1
|
Saadh MJ, Allela OQB, Al-Hussainy AF, Baldaniya L, Rekha MM, Nathiya D, Kaur P, Aminov Z, Sameer HN, Hameed HG, Athab ZH, Adil M. Exosomal non-coding RNAs: gatekeepers of inflammation in autoimmune disease. J Inflamm (Lond) 2025; 22:18. [PMID: 40369549 PMCID: PMC12079953 DOI: 10.1186/s12950-025-00443-z] [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: 11/21/2024] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
Autoimmune diseases (AIDs) are marked by systemic inflammation and immune dysregulation, yet current therapies often fail to target their underlying causes. Emerging evidence positions exosomal non-coding RNAs (ncRNAs)-including miRNAs, lncRNAs, and circRNAs-as key regulators of inflammatory pathways, providing critical insights into AID pathogenesis. This review synthesizes recent advances in how these ncRNAs orchestrate immune cell communication, modulate inflammatory mediators, and drive microglial activation in neuroinflammatory AIDs. It evaluates their dual role as disease amplifiers (e.g., miR-155 in lupus, miR-326 in rheumatoid arthritis) and therapeutic targets, emphasizing their potential to reprogram immune responses or deliver anti-inflammatory agents. In this review, we first provide a glimpse into the pathogenesis of autoimmune diseases and delve into the structure and function of exosomes, emphasizing their role in cell-cell communication. We then discuss the regulatory roles of exosomal ncRNAs in immune modulation, detailing their types, functions, and mechanisms of action. Finally, we examine the implications of exosomes and exosomal ncRNAs in the context of autoimmune diseases, with a particular focus on microglial activation and its contribution to neuroinflammation.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | | | | | - Lalji Baldaniya
- Department of Pharmacy, Faculty of Health Sciences, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Deepak Nathiya
- Department of Pharmacy Practice, Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Zafar Aminov
- Department of Public Health and Healthcare management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Mohaned Adil
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
2
|
Fan R, Liu H, Liang Q. Roles and Therapeutic Targeting of Exosomes in Sepsis-Induced Cardiomyopathy. J Cell Mol Med 2025; 29:e70559. [PMID: 40264381 PMCID: PMC12015131 DOI: 10.1111/jcmm.70559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/31/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025] Open
Abstract
Sepsis-induced cardiomyopathy (SICM) is a complex and fatal manifestation of sepsis, characterised by myocardial dysfunction that exacerbates the clinical prognosis in septic patients. While the pathophysiology of SICM remains incompletely understood, emerging evidence highlights the multifaceted functions of exosomes, small membrane-bound extracellular vesicles, in mediating the inflammatory responses and cardiac dysfunction involved in this condition. During sepsis, exosomes are secreted by various cells, such as cardiomyocytes, endothelial cells and macrophages, which serve as critical messengers, transferring proteins, lipids and RNA molecules that influence recipient cells, thus affecting cellular functions and disease progression. This review summarises the pathophysiology of SICM and the basics of exosomes and focuses on exosome-mediated mechanisms in SICM, including their role in inflammation, oxidative stress, mitochondrial dysfunction and myocardial injury, offering novel insights into the exosome-based therapeutic strategies in SICM.
Collapse
Affiliation(s)
- Rui Fan
- Graduate SchoolHeilongjiang University of Chinese MedicineHarbinChina
| | - Han Liu
- Graduate SchoolUniversity College LondonLondonUK
| | - Qun Liang
- Department of Critical Care MedicineFirst Affiliated Hospital of Heilongjiang University of Chinese MedicineHarbinChina
| |
Collapse
|
3
|
García-Concejo A, Sánchez-Quirós B, Gómez-Sánchez E, Sánchez-de Prada L, Tamayo-Velasco Á, Tovar-Doncel MS, Lorenzo M, Gómez-Pesquera E, Poves-Álvarez R, Bernardo D, Martín-Fernández M, Gonzalo-Benito H, Moreno-Portales P, Prieto-Utrera R, Bardají-Carrillo M, López-Herrero R, Fernández Arranz M, Calaveras-Fernández R, Tomillo-Cebrián F, Aydillo T, Jiménez-Sousa MÁ, Fernández-Rodríguez A, Resino S, Heredia-Rodríguez M, Martínez-Paz P, Tamayo E. Study on the diagnostic role of exosome-derived miRNAs in postoperative septic shock and non-septic shock patients. Crit Care 2025; 29:96. [PMID: 40033446 PMCID: PMC11874436 DOI: 10.1186/s13054-025-05320-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/15/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Diagnosing septic shock promptly is essential but challenging, especially due to its clinical similarity to non-septic shock. Extracellular vesicle-derived miRNAs may serve as biomarkers to distinguish septic shock from non-septic shock, providing a more accurate diagnostic tool for postsurgical patients. This study aims to identify extracellular vesicle-derived miRNA signatures that differentiate septic shock from non-septic shock in postsurgical patients, potentially improving diagnostic accuracy and clinical decision-making. METHODS A multicentre, prospective study was conducted on miRNA profiles in shock patients. Two cohorts were recruited from the Intensive Care Units of two Spanish hospitals: a discovery cohort with 109 patients and a validation cohort with 52 patients. Plasma samples were collected within 24 h of shock diagnosis and subjected to miRNA sequencing. High-throughput sequencing data from the discovery cohort were analysed to identify differentially expressed miRNAs. These findings were validated via qPCR in the validation cohort. RESULTS Thirty miRNAs were identified as significantly differentially expressed between septic and non-septic shock patients. Among these, six miRNAs-miR-100-5p, miR-484, miR-10a-5p, miR-148a-3p, miR-342-3p, and miR-451a-demonstrated strong diagnostic capabilities for septic shock. A combination of miR-100-5p, miR-148a-3p, and miR-451a achieved an area under the curve of 0.894, with qPCR validation in the validation cohort yielding an area under the curve of 0.960. CONCLUSIONS This study highlights extracellular vesicle-derived miRNAs as promising biomarkers for differentiating septic from non-septic shock. The identified three-miRNA signature has significant potential to enhance septic shock diagnosis, thereby aiding in timely and appropriate treatment for postsurgical patients.
Collapse
Affiliation(s)
- Adrián García-Concejo
- Biomedical Research Networking Centre in Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
| | - Belén Sánchez-Quirós
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Department of Anaesthesiology and Critical Care, University Clinical Hospital of Valladolid, Valladolid, Spain
| | - Esther Gómez-Sánchez
- Biomedical Research Networking Centre in Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Department of Anaesthesiology and Critical Care, University Clinical Hospital of Valladolid, Valladolid, Spain
- Department of Surgery, Faculty of Medicine, University of Valladolid, Valladolid, Spain
| | - Laura Sánchez-de Prada
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- National Influenza Centre, Valladolid, Spain
- Department of Microbiology, Río Hortega University Hospital, Valladolid, Spain
| | - Álvaro Tamayo-Velasco
- Biomedical Research Networking Centre in Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Department of Haematology and Hemotherapy, University Clinical Hospital of Valladolid, 47003, Valladolid, Spain
| | - María Sherezade Tovar-Doncel
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Anaesthesiology, Resuscitation and Pain Therapy Service, Unit of Critical Care, University Hospital of Toledo, Toledo, Spain
| | - Mario Lorenzo
- Department of Anaesthesiology and Critical Care, University Clinical Hospital of Valladolid, Valladolid, Spain
| | - Estefanía Gómez-Pesquera
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Department of Anaesthesiology and Critical Care, University Clinical Hospital of Valladolid, Valladolid, Spain
| | - Rodrigo Poves-Álvarez
- Department of Anaesthesiology and Critical Care, University Clinical Hospital of Valladolid, Valladolid, Spain
| | - David Bernardo
- Biomedical Research Networking Centre in Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
- Mucosal Immunology Laboratory, Institute of Biology and Molecular Genetics (IBGM), University of Valladolid - Spanish National Research Council, Valladolid, Spain
| | - Marta Martín-Fernández
- Biomedical Research Networking Centre in Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Department of Pharmacology, Faculty of Medicine, University of Valladolid, Valladolid, Spain
| | - Hugo Gonzalo-Benito
- Biomedical Research Networking Centre in Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Institute of Health Sciences of Castile and Leon (ICSCYL), Soria, Spain
| | - Paula Moreno-Portales
- Institute of Health Sciences of Castile and Leon (ICSCYL), Soria, Spain
- Research Unit, University Clinical Hospital of Valladolid, 47003, Valladolid, Spain
| | - Rosa Prieto-Utrera
- Institute of Health Sciences of Castile and Leon (ICSCYL), Soria, Spain
- Research Unit, University Clinical Hospital of Valladolid, 47003, Valladolid, Spain
| | - Miguel Bardají-Carrillo
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Department of Anaesthesiology and Critical Care, University Clinical Hospital of Valladolid, Valladolid, Spain
| | - Rocío López-Herrero
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Department of Anaesthesiology and Critical Care, University Clinical Hospital of Valladolid, Valladolid, Spain
- Department of Surgery, Faculty of Medicine, University of Valladolid, Valladolid, Spain
| | - María Fernández Arranz
- Department of Anaesthesiology and Critical Care, University Clinical Hospital of Valladolid, Valladolid, Spain
| | - Rosario Calaveras-Fernández
- Department of Anaesthesiology and Critical Care, University Clinical Hospital of Valladolid, Valladolid, Spain
| | - Fé Tomillo-Cebrián
- Department of Anaesthesiology and Critical Care, University Clinical Hospital of Valladolid, Valladolid, Spain
| | - Teresa Aydillo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - María Ángeles Jiménez-Sousa
- Biomedical Research Networking Centre in Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Unit of Viral Infection and Immunity, National Centre for Microbiology (CNM), Carlos III Health Institute, Majadahonda, Spain
| | - Amanda Fernández-Rodríguez
- Biomedical Research Networking Centre in Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Unit of Viral Infection and Immunity, National Centre for Microbiology (CNM), Carlos III Health Institute, Majadahonda, Spain
| | - Salvador Resino
- Biomedical Research Networking Centre in Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Unit of Viral Infection and Immunity, National Centre for Microbiology (CNM), Carlos III Health Institute, Majadahonda, Spain
| | - María Heredia-Rodríguez
- Biomedical Research Networking Centre in Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Department of Anaesthesiology and Critical Care, University Clinical Hospital of Salamanca, Salamanca, Spain
| | - Pedro Martínez-Paz
- Biomedical Research Networking Centre in Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Madrid, Spain.
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain.
- Centre for Experimental Medicine and Rheumatology, Queen Mary University of London, London, UK.
| | - Eduardo Tamayo
- Biomedical Research Networking Centre in Infectious Diseases (CIBERINFEC), Carlos III Health Institute, Madrid, Spain
- Group for Biomedical Research in Critical Care (Biocritic), Avenida Ramón y Cajal 7, 47005, Valladolid, Spain
- Department of Anaesthesiology and Critical Care, University Clinical Hospital of Valladolid, Valladolid, Spain
- Department of Surgery, Faculty of Medicine, University of Valladolid, Valladolid, Spain
| |
Collapse
|
4
|
Zhao B, Zhang Z, Guo X, Liu X, Lei M, Guo S, Yao Q, Zhang F, Peng T, Liu A, Jiang B, Zhu D. Mesenchymal stem cell-derived exosomes in renal ischemia-reperfusion injury: a new therapeutic strategy. Int Urol Nephrol 2025; 57:875-884. [PMID: 39520637 DOI: 10.1007/s11255-024-04258-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Renal ischemia-reperfusion injury (RIRI) is a serious kidney condition that causes significant damage due to lack of blood flow. This injury leads to oxidative stress and inflammation, which can cause acute tubular necrosis and kidney failure. Stem cell-derived exosomes, small vesicles released by stem cells, have shown promise in treating RIRI. Mesenchymal stem cells (MSCs) have been used to mitigate RIRI, and their exosomes have been found to play a crucial role in repairing damaged tissues. This review explores the key roles of exosomes from different sources of MSCs in RIRI, the potential of MSC-derived exosomes in treating this disease, and future research directions.
Collapse
Affiliation(s)
- Bo Zhao
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
- Xianning Central Hospital, First Affiliated Hospital of Hubei University of Science and Technology , 228 Jingui Road, Xian'an District, 437000, Xianning, Hubei Province, People's Republic of China
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, 437000, Xianning, Hubei Province, People's Republic of China
| | - Zhenwang Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Xiying Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Xiufen Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Min Lei
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Shuang Guo
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Qing Yao
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Feixue Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Tie Peng
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China
| | - Aimei Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China.
| | - Botao Jiang
- Xianning Central Hospital, First Affiliated Hospital of Hubei University of Science and Technology , 228 Jingui Road, Xian'an District, 437000, Xianning, Hubei Province, People's Republic of China.
| | - Dan Zhu
- Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, 437000, Xianning, Hubei Province, People's Republic of China.
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, 437000, Xianning, Hubei Province, People's Republic of China.
| |
Collapse
|
5
|
Rayat Pisheh H, Sani M. Mesenchymal stem cells derived exosomes: a new era in cardiac regeneration. Stem Cell Res Ther 2025; 16:16. [PMID: 39849585 PMCID: PMC11756228 DOI: 10.1186/s13287-024-04123-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/18/2024] [Indexed: 01/25/2025] Open
Abstract
Despite significant strides in medical treatments and surgical procedures for cardiovascular diseases, these conditions continue to be a major global health concern. The persistent need for innovative therapeutic approaches to mend damaged heart tissue highlights the complexity and urgency of this medical challenge. In recent years, stem cells have emerged as a promising tool for tissue regeneration, but challenges such as graft rejection and tumor formation have limited their clinical application. Exosomes, extracellular vesicles containing a diverse array of biomolecules, have garnered significant attention for their potential in regenerative medicine. The cardioprotective and reparative properties of mesenchymal stem cell-derived exosomes hold promise for the treatment of heart diseases. These exosomes can modulate various cellular processes, including angiogenesis, apoptosis, and inflammation, thereby enhancing cardiac function. Despite the growing interest, there remains a lack of comprehensive reviews synthesizing the molecular mechanisms, preclinical, and clinical evidence related to the specific role of MSC-derived exosomes in cardiac therapies. This review aims to fill that gap by exploring the potential of MSC-derived exosomes as a therapeutic strategy for cardiac diseases. This review explores the potential of mesenchymal stem cell-derived exosomes as a therapeutic strategy for cardiac diseases. We discuss the molecular mechanisms underlying their cardioprotective effects, summarize preclinical and clinical studies investigating their efficacy, and address the challenges and future perspectives of exosome-based therapies. The collective evidence suggests that MSC-derived exosomes hold promise as a novel and effective therapeutic approach for cardiac diseases.
Collapse
Affiliation(s)
- Hossein Rayat Pisheh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Sani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
- Shiraz Institute for Stem Cell & Regenerative Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
6
|
Li T, Zhao Y, Cao Z, Shen Y, Chen J, Huang X, Shao Z, Zeng Y, Chen Q, Yan X, Li X, Zhang Y, Hu B. Exosomes Derived from Apelin-Pretreated Mesenchymal Stem Cells Ameliorate Sepsis-Induced Myocardial Dysfunction by Alleviating Cardiomyocyte Pyroptosis via Delivery of miR-34a-5p. Int J Nanomedicine 2025; 20:687-703. [PMID: 39845770 PMCID: PMC11750946 DOI: 10.2147/ijn.s498770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/05/2025] [Indexed: 01/24/2025] Open
Abstract
Background Exosomes sourced from mesenchymal stem cells (MSC-EXOs) have become a promising therapeutic tool for sepsis-induced myocardial dysfunction (SMD). Our previous study demonstrated that Apelin pretreatment enhanced the therapeutic benefit of MSCs in myocardial infarction by improving their paracrine effects. This study aimed to determine whether EXOs sourced from Apelin-pretreated MSCs (Apelin-MSC-EXOs) would have potent cardioprotective effects against SMD and elucidate the underlying mechanisms. Methods MSC-EXOs and Apelin-MSC-EXOs were isolated and identified. Mice neonatal cardiomyocytes (NCMs) were treated with MSC-EXOs or Apelin-MSC-EXOs under lipopolysaccharide (LPS) condition in vitro. Cardiomyocyte pyroptosis was determined by TUNEL staining. RNA sequencing was used to identify differentially expressed functional miRNAs between MSC-EXOs and Apelin-MSC-EXOs. MSC-EXOs and Apelin-MSC-EXOs were transplanted into a mouse model of SMD induced by cecal ligation puncture (CLP) via the tail vein. Heart function was evaluated by echocardiography. Results Compared with MSC-EXOs, Apelin-MSC-EXO transplantation greatly enhanced cardiac function in SMD mice. Both MSC-EXOs and Apelin-MSC-EXOs suppressed cardiomyocyte pyroptosis in vivo and in vitro, with the latter exhibiting superior protective effects. miR-34a-5p effectively mediated Apelin-MSC-EXOs to exert their cardioprotective effects in SMD with high mobility group box-1 (HMGB1) as the potential target. Mechanistically, Apelin-MSC-EXOs delivered miR-34a-5p into injured cardiomyocytes, thereby ameliorating cardiomyocyte pyroptosis via regulation of the HMGB1/AMPK axis. These cardioprotective effects were partially abrogated by downregulation of miR-34a-5p in Apelin-MSC-EXOs. Conclusion Our study revealed miR-34a-5p as a key component of Apelin-MSC-EXOs that protected against SMD via mediation of the HMGB1/AMPK signaling pathway.
Collapse
Affiliation(s)
- Ting Li
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Yuechu Zhao
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Zhi Cao
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Ying Shen
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Jiaqi Chen
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Xinran Huang
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Zhuang Shao
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Yi Zeng
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Qi Chen
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Xiaofei Yan
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Xin Li
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Yuelin Zhang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| | - Bei Hu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Emergency Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
7
|
Yuan HJ, Xiang GH, Liu Y, Li Y, Liu WL, Wei JX, Xue YT, Hao H. Exploration and verification of the therapeutic mechanism of shenfu injection in sepsis-induced myocardial injury. PLoS One 2025; 20:e0317738. [PMID: 39823512 PMCID: PMC11741597 DOI: 10.1371/journal.pone.0317738] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Shenfu injection (SFI), derived from a traditional Chinese medicine (TCM) prescription, is an effective drug for the treatment of sepsis-induced myocardial injury (SIMI) with good efficacy, but its exact therapeutic mechanism remains unclear. METHODS SwissTargetPrediction and GeneCards database were used to obtain relevant targets for SFI and SIMI. STRING 11.5 and MCODE were used to analyse potential therapeutic targets for SFI. DAVID 6.8 database was used to perform enrichment analysis. In addition, the SIMI model was constructed by cecal ligation and puncture (CLP) on Sprague Dawley rats and the related protein expression levels were verified by AutoDock Vina 1.1.2 and experiment. RESULTS SFI has a total of 10 main active compounds and treats SIMI through 52 potential targets, among which LGALS3, STAT3, FGF1, and AKT1 were the core targets for treatment. Based on enrichment analysis, STAT3, FGF1, and AKT1 in the core targets were experimentally validated. The experimental results showed that SFI effectively ameliorated the inflammatory response and myocardial injury and inhibited apoptosis in SIMI. And SFI improved SIMI by decreasing caspase-9, STAT3 and phospho-AKT1 (p-AKT1) expression, and enhancing FGF1 expressions. CONCLUSIONS This study showed that SFI effectively reduced the expression of caspase-9, STAT3 and p-AKT1, enhanced the expression of FGF1, reduced the inflammatory response, inhibited apoptosis and attenuated cardiac injury to SIMI.
Collapse
Affiliation(s)
- Hua-jing Yuan
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, PR China
| | - Guo-han Xiang
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, PR China
| | - Yang Liu
- Intensive Care Unit, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, PR China
| | - Yan Li
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, PR China
| | - Wen-li Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, PR China
| | - Jiu-xiang Wei
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, PR China
| | - Yi-tao Xue
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, PR China
| | - Hao Hao
- Intensive Care Unit, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, PR China
- Postdoctoral Mobile Station, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, PR China
| |
Collapse
|
8
|
Kundu D, Shin SY, Chilian WM, Dong F. The Potential of Mesenchymal Stem Cell-Derived Exosomes in Cardiac Repair. Int J Mol Sci 2024; 25:13494. [PMID: 39769256 PMCID: PMC11727646 DOI: 10.3390/ijms252413494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 01/12/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide, and effectively repairing the heart following myocardial injuries remains a significant challenge. Research has increasingly shown that exosomes derived from mesenchymal stem cells (MSC-Exo) can ameliorate myocardial injuries and improve outcomes after such injuries. The therapeutic benefits of MSC-Exo are largely due to their capacity to deliver specific cargo, including microRNAs and proteins. MSC-Exo can modulate various signaling pathways and provide several beneficial effects, including cytoprotection, inflammation modulation, and angiogenesis promotion to help repair the damaged myocardium. In this review, we summarize the cardioprotective effects of MSC-Exo in myocardial injury, the underlying molecular mechanism involved in the process, and various approaches studied to enhance their efficacy based on recent findings.
Collapse
Affiliation(s)
| | | | | | - Feng Dong
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (D.K.); (S.Y.S.); (W.M.C.)
| |
Collapse
|
9
|
Nakazaki M, Yokoyama T, Lankford KL, Hirota R, Kocsis JD, Honmou O. Mesenchymal Stem Cells and Their Extracellular Vesicles: Therapeutic Mechanisms for Blood-Spinal Cord Barrier Repair Following Spinal Cord Injury. Int J Mol Sci 2024; 25:13460. [PMID: 39769223 PMCID: PMC11677717 DOI: 10.3390/ijms252413460] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Spinal cord injury (SCI) disrupts the blood-spinal cord barrier (BSCB) exacerbating damage by allowing harmful substances and immune cells to infiltrate spinal neural tissues from the vasculature. This leads to inflammation, oxidative stress, and impaired axonal regeneration. The BSCB, essential for maintaining spinal cord homeostasis, is structurally similar to the blood-brain barrier. Its restoration is a key therapeutic target for improving outcomes in SCI. Mesenchymal stromal/stem cells (MSCs) and their secreted extracellular vesicles (MSC-EVs) have gained attention for their regenerative, immunomodulatory, and anti-inflammatory properties in promoting BSCB repair. MSCs enhance BSCB integrity by improving endothelial-pericyte association, restoring tight junction proteins, and reducing inflammation. MSC-EVs, which deliver bioactive molecules, replicate many of MSCs' therapeutic effects, and offer a promising cell-free alternative. Preclinical studies have shown that both MSCs and MSC-EVs can reduce BSCB permeability, promote vascular stability, and support functional recovery. While MSC therapy is advancing in clinical trials, MSC-EV therapies require further optimization in terms of production, dosing, and delivery protocols. Despite these challenges, both therapeutic approaches represent significant potential for treating SCI by targeting BSCB repair and improving patient outcomes.
Collapse
Affiliation(s)
- Masahito Nakazaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Hokkaido, Japan
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Takahiro Yokoyama
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Hokkaido, Japan
| | - Karen L. Lankford
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Ryosuke Hirota
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Hokkaido, Japan
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Jeffery D. Kocsis
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060-8556, Hokkaido, Japan
- Department of Neurology, Yale University School of Medicine, New Haven, CT 06510, USA
- Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, CT 06516, USA
| |
Collapse
|
10
|
Miura Y, Fujii S, Ichinohe T. Cell-based and extracellular vesicle-based MSC therapies for acute radiation syndrome affecting organ systems. JOURNAL OF RADIATION RESEARCH 2024; 65:i80-i87. [PMID: 39679884 DOI: 10.1093/jrr/rrae009] [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: 12/05/2023] [Revised: 01/28/2024] [Indexed: 12/17/2024]
Abstract
Exposure to ionizing radiation can induce harmful biological effects on the human body, particularly in cases of high-dose γ-irradiation affecting the gastrointestinal tract, bone marrow, skin and lung. Such exposures lead to lethal outcomes as individuals experience a breakdown in their immune system's ability to defend against pathogens, predisposing them to sepsis-induced multiple organ failures. Mesenchymal stromal/stem cells (MSCs) possess diverse biological characteristics, including immunomodulation, anti-inflammation and tissue regeneration. Off-the-shelf culture-expanded human bone marrow- or adipose tissue-derived MSCs are clinically available to treat graft-versus-host disease following hematopoietic cell transplantation and perianal fistulas in Crohn's disease in Japan. While preclinical studies showcase encouraging outcomes in radiation-induced injuries, the effectiveness of MSC transplantation in addressing acute radiation syndrome affecting organs in irradiated individuals is limited. Recent studies have highlighted MSC-releasing extracellular vesicles as nanoparticle substances responsible for outlining the mechanism of action and have identified various components, including proteins and microRNA, that serve as functional molecules. MSC-releasing extracellular vesicle-based therapy emerges as a promising avenue, offering a potential solution to the challenges posed by radiation-induced injuries. However, further investigation is required, especially regarding whether MSC-releasing extracellular vesicles have regenerative effects on tissue-resident stem cells. These unresolved issues represent key aspects that need to be addressed to optimize the therapeutic potential of cell-based and extracellular vesicle-based MSC therapies for interventions in the context of radiation-induced injuries.
Collapse
Affiliation(s)
- Yasuo Miura
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
- Department of Transfusion Medicine and Cell Therapy, Fujita Health University School of Medicine, 1-93 Dengakugakubo, Kutsukakecho, Toyoake, Aichi 470-1192, Japan
| | - Sumie Fujii
- Department of Transfusion Medicine and Cell Therapy, Fujita Health University School of Medicine, 1-93 Dengakugakubo, Kutsukakecho, Toyoake, Aichi 470-1192, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| |
Collapse
|
11
|
Guo D, Yan J, Yang Z, Chen M, Zhong W, Yuan X, Yu S. The immune regulatory role of exosomal miRNAs and their clinical application potential in heart failure. Front Immunol 2024; 15:1476865. [PMID: 39687609 PMCID: PMC11647038 DOI: 10.3389/fimmu.2024.1476865] [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: 08/06/2024] [Accepted: 11/14/2024] [Indexed: 12/18/2024] Open
Abstract
Heart failure (HF) is a complex and debilitating condition characterized by the heart's inability to pump blood effectively, leading to significant morbidity and mortality. The abnormality of immune response is a key factor in the progression of HF, contributing to adverse cardiac remodeling and dysfunction. Exosomal microRNAs (miRNAs) play a pivotal role in regulating gene expression and cellular function, which are integral to the crosstalk between cardiac and immune cells, influencing immune cell functions, such as macrophage polarization, T cell activity, and cytokine production, thereby modulating various pathological processes of HF, such as inflammation, fibrosis, and cardiac dysfunction. This review emphasizes the immune-regulatory role of exosomal miRNAs in HF and highlights their clinical potential as diagnostic biomarkers and therapeutic agents.
Collapse
Affiliation(s)
- Dandan Guo
- Department of Cardiology, Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
- School of Graduate Studies, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Junchen Yan
- School of Graduate Studies, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhenyu Yang
- School of Graduate Studies, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Mengzhu Chen
- School of Graduate Studies, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Weibo Zhong
- School of Graduate Studies, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xingxing Yuan
- Department of Gastroenterology, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Siming Yu
- School of Graduate Studies, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Nephrology II, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
12
|
Lin L, Liu H, Zhang D, Du L, Zhang H. Nanolevel Immunomodulators in Sepsis: Novel Roles, Current Perspectives, and Future Directions. Int J Nanomedicine 2024; 19:12529-12556. [PMID: 39606559 PMCID: PMC11600945 DOI: 10.2147/ijn.s496456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Sepsis represents a profound challenge in critical care, characterized by a severe systemic inflammatory response which can lead to multi-organ failure and death. The intricate pathophysiology of sepsis involves an overwhelming immune reaction that disrupts normal host defense mechanisms, necessitating innovative approaches to modulation. Nanoscale immunomodulators, with their precision targeting and controlled release capabilities, have emerged as a potent solution to recalibrate immune responses in sepsis. This review explores the recent advancements in nanotechnology for sepsis management, emphasizing the integration of nanoparticulate systems to modulate immune function and inflammatory pathways. Discussions detail the development of the immune system, the distinct inflammatory responses triggered by sepsis, and the scientific principles underpinning nanoscale immunomodulation, including specific targeting mechanisms and delivery systems. The review highlights nanoformulation designs aimed at enhancing bioavailability, stability, and therapeutic efficacy, which shows promise in clinical settings by modulating key inflammatory pathways. Ultimately, this review synthesizes the current state of knowledge and projects future directions for research, underscoring the transformative potential of nanolevel immunomodulators for sepsis treatment through innovative technologies and therapeutic strategies.
Collapse
Affiliation(s)
- Liangkang Lin
- Department of Pediatrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Hanyou Liu
- Department of Pediatrics, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, People’s Republic of China
| | - Dingshan Zhang
- Department of Intensive Care Unit, Public Health Clinical Center of Chengdu, Chengdu, People’s Republic of China
| | - Lijia Du
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| | - Haiyang Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People’s Republic of China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, People’s Republic of China
- NHC Key Laboratory of Chronobiology, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
13
|
Wang D, Xu L, Liu Y, Wang C, Qi S, Li Z, Bai X, Liao Y, Wang Y. Role of mesenchymal stem cells in sepsis and their therapeutic potential in sepsis‑associated myopathy (Review). Int J Mol Med 2024; 54:92. [PMID: 39219272 PMCID: PMC11374154 DOI: 10.3892/ijmm.2024.5416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Sepsis‑induced myopathy (SIM) is one of the leading causes of death in critically ill patients. SIM mainly involves the respiratory and skeletal muscles of patients, resulting in an increased risk of lung infection, aggravated respiratory failure, and prolonged mechanical ventilation and hospital stay. SIM is also an independent risk factor associated with increased mortality in critically ill patients. At present, no effective treatment for SIM has yet been established. However, mesenchymal stem cells (MSCs) have emerged as a promising therapeutic approach and have been utilized in the treatment of various clinical conditions. A significant body of basic and clinical research supports the efficacy of MSCs in managing sepsis and muscle‑related diseases. This literature review aims to explore the relationship between MSCs and sepsis, as well as their impact on skeletal muscle‑associated diseases. Additionally, the present review discusses the potential mechanisms and therapeutic benefits of MSCs in the context of SIM.
Collapse
Affiliation(s)
- Dongfang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Ligang Xu
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yukun Liu
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Chuntao Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Siyuan Qi
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yiliu Liao
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yuchang Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
14
|
Gu Y, Mu Z, Chen Y, Wu C, Shi J, Bai N. Therapeutic potential of ADSCs in diabetic wounds: a proteomics-based approach. Front Cell Dev Biol 2024; 12:1468220. [PMID: 39345337 PMCID: PMC11427884 DOI: 10.3389/fcell.2024.1468220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 08/26/2024] [Indexed: 10/01/2024] Open
Abstract
Background Diabetes mellitus (DM), a chronic metabolic disease characterized by elevated blood sugar, leads to delayed or non-healing wounds, increasing amputation risks, and placing a significant burden on patients and society. While extensive research has been conducted on adipose-derived stem cells (ADSCs) for promoting wound healing, there is a scarcity of studies focusing on diabetic wounds, particularly those employing proteomics and bioinformatics approaches. Objective This study aimed to investigate the mechanisms by which ADSCs promote diabetic wound healing using proteomics and bioinformatics techniques. Methods Healthy rat fat tissue was used to isolate ADSCs. A T2DM rat model with back wounds was established. The experimental group received ADSC injections around the wound, while the control group received PBS injections. Wound healing rates were documented and photographed on days 0, 3, 7, 10, and 14. On day 7, wound tissues were excised for HE and Masson's staining. Additionally, on day 7, tissues were analyzed for protein quantification using 4D-DIA, with subsequent GO and KEGG analyses for differentially expressed proteins (DEPs) and protein-protein interaction (PPI) network analysis using STRING database (String v11.5). Finally, Western blot experiments were performed on day 7 wounds to verify target proteins. Results and Conclusions In all measured days postoperatively, the wound healing rate was significantly higher in the ADSC group than in the PBS group (day 7: p < 0.001, day 10: p = 0.001, day 14: p < 0.01), except on day 3 (p > 0.05). Proteomic analysis identified 474 differentially expressed proteins, with 224 key proteins after PPI analysis (78 upregulated and 146 downregulated in the ADSC group). The main cellular locations of these proteins were "cellular anatomical entity" and "protein-containing complex", while the biological processes were "cellular processes" and "biological regulation". The primary molecular functions were "binding" and "catalytic activity", with GO enrichment focused on "Wnt-protein binding", "neural development", and "collagen-containing extracellular matrix". Further analysis of PPI network nodes using LASSO regression identified Thy1 and Wls proteins, significantly upregulated in the ADSC group, as potentially crucial targets for ADSC application in diabetic wound treatment.
Collapse
Affiliation(s)
- Yuan Gu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Zelan Mu
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China
| | - Yuanzheng Chen
- Department of Burns and Plastic Surgery, Emergency General Hospital, Beijing, China
| | - Can Wu
- Medical Cosmetic Plastic Surgery, Linyi People′s Hospital, Linyi, China
| | - Jie Shi
- Plastic and Cosmetic Surgery, People′s Hospital of Liaoning Province, Shenyang, China
| | - Nan Bai
- Medical Cosmetic Plastic Surgery, Linyi People′s Hospital, Linyi, China
| |
Collapse
|
15
|
Hao Y, Liu R, Wang H, Rui T, Guo J. Research Progress on Mechanisms and Treatment of Sepsis-Induced Myocardial Dysfunction. Int J Gen Med 2024; 17:3387-3393. [PMID: 39130486 PMCID: PMC11313578 DOI: 10.2147/ijgm.s472846] [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: 04/08/2024] [Accepted: 07/25/2024] [Indexed: 08/13/2024] Open
Abstract
Sepsis is a syndrome of organ dysfunction caused by a dysregulated immune response to infection, with high morbidity and mortality. At present, there have been many advances in the study of its pathogenesis, especially the cardiac dysfunction caused by sepsis, namely sepsis-induced myocardial dysfunction, SIMD, which has received widespread attention. The mechanisms of SIMD mainly include excessive release of inflammatory mediators, impaired mitochondrial function, autophagy, apoptosis and myocardial dysfunction. This article reviews the pathogenesis of SIMD and elaborates on the progress in its treatment, aiming to improve the prognosis of patients with SIMD and sepsis.
Collapse
Affiliation(s)
- Yujie Hao
- Division of Cardiology, Department of Medicine, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Runmin Liu
- Division of Cardiology, Department of Medicine, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Hao Wang
- Division of Cardiology, Department of Medicine, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Tao Rui
- Division of Cardiology, Department of Medicine, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Junfang Guo
- Division of Cardiology, Department of Medicine, the Affiliated People’s Hospital of Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| |
Collapse
|
16
|
Seow KS, Ling APK. Mesenchymal stem cells as future treatment for cardiovascular regeneration and its challenges. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:73. [PMID: 39118948 PMCID: PMC11304428 DOI: 10.21037/atm-23-1936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 12/04/2023] [Indexed: 08/10/2024]
Abstract
Cardiovascular diseases (CVDs), particularly stroke and myocardial infarction (MI) contributed to the leading cause of death annually among the chronic diseases globally. Despite the advancement of technology, the current available treatments mainly served as palliative care but not treating the diseases. However, the discovery of mesenchymal stem cells (MSCs) had gained a consideration to serve as promising strategy in treating CVDs. Recent evidence also showed that MSCs are the strong candidate to be used as stem cell therapy involving cardiovascular regeneration due to its cardiomyogenesis, anti-inflammatory and immunomodulatory properties, antifibrotic effects and neovascularization capacity. Besides, MSCs could be used for cellular cardiomyoplasty with its transdifferentiation of MSCs into cardiomyocytes, paracrine effects, microvesicles and exosomes as well as mitochondrial transfer. The safety and efficacy of utilizing MSCs have been described in well-established preclinical and clinical studies in which the accomplishment of MSCs transplantation resulted in further improvement of the cardiac function. Tissue engineering could enhance the desired properties and therapeutic effects of MSCs in cardiovascular regeneration by genome-editing, facilitating the cell delivery and retention, biomaterials-based scaffold, and three-dimensional (3D)-bioprinting. However, there are still obstacles in the use of MSCs due to the complexity and versatility of MSCs, low retention rate, route of administration and the ethical and safety issues of the use of MSCs. The aim of this review is to highlight the details of therapeutic properties of MSCs in treating CVDs, strategies to facilitate the therapeutic effects of MSCs through tissue engineering and the challenges faced using MSCs. A comprehensive review has been done through PubMed and National Center for Biotechnology Information (NCBI) from the year of 2010 to 2021 based on some specific key terms such as 'mesenchymal stem cells in cardiovascular disease', 'mesenchymal stem cells in cardiac regeneration', 'mesenchymal stem cells facilitate cardiac repairs', 'tissue engineering of MSCs' to include relevant literature in this review.
Collapse
Affiliation(s)
- Ke Sin Seow
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| | - Anna Pick Kiong Ling
- Division of Applied Biomedical Sciences and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
17
|
Aghayan AH, Mirazimi Y, Fateh K, Keshtkar A, Rafiee M, Atashi A. Therapeutic Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in sepsis: a Systematic Review and Meta-Analysis of Preclinical Studies. Stem Cell Rev Rep 2024; 20:1480-1500. [PMID: 38814410 DOI: 10.1007/s12015-024-10741-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND Sepsis is a life-threatening disorder with no definitive cure. Preclinical studies suggest that extracellular vesicles derived from mesenchymal stromal cells (EV-MSCs) can mitigate inflammatory conditions, potentially leading to increased survival and reduced organ dysfunction during sepsis. Our aim to conduct this systematic review and meta-analysis is assessing the EV-MSCs therapeutic efficacy in sepsis. METHODS PubMed, Embase, Scopus, WOS and ProQuest databases and also Google Scholar search engine were searched for published articles. We used hazard ratio (HR) and standardized mean difference (SMD) as effect sizes to evaluate the therapeutic effect of EV-MSCs on survival rate and determine their effect on reducing organ dysfunction, respectively. Finally, we employed GRADE tool for preclinical animal studies to evaluate certainty of the evidence. RESULTS 30 studies met the inclusion criteria for our article. Our meta-analysis results demonstrate that animals treated with MSC-EVs have better survival rate than untreated animals (HR = 0.33; 95% CI: 0.27-0.41). Our meta-analysis suggests that EV-MSCs can reduce organ dysfunctions in sepsis, such as the lung, kidney, and liver. Additionally, EV-MSCs decrease pro-inflammatory mediators like TNF-α, IL-1β, and IL-6. CONCLUSION Our results indicate that EV-MSCs can be as promising therapy for sepsis management in animal models and leading to increased survival rate and reduced organ dysfunction. Furthermore, our study introduces a novel tool for risk of bias assessment and provides recommendations based on various analysis. Future studies with aiming to guide clinical translation can utilize the results of this article to establish stronger evidence for EV-MSC effectiveness.
Collapse
Affiliation(s)
- Amir Hossein Aghayan
- Student Research Committee, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Yasin Mirazimi
- Student Research Committee, Department of Medical Laboratory Sciences, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kosar Fateh
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbasali Keshtkar
- Department of Health Sciences Education Development, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Rafiee
- Department of Medical Laboratory Sciences, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Amir Atashi
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran.
| |
Collapse
|
18
|
Xiao Y, Yuan Y, Hu D, Wang H. Exosome-Derived microRNA: Potential Target for Diagnosis and Treatment of Sepsis. J Immunol Res 2024; 2024:4481452. [PMID: 39104595 PMCID: PMC11300089 DOI: 10.1155/2024/4481452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/23/2024] [Accepted: 07/06/2024] [Indexed: 08/07/2024] Open
Abstract
Exosome-derived microRNAs (miRNAs) are emerging as pivotal players in the pathophysiology of sepsis, representing a new frontier in both the diagnosis and treatment of this complex condition. Sepsis, a severe systemic response to infection, involves intricate immune and nonimmune mechanisms, where exosome-mediated communication can significantly influence disease progression and outcomes. During the progress of sepsis, the miRNA profile of exosomes undergoes notable alterations, is reflecting, and may affect the progression of the disease. This review comprehensively explores the biology of exosome-derived miRNAs, which originate from both immune cells (such as macrophages and dendritic cells) and nonimmune cells (such as endothelial and epithelial cells) and play a dynamic role in modulating pathways that affect the course of sepsis, including those related to inflammation, immune response, cell survival, and apoptosis. Taking into account these dynamic changes, we further discuss the potential of exosome-derived miRNAs as biomarkers for the early detection and prognosis of sepsis and advantages over traditional biomarkers due to their stability and specificity. Furthermore, this review evaluates exosome-based therapeutic miRNA delivery systems in sepsis, which may pave the way for targeted modulation of the septic response and personalized treatment options.
Collapse
Affiliation(s)
- Yujie Xiao
- Department of Burns and Cutaneous SurgeryXijing HospitalFourth Military Medical University, 127 West Chang-le Road, Xi'an 710032, Shaanxi, China
| | - Yixuan Yuan
- Department of Burns and Cutaneous SurgeryXijing HospitalFourth Military Medical University, 127 West Chang-le Road, Xi'an 710032, Shaanxi, China
| | - Dahai Hu
- Department of Burns and Cutaneous SurgeryXijing HospitalFourth Military Medical University, 127 West Chang-le Road, Xi'an 710032, Shaanxi, China
| | - Hongtao Wang
- Department of Burns and Cutaneous SurgeryXijing HospitalFourth Military Medical University, 127 West Chang-le Road, Xi'an 710032, Shaanxi, China
| |
Collapse
|
19
|
Li W, Si Y, Wang Y, Chen J, Huo X, Xu P, Jiang B, Li Z, Shang K, Luo Q, Xiong Y. hUCMSC-derived exosomes protect against GVHD-induced endoplasmic reticulum stress in CD4 + T cells by targeting the miR-16-5p/ATF6/CHOP axis. Int Immunopharmacol 2024; 135:112315. [PMID: 38805908 DOI: 10.1016/j.intimp.2024.112315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 05/18/2024] [Accepted: 05/19/2024] [Indexed: 05/30/2024]
Abstract
Exosomes generated from mesenchymal stem cells (MSCs) are thought to be a unique therapeutic strategy for several autoimmune deficiency illnesses. The purpose of this study was to elucidate the protective effects of human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-Exo) on CD4+ T cells dysfunction during graft-versus-host disease (GVHD) and to identify the underlying processes involved. Here, we showed that hUCMSC-Exo treatment can effectively attenuate GVHD injury by alleviating redox metabolism disorders and inflammatory cytokine bursts in CD4+ T cells. Furthermore, hUCMSC-Exo ameliorate ER stress and ATF6/CHOP signaling-mediated apoptosis in CD4+ T cells and promote the development of CD4+IL-10+ T cells during GVHD. Moreover, downregulating miR-16-5p in hUCMSC-Exo impaired their ability to prevent CD4+ T cells apoptosis and weakened their ability to promote the differentiation of CD4+IL-10+ T cells. Collectively, the obtained data suggested that hUCMSC-Exo suppress ATF6/CHOP signaling-mediated ER stress and apoptosis in CD4+ T cells, enhance the differentiation of CD4+IL-10+ T cells, and reverse the imbalance of immune homeostasis in the GVHD process by transferring miR-16-5p. Our study provided further evidence that GVHD patients can benefit from hUCMSC-Exo-mediated therapy.
Collapse
Affiliation(s)
- Weihan Li
- Xu Rongxiang Regenerative Medicine Research Center, Binzhou Medical University, Yantai, PR China; Shanghai Mebo Life Science & Technology Co., Shanghai, PR China
| | - Yaru Si
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Yueming Wang
- Xu Rongxiang Regenerative Medicine Research Center, Binzhou Medical University, Yantai, PR China
| | - Juntong Chen
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Xingyu Huo
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Pengzhan Xu
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Bingzhen Jiang
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Zile Li
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Kangdi Shang
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Qianqian Luo
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China.
| | - Yanlian Xiong
- Xu Rongxiang Regenerative Medicine Research Center, Binzhou Medical University, Yantai, PR China; Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China.
| |
Collapse
|
20
|
Mao L, Liu S, Chen Y, Huang H, Ding F, Deng L. Engineered exosomes: a potential therapeutic strategy for septic cardiomyopathy. Front Cardiovasc Med 2024; 11:1399738. [PMID: 39006168 PMCID: PMC11239395 DOI: 10.3389/fcvm.2024.1399738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/14/2024] [Indexed: 07/16/2024] Open
Abstract
Septic cardiomyopathy, a life-threatening complication of sepsis, can cause acute heart failure and carry a high mortality risk. Current treatments have limitations. Fortunately, engineered exosomes, created through bioengineering technology, may represent a potential new treatment method. These exosomes can both diagnose and treat septic cardiomyopathy, playing a crucial role in its development and progression. This article examines the strategies for using engineered exosomes to protect cardiac function and treat septic cardiomyopathy. It covers three innovative aspects: exosome surface modification technology, the use of exosomes as a multifunctional drug delivery platform, and plant exosome-like nanoparticle carriers. The article highlights the ability of exosomes to deliver small molecules, proteins, and drugs, summarizing several RNA molecules, proteins, and drugs beneficial for treating septic cardiomyopathy. Although engineered exosomes are a promising biotherapeutic carrier, they face challenges in clinical application, such as understanding the interaction mechanism with host cells, distribution within the body, metabolism, and long-term safety. Further research is essential, but engineered exosomes hold promise as an effective treatment for septic cardiomyopathy.
Collapse
Affiliation(s)
- Lixia Mao
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Songtao Liu
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yongxia Chen
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huiyi Huang
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fenghua Ding
- Outpatient Appointment Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Liehua Deng
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
21
|
Zhu Y, Li Y, Cao Z, Xue J, Wang X, Hu T, Han B, Guo Y. Mechanically strained osteocyte-derived exosomes contained miR-3110-5p and miR-3058-3p and promoted osteoblastic differentiation. Biomed Eng Online 2024; 23:44. [PMID: 38705993 PMCID: PMC11070085 DOI: 10.1186/s12938-024-01237-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/11/2024] [Indexed: 05/07/2024] Open
Abstract
BACKGROUND Osteocytes are critical mechanosensory cells in bone, and mechanically stimulated osteocytes produce exosomes that can induce osteogenesis. MicroRNAs (miRNAs) are important constituents of exosomes, and some miRNAs in osteocytes regulate osteogenic differentiation; previous studies have indicated that some differentially expressed miRNAs in mechanically strained osteocytes likely influence osteoblastic differentiation. Therefore, screening and selection of miRNAs that regulate osteogenic differentiation in exosomes of mechanically stimulated osteocytes are important. RESULTS A mechanical tensile strain of 2500 με at 0.5 Hz 1 h per day for 3 days, elevated prostaglandin E2 (PGE2) and insulin-like growth factor-1 (IGF-1) levels and nitric oxide synthase (NOS) activity of MLO-Y4 osteocytes, and promoted osteogenic differentiation of MC3T3-E1 osteoblasts. Fourteen miRNAs differentially expressed only in MLO-Y4 osteocytes which were stimulated with mechanical tensile strain, were screened, and the miRNAs related to osteogenesis were identified. Four differentially expressed miRNAs (miR-1930-3p, miR-3110-5p, miR-3090-3p, and miR-3058-3p) were found only in mechanically strained osteocytes, and the four miRNAs, eight targeted mRNAs which were differentially expressed only in mechanically strained osteoblasts, were also identified. In addition, the mechanically strained osteocyte-derived exosomes promoted the osteoblastic differentiation of MC3T3-E1 cells in vitro, the exosomes were internalized by osteoblasts, and the up-regulated miR-3110-5p and miR-3058-3p in mechanically strained osteocytes, were both increased in the exosomes, which was verified via reverse transcription quantitative polymerase chain reaction (RT-qPCR). CONCLUSIONS In osteocytes, a mechanical tensile strain of 2500 με at 0.5 Hz induced the fourteen differentially expressed miRNAs which probably were in exosomes of osteocytes and involved in osteogenesis. The mechanically strained osteocyte-derived exosomes which contained increased miR-3110-5p and miR-3058-3p (two of the 14 miRNAs), promoted osteoblastic differentiation.
Collapse
Affiliation(s)
- Yingwen Zhu
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
| | - Yanan Li
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
| | - Zhen Cao
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
| | - Jindong Xue
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
| | - Xiaoyan Wang
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
| | - Tingting Hu
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China
| | - Biao Han
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China.
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China.
| | - Yong Guo
- Department of Biomedical Engineering, School of Intelligent Medicine and Biotechnology, Guilin Medical University, No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China.
- Education Department of Guangxi Zhuang Autonomous Region, Key Laboratory of Biochemistry and Molecular Biology (Guilin Medical University), No. 1 Zhiyuan Road, Lingui District, Guilin, 541199, Guangxi, People's Republic of China.
| |
Collapse
|
22
|
Moghaddam MM, Behzadi E, Sedighian H, Goleij Z, Kachuei R, Heiat M, Fooladi AAI. Regulation of immune responses to infection through interaction between stem cell-derived exosomes and toll-like receptors mediated by microRNA cargoes. Front Cell Infect Microbiol 2024; 14:1384420. [PMID: 38756232 PMCID: PMC11096519 DOI: 10.3389/fcimb.2024.1384420] [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: 02/09/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024] Open
Abstract
Infectious diseases are among the factors that account for a significant proportion of disease-related deaths worldwide. The primary treatment approach to combat microbial infections is the use of antibiotics. However, the widespread use of these drugs over the past two decades has led to the emergence of resistant microbial species, making the control of microbial infections a serious challenge. One of the most important solutions in the field of combating infectious diseases is the regulation of the host's defense system. Toll-like receptors (TLRs) play a crucial role in the first primary defense against pathogens by identifying harmful endogenous molecules released from dying cells and damaged tissues as well as invading microbial agents. Therefore, they play an important role in communicating and regulating innate and adaptive immunity. Of course, excessive activation of TLRs can lead to disruption of immune homeostasis and increase the risk of inflammatory reactions. Targeting TLR signaling pathways has emerged as a new therapeutic approach for infectious diseases based on host-directed therapy (HDT). In recent years, stem cell-derived exosomes have received significant attention as factors regulating the immune system. The regulation effects of exosomes on the immune system are based on the HDT strategy, which is due to their cargoes. In general, the mechanism of action of stem cell-derived exosomes in HDT is by regulating and modulating immunity, promoting tissue regeneration, and reducing host toxicity. One of their most important cargoes is microRNAs, which have been shown to play a significant role in regulating immunity through TLRs. This review investigates the therapeutic properties of stem cell-derived exosomes in combating infections through the interaction between exosomal microRNAs and Toll-like receptors.
Collapse
Affiliation(s)
- Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elham Behzadi
- The Academy of Medical Sciences of I.R. Iran, Tehran, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zoleikha Goleij
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Heiat
- Baqiyatallah Research Center for Gastroenterology and Liver Diseases (BRCGL), Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Peng C, Yan J, Jiang Y, Wu L, Li M, Fan X. Exploring Cutting-Edge Approaches to Potentiate Mesenchymal Stem Cell and Exosome Therapy for Myocardial Infarction. J Cardiovasc Transl Res 2024; 17:356-375. [PMID: 37819538 DOI: 10.1007/s12265-023-10438-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/12/2023] [Indexed: 10/13/2023]
Abstract
Cardiovascular diseases (CVDs) continue to be a significant global health concern. Many studies have reported promising outcomes from using MSCs and their secreted exosomes in managing various cardiovascular-related diseases like myocardial infarction (MI). MSCs and exosomes have demonstrated considerable potential in promoting regeneration and neovascularization, as well as exerting beneficial effects against apoptosis, remodeling, and inflammation in cases of myocardial infarction. Nonetheless, ensuring the durability and effectiveness of MSCs and exosomes following in vivo transplantation remains a significant concern. Recently, novel methods have emerged to improve their effectiveness and robustness, such as employing preconditioning statuses, modifying MSC and their exosomes, targeted drug delivery with exosomes, biomaterials, and combination therapy. Herein, we summarize the novel approaches that intensify the therapeutic application of MSC and their derived exosomes in treating MI.
Collapse
Affiliation(s)
- Chendong Peng
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jie Yan
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yu'ang Jiang
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lin Wu
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Cardiology, Peking University First Hospital, Beijing, 100000, China
| | - Miaoling Li
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Key Laboratory of Sichuan Province, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Xinrong Fan
- Department of Cardiology, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
24
|
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
|
25
|
Shi Y, Ji S, Xu Y, Ji J, Yang X, Ye B, Lou J, Tao T. Global trends in research on endothelial cells and sepsis between 2002 and 2022: A systematic bibliometric analysis. Heliyon 2024; 10:e23599. [PMID: 38173483 PMCID: PMC10761786 DOI: 10.1016/j.heliyon.2023.e23599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
Sepsis is a systemic syndrome involving physiological, pathological, and biochemical abnormalities precipitated by infection and is a major global public health problem. Endothelial cells (ECs) dysfunction is a major contributor to sepsis-induced multiple organ failure. This bibliometric analysis aimed to identify and characterize the status, evolution of the field, and new research trends of ECs and sepsis over the past 20 years. For this analysis, the Web of Science Core Collection database was searched to identify relevant publications on ECs in sepsis published between January 1, 2002, and December 31, 2022. Microsoft Excel 2021, VOSviewer software, CiteSpace software, and the online analysis platform of literature metrology (http://bibliometric.com) were used to visualize the trends of publications' countries/regions, institutions, authors, journals, and keywords. In total, 4200 articles were identified and screened, primarily originating from 86 countries/regions and 3489 institutions. The USA was the leading contributor to this research field, providing 1501 articles (35.74 %). Harvard University's scientists were the most prolific, with 129 articles. Overall, 21,944 authors were identified, among whom Bae Jong Sup was the most prolific, contributing 129 publications. Additionally, Levi Marcel was the most frequently co-cited author, appearing 538 times. The journals that published the most articles were SHOCK, CRITICAL CARE MEDICINE, and PLOS ONE, accounting for 10.79 % of the total. The current emerging hotspots are concentrated on "endothelial glycocalyx," "NLRP3 inflammasome," "extracellular vesicle," "biomarkers," and "COVID-19," among others. In conclusion, this study provides a comprehensive overview of the scientific productivity and emerging research trends in the field of ECs in sepsis. The evidence supporting the significant role of ECs in both physiological and pathological responses to sepsis is continuously growing. More in-depth studies of the molecular mechanisms underlying sepsis-induced endothelial dysfunction and EC-targeted therapies are warranted in the future.
Collapse
Affiliation(s)
- Yue Shi
- Department of Anesthesiology, Air Force Medical Center, Beijing, China
- Graduate of China Medical University, Shenyang, China
| | - Shunpan Ji
- Department of Anesthesiology, Air Force Medical Center, Beijing, China
- Graduate of China Medical University, Shenyang, China
| | - Yuhai Xu
- Department of Anesthesiology, Air Force Medical Center, Beijing, China
| | - Jun Ji
- Department of Anesthesiology, Air Force Medical Center, Beijing, China
| | - Xiaoming Yang
- Department of Anesthesiology, Air Force Medical Center, Beijing, China
| | - Bo Ye
- Department of Anesthesiology, Air Force Medical Center, Beijing, China
- Graduate of China Medical University, Shenyang, China
| | - Jingsheng Lou
- Department of Anesthesiology, The General Hospital of the People's Liberation Army, Beijing, China
| | - Tianzhu Tao
- Department of Anesthesiology, Air Force Medical Center, Beijing, China
- Graduate of China Medical University, Shenyang, China
| |
Collapse
|
26
|
He YC, Yuan GD, Li N, Ren MF, Qian-Zhang, Deng KN, Wang LC, Xiao WL, Ma N, Stamm C, Felthaus O, Prantl L, Nie J, Wang G. Recent advances in mesenchymal stem cell therapy for myocardial infarction. Clin Hemorheol Microcirc 2024; 87:383-398. [PMID: 38578884 DOI: 10.3233/ch-249101] [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: 04/07/2024]
Abstract
Myocardial infarction refers to the ischemic necrosis of myocardium, characterized by a sharp reduction or interruption of blood flow in the coronary arteries due to the coronary artery occlusion, resulting in severe and prolonged ischemia in the corresponding myocardium and ultimately leading to ischemic necrosis of the myocardium. Given its high risk, it is considered as one of the most serious health threats today. In current clinical practice, multiple approaches have been explored to diminish myocardial oxygen consumption and alleviate symptoms, but notable success remains elusive. Accumulated clinical evidence has showed that the implantation of mesenchymal stem cell for treating myocardial infarction is both effective and safe. Nevertheless, there persists controversy and variability regarding the standardizing MSC transplantation protocols, optimizing dosage, and determining the most effective routes of administration. Addressing these remaining issues will pave the way of integration of MSCs as a feasible mainstream cardiac treatment.
Collapse
Affiliation(s)
- Yu-Chuan He
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Guo-Dong Yuan
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Nan Li
- Shijiazhuang Obstetrics and Gynecology Hospital, Shijiazhuang, Hebei, China
| | - Mei-Fang Ren
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Qian-Zhang
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Kai-Ning Deng
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Le-Chuan Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Wei-Ling Xiao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Nan Ma
- Helmholtz-Zentrum Hereon, Institute of Active Polymers, Teltow, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Oliver Felthaus
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Jia Nie
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Gang Wang
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| |
Collapse
|
27
|
Alikiaii B, Bagherniya M, Askari G, Rajendram R, Sahebkar A. MicroRNA Profiles in Critically Ill Patients. Curr Med Chem 2024; 31:6801-6825. [PMID: 37496239 DOI: 10.2174/0929867331666230726095222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 05/19/2023] [Accepted: 06/01/2023] [Indexed: 07/28/2023]
Abstract
The use of biomarkers to expedite diagnosis, prognostication, and treatment could significantly improve patient outcomes. The early diagnosis and treatment of critical illnesses can greatly reduce mortality and morbidity. Therefore, there is great interest in the discovery of biomarkers for critical illnesses. Micro-ribonucleic acids (miRNAs) are a highly conserved group of non-coding RNA molecules. They regulate the expression of genes involved in several developmental, physiological, and pathological processes. The characteristics of miRNAs suggest that they could be versatile biomarkers. Assay panels to measure the expression of several miRNAs could facilitate clinical decision-- making for a range of diseases. We have, in this paper, reviewed the current understanding of the role of miRNAs as biomarkers in critically ill patients.
Collapse
Affiliation(s)
- Babak Alikiaii
- Anesthesia and Critical Care Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Bagherniya
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gholamreza Askari
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Rajkumar Rajendram
- Department of Medicine, King Abdulaziz Medical City, King Abdulaziz International Medical Research Center, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University of Health Sciences, Riyadh, Saudi Arabia
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
28
|
Zhang F, Zhang L, Yu H. Potential Druggability of Mesenchymal Stem/Stromal Cell-derived Exosomes. Curr Stem Cell Res Ther 2024; 19:1195-1209. [PMID: 38523514 DOI: 10.2174/011574888x311270240319084835] [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: 02/19/2024] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 03/26/2024]
Abstract
Exosomes secreted by mesenchymal stem/stromal cells (MSC-Exos) are advantageous candidate sources for novel acellular therapy. Despite the current standards of good manufacturing practice (GMP), the deficiency of suitable quality-control methods and the difficulties in large-scale preparation largely restrict the development of therapeutic products and their clinical applications worldwide. Herein, we mainly focus on three dominating issues commonly encountered in exosomal GMP, including issues upstream of the cell culture process, downstream of the purification process, exosomes quality control, and the drug properties of exosomes and their druggability from a corporate perspective. Collectively, in this review article, we put forward the issues of preparing clinical exosome drugs for the treatment of diverse diseases and provide new references for the clinical application of GMP-grade MSC-Exos.
Collapse
Affiliation(s)
- Fan Zhang
- Faculty of Life Sciences and Medicine, Kunming University of Science and Technology, Kunming, 650500, China
| | - Leisheng Zhang
- Science and Technology Innovation Center, The Fourth People's Hospital of Jinan (The Third Affiliated Hospital of Shandong First Medical University), Jinan, 250031, China
- National Health Commission (NHC) Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Hao Yu
- The Postdoctoral Research Station, School of Medicine, Nankai University, Tianjin, 300071, China
| |
Collapse
|
29
|
Tsiapalis D, Floudas A, Tertel T, Boerger V, Giebel B, Veale DJ, Fearon U, O’Driscoll L. Therapeutic Effects of Mesenchymal/Stromal Stem Cells and Their Derived Extracellular Vesicles in Rheumatoid Arthritis. Stem Cells Transl Med 2023; 12:849-862. [PMID: 37934808 PMCID: PMC10726408 DOI: 10.1093/stcltm/szad065] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 08/04/2023] [Indexed: 11/09/2023] Open
Abstract
Currently available therapies for rheumatoid arthritis (RA) are inadequate to alleviate the inflammation and reduce joint damage. While the immune-regulatory effect of human mesenchymal/stromal stem cells (MSCs) extracellular vesicles (EVs) has been tested in many inflammation-related diseases, little is known regarding their effect on patients with RA. Thus, we assessed the effect of human MSCs and MSC-EVs (from naïve or IFN-β-primed MSCs) on CD4+ T cells from patients with RA. Moreover, we investigated the effect of MSC-EVs on RA patients-derived synovial fibroblasts (FLS). MSC-EVs were prepared using a PEG precipitation followed by ultracentrifugation-based protocol. Applied to RA CD4+ T cells, EVs from IFN-β-primed MSCs, suppressed the expression of more key RA-associated cytokines (IL-4, GM-CSF IFN-γ, IL-2, TNF-α), and decreased CD4+ T-cell polyfunctionality than MSCs or EVs from naïve MSCs. MSCs mediated a slight decrease in the frequency of T-regulatory cells, while MSC-EVs rescued the frequency of T-regulatory cells. MSCs significantly inhibited CD4+ T-cell proliferation (P < .05), while no inhibition was observed in response to EV preparations. EVs from IFN-β-primed MSCs inhibited (P < .01) RA FLS migration and downregulated (P < .05) RA FLS surface markers CD34 and HLA-DR. Collectively, we demonstrated the immune-modulatory function of MSCs and their derived EVs in RA CD4+ T cells, which could be further enhanced by priming MSCs with IFN-β. Moreover, EVs from IFN-β-primed MSCs more efficiently inhibit RA FLS migration, and expression of RA FLS-related surface markers, suggesting these EVs as a potent therapy for RA.
Collapse
Affiliation(s)
- Dimitrios Tsiapalis
- School of Pharmacy and Pharmaceutical Sciences & Trinity Biomedical Sciences Institute, Trinity College Dublin and Trinity St. James’s Cancer Institute, Dublin, Ireland
| | - Achilleas Floudas
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent’s University Hospital, UCD, Dublin, Ireland
| | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Verena Boerger
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Douglas J Veale
- EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent’s University Hospital, UCD, Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- EULAR Centre of Excellence, Centre for Arthritis and Rheumatic Diseases, St Vincent’s University Hospital, UCD, Dublin, Ireland
| | - Lorraine O’Driscoll
- School of Pharmacy and Pharmaceutical Sciences & Trinity Biomedical Sciences Institute, Trinity College Dublin and Trinity St. James’s Cancer Institute, Dublin, Ireland
| |
Collapse
|
30
|
Pei XB, Liu B. Research Progress on the Mechanism and Management of Septic Cardiomyopathy: A Comprehensive Review. Emerg Med Int 2023; 2023:8107336. [PMID: 38029224 PMCID: PMC10681771 DOI: 10.1155/2023/8107336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis is defined as a kind of life-threatening organ dysfunction due to a dysregulated host immune response to infection and is a leading cause of mortality in the intensive care unit. Sepsis-induced myocardial dysfunction, also called septic cardiomyopathy, is a common and serious complication in patients with sepsis, which may indicate a bad prognosis. Although efforts have been made to uncover the pathophysiology of septic cardiomyopathy, a number of uncertainties remain. This article sought to review available literature to summarize the existing knowledge on current diagnostic tools and biomarkers, pathogenesis, and treatments for septic cardiomyopathy.
Collapse
Affiliation(s)
- Xue-Bin Pei
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Bo Liu
- Department of Emergency Medicine, Beijing YouAn Hospital, Capital Medical University, Beijing 100069, China
| |
Collapse
|
31
|
Ghassemi K, Inouye K, Takhmazyan T, Bonavida V, Yang JW, de Barros NR, Thankam FG. Engineered Vesicles and Hydrogel Technologies for Myocardial Regeneration. Gels 2023; 9:824. [PMID: 37888397 PMCID: PMC10606880 DOI: 10.3390/gels9100824] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/28/2023] Open
Abstract
Increased prevalence of cardiovascular disease and potentially life-threatening complications of myocardial infarction (MI) has led to emerging therapeutic approaches focusing on myocardial regeneration and restoration of physiologic function following infarction. Extracellular vesicle (EV) technology has gained attention owing to the biological potential to modulate cellular immune responses and promote the repair of damaged tissue. Also, EVs are involved in local and distant cellular communication following damage and play an important role in initiating the repair process. Vesicles derived from stem cells and cardiomyocytes (CM) are of particular interest due to their ability to promote cell growth, proliferation, and angiogenesis following MI. Although a promising candidate for myocardial repair, EV technology is limited by the short retention time of vesicles and rapid elimination by the body. There have been several successful attempts to address this shortcoming, which includes hydrogel technology for the sustained bioavailability of EVs. This review discusses and summarizes current understanding regarding EV technology in the context of myocardial repair.
Collapse
Affiliation(s)
- Kaitlyn Ghassemi
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| | - Keiko Inouye
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| | - Tatevik Takhmazyan
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| | - Victor Bonavida
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| | - Jia-Wei Yang
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; (J.-W.Y.); (N.R.d.B.)
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; (J.-W.Y.); (N.R.d.B.)
| | - Finosh G. Thankam
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (K.G.); (K.I.); (T.T.); (V.B.)
| |
Collapse
|
32
|
An N, Chen Z, Zhao P, Yin W. Extracellular Vesicles in Sepsis: Pathogenic Roles, Organ Damage, and Therapeutic Implications. Int J Med Sci 2023; 20:1722-1731. [PMID: 37928875 PMCID: PMC10620861 DOI: 10.7150/ijms.86832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/05/2023] [Indexed: 11/07/2023] Open
Abstract
Despite significant advances in anti-infective treatment and organ function support technology in recent years, the mortality rate of sepsis remains high. In addition to the high costs of sepsis treatment, the increasing consumption of medical resources also aggravates economic pressure and social burden. Extracellular vesicles (EVs) are membrane vesicles released from different types of activated or apoptotic cells to mediate intercellular communication, which can be detected in both human and animal body fluids. A growing body of researches suggest that EVs play an important role in the pathogenesis of sepsis. In this review, we summarize the predominant roles of EVs in various pathological processes during sepsis and its related organ dysfunction.
Collapse
Affiliation(s)
- Ni An
- Department of Emergency, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Zhe Chen
- University College London, London, UK
| | - Peng Zhao
- Department of Emergency, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Wen Yin
- Department of Emergency, Xijing Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
33
|
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
|
34
|
Wang C, Wang X, Zhang D, Sun X, Wu Y, Wang J, Li Q, Jiang G. The macrophage polarization by miRNAs and its potential role in the treatment of tumor and inflammation (Review). Oncol Rep 2023; 50:190. [PMID: 37711048 PMCID: PMC10523439 DOI: 10.3892/or.2023.8627] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 08/18/2023] [Indexed: 09/16/2023] Open
Abstract
The characteristics of monocyte/macrophage lineage are diversity and plasticity, mainly manifested by M1 and M2 subtypes in the body tissues, and playing different roles in the immunity. In the polarization process of macrophages, the classic molecular mechanism is related to sequential transcription factors. Whether in tumor or inflammatory local microenvironment, the pathological factors of the local microenvironment often affect the polarization of M1 and M2 macrophages, and participate in the occurrence and development of these pathological processes. In recent years, a growing number of research results demonstrated that non‑coding RNA (ncRNA) also participates in the polarization process of macrophages, in addition to traditional cytokines and transcriptional regulation signal pathway molecules. Among numerous ncRNAs, microRNAs (miRNAs) have attracted more attention from scholars both domestically and internationally, and significant progress has been made in basic and clinical research. Therefore, for improved understanding of the molecular mechanism of miRNAs in macrophage polarization and analysis of the potential value of this regulatory pathway in tumor and inflammatory intervention therapy, a comprehensive review of the progress of relevant literature research was conducted and some viewpoints and perspectives were proposed.
Collapse
Affiliation(s)
- Chaozhe Wang
- Department of Immunology, College of Basic Medicine, Binzhou Medical University, Yantai, Shandong 2640032, P.R. China
| | - Xidi Wang
- Department of Laboratory Medicine, Zhangqiu People's Hospital, Jinan, Shandong 250200, P.R. China
| | - Danfeng Zhang
- Department of Laboratory Medicine, Lixia People's Hospital, Jinan, Shandong 250013, P.R. China
| | - Xiaolin Sun
- Department of Laboratory Medicine, Zibo First Hospital, Zibo, Shandong 255200, P.R. China
| | - Yunhua Wu
- Department of Immunology, College of Basic Medicine, Binzhou Medical University, Yantai, Shandong 2640032, P.R. China
| | - Jing Wang
- Department of Immunology, Shandong Yinfeng Academy of Life Science, Jinan, Shandong 250013, P.R. China
| | - Qing Li
- Department of Laboratory Medicine, Zibo First Hospital, Zibo, Shandong 255200, P.R. China
| | - Guosheng Jiang
- Department of Immunology, College of Basic Medicine, Binzhou Medical University, Yantai, Shandong 2640032, P.R. China
- Department of Laboratory Medicine, Zibo First Hospital, Zibo, Shandong 255200, P.R. China
| |
Collapse
|
35
|
Moosazadeh Moghaddam M, Fazel P, Fallah A, Sedighian H, Kachuei R, Behzadi E, Imani Fooladi AA. Host and Pathogen-Directed Therapies against Microbial Infections Using Exosome- and Antimicrobial Peptide-derived Stem Cells with a Special look at Pulmonary Infections and Sepsis. Stem Cell Rev Rep 2023; 19:2166-2191. [PMID: 37495772 DOI: 10.1007/s12015-023-10594-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 07/28/2023]
Abstract
Microbial diseases are a great threat to global health and cause considerable mortality and extensive economic losses each year. The medications for treating this group of diseases (antibiotics, antiviral, antifungal drugs, etc.) directly attack the pathogenic agents by recognizing the target molecules. However, it is necessary to note that excessive use of any of these drugs can lead to an increase in microbial resistance and infectious diseases. New therapeutic methods have been studied recently using emerging drugs such as mesenchymal stem cell-derived exosomes (MSC-Exos) and antimicrobial peptides (AMPs), which act based on two completely different strategies against pathogens including Host-Directed Therapy (HDT) and Pathogen-Directed Therapy (PDT), respectively. In the PDT approach, AMPs interact directly with pathogens to interrupt their intrusion, survival, and proliferation. These drugs interact directly with the cell membrane or intracellular components of pathogens and cause the death of pathogens or inhibit their replication. The mechanism of action of MSC-Exos in HDT is based on immunomodulation and regulation, promotion of tissue regeneration, and reduced host toxicity. This review studies the potential of mesenchymal stem cell-derived exosomes/ATPs therapeutic properties against microbial infectious diseases especially pulmonary infections and sepsis.
Collapse
Affiliation(s)
- Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Parvindokht Fazel
- Department of Microbiology, Fars Science and Research Branch, Islamic Azad University, Shiraz, Iran
| | - Arezoo Fallah
- Department of Bacteriology and Virology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Sedighian
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Kachuei
- Molecular Biology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Elham Behzadi
- Academy of Medical Sciences of the I.R. of Iran, Tehran, Iran
| | - Abbas Ali Imani Fooladi
- Applied Microbiology Research Center, Systems Biology and Poisonings Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
36
|
Li N, Zhang T, Zhu L, Sun L, Shao G, Gao J. Recent Advances of Using Exosomes as Diagnostic Markers and Targeting Carriers for Cardiovascular Disease. Mol Pharm 2023; 20:4354-4372. [PMID: 37566627 DOI: 10.1021/acs.molpharmaceut.3c00268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2023]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of human death worldwide. Exosomes act as endogenous biological vectors; they possess advantages of low immunogenicity and low safety risks, also providing tissue selectivity, including the inherent targeting the to heart. Therefore, exosomes not only have been applied as biomarkers for diagnosis and therapeutic outcome confirmation but also showed potential as drug carriers for cardiovascular targeting delivery. This review aims to summarize the progress and challenges of exosomes as novel biomarkers, especially many novel exosomal noncoding RNAs (ncRNAs), and also provides an overview of the improved targeting functions of exosomes by unique engineered approaches, the latest developed administration methods, and the therapeutic effects of exosomes used as the biocarriers of medications for cardiovascular disease treatment. Also, the possible therapeutic mechanisms and the potentials for transferring exosomes to the clinic for CVD treatment are discussed. The advances, in vivo and in vitro applications, modifications, mechanisms, and challenges summarized in this review will provide a general understanding of this promising strategy for CVD treatment.
Collapse
Affiliation(s)
- Ni Li
- Department of Cardiothoracic Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, Zhejiang 315041, China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Tianyuan Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Linwen Zhu
- Department of Cardiothoracic Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, Zhejiang 315041, China
| | - Lebo Sun
- Department of Cardiothoracic Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, Zhejiang 315041, China
| | - Guofeng Shao
- Department of Cardiothoracic Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, Zhejiang 315041, China
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
37
|
Lu D, Jiao X, Jiang W, Yang L, Gong Q, Wang X, Wei M, Gong S. Mesenchymal stem cells influence monocyte/macrophage phenotype: Regulatory mode and potential clinical applications. Biomed Pharmacother 2023; 165:115042. [PMID: 37379639 DOI: 10.1016/j.biopha.2023.115042] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 06/30/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are pluripotent stem cells derived from a variety of tissues, such as umbilical cord, fat, and bone marrow. Today, MSCs are widely recognized for their prominent anti-inflammatory properties in a variety of acute and chronic inflammatory diseases. In inflammatory diseases, monocytes/macrophages are an important part of the innate immune response in the body, and the alteration of the inflammatory phenotype plays a crucial role in the secretion of pro-inflammatory/anti-inflammatory factors, the repair of injured sites, and the infiltration of inflammatory cells. In this review, starting from the effect of MSCs on the monocyte/macrophage phenotype, we have outlined in detail the process by which MSCs influence the transformation of the monocyte/macrophage inflammatory phenotype, emphasizing the central role of monocytes/macrophages in MSC-mediated anti-inflammatory and damage site repair. MSCs are phagocytosed by monocytes/macrophages in various physiological states, the paracrine effect of MSCs and mitochondrial transfer of MSCs to macrophages to promote the transformation of monocytes/macrophages into anti-inflammatory phenotypes. We also review the clinical applications of the MSCs-monocytes/macrophages system and describe novel pathways between MSCs and tissue repair, the effects of MSCs on the adaptive immune system, and the effects of energy metabolism levels on monocyte/macrophage phenotypic changes.
Collapse
Affiliation(s)
- Dejin Lu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xue Jiao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Wenjian Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Li Yang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Qian Gong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xiaobin Wang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| | - Shiqiang Gong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
38
|
Chen J, Ma S, Luo B, Hao H, Li Y, Yang H, Zhu F, Zhang P, Niu R, Pan P. Human umbilical cord mesenchymal stromal cell small extracellular vesicle transfer of microRNA-223-3p to lung epithelial cells attenuates inflammation in acute lung injury in mice. J Nanobiotechnology 2023; 21:295. [PMID: 37626408 PMCID: PMC10464265 DOI: 10.1186/s12951-023-02038-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Acute lung injury (ALI), manifested as strong pulmonary inflammation and alveolar epithelial damage, is a life-threatening disease with high morbidity and mortality. Small extracellular vesicles (sEVs), secreted by multiple types of cells, are critical cellular communication mediators and can inhibit inflammation by transferring bioactive molecules, such as microRNAs (miRNAs). Thus, we hypothesized that sEVs derived from mesenchymal stromal cells (MSC sEVs) could transfer miRNAs to attenuate inflammation of lung epithelial cells during ALI. METHODS C57BL/6 male mice were intratracheally administered LPS (10 mg/kg). Six hours later, the mice were randomly administered with MSC sEVs (40 µg per mouse in 150 µl of saline), which were collected by ultracentrifugation. Control group received saline administration. After 48 h, the mice were sacrificed to evaluate pulmonary microvascular permeability and inflammatory responses. In vitro, A549 cells and primary human small airway epithelial cells (SAECs) were stimulated with LPS with or without MSC sEVs treatment. RESULTS In vitro, MSC sEVs could also inhibit the inflammation induced by LPS in A549 cells and SAECs (reducing TNF-α, IL-1β, IL-6 and MCP-1). Moreover, MSC sEV treatment improved the survival rate, alleviated pulmonary microvascular permeability, and inhibited proinflammatory responses (reducing TNF-α, IL-1β, IL-6 and JE-1) in ALI mice. Notably, miR-223-3p was found to be served as a critical mediator in MSC sEV-induced regulatory effects through inhibition of poly (adenosine diphosphate-ribose) polymerase-1 (PARP-1) in lung epithelial cells. CONCLUSIONS Overall, these findings suggest that MSC sEVs may offer a novel promising strategy for ALI.
Collapse
Affiliation(s)
- Jie Chen
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China
| | - Shiyang Ma
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China
| | - Baihua Luo
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Haojie Hao
- Institute of Basic Medicine Science, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Medical College, Beijing, China
| | - Yanqin Li
- Center of Pulmonary & Critical Care Medicine, Chinese People's Liberation Army (PLA) General Hospital, Chinese PLA Medical College, Beijing, China
| | - Hang Yang
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China
| | - Fei Zhu
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China
| | - Peipei Zhang
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China
| | - Ruichao Niu
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China.
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China.
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China.
- Department of Respiratory Medicine, The Second Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi, China.
| | - Pinhua Pan
- Department of Respiratory Medicine, Clinical Research Center for Respiratory Disease, Xiangya Hospital, National Key Clinical Specialty, Branch of National, Central South University, No.28 Xiangya Road, Kai-Fu District, Changsha, 410008, Hunan, China.
- Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, 410008, Hunan, China.
- Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Chang-sha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Hunan, P.R. China.
| |
Collapse
|
39
|
Khan S, Mahgoub S, Fallatah N, Lalor PF, Newsome PN. Liver Disease and Cell Therapy: Advances Made and Remaining Challenges. Stem Cells 2023; 41:739-761. [PMID: 37052348 PMCID: PMC10809282 DOI: 10.1093/stmcls/sxad029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 02/27/2023] [Indexed: 04/14/2023]
Abstract
The limited availability of organs for liver transplantation, the ultimate curative treatment for end stage liver disease, has resulted in a growing and unmet need for alternative therapies. Mesenchymal stromal cells (MSCs) with their broad ranging anti-inflammatory and immunomodulatory properties have therefore emerged as a promising therapeutic agent in treating inflammatory liver disease. Significant strides have been made in exploring their biological activity. Clinical application of MSC has shifted the paradigm from using their regenerative potential to one which harnesses their immunomodulatory properties. Reassuringly, MSCs have been extensively investigated for over 30 years with encouraging efficacy and safety data from translational and early phase clinical studies, but questions remain about their utility. Therefore, in this review, we examine the translational and clinical studies using MSCs in various liver diseases and their impact on dampening immune-mediated liver damage. Our key observations include progress made thus far with use of MSCs for clinical use, inconsistency in the literature to allow meaningful comparison between different studies and need for standardized protocols for MSC manufacture and administration. In addition, the emerging role of MSC-derived extracellular vesicles as an alternative to MSC has been reviewed. We have also highlighted some of the remaining clinical challenges that should be addressed before MSC can progress to be considered as therapy for patients with liver disease.
Collapse
Affiliation(s)
- Sheeba Khan
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, West Midlands, UK
| | - Sara Mahgoub
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, West Midlands, UK
| | - Nada Fallatah
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Patricia F Lalor
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
| | - Philip N Newsome
- National Institute for Health Research, Biomedical Research Centre at University Hospitals Birmingham NHS Foundation Trust and the University of Birmingham, Birmingham, West Midlands, UK
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, West Midlands, UK
- Liver Unit, University Hospitals Birmingham NHS Foundation Trust, Birmingham, Birmingham, West Midlands, UK
| |
Collapse
|
40
|
Kandeel M, Morsy MA, Alkhodair KM, Alhojaily S. Mesenchymal Stem Cell-Derived Extracellular Vesicles: An Emerging Diagnostic and Therapeutic Biomolecules for Neurodegenerative Disabilities. Biomolecules 2023; 13:1250. [PMID: 37627315 PMCID: PMC10452295 DOI: 10.3390/biom13081250] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are a type of versatile adult stem cells present in various organs. These cells give rise to extracellular vesicles (EVs) containing a diverse array of biologically active elements, making them a promising approach for therapeutics and diagnostics. This article examines the potential therapeutic applications of MSC-derived EVs in addressing neurodegenerative disorders such as Alzheimer's disease (AD), multiple sclerosis (MS), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD). Furthermore, the present state-of-the-art for MSC-EV-based therapy in AD, HD, PD, ALS, and MS is discussed. Significant progress has been made in understanding the etiology and potential treatments for a range of neurodegenerative diseases (NDs) over the last few decades. The contents of EVs are carried across cells for intercellular contact, which often results in the control of the recipient cell's homeostasis. Since EVs represent the therapeutically beneficial cargo of parent cells and are devoid of many ethical problems connected with cell-based treatments, they offer a viable cell-free therapy alternative for tissue regeneration and repair. Developing innovative EV-dependent medicines has proven difficult due to the lack of standardized procedures in EV extraction processes as well as their pharmacological characteristics and mechanisms of action. However, recent biotechnology and engineering research has greatly enhanced the content and applicability of MSC-EVs.
Collapse
Affiliation(s)
- Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Mohamed A. Morsy
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
- Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia 61511, Egypt
| | - Khalid M. Alkhodair
- Department of Anatomy, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| | - Sameer Alhojaily
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| |
Collapse
|
41
|
Li Y, Wang W, Zhang B, Li L, Zhou D. A bibliometric analysis of exosomes in sepsis from 2004 to 2022. Medicine (Baltimore) 2023; 102:e34613. [PMID: 37543762 PMCID: PMC10403034 DOI: 10.1097/md.0000000000034613] [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: 04/05/2023] [Revised: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 08/07/2023] Open
Abstract
The study aims to summarize topical and frontier issues in sepsis and exosomes and provide advice and resources for researchers working in related disciplines. Publications on exosomes in sepsis from 2004 to 2022 were extracted from the Web of Science Core Collection database. VOSviewer 1.6.18 and CiteSpace 6.1.3 were used to conduct the bibliometric analysis. The number of publications on exosomes in sepsis showed a rapidly rising trend globally. China and the United States were the most published countries. Shanghai Jiao Tong University is the most prolific institution. Frontiers in Immunology was one of the journals with the highest number of papers. Journal of Immunology was the most co-cited journal. Ping Wang was the most productive author. Clotilde Thery was the author who has been cited the most times among co-cited authors. Singer m, 2016, Jama-j am med assoc was the most co-cited reference. "Mesenchymal stem cells derived exosomes," "microRNAs," "apoptosis," and "immunomodulatory therapy" are the current research hot spots and frontiers. This study provides a comprehensive overview of the current status and trends in sepsis and exosomal research. Researchers working in this area will benefit from the hot spots and trends of exosomes in sepsis discovered through this study.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, China
| | - Weina Wang
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, China
| | - Bo Zhang
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, China
| | - Lili Li
- Department of Gastroenterology, Wuhan Fourth Hospital, Wuhan, China
| | - Dengfeng Zhou
- Department of Respiratory and Critical Care Medicine, Wuhan Fourth Hospital, Wuhan, China
| |
Collapse
|
42
|
Pan LF, Niu ZQ, Ren S, Pei HH, Gao YX, Feng H, Sun JL, Zhang ZL. Could extracellular vesicles derived from mesenchymal stem cells be a potential therapy for acute pancreatitis-induced cardiac injury? World J Stem Cells 2023; 15:654-664. [PMID: 37545754 PMCID: PMC10401421 DOI: 10.4252/wjsc.v15.i7.654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/11/2023] [Accepted: 06/27/2023] [Indexed: 07/25/2023] Open
Abstract
Acute pancreatitis (AP) often leads to a high incidence of cardiac injury, posing significant challenges in the treatment of severe AP and contributing to increased mortality rates. Mesenchymal stem cells (MSCs) release bioactive molecules that participate in various inflammatory diseases. Similarly, extracellular vesicles (EVs) secreted by MSCs have garnered extensive attention due to their comparable anti-inflammatory effects to MSCs and their potential to avoid risks associated with cell transplantation. Recently, the therapeutic potential of MSCs-EVs in various inflammatory diseases, including sepsis and AP, has gained increasing recognition. Although preclinical research on the utilization of MSCs-EVs in AP-induced cardiac injury is limited, several studies have demonstrated the positive effects of MSCs-EVs in regulating inflammation and immunity in sepsis-induced cardiac injury and cardiovascular diseases. Furthermore, clinical studies have been conducted on the therapeutic application of MSCs-EVs for some other diseases, wherein the contents of these EVs could be deliberately modified through prior modulation of MSCs. Consequently, we hypothesize that MSCs-EVs hold promise as a potential therapy for AP-induced cardiac injury. This paper aims to discuss this topic. However, additional research is essential to comprehensively elucidate the underlying mechanisms of MSCs-EVs in treating AP-induced cardiac injury, as well as to ascertain their safety and efficacy.
Collapse
Affiliation(s)
- Long-Fei Pan
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China.
| | - Ze-Qun Niu
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Song Ren
- Department of Geriatric Digestive Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Hong-Hong Pei
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Yan-Xia Gao
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Hui Feng
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Jiang-Li Sun
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| | - Zheng-Liang Zhang
- Emergency Department, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China
| |
Collapse
|
43
|
Wu M, Li G, Wang W, Ren H. Emerging roles of microRNAs in septic cardiomyopathy. Front Pharmacol 2023; 14:1181372. [PMID: 37475718 PMCID: PMC10354437 DOI: 10.3389/fphar.2023.1181372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 06/27/2023] [Indexed: 07/22/2023] Open
Abstract
As one of the serious complications of sepsis, septic cardiomyopathy has gained more and more attention, because of its high morbidity and mortality. With the in-depth study of septic cardiomyopathy, several methods have been adopted clinically but have poor therapeutic effects due to failure to find precise therapeutic targets. In recent years, microRNAs have been found to be related to the pathogenesis, diagnosis, and treatment of septic cardiomyopathy via regulating immunity and programmed cell death. This paper reviews the role of microRNAs in septic cardiomyopathy, aiming to provide new targets for the diagnosis and treatment of septic cardiomyopathy.
Collapse
Affiliation(s)
| | | | - Wenjun Wang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Hongsheng Ren
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
44
|
Zhu Z, Zhu P, Fan X, Mo X, Wu X. Mesenchymal stem cell-derived exosomes: a possible therapeutic strategy for repairing heart injuries. Front Cell Dev Biol 2023; 11:1093113. [PMID: 37457298 PMCID: PMC10348815 DOI: 10.3389/fcell.2023.1093113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are one of the most potent therapeutic strategies for repairing cardiac injury. It has been shown in the latest studies that MSCs cannot survive in the heart for a long time. Consequently, the exosomes secreted by MSCs may dominate the repair of heart injury and promote the restoration of cardiac cells, vascular proliferation, immune regulation, etc. Based on the current research, the progress of the acting mechanism, application prospects and challenges of exosomes, including non-coding RNA, in repairing cardiac injuries are summarised in this article.
Collapse
Affiliation(s)
- Zeshu Zhu
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou, Guangdong, China
| | - Xiongwei Fan
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Xiaoyang Mo
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Xiushan Wu
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou, Guangdong, China
| |
Collapse
|
45
|
Lin L, Wei J, Zhu C, Hao G, Xue J, Zhu Y, Wu R. Sema3A alleviates viral myocarditis by modulating SIRT1 to regulate cardiomyocyte mitophagy. ENVIRONMENTAL TOXICOLOGY 2023; 38:1305-1317. [PMID: 36880403 DOI: 10.1002/tox.23765] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/25/2022] [Accepted: 02/20/2023] [Indexed: 05/18/2023]
Abstract
Viral myocarditis (VMC) is a common myocardial inflammatory disease characterized by inflammatory cell infiltration and cardiomyocyte necrosis. Sema3A was reported to reduce cardiac inflammation and improve cardiac function after myocardial infarction, but its role in VMC remains to be explored. Here, a VMC mouse model was established by infection with CVB3, and Sema3A was overexpressed in vivo by intraventricular injection of an adenovirus-mediated Sema3A expression vector (Ad-Sema3A). We found that Sema3A overexpression attenuated CVB3-induced cardiac dysfunction and tissue inflammation. And Sema3A also reduced macrophage accumulation and NLRP3 inflammasome activation in the myocardium of VMC mice. In vitro, LPS was used to stimulate primary splenic macrophages to mimic the macrophage activation state in vivo. Activated macrophages were co-cultured with primary mouse cardiomyocytes to evaluate macrophage infiltration-induced cardiomyocyte damage. Ectopic expression of Sema3A in cardiomyocytes effectively protected cardiomyocytes from activated macrophage-induced inflammation, apoptosis, and ROS accumulation. Mechanistically, cardiomyocyte-expressed Sema3A mitigated macrophage infiltration-caused cardiomyocyte dysfunction by promoting cardiomyocyte mitophagy and hindering NLRP3 inflammasome activation. Furthermore, NAM (a SIRT1 inhibitor) reversed the protective effect of Sema3A against activated macrophage-induced cardiomyocyte dysfunction by suppressing cardiomyocyte mitophagy. In conclusion, Sema3A promoted cardiomyocyte mitophagy and suppressed inflammasome activation by regulating SIRT1, thereby attenuating macrophage infiltration-induced cardiomyocyte injury in VMC.
Collapse
Affiliation(s)
- Lin Lin
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Wei
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Canzhan Zhu
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Guanghua Hao
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiahong Xue
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanhe Zhu
- Department of Medicine, School of Public Health, Institute of Endemic Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Ruiyun Wu
- Department of Medicine, School of Public Health, Institute of Endemic Diseases, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
46
|
Wang L, Wu Y, Yao R, Li Y, Wei Y, Cao Y, Zhang Z, Wu M, Zhu H, Yao Y, Kang H. The role of mesenchymal stem cell-derived extracellular vesicles in inflammation-associated programmed cell death. NANO TODAY 2023; 50:101865. [DOI: 10.1016/j.nantod.2023.101865] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
47
|
Homma K, Bazhanov N, Hashimoto K, Shimizu M, Heathman T, Hao Q, Nawgiri R, Muthukumarana V, Lee JW, Prough DS, Enkhbaatar P. Mesenchymal stem cell-derived exosomes for treatment of sepsis. Front Immunol 2023; 14:1136964. [PMID: 37180159 PMCID: PMC10169690 DOI: 10.3389/fimmu.2023.1136964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/31/2023] [Indexed: 05/15/2023] Open
Abstract
Introduction The pathogenesis of sepsis is an imbalance between pro-inflammatory and anti-inflammatory responses. At the onset of sepsis, the lungs are severely affected, and the injury progresses to acute respiratory distress syndrome (ARDS), with a mortality rate of up to 40%. Currently, there is no effective treatment for sepsis. Cellular therapies using mesenchymal stem cells (MSCs) have been initiated in clinical trials for both ARDS and sepsis based on a wealth of pre-clinical data. However, there remains concern that MSCs may pose a tumor risk when administered to patients. Recent pre-clinical studies have demonstrated the beneficial effects of MSC-derived extracellular vesicles (EVs) for the treatment of acute lung injury (ALI) and sepsis. Methods After recovery of initial surgical preparation, pneumonia/sepsis was induced in 14 adult female sheep by the instillation of Pseudomonas aeruginosa (~1.0×1011 CFU) into the lungs by bronchoscope under anesthesia and analgesia. After the injury, sheep were mechanically ventilated and continuously monitored for 24 h in a conscious state in an ICU setting. After the injury, sheep were randomly allocated into two groups: Control, septic sheep treated with vehicle, n=7; and Treatment, septic sheep treated with MSC-EVs, n=7. MSC-EVs infusions (4ml) were given intravenously one hour after the injury. Results The infusion of MSCs-EVs was well tolerated without adverse events. PaO2/FiO2 ratio in the treatment group tended to be higher than the control from 6 to 21 h after the lung injury, with no significant differences between the groups. No significant differences were found between the two groups in other pulmonary functions. Although vasopressor requirement in the treatment group tended to be lower than in the control, the net fluid balance was similarly increased in both groups as the severity of sepsis progressed. The variables reflecting microvascular hyperpermeability were comparable in both groups. Conclusion We have previously demonstrated the beneficial effects of bone marrow-derived MSCs (10×106 cells/kg) in the same model of sepsis. However, despite some improvement in pulmonary gas exchange, the present study demonstrated that EVs isolated from the same amount of bone marrow-derived MSCs failed to attenuate the severity of multiorgan dysfunctions.
Collapse
Affiliation(s)
- Kento Homma
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Nikolay Bazhanov
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Kazuki Hashimoto
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Masaru Shimizu
- Department of Anesthesiology, University of California, San Francisco, CA, United States
| | - Thomas Heathman
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Qi Hao
- Department of Anesthesiology, University of California, San Francisco, CA, United States
| | - Ranjana Nawgiri
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Vidarshi Muthukumarana
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jae Woo Lee
- Department of Anesthesiology, University of California, San Francisco, CA, United States
| | - Donald S. Prough
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Perenlei Enkhbaatar
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
48
|
Yang S, Zhang K, Hou J, Liu X, Xu D, Chen X, Li S, Hong Y, Zhou C, Wu H, Zheng G, Zeng C, Wu H, Fu J, Wang T. Protective properties of extracellular vesicles in sepsis models: a systematic review and meta-analysis of preclinical studies. J Transl Med 2023; 21:262. [PMID: 37069645 PMCID: PMC10108460 DOI: 10.1186/s12967-023-04121-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/09/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Multiple preclinical studies have reported a beneficial effect of extracellular vesicles (EVs), especially mesenchymal stem cells derived EVs (MSC-EVs), in the treatment of sepsis. However, the therapeutic effect of EVs is still not universally recognized. Therefore, we conducted this meta-analysis by summarizing data from all published studies that met certain criteria to systematically review the association between EVs treatment and mortality in animal models of sepsis. METHODS Systematic retrieval of all studies in PubMed, Cochrane and Web of Science that reported the effects of EVs on sepsis models up to September 2022. The primary outcome was animal mortality. After screening the eligible articles according to inclusion and exclusion criteria, the inverse variance method of fixed effect model was used to calculate the joint odds ratio (OR) and 95% confidence interval (CI). Meta-analysis was performed by RevMan version 5.4. RESULTS In total, 17 studies met the inclusion criteria. Meta-analysis of those studies showed that EVs treatment was associated with reduced mortality in animal models of sepsis (OR 0.17 95% CI: 0.11,0.26, P < 0.001). Further subgroup analysis showed that the mode of sepsis induction, the source, dose, time and method of injection, and the species and gender of mice had no significant effect on the therapeutic effect of EVs. CONCLUSION This meta-analysis showed that MSC-EVs treatment may be associated with lower mortality in animal models of sepsis. Subsequent preclinical studies will need to address the standardization of dose, source, and timing of EVs to provide comparable data. In addition, the effectiveness of EVs in treating sepsis must be studied in large animal studies to provide important clues for human clinical trials.
Collapse
Affiliation(s)
- Shujun Yang
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518003, Guangdong, People's Republic of China
| | - Kanglong Zhang
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518003, Guangdong, People's Republic of China
| | - Jingyu Hou
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518003, Guangdong, People's Republic of China
| | - Xin Liu
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518003, Guangdong, People's Republic of China
| | - Daishi Xu
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518003, Guangdong, People's Republic of China
| | - Xuxiang Chen
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518003, Guangdong, People's Republic of China
| | - Shuangmei Li
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518003, Guangdong, People's Republic of China
| | - Yinghui Hong
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518003, Guangdong, People's Republic of China
| | - Changqing Zhou
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518003, Guangdong, People's Republic of China
| | - Hao Wu
- Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Guanghui Zheng
- Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Chaotao Zeng
- Department of Emergency, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, Guangdong, People's Republic of China
| | - Haidong Wu
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518003, Guangdong, People's Republic of China
| | - Jiaying Fu
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518003, Guangdong, People's Republic of China
| | - Tong Wang
- Department of Emergency, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518003, Guangdong, People's Republic of China.
| |
Collapse
|
49
|
Niu R, Pan P, Li C, Luo B, Ma H, Hao H, Zhao Z, Yang H, Ma S, Zhu F, Chen J. Bone mesenchymal stromal cell-derived small extracellular vesicles inhibit inflammation and ameliorate sepsis via delivery of microRNA-21a-5p. Cytotherapy 2023; 25:625-639. [PMID: 36868991 DOI: 10.1016/j.jcyt.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/17/2023] [Accepted: 02/04/2023] [Indexed: 03/05/2023]
Abstract
BACKGROUND AIMS Sepsis is a potentially life-threatening disease that results from a severe systemic inflammatory response due to infection. Mesenchymal stromal cell-derived small extracellular vesicles (MSC sEVs) are able to transfer bioactive molecules and have been demonstrated to play an important role in the pathophysiological process of sepsis. Herein the authors aimed to investigate the potential role and downstream molecular mechanism of MSC sEVs in sepsis. METHODS MSC sEVs were acquired by ultracentrifugation and then injected into a cecal ligation and puncture mouse model. The efficacy of MSC sEVs in both in vitro and in vivo models of sepsis was evaluated. RESULTS MSC sEV therapy improved survival, reduced sepsis-induced inflammation, attenuated pulmonary capillary permeability and improved liver and kidney function in septic mice. In addition, the authors found that microRNA-21a-5p (miR-21a-5p) was highly enriched in MSC sEVs, could be transferred to recipient cells, inhibited inflammation and increased survival in septic mice. Furthermore, the authors demonstrated that MSC sEV miR-21a-5p suppressed inflammation by targeting toll-like receptor 4 and programmed cell death 4. The therapeutic efficacy of MSC sEVs was partially abrogated by transfection with miR-21a-5p inhibitors. CONCLUSIONS Collectively, the authors' data suggest that miR-21a-5p-bearing MSC sEVs may be a prospective and effective sepsis therapeutic strategy.
Collapse
Affiliation(s)
- Ruichao Niu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China; Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Department of Respiratory Medicine, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China; Department of Hepatobiliary Surgery, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Medical College, Beijing, China
| | - Pinhua Pan
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China; Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Department of Respiratory Medicine, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Chonghui Li
- Institute of Hepatobiliary Surgery, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Medical College, Beijing, China
| | - Baihua Luo
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Hua Ma
- Department of Infectious Disease, People's Hospital of Liuyang City, Liuyang, China
| | - Haojie Hao
- Institute of Basic Medicine Science, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Medical College, Beijing, China
| | - Zhigang Zhao
- Center of Pulmonary and Critical Care Medicine, Chinese People's Liberation Army General Hospital, Chinese People's Liberation Army Medical College, Beijing, China
| | - Hang Yang
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China; Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Department of Respiratory Medicine, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Shiyang Ma
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China; Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Department of Respiratory Medicine, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Fei Zhu
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China; Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Department of Respiratory Medicine, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jie Chen
- Department of Respiratory Medicine, National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China; Center of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China; Clinical Research Center for Respiratory Diseases in Hunan Province, Changsha, China; Hunan Engineering Research Center for Intelligent Diagnosis and Treatment of Respiratory Disease, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China; Department of Respiratory Medicine, Second Affiliated Hospital of Xinjiang Medical University, Urumqi, China.
| |
Collapse
|
50
|
Manzoor T, Saleem A, Farooq N, Dar LA, Nazir J, Saleem S, Ismail S, Gugjoo MB, Shiekh PA, Ahmad SM. Extracellular vesicles derived from mesenchymal stem cells - a novel therapeutic tool in infectious diseases. Inflamm Regen 2023; 43:17. [PMID: 36849892 PMCID: PMC9970864 DOI: 10.1186/s41232-023-00266-6] [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: 12/11/2022] [Accepted: 02/04/2023] [Indexed: 03/01/2023] Open
Abstract
Extracellular vesicles (EVs) are nano-sized lipid-bilayer encapsulated vesicles produced by the cells. These EVs are released into the surrounding space by almost all cell types. The EVs help in intercellular communication via their payloads which contain various proteins, lipids, and nucleic acids generated from the donor cells and allow for synergistic responses in surrounding cells. In recent years, EVs have been increasingly important in treating infectious diseases, including respiratory tract infections, urinary tract infections, wound infections, sepsis, and intestinal infections. Studies have confirmed the therapeutic value of mesenchymal stem cell-derived EVs (MSC-EVs) for treating infectious diseases to eliminate the pathogen, modulate the resistance, and restore tissue damage in infectious diseases. This can be achieved by producing antimicrobial substances, inhibiting pathogen multiplication, and activating macrophage phagocytic activity. Pathogen compounds can be diffused by inserting them into EVs produced and secreted by host cells or by secreting them as microbial cells producing EVs carrying signalling molecules and DNA shielding infected pathogens from immune attack. EVs play a key role in infectious pathogenesis and hold great promise for developing innovative treatments. In this review, we discuss the role of MSC-EVs in treating various infectious diseases.
Collapse
Affiliation(s)
- Tasaduq Manzoor
- grid.444725.40000 0004 0500 6225Division of Animal Biotechnology, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-Kashmir, Srinagar, 190006 India
| | - Afnan Saleem
- grid.444725.40000 0004 0500 6225Division of Animal Biotechnology, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-Kashmir, Srinagar, 190006 India
| | - Nida Farooq
- grid.444725.40000 0004 0500 6225Division of Animal Biotechnology, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-Kashmir, Srinagar, 190006 India
| | - Lateef Ahmad Dar
- grid.444725.40000 0004 0500 6225Division of Animal Biotechnology, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-Kashmir, Srinagar, 190006 India
| | - Junaid Nazir
- grid.444725.40000 0004 0500 6225Division of Animal Biotechnology, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-Kashmir, Srinagar, 190006 India
| | - Sahar Saleem
- grid.444725.40000 0004 0500 6225Division of Animal Biotechnology, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-Kashmir, Srinagar, 190006 India
| | - Sameena Ismail
- grid.412997.00000 0001 2294 5433Government Degree College, Khanabal Kashmir, India
| | - Mudasir Bashir Gugjoo
- grid.444725.40000 0004 0500 6225Veterinary Clinical Services Complex, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-Kashmir, Srinagar, India
| | - Parvaiz A. Shiekh
- grid.417967.a0000 0004 0558 8755Centre for Biomedical Engineering, Indian Institute of Technology-Delhi, New Delhi, 110016 India
| | - Syed Mudasir Ahmad
- Division of Animal Biotechnology, Faculty of Veterinary Sciences & Animal Husbandry, SKUAST-Kashmir, Srinagar, 190006, India.
| |
Collapse
|