examine the immune response underlying the pathophysiology of SCI and assess both current and future therapies including pharmaceutical therapies, stem cell therapy, and the exciting potential of extracellular vesicle therapy.
Collapse
Affiliation(s)
- Robert C. Sterner
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, United States
| | - Rosalie M. Sterner
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States,*Correspondence: Rosalie M. Sterner,
| |
Collapse
|
35
|
Raggi F, Bartolucci M, Cangelosi D, Rossi C, Pelassa S, Trincianti C, Petretto A, Filocamo G, Civino A, Eva A, Ravelli A, Consolaro A, Bosco MC. Proteomic profiling of extracellular vesicles in synovial fluid and plasma from Oligoarticular Juvenile Idiopathic Arthritis patients reveals novel immunopathogenic biomarkers. Front Immunol 2023; 14:1134747. [PMID: 37205098 PMCID: PMC10186353 DOI: 10.3389/fimmu.2023.1134747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/28/2023] [Indexed: 05/21/2023] Open
Abstract
Introduction New early low-invasive biomarkers are demanded for the management of Oligoarticular Juvenile Idiopathic Arthritis (OJIA), the most common chronic pediatric rheumatic disease in Western countries and a leading cause of disability. A deeper understanding of the molecular basis of OJIA pathophysiology is essential for identifying new biomarkers for earlier disease diagnosis and patient stratification and to guide targeted therapeutic intervention. Proteomic profiling of extracellular vesicles (EVs) released in biological fluids has recently emerged as a minimally invasive approach to elucidate adult arthritis pathogenic mechanisms and identify new biomarkers. However, EV-prot expression and potential as biomarkers in OJIA have not been explored. This study represents the first detailed longitudinal characterization of the EV-proteome in OJIA patients. Methods Fourty-five OJIA patients were recruited at disease onset and followed up for 24 months, and protein expression profiling was carried out by liquid chromatography-tandem mass spectrometry in EVs isolated from plasma (PL) and synovial fluid (SF) samples. Results We first compared the EV-proteome of SF vs paired PL and identified a panel of EV-prots whose expression was significantly deregulated in SF. Interaction network and GO enrichment analyses performed on deregulated EV-prots through STRING database and ShinyGO webserver revealed enrichment in processes related to cartilage/bone metabolism and inflammation, suggesting their role in OJIA pathogenesis and potential value as early molecular indicators of OJIA development. Comparative analysis of the EV-proteome in PL and SF from OJIA patients vs PL from age/gender-matched control children was then carried out. We detected altered expression of a panel of EV-prots able to differentiate new-onset OJIA patients from control children, potentially representing a disease-associated signature measurable at both the systemic and local levels with diagnostic potential. Deregulated EV-prots were significantly associated with biological processes related to innate immunity, antigen processing and presentation, and cytoskeleton organization. Finally, we ran WGCNA on the SF- and PL-derived EV-prot datasets and identified a few EV-prot modules associated with different clinical parameters stratifying OJIA patients in distinct subgroups. Discussion These data provide novel mechanistic insights into OJIA pathophysiology and an important contribution in the search of new candidate molecular biomarkers for the disease.
Collapse
Affiliation(s)
- Federica Raggi
- Laboratory of Molecular Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
- Unit of Autoinflammatory Diseases and Immunodeficiences, Pediatric Rheumatology Clinic, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Martina Bartolucci
- Core Facilities, Clinical Proteomics and Metabolomics, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Davide Cangelosi
- Laboratory of Molecular Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
- Clinical Bioinformatics Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Chiara Rossi
- Laboratory of Molecular Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
- Unit of Autoinflammatory Diseases and Immunodeficiences, Pediatric Rheumatology Clinic, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Simone Pelassa
- Laboratory of Molecular Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
- Unit of Autoinflammatory Diseases and Immunodeficiences, Pediatric Rheumatology Clinic, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Chiara Trincianti
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal-Infantile Sciences (DiNOGMI), University of Genova, Genova, Italy
| | - Andrea Petretto
- Core Facilities, Clinical Proteomics and Metabolomics, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Giovanni Filocamo
- Division of Pediatric Immunology and Rheumatology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Cà Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Adele Civino
- Pediatric Rheumatology and Immunology, Ospedale “Vito Fazzi”, Lecce, Italy
| | - Alessandra Eva
- Laboratory of Molecular Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Angelo Ravelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal-Infantile Sciences (DiNOGMI), University of Genova, Genova, Italy
- Scientific Direction, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Alessandro Consolaro
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics and Maternal-Infantile Sciences (DiNOGMI), University of Genova, Genova, Italy
- Pediatric Rheumatology Clinic, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
| | - Maria Carla Bosco
- Laboratory of Molecular Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
- Unit of Autoinflammatory Diseases and Immunodeficiences, Pediatric Rheumatology Clinic, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genova, Italy
- *Correspondence: Maria Carla Bosco,
| |
Collapse
|
36
|
Anselmo A, Boselli D. Flow Cytometry Analysis of IL-1 Receptors and Toll-Like Receptors on Platelets and Platelet-Derived Extracellular Vesicles. Methods Mol Biol 2023; 2700:117-137. [PMID: 37603177 DOI: 10.1007/978-1-0716-3366-3_6] [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: 08/22/2023]
Abstract
Flow cytometry is largely used for the immunophenotyping and quantification of several cell types or related components including platelets and extracellular vesicles. Platelets and platelet-derived extracellular vesicles (PEVs) are receiving increased interest in inflammatory diseases including sepsis. Thus, in this chapter, we will describe protocols for the flow cytometry analysis of platelets, platelet/neutrophils hetero aggregates, and PEVs mainly focusing on the evaluation of the surface expression of some IL-1 receptor (ILR) and Toll-like receptor (TLR) family members.
Collapse
Affiliation(s)
- Achille Anselmo
- Flow Cytometry Resource, Advanced Cytometry Technical Applications Laboratory, San Raffaele Scientific Institute, Milan, Italy.
| | - Daniela Boselli
- Flow Cytometry Resource, Advanced Cytometry Technical Applications Laboratory, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
37
|
Membrane-Binding Biomolecules Influence the Rate of Vesicle Exchange between Bacteria. Appl Environ Microbiol 2022; 88:e0134622. [PMID: 36342184 PMCID: PMC9746307 DOI: 10.1128/aem.01346-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The exchange of bacterial extracellular vesicles facilitates molecular exchange between cells, including the horizontal transfer of genetic material. Given the implications of such transfer events on cell physiology and adaptation, some bacterial cells have likely evolved mechanisms to regulate vesicle exchange. Past work has identified mechanisms that influence the formation of extracellular vesicles, including the production of small molecules that modulate membrane structure; however, whether these mechanisms also modulate vesicle uptake and have an overall impact on the rate of vesicle exchange is unknown. Here, we show that membrane-binding molecules produced by microbes influence both the formation and uptake of extracellular vesicles and have the overall impact of increasing the vesicle exchange rate within a bacterial coculture. In effect, production of compounds that increase vesicle exchange rates encourage gene exchange between neighboring cells. The ability of several membrane-binding compounds to increase vesicle exchange was demonstrated. Three of these compounds, nisin, colistin, and polymyxin B, are antimicrobial peptides added at sub-inhibitory concentrations. These results suggest that a potential function of exogenous compounds that bind to membranes may be the regulation of vesicle exchange between cells. IMPORTANCE The exchange of bacterial extracellular vesicles is one route of gene transfer between bacteria, although it was unclear if bacteria developed strategies to modulate the rate of gene transfer within vesicles. In eukaryotes, there are many examples of specialized molecules that have evolved to facilitate the production, loading, and uptake of vesicles. Recent work with bacteria has shown that some small molecules influence membrane curvature and induce vesicle formation. Here, we show that similar compounds facilitate vesicle uptake, thereby increasing the overall rate of vesicle exchange within bacterial populations. The addition of membrane-binding compounds, several of them antibiotics at subinhibitory concentrations, to a bacterial coculture increased the rate of horizontal gene transfer via vesicle exchange.
Collapse
|
38
|
Han D, Lu D, Huang S, Pang J, Wu Y, Hu J, Zhang X, Pi Y, Zhang G, Wang J. Small extracellular vesicles from Ptpn1-deficient macrophages alleviate intestinal inflammation by reprogramming macrophage polarization via lactadherin enrichment. Redox Biol 2022; 58:102558. [PMID: 36462232 PMCID: PMC9712762 DOI: 10.1016/j.redox.2022.102558] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/22/2022] [Accepted: 11/26/2022] [Indexed: 11/29/2022] Open
Abstract
Tyrosine-protein phosphatase non-receptor type 1 (Ptpn1) is known to be involved in macrophage polarization. However, whether and how Ptpn1 regulates macrophage phenotype to affect intestinal epithelial barrier function remains largely unexplored. Herein, we investigated the impact of Ptpn1 and macrophage-derived small extracellular vesicles (sEVs) on macrophage-intestinal epithelial cell (IEC) interactions in the context of intestinal inflammation. We found that Ptpn1 knockdown shifts macrophages toward the anti-inflammatory M2 phenotype, thereby promoting intestinal barrier integrity and suppressing inflammatory response in the macrophage-IEC co-culture model. We further revealed that conditioned medium or sEVs isolated from Ptp1b knockdown macrophages are the primary factor driving the beneficial outcomes. Consistently, administration of the sEVs from Ptpn1-knockdown macrophages reduced disease severity and ameliorated intestinal inflammation in LPS-challenged mice. Furthermore, depletion of macrophages in mice abrogated the protective effect of Ptpn1-knockdown macrophage sEVs against Salmonella Typhimurium infection. Importantly, we found lactadherin to be highly enriched in the sEVs of Ptpn1-knockdown macrophages. Administration of recombinant lactadherin alleviated intestinal inflammation and barrier dysfunction by inducing macrophage M2 polarization. Interestingly, sEVs lactadherin was also internalized by macrophages and IECs, leading to macrophage M2 polarization and enhanced intestinal barrier integrity. Mechanistically, the anti-inflammatory and barrier-enhancing effect of lactadherin was achieved by reducing TNF-α and NF-κB activation. Thus, we demonstrated that sEVs from Ptpn1-knockdown macrophages mediate the communication between IECs and macrophages through enrichment of lactadherin. The outcome could potentially lead to the development of novel therapies for intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Dongdong Lu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Shimeng Huang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jiaman Pang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yujun Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jie Hu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiangyu Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yu Pi
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Guolong Zhang
- Department of Animal and Food Sciences, Oklahoma State University, Stillwater, Oklahoma, 74078, USA
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
39
|
Mesenchymal Stem Cell-Derived Extracellular Vesicles Therapy for Pulmonary Hypertension: A Comprehensive Review of Preclinical Studies. J Interv Cardiol 2022; 2022:5451947. [PMID: 36419957 PMCID: PMC9652076 DOI: 10.1155/2022/5451947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 10/09/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
Pulmonary hypertension (PH) is a type of clinical pathophysiological syndrome characterized by a progressive increase in pulmonary vascular resistance and subsequent progressive failure of the right heart function, and is a common complication of many diseases. Mesenchymal stem cells (MSCs) autonomously home to sites damaged by disease, repair damaged tissues, and participate in the regulation of systemic inflammation and immune responses, which have good clinical application prospects. Extracellular vesicles (EVs), such as exosomes and microvesicles, participate in various biological activities by regulating intercellular communication. Exosomes secreted into the extracellular environment also affect the host immune system. MSC-derived extracellular vesicles (MSC-EVs), as a mediator in the paracrine processes of MSCs, carry biologically active substances such as proteins, lipids, mRNA, and micro-RNA. MSC-EVs therapies, safer than cell-based treatments, have been shown to be effective in modulating macrophages to support anti-inflammatory phenotypes, which are strongly related to histological and functional benefits in preclinical models of pulmonary hypertension. The main effects of active substances and their potential medical value have attracted wide attention from researchers. This article reviews the role and relevant mechanisms of MSC-EVs in the treatment of pulmonary hypertension in recent studies and provides a basis for their future clinical applications.
Collapse
|
40
|
Wu HM, Chen LH, Hsu LT, Lai CH. Immune Tolerance of Embryo Implantation and Pregnancy: The Role of Human Decidual Stromal Cell- and Embryonic-Derived Extracellular Vesicles. Int J Mol Sci 2022; 23:ijms232113382. [PMID: 36362169 PMCID: PMC9658721 DOI: 10.3390/ijms232113382] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/30/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022] Open
Abstract
Embryo–endometrial communication plays a critical role in embryo implantation and the establishment of a successful pregnancy. Successful pregnancy outcomes involve maternal immune modulation during embryo implantation. The endometrium is usually primed and immunomodulated by steroid hormones and embryo signals for subsequent embryo implantation and the maintenance of pregnancy. The roles of extracellular vesicles (EVs) and microRNAs for the embryo–maternal interactions have been elucidated recently. New evidence shows that endometrial EVs and trophectoderm-originated EV cargo, including microRNAs, proteins, and lipids in the physiological microenvironment, regulate maternal immunomodulation for embryo implantation and subsequent pregnancy. On the other hand, trophoblast-derived EVs also control the cross-communication between the trophoblasts and immune cells. The exploration of EV functions and mechanisms in the processes of embryo implantation and pregnancy will shed light on a practical tool for the diagnostic or therapeutic approaches to reproductive medicine and infertility.
Collapse
Affiliation(s)
- Hsien-Ming Wu
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Liang-Hsuan Chen
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Le-Tien Hsu
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Chyong-Huey Lai
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
- Gynecologic Cancer Research Center, Linkou Medical Center, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
- Correspondence: ; Tel.: +886-3-328-1200 (ext. 8254)
| |
Collapse
|
41
|
Badimon L, Padro T, Arderiu G, Vilahur G, Borrell-Pages M, Suades R. Extracellular vesicles in atherothrombosis: From biomarkers and precision medicine to therapeutic targets. Immunol Rev 2022; 312:6-19. [PMID: 35996799 DOI: 10.1111/imr.13127] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of global mortality. Extracellular vesicles (EVs) are small phospholipid vesicles that convey molecular bioactive cargoes and play essential roles in intercellular communication and, hence, a multifaceted role in health and disease. The present review offers a glimpse into the current state and up-to-date concepts on EV field. It also covers their association with several cardiovascular risk factors and ischemic conditions, being subclinical atherosclerosis of utmost relevance for prevention. Interestingly, we show that EVs hold promise as prognostic and diagnostic as well as predictive markers of ASCVD in the precision medicine era. We then report on the role of EVs in atherothrombosis, disentangling the mechanisms involved in the initiation, progression, and complication of atherosclerosis and showing their direct effect in the context of arterial thrombosis. Finally, their potential use for therapeutic intervention is highlighted.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain.,Cardiovascular Research Chair, UAB, Barcelona, Spain
| | - Teresa Padro
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Arderiu
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Maria Borrell-Pages
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Suades
- Cardiovascular Program ICCC, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain.,CIBERCV Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
42
|
A Tale of Two: When Neural Stem Cells Encounter Hypoxia. Cell Mol Neurobiol 2022:10.1007/s10571-022-01293-6. [DOI: 10.1007/s10571-022-01293-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/02/2022] [Indexed: 11/12/2022]
|
43
|
Rajput A, Varshney A, Bajaj R, Pokharkar V. Exosomes as New Generation Vehicles for Drug Delivery: Biomedical Applications and Future Perspectives. Molecules 2022; 27:7289. [PMID: 36364116 PMCID: PMC9658823 DOI: 10.3390/molecules27217289] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/30/2022] Open
Abstract
Currently, particular interest among the scientific community is focused on exploring the use of exosomes for several pharmaceutical and biomedical applications. This is due to the identification of the role of exosomes as an excellent intercellular communicator by delivering the requisite cargo comprising of functional proteins, metabolites and nucleic acids. Exosomes are the smallest extracellular vesicles (EV) with sizes ranging from 30-100 nm and are derived from endosomes. Exosomes have similar surface morphology to cells and act as a signal transduction channel between cells. They encompass different biomolecules, such as proteins, nucleic acids and lipids, thus rendering them naturally as an attractive drug delivery vehicle. Like the other advanced drug delivery systems, such as polymeric nanoparticles and liposomes to encapsulate drug substances, exosomes also gained much attention in enhancing therapeutic activity. Exosomes present many advantages, such as compatibility with living tissues, low toxicity, extended blood circulation, capability to pass contents from one cell to another, non-immunogenic and special targeting of various cells, making them an excellent therapeutic carrier. Exosome-based molecules for drug delivery are still in the early stages of research and clinical trials. The problems and clinical transition issues related to exosome-based drugs need to be overcome using advanced tools for better understanding and systemic evaluation of exosomes. In this current review, we summarize the most up-to-date knowledge about the complex biological journey of exosomes from biogenesis and secretion, isolation techniques, characterization, loading methods, pharmaceutical and therapeutic applications, challenges and future perspectives of exosomes.
Collapse
Affiliation(s)
| | | | | | - Varsha Pokharkar
- Department of Pharmaceutics, Poona College of Pharmacy, Bharti Vidyapeeth Deemed University, Erandwane, Pune 411038, Maharashtra, India
| |
Collapse
|
44
|
Kell DB, Pretorius E. The potential role of ischaemia-reperfusion injury in chronic, relapsing diseases such as rheumatoid arthritis, Long COVID, and ME/CFS: evidence, mechanisms, and therapeutic implications. Biochem J 2022; 479:1653-1708. [PMID: 36043493 PMCID: PMC9484810 DOI: 10.1042/bcj20220154] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 02/07/2023]
Abstract
Ischaemia-reperfusion (I-R) injury, initiated via bursts of reactive oxygen species produced during the reoxygenation phase following hypoxia, is well known in a variety of acute circumstances. We argue here that I-R injury also underpins elements of the pathology of a variety of chronic, inflammatory diseases, including rheumatoid arthritis, ME/CFS and, our chief focus and most proximally, Long COVID. Ischaemia may be initiated via fibrin amyloid microclot blockage of capillaries, for instance as exercise is started; reperfusion is a necessary corollary when it finishes. We rehearse the mechanistic evidence for these occurrences here, in terms of their manifestation as oxidative stress, hyperinflammation, mast cell activation, the production of marker metabolites and related activities. Such microclot-based phenomena can explain both the breathlessness/fatigue and the post-exertional malaise that may be observed in these conditions, as well as many other observables. The recognition of these processes implies, mechanistically, that therapeutic benefit is potentially to be had from antioxidants, from anti-inflammatories, from iron chelators, and via suitable, safe fibrinolytics, and/or anti-clotting agents. We review the considerable existing evidence that is consistent with this, and with the biochemical mechanisms involved.
Collapse
Affiliation(s)
- Douglas B. Kell
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Kemitorvet 200, 2800 Kgs Lyngby, Denmark
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| | - Etheresia Pretorius
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 7ZB, U.K
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1 Matieland 7602, South Africa
| |
Collapse
|
45
|
Yan J, Zhen Y, Wang R, Li X, Huang S, Zhong H, Wen H, Sun Q. Downregulation of miRNA miR-1305 and upregulation of miRNA miR-6785-5p may be associated with psoriasis. Front Genet 2022; 13:891465. [PMID: 36035154 PMCID: PMC9399421 DOI: 10.3389/fgene.2022.891465] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The role of serum extracellular vesicles (EVs) is less known in psoriasis. Objectives: To explore the transcriptomic profile of serum EVs and the potential biomarkers in psoriasis. Methods: EVs were isolated by differential ultracentrifugation and identified by transmission electron microscope. The diameters of EVs were detected using nanoparticle tracking analysis. Serum EVs-keratinocyte interaction was observed through confocal fluorescence microscopy. miRNA microarray and mRNA microarray were performed in serum EVs (n = 4) and skin lesions (n = 3), respectively. Quantitative reverse-transcriptase polymerase chain reaction (qRT-PCR) and fluorescence in situ hybridization were used to detect the expression of miRNAs in serum EVs and skin lesions (n = 15). Bioinformatics analysis was performed to predict the potential target genes and functions of miR-1305 and miR-6785-5p. Western blot, CCK-8 and enzyme-linked immunosorbent assay (ELISA) were used to detect the EVs’ biomarkers, keratinocytes proliferation and cytokines secretion. Results: A total of 16 miRNAs and 1,725 mRNAs were significantly dysregulated in serum EVs and skin lesions, respectively. miR-1305 was down-regulated and miR-6785-5p was upregulated in both serum EVs and skin lesions. Serum EVs could be taken up by keratinocytes. miR-1305 was downregulated and miR-6785-5p were upregulated in keratinocytes after co-cultured with psoriasis serum EVs compared with controls. Psoriasis serum EVs promoted keratinocyte proliferation and the secretion of CCL20 and IL-8. Serum EVs miR-1305 and miR-6785-5p were associated with disease severity. Conclusion: Serum EVs might be involved in the activation of keratinocytes through loaded miRNAs in psoriasis. Serum EVs miR-1305 and miR-6785-5p may be associated with psoriasis.
Collapse
Affiliation(s)
- Jianjun Yan
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Yunyue Zhen
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Ruijie Wang
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Xueqing Li
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Shan Huang
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Hua Zhong
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - He Wen
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
| | - Qing Sun
- Department of Dermatology, Qilu Hospital of Shandong University, Jinan, China
- Laboratory of Basic Medical Science, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Qing Sun,
| |
Collapse
|
46
|
Lu B, Ku J, Flojo R, Olson C, Bengford D, Marriott G. Exosome- and extracellular vesicle-based approaches for the treatment of lysosomal storage disorders. Adv Drug Deliv Rev 2022; 188:114465. [PMID: 35878794 DOI: 10.1016/j.addr.2022.114465] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 06/22/2022] [Accepted: 07/19/2022] [Indexed: 12/16/2022]
Abstract
Cell-generated extracellular vesicles (EVs) are being engineered as biologically-inspired vehicles for targeted delivery of therapeutic agents to treat difficult-to-manage human diseases, including lysosomal storage disorders (LSDs). Engineered EVs offer distinct advantages for targeted delivery of therapeutics compared to existing synthetic and semi-synthetic nanoscale systems, for example with regard to their biocompatibility, circulation lifetime, efficiencies in delivery of drugs and biologics to target cells, and clearance from the body. Here, we review literature related to the design and preparation of EVs as therapeutic carriers for targeted delivery and therapy of drugs and biologics with a focus on LSDs. First, we introduce the basic pathophysiology of LDSs and summarize current approaches to diagnose and treat LSDs. Second, we will provide specific details about EVs, including subtypes, biogenesis, biological properties and their potential to treat LSDs. Third, we review state-of-the-art approaches to engineer EVs for treatments of LSDs. Finally, we summarize explorative basic research and applied applications of engineered EVs for LSDs, and highlight current challenges, and new directions in developing EV-based therapies and their potential impact on clinical medicine.
Collapse
Affiliation(s)
- Biao Lu
- Department of Bioengineering, School of Engineering, Santa Clara University, 500 El Camino Real, Santa Clara, California 95053, USA
| | - Joy Ku
- Department of Bioengineering, School of Engineering, Santa Clara University, 500 El Camino Real, Santa Clara, California 95053, USA
| | - Renceh Flojo
- Department of Bioengineering, School of Engineering, Santa Clara University, 500 El Camino Real, Santa Clara, California 95053, USA
| | - Chris Olson
- Department of Bioengineering, School of Engineering, Santa Clara University, 500 El Camino Real, Santa Clara, California 95053, USA
| | - David Bengford
- Department of Bioengineering, School of Engineering, Santa Clara University, 500 El Camino Real, Santa Clara, California 95053, USA
| | - Gerard Marriott
- Department of Bioengineering, University of California at Berkeley, California 94720, USA.
| |
Collapse
|
47
|
Extracellular Vesicles, Inflammation, and Cardiovascular Disease. Cells 2022; 11:cells11142229. [PMID: 35883672 PMCID: PMC9320258 DOI: 10.3390/cells11142229] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular disease is a leading cause of death worldwide. The underlying mechanisms of most cardiovascular disorders involve innate and adaptive immune responses, and extracellular vesicles are implicated in both. In this review, we describe the mechanistic role of extracellular vesicles at the intersection of inflammatory processes and cardiovascular disease. Our discussion focuses on atherosclerosis, myocardial ischemia and ischemic heart disease, heart failure, aortic aneurysms, and valvular pathology.
Collapse
|
48
|
Yang X, Zheng E, Chatterjee V, Ma Y, Reynolds A, Villalba N, Wu MH, Yuan SY. Protein palmitoylation regulates extracellular vesicle production and function in sepsis. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e50. [PMID: 38419739 PMCID: PMC10901530 DOI: 10.1002/jex2.50] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/16/2022] [Accepted: 06/19/2022] [Indexed: 03/02/2024]
Abstract
Extracellular vesicles (EVs) are bioactive membrane-encapsulated particles generated by a series of events involving membrane budding, fission and fusion. Palmitoylation, mediated by DHHC palmitoyl acyltransferases, is a lipidation reaction that increases protein lipophilicity and membrane localization. Here, we report palmitoylation as a novel regulator of EV formation and function during sepsis. Our results showed significantly decreased circulating EVs in mice with DHHC21 functional deficiency (Zdhhc21dep/dep), compared to wild-type (WT) mice 24 h after septic injury. Furthermore, WT and Zdhhc21dep/dep EVs displayed distinct palmitoyl-proteomic profiles. Ingenuity pathway analysis indicated that sepsis altered several inflammation related pathways expressed in EVs, among which the most significantly activated was the complement pathway; however, this sepsis-induced complement enrichment in EVs was greatly blunted in Zdhhc21dep/dep EVs. Functionally, EVs isolated from WT mice with sepsis promoted neutrophil adhesion, transmigration, and neutrophil extracellular trap production; these effects were significantly attenuated by DHHC21 loss-of-function. Furthermore, Zdhhc21dep/dep mice displayed reduced neutrophil infiltration in lungs and improved survival after CLP challenges. These findings indicate that blocking palmitoylation via DHHC21 functional deficiency can reduce sepsis-stimulated production of complement-enriched EVs and attenuates their effects on neutrophil activity.
Collapse
Affiliation(s)
- Xiaoyuan Yang
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Ethan Zheng
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Victor Chatterjee
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Yonggang Ma
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Amanda Reynolds
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Nuria Villalba
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Mack H. Wu
- Department of SurgeryUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Sarah Y. Yuan
- Department of Molecular Pharmacology and PhysiologyUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
- Department of SurgeryUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| |
Collapse
|
49
|
Li C, Zheng X, Hu M, Jia M, Jin R, Nie Y. Recent progress in therapeutic strategies and biomimetic nanomedicines for rheumatoid arthritis treatment. Expert Opin Drug Deliv 2022; 19:883-898. [PMID: 35760767 DOI: 10.1080/17425247.2022.2094364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Rheumatoid arthritis (RA) is an autoimmune systemic disease in which inflammatory and immune cells accumulate in inflamed joints. Researchers aimed at the characteristics of RA to achieve the effect of treating RA through different therapeutic strategies, and have used various endogenous materials to design drug-loaded nanoparticles that can target RA by binding to cell adhesion molecules or chemokines. In some cases, the nanoparticles can respond to the characteristics of the microenvironment. AREAS COVERED This article reviews the recent advances in the treatment of RA from two aspects of therapeutic strategies and delivery strategies. Therapeutic strategies mainly include neutralization of inflammatory factors, promotion of inflammatory cell apoptosis, ROS scavenger, immunosuppression, and bone tissue repair. The drug delivery strategy is mainly described from two aspects: chemically functionalized biomimetic nanoparticles and endogenous nanoparticles. EXPERT OPINION Biomimetic NPs may be effective drug carriers for targeted RA treatment. NPs can reduce the clearance of mononuclear phagocytes, prolong the blood circulation time, and improve the targeting ability. With the deepening of research, more and more biomimetic NPs have entered the clinical trial stage. However, safe and scalable preparation methods are needed to improve their clinical applicability.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China.,National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Xiu Zheng
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Mei Hu
- Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Ming Jia
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Rongrong Jin
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| | - Yu Nie
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, P. R. China
| |
Collapse
|
50
|
Thome AD, Thonhoff JR, Zhao W, Faridar A, Wang J, Beers DR, Appel SH. Extracellular Vesicles Derived From Ex Vivo Expanded Regulatory T Cells Modulate In Vitro and In Vivo Inflammation. Front Immunol 2022; 13:875825. [PMID: 35812435 PMCID: PMC9258040 DOI: 10.3389/fimmu.2022.875825] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vehicles (EVs) are efficient biomarkers of disease and participate in disease pathogenesis; however, their use as clinical therapies to modify disease outcomes remains to be determined. Cell-based immune therapies, including regulatory T cells (Tregs), are currently being clinically evaluated for their usefulness in suppressing pro-inflammatory processes. The present study demonstrates that ex vivo expanded Tregs generate a large pool of EVs that express Treg-associated markers and suppress pro-inflammatory responses in vitro and in vivo. Intravenous injection of Treg EVs into an LPS-induced mouse model of inflammation reduced peripheral pro-inflammatory transcripts and increased anti-inflammatory transcripts in myeloid cells as well as Tregs. Intranasal administration of enriched Treg EVs in this model also reduced pro-inflammatory transcripts and the associated neuroinflammatory responses. In a mouse model of amyotrophic lateral sclerosis, intranasal administration of enriched Treg EVs slowed disease progression, increased survival, and modulated inflammation within the diseased spinal cord. These findings support the therapeutic potential of expanded Treg EVs to suppress pro-inflammatory responses in human disease.
Collapse
Affiliation(s)
- Aaron D Thome
- Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Jason R Thonhoff
- Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Weihua Zhao
- Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Alireza Faridar
- Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Jinghong Wang
- Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - David R Beers
- Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| | - Stanley H Appel
- Department of Neurology, Houston Methodist Neurological Institute, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|