1
|
Oliveira Dias J, Sampaio Fagundes I, Bisio MDC, da Silva Barboza V, Jacinto AA, Altei WF. Extracellular vesicles as the common denominator among the 7 Rs of radiobiology: From the cellular level to clinical practice. Biochim Biophys Acta Rev Cancer 2025; 1880:189315. [PMID: 40216093 DOI: 10.1016/j.bbcan.2025.189315] [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: 11/22/2024] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 04/17/2025]
Abstract
Extracellular vesicles (EVs) are lipid-bound particles released by tumor cells and widely explored in cancer development, progression, and treatment response, being considered as valuable components to be explored as biomarkers or cellular targets to modulate the effect of therapies. The mechanisms underlying the production and profile of EVs during radiotherapy (RT) require addressing radiobiological aspects to determine cellular responses to specific radiation doses and fractionation. In this review, we explore the role of EVs in the 7 Rs of radiobiology, known as the molecular basis of a biological tissue response to radiation, supporting EVs as a shared player in all the seven processes. We also highlight the relevance of EVs in the context of liquid biopsy and resistance to immunotherapy, aiming to establish the connection and utility of EVs as tools in contemporary and precision radiotherapy.
Collapse
Affiliation(s)
- Júlia Oliveira Dias
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | | | | | | | | | - Wanessa Fernanda Altei
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil; Radiation Oncology Department, Barretos Cancer Hospital, Barretos, Brazil.
| |
Collapse
|
2
|
Huang C, Li H, Zhang Z, Mou T, Wang D, Li C, Tian L, Zong C. From Mechanism to Therapy: The Role of MSC-EVs in Alleviating Radiation-Induced Injuries. Pharmaceutics 2025; 17:652. [PMID: 40430942 PMCID: PMC12114651 DOI: 10.3390/pharmaceutics17050652] [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: 04/03/2025] [Revised: 05/08/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Radiation injury is a severe issue in both nuclear accidents and cancer radiotherapy. Ionizing radiation impairs the regenerative and repair capabilities of tissues and organs, resulting in a scarcity of effective therapeutic approaches to prevent or mitigate such injuries. Mesenchymal stem cells (MSCs) possess favorable biological characteristics and have emerged as ideal candidates for the treatment of radiation injury. However, the use of MSCs as therapeutic agents is associated with uncertainties in therapeutic efficacy, transient effects, and the risk of immune rejection. Recent advances in research have revealed that extracellular vesicles (EVs) derived from mesenchymal stem cells (MSC-EVs) exhibit similar beneficial properties to MSCs and represent a promising cell-free therapy for mitigating radiation injuries. MSC-EVs are enriched with microRNAs (miRNAs), proteins, and lipids, which can modulate immune responses, inflammatory reactions, cell survival, and proliferation in irradiated tissues. This review synthesizes recent studies on the application of MSC-EVs in radiation injury, focusing on the therapeutic effects and mechanisms of MSC-EVs derived from various sources in radiation-induced diseases of different organs. The therapeutic potential of MSC-EVs for radiation injury provides valuable insights for addressing ionizing radiation-induced injuries and offers a reference for future clinical applications.
Collapse
Affiliation(s)
- Chong Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China; (C.H.); (H.L.); (Z.Z.); (D.W.); (C.L.)
| | - Heng Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China; (C.H.); (H.L.); (Z.Z.); (D.W.); (C.L.)
| | - Zhiyue Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China; (C.H.); (H.L.); (Z.Z.); (D.W.); (C.L.)
| | - Ting Mou
- School of Stomatology, Jiamusi University, Jiamusi 154007, China;
| | - Dandan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China; (C.H.); (H.L.); (Z.Z.); (D.W.); (C.L.)
| | - Chenlu Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China; (C.H.); (H.L.); (Z.Z.); (D.W.); (C.L.)
| | - Lei Tian
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China; (C.H.); (H.L.); (Z.Z.); (D.W.); (C.L.)
| | - Chunlin Zong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China; (C.H.); (H.L.); (Z.Z.); (D.W.); (C.L.)
| |
Collapse
|
3
|
Saadh MJ, Hussein A, Bayani A, Dastafkan S, Amiri M, Akbari A, Shahsavan S, Soleimani Samarkhazan H, Shirani Asl V. Mesenchymal stem cell-derived exosomes: a novel therapeutic frontier in hematological disorders. Med Oncol 2025; 42:199. [PMID: 40327167 DOI: 10.1007/s12032-025-02742-0] [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: 02/11/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells valued for their immunomodulatory and regenerative properties, positioning them as a cornerstone of regenerative medicine. Their derived exosomes small extracellular vesicles laden with bioactive molecules such as proteins, lipids, and nucleic acids have emerged as critical mediators of MSC therapeutic effects. This review systematically explores the biology of MSC-derived exosomes, detailing their biogenesis, molecular composition, and pivotal roles in hematopoiesis, inflammation, and immune regulation. In hematological disorders, including leukemia, lymphoma, and myelodysplastic syndromes, these exosomes exhibit significant therapeutic potential by modulating the tumor microenvironment, enhancing hematopoietic recovery, and suppressing malignant cell proliferation. Notable findings include their ability to induce cell cycle arrest in leukemia cells via the p53 pathway and to reduce chemoresistance through targeted signaling mechanisms, such as the IRF2/INPP4B axis. However, clinical translation is hindered by several challenges, including the standardization of isolation techniques such as ultracentrifugation which are costly and susceptible to contamination as well as difficulties in optimizing large-scale production and ensuring long-term safety and efficacy. Despite these obstacles, MSC-derived exosomes offer a promising, cell-free therapeutic alternative that minimizes risks such as immune rejection and tumorigenicity associated with whole-cell therapies. Future research must prioritize the refinement of isolation and production protocols, the development of precise delivery strategies, and the execution of comprehensive safety evaluations to unlock their full clinical potential in treating hematological disorders and beyond. This review integrates recent advancements to provide a clearer understanding of their multifaceted contributions and highlights the critical gaps that remain.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Ahmed Hussein
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Alireza Bayani
- Division of Hematology and Blood Bank, Department of Laboratory Science, School of Paramedical Science, Shiraz University of Med1ical Sciences, Shiraz, Iran
| | - Shayan Dastafkan
- Student Research Committee, Guilan University of Medical Sciences, Rasht, Iran
| | - Mahdie Amiri
- Department of Laboratory Sciences, Lahijan Branch, Islamic Azad University, Lahijan, Iran
| | - Atie Akbari
- Department of Family Medicine, School of Medicine, Ziaeian Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Shaghayegh Shahsavan
- HSCT Research Center, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Soleimani Samarkhazan
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Vida Shirani Asl
- Division of Hematology and Blood Bank, Department of Laboratory Science, School of Paramedical Science, Shiraz University of Med1ical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Wen S, Dooner M, Pereira M, Del Tatto M, Quesenberry P. Mesenchymal Stem Cell-Derived Extracellular Vesicles Improve Survival and Enhance Hematopoietic Recovery in Mice Exposed to High-Dose Irradiation. Stem Cells Dev 2025; 34:189-200. [PMID: 40135580 DOI: 10.1089/scd.2025.0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025] Open
Abstract
Exposure to high-dose radiation often results in hematopoietic acute radiation syndromes, leading to early mortality, while current therapies for patients exposed to lethal radiation doses are limited. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have shown promise in tissue repair and regeneration but have not been well investigated for mitigating high-dose radiation damage. We previously demonstrated that human or murine MSC-EVs can reverse bone marrow injury caused by mild or moderate radiation. The current study evaluated the therapeutic potential of human MSC-EVs in mice exposed to high-dose total body irradiation (TBI). Mice were exposed to 0, 700, or 950 cGy TBI and subsequently received daily intravenous MSC-EV injections (1 × 109 particles) for 3 days postirradiation. We evaluated survival rates, peripheral blood recovery, bone marrow engraftment, and bone marrow gene expression profiles at various intervals following treatment. MSC-EV administration significantly enhanced survival, with 70% of treated mice surviving 120 days after 950 cGy TBI exposure, compared with 0% survival in untreated controls by day 30. Although early peripheral blood recovery was not observed within 14 days, MSC-EV treatment facilitated substantial recovery at 3 months postirradiation, with significant increases in red blood cell, platelet, white blood cell, and hemoglobin levels, despite white blood cell and hemoglobin levels remaining slightly below normal. Furthermore, the engraftment capacity of bone marrow stem cells was significantly improved. The changes in hematopoietic-related gene expression presented at 14 days postirradiation returned to normal levels by 120 days in MSC-EV-treated mice. These results highlight the potential of MSC-EVs as a therapeutic strategy for high-dose radiation injuries by promoting hematopoietic recovery and improving survival. Our future research will focus on elucidating the radioprotective mechanisms and investigating their integration with existing therapies.
Collapse
Affiliation(s)
- Sicheng Wen
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Mark Dooner
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Mandy Pereira
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Michael Del Tatto
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Peter Quesenberry
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| |
Collapse
|
5
|
Li L, Liu Y, Wang K, Mo J, Weng Z, Jiang H, Jin C. Stem cell exosomes: new hope and future potential for relieving liver fibrosis. Clin Mol Hepatol 2025; 31:333-349. [PMID: 39510097 PMCID: PMC12016649 DOI: 10.3350/cmh.2024.0854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024] Open
Abstract
Liver fibrosis is a chronic liver injury resulting from factors like viral hepatitis, autoimmune hepatitis, non-alcoholic steatohepatitis, fatty liver disease, and cholestatic liver disease. Liver transplantation is currently the gold standard for treating severe liver diseases. However, it is limited by a shortage of donor organs and the necessity for lifelong immunosuppressive therapy. Mesenchymal stem cells (MSCs) can differentiate into various liver cells and enhance liver function when transplanted into patients due to their differentiation and proliferation capabilities. Therefore, it can be used as an alternative therapy for treating liver diseases, especially for liver cirrhosis, liver failure, and liver transplant complications. However, due to the potential tumorigenic effects of MSCs, researchers are exploring a new approach to treating liver fibrosis using extracellular vesicles (exosomes) secreted by stem cells. Many studies show that exosomes released by stem cells can promote liver injury repair through various pathways, contributing to the treatment of liver fibrosis. In this review, we focus on the molecular mechanisms by which stem cell exosomes affect liver fibrosis through different pathways and their potential therapeutic targets. Additionally, we discuss the advantages of exosome therapy over stem cell therapy and the possible future directions of exosome research, including the prospects for clinical applications and the challenges to be overcome.
Collapse
Affiliation(s)
- Lihua Li
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Yongjie Liu
- Department of Cell biology, School of Medicine, Taizhou University, Taizhou, Zhejiang Province, P. R. China
- Department of Pathophysiology, School of Basic Medicine, Shenyang Medical College, Shenyang, Liaoning Province, P. R. China
| | - Kunpeng Wang
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Jinggang Mo
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Zhiyong Weng
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Hao Jiang
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| | - Chong Jin
- 1 Department of General Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou, Zhejiang Province, P. R. China
| |
Collapse
|
6
|
Sun H, Xia T, Ma S, Lv T, Li Y. Intercellular communication is crucial in the regulation of healthy aging via exosomes. Pharmacol Res 2025; 212:107591. [PMID: 39800177 DOI: 10.1016/j.phrs.2025.107591] [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: 11/14/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025]
Abstract
The hallmarks of aging encompass a variety of molecular categories (genomic, telomeric, and epigenetic), organelles (proteostasis, autophagy, and mitochondria), cellular components (including stem cells), systems (such as intercellular communication and chronic inflammation), and environmental factors (dysbiosis and nutrient sensing). These hallmarks play a crucial role in the aging process. Despite their intricate interconnections, the relationships among the hallmarks of aging remain unclear. Although the boundaries between these hallmarks may be indistinct, they exhibit interdependence, with the influence of one hallmark extending to others. Building on this foundation, we investigated the interrelations among the various hallmarks of aging and provided a systematic overview of their logical relationships, proposing that cellular communication plays a crucial role in the aging process. Exosomes function as a primary mode of cellular communication and significantly impact the aging process. Therefore, we propose utilizing exosomes as valuable tools for understanding the mechanisms of aging and addressing age-related concerns. Exosomes may represent a novel approach for the treatment and diagnosis of aging-related conditions in animals. Furthermore, our research reveals that exocytosis in young nematodes slows the aging process, while exocytosis in aged nematodes has the opposite effect, accelerating aging. In conclusion, exosomes act as intercellular messengers that influence the maintenance of a healthy aging process and link the hallmarks of aging with indicators of well-being.
Collapse
Affiliation(s)
- Huifang Sun
- College of Biological and Food Engineering, Qujing Normal University, Qujing, Yunnan 655011, China
| | - Tengyuan Xia
- College of Biological and Food Engineering, Qujing Normal University, Qujing, Yunnan 655011, China
| | - Shuting Ma
- College of Biological and Food Engineering, Qujing Normal University, Qujing, Yunnan 655011, China
| | - Tao Lv
- College of Biological and Food Engineering, Qujing Normal University, Qujing, Yunnan 655011, China.
| | - Yuhong Li
- College of Biological and Food Engineering, Qujing Normal University, Qujing, Yunnan 655011, China.
| |
Collapse
|
7
|
Han J, Xu K, Xu T, Song Q, Duan T, Yang J. The functional regulation between extracellular vesicles and the DNA damage responses. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2025; 795:108532. [PMID: 39828141 DOI: 10.1016/j.mrrev.2025.108532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 01/04/2025] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
The DNA damage response (DDR) is a crucial regulatory mechanism for the survival of organisms, and irregularity of DDR may contribute to the development of various diseases, including tumors, making it is a prominent topic in therapeutic research. Extracellular vesicles (EVs), as important mediators of intercellular communication, have been extensively studied in recent years. Notably, an increasing number of studies have revealed a strong connection between DDR and EVs. On one hand, DNA damage affects the release of EVs and their compositional content; on the other hand, EVs can dictate cell survival or death by modulating DDR in both the parental and the recipient cells. This review outlines current progress in the inter-regulatory relationship between EVs and DDR, with special emphasis on the effects of EVs derived from various sources on DDR in recipient cells. In addition, the potential applications of EVs in research and tumor therapy are discussed.
Collapse
Affiliation(s)
- Jinyi Han
- Department of Nutrition and Toxicology, Hangzhou Normal University School of Public Health, Hangzhou, China
| | - Kexin Xu
- Department of Nutrition and Toxicology, Hangzhou Normal University School of Public Health, Hangzhou, China
| | - Ting Xu
- Department of Nutrition and Toxicology, Hangzhou Normal University School of Public Health, Hangzhou, China
| | - Qin Song
- Department of Nutrition and Toxicology, Hangzhou Normal University School of Public Health, Hangzhou, China
| | - Ting Duan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Jun Yang
- Department of Nutrition and Toxicology, Hangzhou Normal University School of Public Health, Hangzhou, China; Zhejiang Provincial Center for Uterine Cancer Diagnosis and Therapy Research, The Affiliated Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
8
|
Wang N, Ma F, Song H, He N, Zhang H, Li J, Liu Q, Xu C. Mesenchymal Stem Cell-Derived Extracellular Vesicles for Regenerative Applications and Radiotherapy. Cell Transplant 2025; 34:9636897241311019. [PMID: 39780320 PMCID: PMC11713979 DOI: 10.1177/09636897241311019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Tissue repair is an extremely crucial part of clinical treatment. During the course of disease treatment, surgery, chemotherapy, and radiotherapy cause tissue damage. On the other hand, Normal tissue from accidental or therapeutic exposure to high-dose radiation can cause severe tissue damage. There is an urgent need for developing medical countermeasures against radiation injury for tissue repair. Tissue repair involves the regeneration, proliferation, differentiation, and migration of tissue cells; imbalance of local tissue homeostasis, progressive chronic inflammation; decreased cell activity and stem cell function; and wound healing. Although many clinical treatments are currently available for tissue repair, they are expensive. The long recovery time and some unavoidable complications such as cell damage and the inflammatory reaction caused by radiotherapy have led to unsatisfactory results. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) have similar tissue repair functions as MSCs. In tissue damage, EVs can be used as an alternative to stem cell therapy, thereby avoiding related complications such as immunological rejection. EVs play a major role in regulating tissue damage, anti-inflammation, pro-proliferation, and immune response, thus providing a diversified and efficient solution for the repair of disease- and radiotherapy-induced tissue damage. This article reviews the research progress of mesenchymal stem cell-derived EVs in promoting the repair of tissue including heart, lung, liver, intestine, skin, blood system, central nervous system, and tissue damage caused by radiotherapy, thereby aiming to offer new directions and ideas for the radiotherapy and regenerative applications.
Collapse
Affiliation(s)
- Ning Wang
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
| | - Feifei Ma
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
| | - Huijuan Song
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Ningning He
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Huanteng Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Jianguo Li
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
| | - Qiang Liu
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Chang Xu
- School of Basic Medicine Sciences, Shandong Second Medical University, Weifang, China
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| |
Collapse
|
9
|
Karimi N, Dinçsoy AB. The Role of Mesenchymal Stem Cell-Derived Exosomes in Skin Regeneration, Tissue Repair, and the Regulation of Hair Follicle Growth. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1479:1-17. [PMID: 39841379 DOI: 10.1007/5584_2024_839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Skin regeneration, repair, and the promotion of hair growth are intricate and dynamic processes essential for preserving the overall health, functionality, and appearance of both skin and hair. These processes involve a coordinated interplay of cellular activities and molecular signaling pathways that ensure the maintenance and restoration of skin integrity and hair vitality. Recent advancements in regenerative medicine have underscored the significant role of mesenchymal stem cell (MSC)-derived exosomes as key mediators in these processes. Exosomes, emerging as a promising cell-free therapy in tissue engineering, hold substantial potential due to their ability to influence various biological functions. This review explores the mechanisms by which MSC-derived exosomes facilitate skin regeneration and repair, and hair growth, their therapeutic applications, and the future research directions in this emerging field.
Collapse
Affiliation(s)
- Nazli Karimi
- Department of Physiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey.
| | - Adnan Berk Dinçsoy
- Department of Physiology, Faculty of Medicine, Muğla Sıtkı Koçman University, Muğla, Turkey
| |
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
|
Buitrago JC, Cruz-Barrera M, Dorsant-Ardón V, Medina C, Hernández-Mejía DG, Beltrán K, Flórez N, Camacho B, Gruber J, Salguero G. Large and small extracellular vesicles from Wharton's jelly MSCs: Biophysics, function, and strategies to improve immunomodulation. Mol Ther Methods Clin Dev 2024; 32:101353. [PMID: 39512906 PMCID: PMC11541841 DOI: 10.1016/j.omtm.2024.101353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/04/2024] [Indexed: 11/15/2024]
Abstract
Extracellular vesicles (EVs) have emerged as mediators of immunosuppression and pro-regenerative processes, particularly through mesenchymal stromal cells (MSCs) across various disease models. Despite significant progress, there is still a need for a deeper understanding of EV content and functionality to fully harness their biomedical potential. Moreover, strategies to enhance EV production for clinical scalability are still under development. This study aimed to characterize two distinct types of EV-large EV (lgEV) and small EV (smEV)-secreted by Wharton's jelly MSCs (WJ-MSCs). Strategies were explored to augment both EV production and their immunoregulatory effects. Both lgEV and smEV displayed typical EV markers and demonstrated inhibition of human lymphocyte proliferation. Furthermore, analysis of IsomiR content revealed a pronounced immunomodulating signature within MSC-derived EVs, validated by a dual-fluorescence reporter system. MSC primed with pro-inflammatory cytokines yielded increased production of lgEV and smEV, enhancing their immunomodulatory potency. Finally, genetically engineering WJ-MSC to express CD9 resulted in lgEV and smEV with heightened efficacy in suppressing lymphocyte proliferation. This study successfully isolated, characterized, and demonstrated the potent immunosuppressive effect of WJ-MSC-derived lgEV and smEV. We propose cytokine preconditioning and genetic manipulation as viable strategies to enhance the therapeutic potential of WJ-MSC-derived EV in inflammatory conditions.
Collapse
Affiliation(s)
- July Constanza Buitrago
- Advanced Therapies Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud – IDCBIS, Bogotá, Colombia
- Curexsys GmbH, Göttingen, Germany
- PhD Biomedical and Biological Sciences Program, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Mónica Cruz-Barrera
- Advanced Therapies Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud – IDCBIS, Bogotá, Colombia
| | - Valerie Dorsant-Ardón
- Advanced Therapies Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud – IDCBIS, Bogotá, Colombia
| | - Carlos Medina
- Advanced Therapies Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud – IDCBIS, Bogotá, Colombia
| | - David G. Hernández-Mejía
- Advanced Therapies Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud – IDCBIS, Bogotá, Colombia
| | - Karl Beltrán
- Advanced Therapies Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud – IDCBIS, Bogotá, Colombia
| | - Natalia Flórez
- Faculty of Medicine, Universidad EAN, Medellín, Colombia
| | - Bernardo Camacho
- Advanced Therapies Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud – IDCBIS, Bogotá, Colombia
| | | | - Gustavo Salguero
- Advanced Therapies Unit, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud – IDCBIS, Bogotá, Colombia
| |
Collapse
|
12
|
Yang BL, Long YY, Lei Q, Gao F, Ren WX, Cao YL, Wu D, Xu LY, Qu J, Li H, Yu YL, Zhang AY, Wang S, Wang HX, Chen ZC, Li QB. Lethal pulmonary thromboembolism in mice induced by intravenous human umbilical cord mesenchymal stem cell-derived large extracellular vesicles in a dose- and tissue factor-dependent manner. Acta Pharmacol Sin 2024; 45:2300-2312. [PMID: 38914677 PMCID: PMC11489411 DOI: 10.1038/s41401-024-01327-3] [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: 01/30/2024] [Accepted: 05/27/2024] [Indexed: 06/26/2024]
Abstract
Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have obvious advantages over MSC therapy. But the strong procoagulant properties of MSC-EVs pose a potential risk of thromboembolism, an issue that remains insufficiently explored. In this study, we systematically investigated the procoagulant activity of large EVs derived from human umbilical cord MSCs (UC-EVs) both in vitro and in vivo. UC-EVs were isolated from cell culture supernatants. Mice were injected with UC-EVs (0.125, 0.25, 0.5, 1, 2, 4 μg/g body weight) in 100 μL PBS via the tail vein. Behavior and mortality were monitored for 30 min after injection. We showed that these UC-EVs activated coagulation in a dose- and tissue factor-dependent manner. UC-EVs-induced coagulation in vitro could be inhibited by addition of tissue factor pathway inhibitor. Notably, intravenous administration of high doses of the UC-EVs (1 μg/g body weight or higher) led to rapid mortality due to multiple thrombus formations in lung tissue, platelets, and fibrinogen depletion, and prolonged prothrombin and activated partial thromboplastin times. Importantly, we demonstrated that pulmonary thromboembolism induced by the UC-EVs could be prevented by either reducing the infusion rate or by pre-injection of heparin, a known anticoagulant. In conclusion, this study elucidates the procoagulant characteristics and mechanisms of large UC-EVs, details the associated coagulation risk during intravenous delivery, sets a safe upper limit for intravenous dose, and offers effective strategies to prevent such mortal risks when high doses of large UC-EVs are needed for optimal therapeutic effects, with implications for the development and application of large UC-EV-based as well as other MSC-EV-based therapies.
Collapse
Affiliation(s)
- Bian-Lei Yang
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yao-Ying Long
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qian Lei
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fei Gao
- Department of Hematology, Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Wen-Xiang Ren
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu-Lin Cao
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Di Wu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Liu-Yue Xu
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiao Qu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - He Li
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ya-Li Yu
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - An-Yuan Zhang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shan Wang
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hong-Xiang Wang
- Department of Hematology, Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Zhi-Chao Chen
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiu-Bai Li
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Engineering Research Center for Application of Extracellular Vesicles, Hubei University of Science and Technology, Xianning, 437100, China.
| |
Collapse
|
13
|
Mehrvar A, Akbari M, Khosroshahi EM, Nekavand M, Mokhtari K, Baniasadi M, Aghababaian M, Karimi M, Amiri S, Moazen A, Maghsoudloo M, Alimohammadi M, Rahimzadeh P, Farahani N, Vaghar ME, Entezari M, Hashemi M. The impact of exosomes on bone health: A focus on osteoporosis. Pathol Res Pract 2024; 263:155618. [PMID: 39362132 DOI: 10.1016/j.prp.2024.155618] [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: 08/29/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
Osteoporosis is a widespread chronic condition. Although standard treatments are generally effective, they are frequently constrained by side effects and the risk of developing drug resistance. A promising area of research is the investigation of extracellular vesicles (EVs), including exosomes, microvesicles, and apoptotic bodies, which play a crucial role in bone metabolism. Exosomes, in particular, have shown significant potential in both the diagnosis and treatment of osteoporosis. EVs derived from osteoclasts, osteoblasts, mesenchymal stem cells, and other sources can influence bone metabolism, while exosomes from inflammatory and tumor cells may exacerbate bone loss, highlighting their dual role in osteoporosis pathology. This review offers a comprehensive overview of EV biogenesis, composition, and function in osteoporosis, focusing on their diagnostic and therapeutic potential. We examine the roles of various types of EVs and their cargo-proteins, RNAs, and lipids-in bone metabolism. Additionally, we explore the emerging applications of EVs as biomarkers and therapeutic agents, emphasizing the need for further research to address current challenges and enhance EV-based strategies for managing osteoporosis.
Collapse
Affiliation(s)
- Amir Mehrvar
- Assistant Professor, Department of Orthopedics, Taleghani Hospital Research Development Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadarian Akbari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrandokht Nekavand
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Midwifery, Faculty of nursing and midwifery, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Khatere Mokhtari
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mojtaba Baniasadi
- Department of Orthopedic Surgery, Isfahan University of Medical Sciences, Isfahan, Iran; MD, Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Aghababaian
- Department of Orthopedic Surgery, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansour Karimi
- MD, Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Amiri
- MD, Assistant Professor of Orthopaedic Surgery, Shohadaye Haftom-e-Tir Hospital, Department of Orthopedic, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Moazen
- Department of Orthopedics, Bone and Joint Reconstruction Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, PR China
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mohammad Eslami Vaghar
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of gynecology, Faculty of Medicine, Tehran Medical sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
14
|
Buitrago JC, Morris SL, Backhaus A, Kaltenecker G, Kaipa JM, Girard C, Schneider S, Gruber J. Unveiling the Immunomodulatory and regenerative potential of iPSC-derived mesenchymal stromal cells and their extracellular vesicles. Sci Rep 2024; 14:24098. [PMID: 39407038 PMCID: PMC11480492 DOI: 10.1038/s41598-024-75956-3] [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: 06/27/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
Induced pluripotent stem cell (iPSC)-derived mesenchymal stromal cells (iMSCs) offer a promising alternative to primary mesenchymal stromal cells (MSCs) and their derivatives, particularly extracellular vesicles (EVs), for use in advanced therapy medicinal products. In this study we evaluated the immunomodulatory and regenerative potential of iMSCs as well as iMSC-EVs, alongside primary human umbilical cord-derived mesenchymal stromal cells (hUCMSCs). Our findings demonstrate that iMSCs exhibit comparable abilities to hUCMSCs in regulating lymphocyte proliferation and inducing an anti-inflammatory phenotype in monocytes. We also observed decreased TNFα levels and increased IL-10 induction, indicating a potential mechanism for their immunomodulatory effects. Furthermore, iMSC-EVs also showed effective immunomodulation by inhibiting T cell proliferation and inducing macrophage polarization similar to their parental cells. Additionally, iMSC-EVs exhibited pro-regenerative potential akin to hUCMSC-EVs in in vitro scratch assays. Notably, priming iMSCs with pro-inflammatory cytokines significantly enhanced the immunomodulatory potential of iMSC-EVs. These results underscore the considerable promise of iMSCs and iMSCs-EVs as an alternate source for MSC-derived therapeutics, given their potent immunomodulatory and regenerative properties.
Collapse
Affiliation(s)
- July Constanza Buitrago
- Curexsys GmbH, Göttingen, Germany.
- PhD Biomedical and Biological Sciences Program, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia.
- Life Science Factory, Curexsys GmbH, Annastraβe 27, Göttingen, Germany, D-37075.
| | | | | | | | | | | | | | - Jens Gruber
- Curexsys GmbH, Göttingen, Germany.
- Life Science Factory, Curexsys GmbH, Annastraβe 27, Göttingen, Germany, D-37075.
| |
Collapse
|
15
|
Fathi E, Valipour B, Jafari S, Kazemi A, Montazersaheb S, Farahzadi R. The role of the hematopoietic stem/progenitor cells-derived extracellular vesicles in hematopoiesis. Heliyon 2024; 10:e35051. [PMID: 39157371 PMCID: PMC11327835 DOI: 10.1016/j.heliyon.2024.e35051] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 07/20/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are tightly regulated by specific microenvironments called niches to produce an appropriate number of mature blood cell types. Self-renewal and differentiation are two hallmarks of hematopoietic stem and progenitor cells, and their balance is critical for proper functioning of blood and immune cells throughout life. In addition to cell-intrinsic regulation, extrinsic cues within the bone marrow niche and systemic factors also affect the fate of HSCs. Despite this, many paracrine and endocrine factors that influence the function of hematopoietic cells remain unknown. In hematological malignancies, malignant cells remodel their niche into a permissive environment to enhance the survival of leukemic cells. These events are accompanied by loss of normal hematopoiesis. It is well known that extracellular vehicles (EVs) mediate intracellular interactions under physiological and pathological conditions. In other words, EVs transfer biological information to surrounding cells and contribute not only to physiological functions but also to the pathogenesis of some diseases, such as cancers. Therefore, a better understanding of cell-to-cell interactions may lead to identification of potential therapeutic targets. Recent reports have suggested that EVs are evolutionarily conserved constitutive mediators that regulate hematopoiesis. Here, we focus on the emerging roles of EVs in normal and pathological conditions, particularly in hematological malignancies. Owing to the high abundance of EVs in biological fluids, their potential use as biomarkers and therapeutic tools is discussed.
Collapse
Affiliation(s)
- Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Behnaz Valipour
- Department of Basic Sciences and Health, Sarab Faculty of Medical Sciences, Sarab, Iran
| | - Sevda Jafari
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abdolhassan Kazemi
- Medical Philosophy and History Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Parasitology and Mycology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Li Z, Yao X, Zhang J, Yang J, Ni J, Wang Y. Exploring the bone marrow micro environment in thalassemia patients: potential therapeutic alternatives. Front Immunol 2024; 15:1403458. [PMID: 39161767 PMCID: PMC11330836 DOI: 10.3389/fimmu.2024.1403458] [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: 03/19/2024] [Accepted: 07/22/2024] [Indexed: 08/21/2024] Open
Abstract
Genetic mutations in the β-globin gene lead to a decrease or removal of the β-globin chain, causing the build-up of unstable alpha-hemoglobin. This condition is referred to as beta-thalassemia (BT). The present treatment strategies primarily target the correction of defective erythropoiesis, with a particular emphasis on gene therapy and hematopoietic stem cell transplantation. However, the presence of inefficient erythropoiesis in BT bone marrow (BM) is likely to disturb the previously functioning BM microenvironment. This includes accumulation of various macromolecules, damage to hematopoietic function, destruction of bone cell production and damage to osteoblast(OBs), and so on. In addition, the changes of BT BM microenvironment may have a certain correlation with the occurrence of hematological malignancies. Correction of the microenvironment can be achieved through treatments such as iron chelation, antioxidants, hypoglycemia, and biologics. Hence, This review describes damage in the BT BM microenvironment and some potential remedies.
Collapse
Affiliation(s)
- Zengzheng Li
- Department of Hematology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China
| | - Xiangmei Yao
- Department of Hematology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China
| | - Jie Zhang
- Department of Medical Genetics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jinghui Yang
- Department of Pediatrics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Junxue Ni
- Hospital Office, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yajie Wang
- Department of Hematology, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Province Clinical Research Center for Hematologic Disease, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- Yunnan Provincial Clinical Medical Center for Blood Diseases and Thrombosis Prevention and Treatment, Kunming, Yunnan, China
| |
Collapse
|
17
|
Lyu Z, Xin M, Oyston DR, Xue T, Kang H, Wang X, Wang Z, Li Q. Cause and consequence of heterogeneity in human mesenchymal stem cells: Challenges in clinical application. Pathol Res Pract 2024; 260:155354. [PMID: 38870711 DOI: 10.1016/j.prp.2024.155354] [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: 02/22/2024] [Revised: 04/25/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024]
Abstract
Human mesenchymal stem cells (hMSCs) are mesoderm-derived adult stem cells with self-proliferation capacity, pluripotent differentiation potency, and excellent histocompatibility. These advantages make hMSCs a promising tool in clinical application. However, the majority of clinical trials using hMSC therapy for diverse human diseases do not achieve expectations, despite the prospective pre-clinical outcomes in animal models. This is partly attributable to the intrinsic heterogeneity of hMSCs. In this review, the cause of heterogeneity in hMSCs is systematically discussed at multiple levels, including isolation methods, cultural conditions, donor-to-donor variation, tissue sources, intra-tissue subpopulations, etc. Additionally, the effect of hMSCs heterogeneity on the contrary role in tumor progression and immunomodulation is also discussed. The attempts to understand the cellular heterogeneity of hMSCs and its consequences are important in supporting and improving therapeutic strategies for hMSCs.
Collapse
Affiliation(s)
- Zhao Lyu
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Miaomiao Xin
- Assisted Reproductive Center, Women's & Children's Hospital of Northwest, Xi'an, Shaanxi, China; University of South Bohemia in Ceske Budejovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Research Institute of Fish Culture and Hydrobiology, Vodnany, Czech Republic
| | - Dale Reece Oyston
- Department of Evolution, Ecology and Behaviour, University of Liverpool, Liverpool, UK
| | - Tingyu Xue
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Hong Kang
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Xiangling Wang
- Department of Clinical Laboratory, Xi'an International Medical Center Hospital, Xi'an, Shaanxi, China
| | - Zheng Wang
- Medical Center of Hematology, the Second Affiliated Hospital, Army Medical University, Chongqing, Sichuan, China.
| | - Qian Li
- Changsha Medical University, Changsha, Hunan, China.
| |
Collapse
|
18
|
Christy BA, Herzig MC, Wu X, Mohammadipoor A, McDaniel JS, Bynum JA. Cell Therapies for Acute Radiation Syndrome. Int J Mol Sci 2024; 25:6973. [PMID: 39000080 PMCID: PMC11241804 DOI: 10.3390/ijms25136973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024] Open
Abstract
The risks of severe ionizing radiation exposure are increasing due to the involvement of nuclear powers in combat operations, the increasing use of nuclear power, and the existence of terrorist threats. Exposure to a whole-body radiation dose above about 0.7 Gy results in H-ARS (hematopoietic acute radiation syndrome), which is characterized by damage to the hematopoietic system; higher doses result in further damage to the gastrointestinal and nervous systems. Only a few medical countermeasures for ARS are currently available and approved for use, although others are in development. Cell therapies (cells or products produced by cells) are complex therapeutics that show promise for the treatment of radiation injury and have been shown to reduce mortality and morbidity in animal models. Since clinical trials for ARS cannot be ethically conducted, animal testing is extremely important. Here, we describe cell therapies that have been tested in animal models. Both cells and cell products appear to promote survival and lessen tissue damage after whole-body irradiation, although the mechanisms are not clear. Because radiation exposure often occurs in conjunction with other traumatic injuries, animal models of combined injury involving radiation and future countermeasure testing for these complex medical problems are also discussed.
Collapse
Affiliation(s)
- Barbara A Christy
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
- Department of Molecular Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Maryanne C Herzig
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
| | - Xiaowu Wu
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
| | - Arezoo Mohammadipoor
- Hemorrhage and Vascular Dysfunction, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
| | - Jennifer S McDaniel
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
| | - James A Bynum
- Blood and Shock Resuscitation, US Army Institute of Surgical Research, Joint Base San Antonio, Fort Sam Houston, TX 78234, USA
- Department of Surgery, UT Health San Antonio, San Antonio, TX 78229, USA
- Trauma Research and Combat Casualty Care Collaborative, UT Health San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
19
|
Wang H, Zhang N, Wang X, Tian J, Yi J, Yao L, Huang G. Emerging role of mesenchymal stem cell-derived exosome microRNA in radiation injury. Int J Radiat Biol 2024; 100:996-1008. [PMID: 38776447 DOI: 10.1080/09553002.2024.2347348] [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: 01/18/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE Radiation injury (RI) is a common occurrence in malignant tumors patients receiving radiation therapy. While killing tumor cells, normal tissue surrounding the target area is inevitably irradiated at a certain dose, which can cause varying results of radiation injury. Currently, there are limited clinical treatments available for radiation injuries. In recent years, the negative effects of stem cell therapy have been reported more clearly and non-cellular therapies such as exosomes have become a focus of attention for researchers. As a type of vesicle-like substances secreted by mesenchymal stem cells (MSC), MSC derived exosomes (MSC-exo) carry DNA, mRNA, microRNA (miRNAs), specific proteins, lipids, and other active substances involved in intercellular information exchange. miRNAs released by MSC-exo are capable of alleviating and repairing damaged tissues through anti-apoptosis, modulating immune response, regulating inflammatory response and promoting angiogenesis, which indicates that MSC-exo miRNAs have great potential for application in the prevention and treatment of radiation injury. Therefore, it is necessary to explore the underlying therapeutic mechanisms of MSC-exo miRNAs in this process, which may shed new lights on the treatment of radiation injury. CONCLUSIONS Increasing evidence confirms that MSC-exo has shown encouraging applications in tissue repair due to the anti-apoptotic, immunoreactive, and pro-angiogenesis effects of the miRNAs it carries as intercellular communication carriers. However, miRNA-based therapeutics are still in their infancy and many practical issues remain to be addressed for clinical applications.
Collapse
Affiliation(s)
- Huike Wang
- School of Stomatology, ZunYi Medical University, Zunyi, Guizhou, China
| | - Nini Zhang
- School of Stomatology, ZunYi Medical University, Zunyi, Guizhou, China
| | - Xue Wang
- School of Stomatology, ZunYi Medical University, Zunyi, Guizhou, China
| | - Jia Tian
- School of Stomatology, ZunYi Medical University, Zunyi, Guizhou, China
| | - Jie Yi
- School of Stomatology, ZunYi Medical University, Zunyi, Guizhou, China
| | | | - Guilin Huang
- School of Stomatology, ZunYi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
20
|
Ratajczak MZ, Ratajczak J. Leukemogenesis occurs in a microenvironment enriched by extracellular microvesicles/exosomes: recent discoveries and questions to be answered. Leukemia 2024; 38:692-698. [PMID: 38388648 PMCID: PMC10997496 DOI: 10.1038/s41375-024-02188-9] [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: 01/16/2024] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/24/2024]
Abstract
In single-cell organisms, extracellular microvesicles (ExMVs) were one of the first cell-cell communication platforms that emerged very early during evolution. Multicellular organisms subsequently adapted this mechanism. Evidence indicates that all types of cells secrete these small circular structures surrounded by a lipid membrane that may be encrusted by ligands and receptors interacting with target cells and harboring inside a cargo comprising RNA species, proteins, bioactive lipids, signaling nucleotides, and even entire organelles "hijacked" from the cells of origin. ExMVs are secreted by normal cells and at higher levels by malignant cells, and there are some differences in their cargo. On the one hand, ExMVs secreted from malignant cells interact with cells in the microenvironment, and in return, they are exposed by a "two-way mechanism" to ExMVs secreted by non-leukemic cells. Therefore, leukemogenesis occurs and progresses in ExMVs enriched microenvironments, and this biological fact has pathologic, diagnostic, and therapeutic implications. We are still trying to decipher this intriguing cell-cell communication language better. We will present a current point of view on this topic and review some selected most recent discoveries and papers.
Collapse
Affiliation(s)
- Mariusz Z Ratajczak
- Stem Cell Institute at Brown Cancer Center, University of Louisville, Louisville, KY, USA.
- Department of Regenerative Medicine, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland.
| | - Janina Ratajczak
- Stem Cell Institute at Brown Cancer Center, University of Louisville, Louisville, KY, USA
| |
Collapse
|
21
|
Padinharayil H, Varghese J, Wilson C, George A. Mesenchymal stem cell-derived exosomes: Characteristics and applications in disease pathology and management. Life Sci 2024; 342:122542. [PMID: 38428567 DOI: 10.1016/j.lfs.2024.122542] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
Mesenchymal stem cells (MSCs) possess a role in tissue regeneration and homeostasis because of inherent immunomodulatory capacity and the production of factors that encourage healing. There is substantial evidence that MSCs' therapeutic efficacy is primarily determined by their paracrine function including in cancers. Extracellular vesicles (EVs) are basic paracrine effectors of MSCs that reside in numerous bodily fluids and cell homogenates and play an important role in bidirectional communication. MSC-derived EVs (MSC-EVs) offer a wide range of potential therapeutic uses that exceed cell treatment, while maintaining protocell function and having less immunogenicity. We describe characteristics and isolation methods of MSC-EVs, and focus on their therapeutic potential describing its roles in tissue repair, anti-fibrosis, and cancer with an emphasis on the molecular mechanism and immune modulation and clinical trials. We also explain current understanding and challenges in the clinical applications of MSC-EVs as a cell free therapy.
Collapse
Affiliation(s)
- Hafiza Padinharayil
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 05, Kerala, India; PG & Research Department of Zoology, St. Thomas College, Kozhencherry, Pathanamthitta, Kerala 689641, India
| | - Jinsu Varghese
- PG & Research Department of Zoology, St. Thomas College, Kozhencherry, Pathanamthitta, Kerala 689641, India
| | - Cornelia Wilson
- Canterbury Christ Church University, Natural Applied Sciences, Life Science Industry Liaison Lab, Discovery Park, Sandwich CT139FF, United Kingdom.
| | - Alex George
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur 05, Kerala, India.
| |
Collapse
|
22
|
Li Z, Hou D, Tang Z, Xiong L, Yan Y. The potential role of stem cells-derived extracellular vesicles in the treatment of musculoskeletal system diseases. Cell Biol Int 2024; 48:237-252. [PMID: 38100269 DOI: 10.1002/cbin.12107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/29/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023]
Abstract
The therapeutic potential of stem cells-derived extracellular vesicles (EVs) has shown a great progress in the regenerative medicine. EVs are rich in a variety of bioactive substances, which are important carriers of signal transmission and interactions between cells, and they play an important role in the processes of tissue repair and regeneration. Several studies have shown that stem cells-derived EVs regulate immunity, promote cell proliferation and differentiation, enhance bone and vascular regeneration, and play an increasingly important role in musculoskeletal system. This review aimed to describe the biological characteristics of stem cells-derived EVs and discuss their potential role in the therapy of musculoskeletal system diseases.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Demiao Hou
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Zijin Tang
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Lishun Xiong
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| | - Yiguo Yan
- Department of Spine Surgery, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, Hunan, China
| |
Collapse
|
23
|
Liu H, Kuang H, Wang Y, Bao L, Cao W, Yu L, Qi M, Wang R, Yang X, Ye Q, Ding F, Ren L, Liu S, Ma F, Liu S. MSC-derived exosomes protect auditory hair cells from neomycin-induced damage via autophagy regulation. Biol Res 2024; 57:3. [PMID: 38217055 PMCID: PMC10787390 DOI: 10.1186/s40659-023-00475-w] [Citation(s) in RCA: 6] [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: 06/27/2023] [Accepted: 11/10/2023] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND Sensorineural hearing loss (SNHL) poses a major threat to both physical and mental health; however, there is still a lack of effective drugs to treat the disease. Recently, novel biological therapies, such as mesenchymal stem cells (MSCs) and their products, namely, exosomes, are showing promising therapeutic potential due to their low immunogenicity, few ethical concerns, and easy accessibility. Nevertheless, the precise mechanisms underlying the therapeutic effects of MSC-derived exosomes remain unclear. RESULTS Exosomes derived from MSCs reduced hearing and hair cell loss caused by neomycin-induced damage in models in vivo and in vitro. In addition, MSC-derived exosomes modulated autophagy in hair cells to exert a protective effect. Mechanistically, exogenously administered exosomes were internalized by hair cells and subsequently upregulated endocytic gene expression and endosome formation, ultimately leading to autophagy activation. This increased autophagic activity promoted cell survival, decreased the mitochondrial oxidative stress level and the apoptosis rate in hair cells, and ameliorated neomycin-induced ototoxicity. CONCLUSIONS In summary, our findings reveal the otoprotective capacity of exogenous exosome-mediated autophagy activation in hair cells in an endocytosis-dependent manner, suggesting possibilities for deafness treatment.
Collapse
Affiliation(s)
- Huan Liu
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, China
| | - Huijuan Kuang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yiru Wang
- Anesthesiology Department, Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Lili Bao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wanxin Cao
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, China
| | - Lu Yu
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University and Shandong Key Laboratory of Oral Tissue Regeneration and Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration and Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong, China
| | - Meihao Qi
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Air Force Military, Xi'an, Shaanxi, China
| | - Renfeng Wang
- Department of Otolaryngology-Head and Neck Surgery, Xijing Hospital, Air Force Military, Xi'an, Shaanxi, China
| | - Xiaoshan Yang
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Qingyuan Ye
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Digital Dentistry Center, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Feng Ding
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lili Ren
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Siying Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Furong Ma
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, China.
| | - Shiyu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology,, The Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
24
|
Wei W, Bai H, Zhang T, Cai S, Zhou Y, Liu M, Zhang Y, Chen Y, Hua J, He J, Ding N, Miao G, Wang J. Regulation of Circulating miR-342-3p Alleviates the Radiation-Induced Immune System Injury. Radiat Res 2023; 200:556-568. [PMID: 37874034 DOI: 10.1667/rade-23-00125.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/12/2023] [Indexed: 10/25/2023]
Abstract
Ionizing radiation in space, radiation devices or nuclear disasters are major threats to human health and public security. Expanding countermeasures for dealing with accidental or occupational radiation exposure is crucial for the protection of radiation injuries. Circulating microRNAs (miRNAs) have emerged as promising radiation biomarkers in recent years. However, the origin, distribution and functions of radiosensitive circulating miRNAs remain unclear, which obstructs their clinical applications in the future. In this study, we found that mmu-miR-342-3p (miR-342) in mouse serum presents a stable and significant decrease after X-ray total-body irradiation (TBI). Focusing on this miRNA, we investigated the influences of circulating miR-342 on the radiation-induced injury. Through tail vein injection of Cy5-labeled synthetic miR-342, we found the exogenous miR-342-Cy5 was mainly enriched in metabolic and immune organs. Besides, the bioinformatic analysis predicted that miR-342 might involve in immune-related processes or pathways. Further, mice were tail vein injected with synthetic miR-342 mimetics (Ago-miR-342) after irradiation to upregulate the level of miR-342 in circulating blood. The results showed that the upregulation of circulating miR-342 alleviated the radiation-induced depletion of CD3+CD4+ T lymphocytes and influenced the levels of IL-2 and IL-6 in irradiated mice. Moreover, the injection of Ago-miR-342 improved the survival rates of mice with acute radiation injury. Our findings demonstrate that upregulation of circulating miR-342 alleviates the radiation-induced immune system injury, which provides us new insights into the functions of circulating miRNAs and the prospect as the targets for mitigation of radiation injuries.
Collapse
Affiliation(s)
- Wenjun Wei
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Hao Bai
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Tianyi Zhang
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Shufan Cai
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
- School of Nuclear Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Yumeng Zhou
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Min Liu
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Yanan Zhang
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Yaxiong Chen
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Junrui Hua
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Jinpeng He
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Nan Ding
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - GuoYing Miao
- Department of Radiation Oncology, Gansu Provincial Central Hospital, Lanzhou 730000, China
| | - Jufang Wang
- Biomedical Research Center, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| |
Collapse
|
25
|
Harrell CR, Djonov V, Volarevic V. Mesenchymal stem cell-derived microRNAs: Friends or foes of tumor cells? Histol Histopathol 2023; 38:1373-1379. [PMID: 37306386 DOI: 10.14670/hh-18-633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Mesenchymal stem cell (MSC)-dependent biological effects in the tumor microenvironment mainly rely on the activity of MSC-sourced microRNAs (MSC-miRNAs) which modulate protein synthesis in target tumor cells, endothelial cells and tumor-infiltrated immune cells, regulating their phenotype and function. Several MSC-sourced miRNAs (miR-221, miR-23b, miR-21-5p, miR-222/223, miR-15a miR-424, miR-30b, miR-30c) possess tumor-promoting properties and are able to enhance viability, invasiveness and metastatic potential of malignant cells, induce proliferation and sprouting of tumor endothelial cells and suppress effector functions of cytotoxic tumor-infiltrated immune cells, crucially contributing to the rapid growth and progression of tumor tissue. On the contrary, MSCs also produce "anti-tumorigenic" miRNAs (miR-100, miR-222-3p, miR-146b miR-302a, miR-338-5p, miR-100-5p and miR-1246) which suppress tumor growth and progression by: Up-regulating expression of chemoresistance-related genes in tumor cells, by suppressing neo-angiogenesis and by inducing generation of tumorotoxic phenotypes in tumor-infiltrated lymphocytes. In this review article, we summarize the current knowledge about molecular mechanisms that are responsible for MSC-miRNA-dependent alterations of intracellular signaling in tumor and immune cells and we discuss different insights regarding the therapeutic potential of MSC-derived miRNAs in cancer treatment.
Collapse
Affiliation(s)
| | | | - Vladislav Volarevic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.
- Department of Microbiology and Immunology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
26
|
Psaraki A, Zagoura D, Ntari L, Makridakis M, Nikokiraki C, Trohatou O, Georgila K, Karakostas C, Angelioudaki I, Kriebardis AG, Gramignioli R, Sakellariou S, Xilouri M, Eliopoulos AG, Vlahou A, Roubelakis MG. MFGE-8 identified in fetal mesenchymal-stromal-cell-derived exosomes ameliorates acute hepatic failure pathology. iScience 2023; 26:108100. [PMID: 37915594 PMCID: PMC10616317 DOI: 10.1016/j.isci.2023.108100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/03/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023] Open
Abstract
Liver transplantation is the gold-standard therapy for acute hepatic failure (AHF) with limitations related to organ shortage and life-long immunosuppressive therapy. Cell therapy emerges as a promising alternative to transplantation. We have previously shown that IL-10 and Annexin-A1 released by amniotic fluid human mesenchymal stromal cells (AF-MSCs) and their hepatocyte progenitor-like (HPL) or hepatocyte-like (HPL) cells induce liver repair and downregulate systemic inflammation in a CCl4-AHF mouse model. Herein, we demonstrate that exosomes (EXO) derived from these cells improve liver phenotype in CCl4-induced mice and promote oval cell proliferation. LC-MS/MS proteomic analysis identified MEFG-8 in EXO cargo that facilitates rescue of AHF by suppressing PI3K signaling. Administration of recombinant MFGE-8 protein also reduced liver damage in CCl4-induced mice. Clinically, MEFG-8 expression was decreased in liver biopsies from AHF patients. Collectively, our study provides proof-of-concept for an innovative, cell-free, less immunogenic, and non-toxic alternative strategy for AHF.
Collapse
Affiliation(s)
- Adriana Psaraki
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), Athens, Greece
- Cell and Gene Therapy Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Dimitra Zagoura
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Lydia Ntari
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Manousos Makridakis
- Biotechnology Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Christina Nikokiraki
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), Athens, Greece
- Cell and Gene Therapy Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Ourania Trohatou
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Konstantina Georgila
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Christos Karakostas
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Ioanna Angelioudaki
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Anastasios G. Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, Section of Medical Laboratories, School of Health & Caring Sciences, University of West Attica (UniWA), Ag. Spyridonos Str, 12243 Egaleo, Greece
| | - Roberto Gramignioli
- Clinical Pathology and Cancer Diagnosis Unit, Karolinska Institute, 141 57 Huddinge, Sweden
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Stratigoula Sakellariou
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Maria Xilouri
- Center of Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Aristides G. Eliopoulos
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), Athens, Greece
| | - Antonia Vlahou
- Biotechnology Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| | - Maria G. Roubelakis
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), Athens, Greece
- Cell and Gene Therapy Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), Athens, Greece
| |
Collapse
|
27
|
Ng J, Marneth AE, Griffith A, Younger D, Ghanta S, Jiao A, Willis G, Han J, Imani J, Niu B, Keegan JW, Hancock B, Guo F, Shi Y, Perrella MA, Lederer JA. Mesenchymal Stromal Cells Facilitate Neutrophil-Trained Immunity by Reprogramming Hematopoietic Stem Cells. J Innate Immun 2023; 15:765-781. [PMID: 37797588 PMCID: PMC10622164 DOI: 10.1159/000533732] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 07/20/2023] [Indexed: 10/07/2023] Open
Abstract
Novel therapeutics are urgently needed to prevent opportunistic infections in immunocompromised individuals undergoing cancer treatments or other immune-suppressive therapies. Trained immunity is a promising strategy to reduce this burden of disease. We previously demonstrated that mesenchymal stromal cells (MSCs) preconditioned with a class A CpG oligodeoxynucleotide (CpG-ODN), a Toll-like receptor 9 (TLR9) agonist, can augment emergency granulopoiesis in a murine model of neutropenic sepsis. Here, we used a chimeric mouse model to demonstrate that MSCs secrete paracrine factors that act on lineage-negative c-kit+ hematopoietic stem cells (HSCs), leaving them "poised" to enhance emergency granulopoiesis months after transplantation. Chimeric mice developed from HSCs exposed to conditioned media from MSCs and CpG-ODN-preconditioned MSCs showed significantly higher bacterial clearance and increased neutrophil granulopoiesis following lung infection than control mice. By Cleavage Under Targets and Release Using Nuclease (CUT&RUN) chromatin sequencing, we identified that MSC-conditioned media leaves H3K4me3 histone marks in HSCs at genes involved in myelopoiesis and in signaling persistence by the mTOR pathway. Both soluble factors and extracellular vesicles from MSCs mediated these effects on HSCs and proteomic analysis by mass spectrometry revealed soluble calreticulin as a potential mediator. In summary, this study demonstrates that trained immunity can be mediated by paracrine factors from MSCs to induce neutrophil-trained immunity by reprogramming HSCs for long-lasting functional changes in neutrophil-mediated antimicrobial immunity.
Collapse
Affiliation(s)
- Julie Ng
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Anna E. Marneth
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Alec Griffith
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Daniel Younger
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Sailaja Ghanta
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Alan Jiao
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Gareth Willis
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Junwen Han
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Jewel Imani
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Bailin Niu
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Joshua W. Keegan
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Brandon Hancock
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Fei Guo
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Yang Shi
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Mark A. Perrella
- Division of Pulmonary and Critical Care, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - James A. Lederer
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
28
|
Lu J, Yu N, Liu Q, Xie Y, Zhen L. Periodontal Ligament Stem Cell Exosomes Key to Regulate Periodontal Regeneration by miR-31-5p in Mice Model. Int J Nanomedicine 2023; 18:5327-5342. [PMID: 37746047 PMCID: PMC10516219 DOI: 10.2147/ijn.s409664] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/06/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Periodontitis is a chronic inflammatory disease that causes alveolar bone loss. Diabetes is one of the most important factors contributing to periodontitis. Exosomes derived from mesenchymal stem cells (MSCs-Exo) have been reported to promote bone regeneration. This study aimed to examine the function and mechanism of exosomes derived from periodontal ligament stem cells (PDLSCs-Exo) in regulating periodontal regeneration in diabetic periodontitis. Methods Exosomes derived from normal-glucose-cultured PDLSCs (NG-PDLSCs-Exo) and high-glucose-preconditioned PDLSCs (HG-PDLSCs-Exo) were used. Their effects on RAW264.7 cells were investigated by TRAP staining and quantitative real time-polymerase chain reaction (qRT-PCR). The role of exosomal miR-31-5p in osteoclast differentiation was tested using qRT-PCR, double luciferase analysis, and Western blotting. We investigated the effects of these two types of PDLSCs-Exo on alveolar bone loss in vivo in mice with experimental periodontitis. Results PDLSCs-Exo were transferred to RAW264.7, and HG-PDLSCs-Exo inhibited osteoclast formation to a lesser extent than NG-PDLSCs-Exo. Further studies revealed the effect of PDLSCs-Exo on osteoclastogenesis via the miR-31-5p/eNOS signaling pathway. In mice with experimental periodontitis, PDLSCs-Exo reduced alveolar bone destruction and decreased the number of osteoclasts on the alveolar bone surface. Conclusion Our results suggest that exosomal miR-31-5p derived from PDLSCs regulates alveolar bone regeneration by targeting eNOS.
Collapse
Affiliation(s)
- Jiuqing Lu
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, People’s Republic of China
- Department of Stomatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Nijia Yu
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Qian Liu
- Department of Stomatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Yajia Xie
- Oral Biomedical Engineering Laboratory, Shanghai Stomatological Hospital, Fudan University, Shanghai, People’s Republic of China
| | - Lei Zhen
- Department of Stomatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| |
Collapse
|
29
|
Zhong L, Wang J, Wang P, Liu X, Liu P, Cheng X, Cao L, Wu H, Chen J, Zhou L. Neural stem cell-derived exosomes and regeneration: cell-free therapeutic strategies for traumatic brain injury. Stem Cell Res Ther 2023; 14:198. [PMID: 37553595 PMCID: PMC10408078 DOI: 10.1186/s13287-023-03409-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 07/06/2023] [Indexed: 08/10/2023] Open
Abstract
Regenerative repair of the brain after traumatic brain injury (TBI) remains an extensive clinical challenge, inspiring intensified interest in therapeutic approaches to explore superior repair strategies. Exosome therapy is another research hotspot following stem cell alternative therapy. Prior research verified that exosomes produced by neural stem cells can participate in the physiological and pathological changes associated with TBI and have potential neuroregulatory and repair functions. In comparison with their parental stem cells, exosomes have superior stability and immune tolerance and lower tumorigenic risk. In addition, they can readily penetrate the blood‒brain barrier, which makes their treatment efficiency superior to that of transplanted stem cells. Exosomes secreted by neural stem cells present a promising strategy for the development of novel regenerative therapies. Their tissue regeneration and immunomodulatory potential have made them encouraging candidates for TBI repair. The present review addresses the challenges, applications and potential mechanisms of neural stem cell exosomes in regenerating damaged brains.
Collapse
Affiliation(s)
- Lin Zhong
- Department of Hematology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Jingjing Wang
- Tianjin Key Laboratory of Neurotrauma Repair, Institute of Neurotrauma Repair, Characteristic Medical Center of People's Armed Police Forces, Tianjin, 300162, China
| | - Peng Wang
- Department of Health Management, Tianjin Hospital, Tianjin, 300211, China
| | - Xiaoyin Liu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Peng Liu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xu Cheng
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Lujia Cao
- Department of Hematology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, China
| | - Hongwei Wu
- Department of Hematology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, 610500, Sichuan, China.
| | - Jing Chen
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Liangxue Zhou
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
30
|
Seim RF, Herring LE, Mordant AL, Willis ML, Wallet SM, Coleman LG, Maile R. Involvement of extracellular vesicles in the progression, diagnosis, treatment, and prevention of whole-body ionizing radiation-induced immune dysfunction. Front Immunol 2023; 14:1188830. [PMID: 37404812 PMCID: PMC10316130 DOI: 10.3389/fimmu.2023.1188830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/23/2023] [Indexed: 07/06/2023] Open
Abstract
Acute radiation syndrome (ARS) develops after exposure to high doses of ionizing radiation and features immune suppression and organ failure. Currently, there are no diagnostics to identify the occurrence or severity of exposure and there are limited treatments and preventative strategies to mitigate ARS. Extracellular vesicles (EVs) are mediators of intercellular communication that contribute to immune dysfunction across many diseases. We investigated if EV cargo can identify whole body irradiation (WBIR) exposure and if EVs promote ARS immune dysfunction. We hypothesized that beneficial EVs derived from mesenchymal stem cells (MSC-EVs) would blunt ARS immune dysfunction and might serve as prophylactic radioprotectants. Mice received WBIR (2 or 9 Gy) with assessment of EVs at 3 and 7 days after exposure. LC-MS/MS proteomic analysis of WBIR-EVs found dose-related changes as well as candidate proteins that were increased with both doses and timepoints (34 total) such as Thromboxane-A Synthase and lymphocyte cytosolic protein 2. Suprabasin and Sarcalumenin were increased only after 9 Gy suggesting these proteins may indicate high dose/lethal exposure. Analysis of EV miRNAs identified miR-376 and miR-136, which were increased up to 200- and 60-fold respectively by both doses of WBIR and select miRNAs such as miR-1839 and miR-664 were increased only with 9 Gy. WBIR-EVs (9 Gy) were biologically active and blunted immune responses to LPS in RAW264.7 macrophages, inhibiting canonical signaling pathways associated with wound healing and phagosome formation. When given 3 days after exposure, MSC-EVs slightly modified immune gene expression changes in the spleens of mice in response to WBIR and in a combined radiation plus burn injury exposure (RCI). MSC-EVs normalized the expression of certain key immune genes such as NFκBia and Cxcr4 (WBIR), Map4k1, Ccr9 and Cxcl12 (RCI) and lowered plasma TNFα cytokine levels after RCI. When given prophylactically (24 and 3 hours before exposure), MSC-EVs prolonged survival to the 9 Gy lethal exposure. Thus, EVs are important participants in ARS. EV cargo might be used to diagnose WBIR exposure, and MSC-EVs might serve as radioprotectants to blunt the impact of toxic radiation exposure.
Collapse
Affiliation(s)
- Roland F. Seim
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Laura E. Herring
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Angie L. Mordant
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Micah L. Willis
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Oral Biology, University of Florida, Gainesville, FL, United States
| | - Shannon M. Wallet
- Department of Oral Biology, University of Florida, Gainesville, FL, United States
| | - Leon G. Coleman
- Curriculum in Toxicology & Environmental Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, United States
| | - Robert Maile
- Department of Surgery, University of Florida, Gainesville, FL, United States
| |
Collapse
|
31
|
Yamaguchi M, Kashiwakura I. The Acute Radiation Syndrome-Mitigator Romiplostim and Secreted Extracellular Vesicles Improved Survival in Mice Acutely Exposed to Myelosuppressive Doses of Ionizing Radiation. Biomolecules 2023; 13:biom13050837. [PMID: 37238707 DOI: 10.3390/biom13050837] [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: 04/17/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
In cases of accidental high-dose total-body irradiation (TBI), acute radiation syndrome (ARS) can cause death. We reported that the thrombopoietin receptor agonist romiplostim (RP) has the potential to completely rescue mice exposed to lethal TBI. Extracellular vesicles (EVs) are involved in cell-to-cell communication, and the mechanism of RP action may be related to EVs that reflect the radio-mitigative information. We investigated the radio-mitigative effects of EVs on mice with severe ARS. C57BL/6 mice exposed to lethal TBI were treated with RP, and the EVs were isolated from the serum and intraperitoneally injected into other mice with severe ARS. The 30-day survival rate of lethal TBI mice drastically improved by 50-100% with the administration of EVs in the sera collected weekly from the mice in which radiation damage was alleviated and mortality was avoided by the administration of RP. Four responsive miRNAs, namely, miR-144-5p, miR-3620-5p, miR-6354, and miR-7686-5p showed significant expression changes in an array analysis. In particular, miR-144-5p was expressed only in the EVs of RP-treated TBI mice. Specific EVs may exist in the circulating blood of mice that escaped mortality with an ARS mitigator, and their membrane surface and endogenous molecules may be the key to the survival of mice with severe ARS.
Collapse
Affiliation(s)
- Masaru Yamaguchi
- Graduate School of Health Sciences, Hirosaki University, 66-1 Hon-cho, Hirosaki 036-8564, Aomori, Japan
| | - Ikuo Kashiwakura
- Graduate School of Health Sciences, Hirosaki University, 66-1 Hon-cho, Hirosaki 036-8564, Aomori, Japan
| |
Collapse
|
32
|
Hayashi Y, Nishimura K, Tanaka A, Inoue D. Extracellular vesicle-mediated remodeling of the bone marrow microenvironment in myeloid malignancies. Int J Hematol 2023; 117:821-829. [PMID: 37041345 DOI: 10.1007/s12185-023-03587-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/07/2023] [Accepted: 03/22/2023] [Indexed: 04/13/2023]
Abstract
Hematopoiesis is maintained and regulated by a bone marrow-specific microenvironment called a niche. In hematological malignancies, tumor cells induce niche remodeling, and the reconstructed niche is closely linked to disease pathogenesis. Recent studies have suggested that extracellular vesicles (EVs) secreted from tumor cells play a principal role in niche remodeling in hematological malignancies. Although EVs are emerging as potential therapeutic targets, the underlying mechanism of action remains unclear, and selective inhibition remains a challenge. This review summarizes remodeling of the bone marrow microenvironment in hematological malignancies and its contribution to pathogenesis, as well as roles of tumor-derived EVs, and provides a perspective on future research in this field.
Collapse
Affiliation(s)
- Yasutaka Hayashi
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, 6-3-7, Minatojimaminami-machi, Chuo-ku, Kobe, 650-0047, Japan.
| | - Koutarou Nishimura
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, 6-3-7, Minatojimaminami-machi, Chuo-ku, Kobe, 650-0047, Japan
| | - Atsushi Tanaka
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, 6-3-7, Minatojimaminami-machi, Chuo-ku, Kobe, 650-0047, Japan
- Laboratory of Immunology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Daichi Inoue
- Department of Hematology-Oncology, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, 6-3-7, Minatojimaminami-machi, Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
33
|
Zhao Q, Bae EH, Zhang Y, Shahsavari A, Lotey P, Lee RH, Liu F. Inhibitory Effects of Extracellular Vesicles from iPS-Cell-Derived Mesenchymal Stem Cells on the Onset of Sialadenitis in Sjögren's Syndrome Are Mediated by Immunomodulatory Splenocytes and Improved by Inhibiting miR-125b. Int J Mol Sci 2023; 24:5258. [PMID: 36982329 PMCID: PMC10049013 DOI: 10.3390/ijms24065258] [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: 01/26/2023] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Extracellular vesicles (EVs) from allogeneic-tissue-derived mesenchymal stem cells (MSCs) are promising to improve Sjögren's syndrome (SS) treatment, but their application is hindered by high variations in and limited expandability of tissue MSCs. We derived standardized and scalable MSCs from iPS cells (iMSCs) and reported that EVs from young but not aging iMSCs (iEVs) inhibited sialadenitis onset in SS mouse models. Here, we aim to determine cellular mechanisms and optimization approaches of SS-inhibitory effects of iEVs. In NOD.B10.H2b mice at the pre-disease stage of SS, we examined the biodistribution and recipient cells of iEVs with imaging, flow cytometry, and qRT-PCR. Intravenously infused iEVs accumulated in the spleen but not salivary glands or cervical lymph nodes and were mainly taken up by macrophages. In the spleen, young but not aging iEVs increased M2 macrophages, decreased Th17 cells, and changed expression of related immunomodulatory molecules. Loading miR-125b inhibitors into aging iEVs significantly improved their effects on repressing sialadenitis onset and regulating immunomodulatory splenocytes. These data indicated that young but not aging iEVs suppress SS onset by regulating immunomodulatory splenocytes, and inhibiting miR-125b in aging iEVs restores such effects, which is promising to maximize production of effective iEVs from highly expanded iMSCs for future clinical application.
Collapse
Affiliation(s)
| | | | | | | | | | - Ryang Hwa Lee
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| | - Fei Liu
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, College Station, TX 77843, USA
| |
Collapse
|
34
|
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
|
35
|
Meng Z, Liao Y, Peng Z, Zhou X, Zhou H, Nüssler AK, Liu L, Yang W. Bone Marrow Mesenchymal Stem-Cell-Derived Exosomes Ameliorate Deoxynivalenol-Induced Mice Liver Damage. Antioxidants (Basel) 2023; 12:588. [PMID: 36978835 PMCID: PMC10045494 DOI: 10.3390/antiox12030588] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/20/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Deoxynivalenol (DON) is a kind of Fusarium toxin that can cause a variety of toxic effects. DON is mainly metabolized and detoxified by the liver. When the concentration of DON exceeds the metabolic capacity of the liver, it will trigger acute or chronic damage to the liver tissue. Previous studies demonstrated that bone marrow mesenchymal stem-cell-secreted exosomes (BMSC-exos) reduce liver injury. Therefore, we issue a hypothesis that in vitro-cultured rat BMSC-secreted exos could ameliorate liver damage after 2 mg/kg bw/day of DON exposure. In total, 144 lipids were identified in BMEC-exos, including high polyunsaturated fatty acid (PUFA) levels. BMSC-exos treatment alleviated liver pathological changes and decreased levels of alanine aminotransferase, aspartate aminotransferase, inflammatory factors interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α), and lipid peroxidation. Otherwise, low or high BMSC-exos treatment obviously changes DON-induced hepatic oxylipin patterns. According to the results from our correlation network analysis, Pearson correlation coefficient analysis, and hierarchical clustering analysis, the top 10% oxidized lipids can be classified into two categories: one that was positively correlated with copper-zinc superoxide dismutase (Cu/Zn SOD) and another that was positively correlated with liver injury indicators. Altogether, BMSC-exos administration maintained normal liver function and reduced oxidative damage in liver tissue. Moreover, it could also significantly change the oxylipin profiles under DON conditions.
Collapse
Affiliation(s)
- Zitong Meng
- Hubei Key Laboratory of Food Nutrition and Safety, Department of Nutrition and Food Hygiene, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
- MOE Key Lab of Environment and Health, Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Yuxiao Liao
- Hubei Key Laboratory of Food Nutrition and Safety, Department of Nutrition and Food Hygiene, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
- MOE Key Lab of Environment and Health, Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Zhao Peng
- Hubei Key Laboratory of Food Nutrition and Safety, Department of Nutrition and Food Hygiene, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
- MOE Key Lab of Environment and Health, Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Xiaolei Zhou
- Hubei Key Laboratory of Food Nutrition and Safety, Department of Nutrition and Food Hygiene, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
- MOE Key Lab of Environment and Health, Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Huanhuan Zhou
- Hubei Key Laboratory of Food Nutrition and Safety, Department of Nutrition and Food Hygiene, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
- MOE Key Lab of Environment and Health, Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Andreas K. Nüssler
- Department of Traumatology, BG Trauma Center, University of Tübingen, Schnarrenbergstr. 95, 72076 Tübingen, Germany
| | - Liegang Liu
- Hubei Key Laboratory of Food Nutrition and Safety, Department of Nutrition and Food Hygiene, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
- MOE Key Lab of Environment and Health, Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| | - Wei Yang
- Hubei Key Laboratory of Food Nutrition and Safety, Department of Nutrition and Food Hygiene, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
- MOE Key Lab of Environment and Health, Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan 430030, China
| |
Collapse
|
36
|
Li G, Chen T, Dahlman J, Eniola‐Adefeso L, Ghiran IC, Kurre P, Lam WA, Lang JK, Marbán E, Martín P, Momma S, Moos M, Nelson DJ, Raffai RL, Ren X, Sluijter JPG, Stott SL, Vunjak‐Novakovic G, Walker ND, Wang Z, Witwer KW, Yang PC, Lundberg MS, Ochocinska MJ, Wong R, Zhou G, Chan SY, Das S, Sundd P. Current challenges and future directions for engineering extracellular vesicles for heart, lung, blood and sleep diseases. J Extracell Vesicles 2023; 12:e12305. [PMID: 36775986 PMCID: PMC9923045 DOI: 10.1002/jev2.12305] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/19/2022] [Accepted: 01/09/2022] [Indexed: 02/14/2023] Open
Abstract
Extracellular vesicles (EVs) carry diverse bioactive components including nucleic acids, proteins, lipids and metabolites that play versatile roles in intercellular and interorgan communication. The capability to modulate their stability, tissue-specific targeting and cargo render EVs as promising nanotherapeutics for treating heart, lung, blood and sleep (HLBS) diseases. However, current limitations in large-scale manufacturing of therapeutic-grade EVs, and knowledge gaps in EV biogenesis and heterogeneity pose significant challenges in their clinical application as diagnostics or therapeutics for HLBS diseases. To address these challenges, a strategic workshop with multidisciplinary experts in EV biology and U.S. Food and Drug Administration (USFDA) officials was convened by the National Heart, Lung and Blood Institute. The presentations and discussions were focused on summarizing the current state of science and technology for engineering therapeutic EVs for HLBS diseases, identifying critical knowledge gaps and regulatory challenges and suggesting potential solutions to promulgate translation of therapeutic EVs to the clinic. Benchmarks to meet the critical quality attributes set by the USFDA for other cell-based therapeutics were discussed. Development of novel strategies and approaches for scaling-up EV production and the quality control/quality analysis (QC/QA) of EV-based therapeutics were recognized as the necessary milestones for future investigations.
Collapse
Affiliation(s)
- Guoping Li
- Cardiovascular Research CenterMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Tianji Chen
- Department of Pediatrics, College of MedicineUniversity of Illinois at ChicagoChicagoIllinoisUSA
| | - James Dahlman
- Department of Biomedical EngineeringGeorgia Institute of Technology and Emory University School of MedicineAtlantaGeorgiaUSA
| | - Lola Eniola‐Adefeso
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Ionita C. Ghiran
- Department of Anesthesia and Pain MedicineBeth Israel Deaconess Medical Center, and Harvard Medical SchoolBostonMassachusettsUSA
| | - Peter Kurre
- Children's Hospital of Philadelphia, Comprehensive Bone Marrow Failure Center, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Wilbur A. Lam
- Wallace H. Coulter Department of Biomedical Engineering, Department of PediatricsEmory School of MedicineAflac Cancer and Blood Disorders Center of Children's Healthcare of Atlanta, Emory University and Georgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Jennifer K. Lang
- Department of Medicine, Division of Cardiology, Jacobs School of Medicine and Biomedical SciencesVeterans Affairs Western New York Healthcare SystemBuffaloNew YorkUSA
| | - Eduardo Marbán
- Smidt Heart InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Pilar Martín
- Centro Nacional de Investigaciones Cardiovasculares (CNIC)Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV)MadridSpain
| | - Stefan Momma
- Institute of Neurology (Edinger Institute)University HospitalGoethe UniversityFrankfurt am MainGermany
| | - Malcolm Moos
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and ResearchUnited States Food and Drug AdministrationSilver SpringMarylandUSA
| | - Deborah J. Nelson
- Department of Pharmacological and Physiological SciencesThe University of ChicagoChicagoIllinoisUSA
| | - Robert L. Raffai
- Department of Veterans Affairs, Surgical Service (112G)San Francisco VA Medical CenterSan FranciscoCaliforniaUSA
- Department of Surgery, Division of Vascular and Endovascular SurgeryUniversity of CaliforniaSan FranciscoCaliforniaUSA
| | - Xi Ren
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Joost P. G. Sluijter
- Department of Experimental Cardiology, Circulatory Health LaboratoryRegenerative Medicine Centre, UMC Utrecht, University UtrechtUtrechtThe Netherlands
| | - Shannon L. Stott
- Massachusetts General Hospital Cancer Center and Harvard Medical SchoolBostonMassachusettsUSA
| | - Gordana Vunjak‐Novakovic
- Department of Biomedical Engineering, Department of MedicineColumbia UniversityNew YorkNew YorkUSA
| | - Nykia D. Walker
- Department of Biological SciencesUniversity of Maryland Baltimore CountyBaltimoreMarylandUSA
| | - Zhenjia Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical SciencesWashington State UniversitySpokaneWashingtonUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiology, Department of Neurology and Neurosurgeryand The Richman Family Precision Medicine Center of Excellence in Alzheimer's DiseaseThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Phillip C. Yang
- Division of Cardiovascular Medicine, Department of MedicineStanford University School of MedicineStanfordCaliforniaUSA
| | - Martha S. Lundberg
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Margaret J. Ochocinska
- Division of Blood Diseases and Resources, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Renee Wong
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Guofei Zhou
- Division of Lung Diseases, National Heart, Lung, and Blood InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Stephen Y. Chan
- Pittsburgh Heart, Lung and Blood Vascular Medicine InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Division of Cardiology and Department of MedicineUniversity of Pittsburgh School of Medicine and University of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Saumya Das
- Cardiovascular Research CenterMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Prithu Sundd
- Pittsburgh Heart, Lung and Blood Vascular Medicine InstituteUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Division of Pulmonary Allergy and Critical Care Medicine and Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
37
|
Yang P, Zhang S, Yan T, Li F, Zhang S. The Therapeutic Application of Stem Cells and Their Derived Exosomes in the Treatment of Radiation-Induced Skin Injury. Radiat Res 2023; 199:182-201. [PMID: 36630584 DOI: 10.1667/rade-22-00023.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 12/05/2022] [Indexed: 01/13/2023]
Abstract
Radiation-induced skin injury (RISI) is a serious concern for nuclear accidents and cancer radiotherapy, which seriously affects the quality of life of patients. This injury differs from traditional wounds due to impaired healing and the propensity to recurrence and is divided into acute and chronic phases on the basis of the injury time. Unfortunately, there are few effective therapies for preventing or mitigating this injury. Over the last few decades, various studies have focused on the effects of stem cell-based therapies to address the tissue repair and regeneration of irradiated skin. These stem cells modulate inflammation and instigate tissue repair by differentiating into specific kinds of cells or releasing paracrine factors. Stem cell-based therapies, including bone marrow-derived stem cells (BMSCs), adipose-derived stem cells (ADSCs) and stromal vascular fraction (SVF), have been reported to facilitate wound healing after radiation exposure. Moreover, stem cell-derived exosomes have recently been suggested as an effective and cell-free approach to support skin regeneration, circumventing the concerns respecting direct application of stem cells. Based on the literature on stem cell-based therapies for radiation-induced skin injury, we summarize the characteristics of different stem cells and describe their latest animal and clinical applications, as well as potential mechanisms. The promise of stem-cell based therapies against radiation-induced skin injury contribute to our response to nuclear events and smooth progress of cancer radiotherapy.
Collapse
Affiliation(s)
- Ping Yang
- Laboratory of Radiation Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Shuaijun Zhang
- Laboratory of Radiation Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Yan
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China.,Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China
| | - Fengsheng Li
- PLA Rocket Rorce Characteristic Medical Center, Beijing 100088, China
| | - Shuyu Zhang
- Laboratory of Radiation Medicine, West China Second University Hospital, Sichuan University, Chengdu 610041, China.,Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China.,Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China.,NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang 621099, China
| |
Collapse
|
38
|
Romero-García N, Huete-Acevedo J, Mas-Bargues C, Sanz-Ros J, Dromant M, Borrás C. The Double-Edged Role of Extracellular Vesicles in the Hallmarks of Aging. Biomolecules 2023; 13:165. [PMID: 36671550 PMCID: PMC9855573 DOI: 10.3390/biom13010165] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
The exponential growth in the elderly population and their associated socioeconomic burden have recently brought aging research into the spotlight. To integrate current knowledge and guide potential interventions, nine biochemical pathways are summarized under the term hallmarks of aging. These hallmarks are deeply inter-related and act together to drive the aging process. Altered intercellular communication is particularly relevant since it explains how damage at the cellular level translates into age-related loss of function at the organismal level. As the main effectors of intercellular communication, extracellular vesicles (EVs) might play a key role in the aggravation or mitigation of the hallmarks of aging. This review aims to summarize this role and to provide context for the multiple emerging EV-based gerotherapeutic strategies that are currently under study.
Collapse
Affiliation(s)
- Nekane Romero-García
- Department of Anesthesiology and Surgical Trauma Intensive Care, Hospital Clinic Universitari Valencia, University of Valencia, 46010 Valencia, Spain
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Javier Huete-Acevedo
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Cristina Mas-Bargues
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Jorge Sanz-Ros
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
- Cardiology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain
| | - Mar Dromant
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| | - Consuelo Borrás
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable-Instituto de Salud Carlos III (CIBERFES-ISCIII), INCLIVA, 46010 Valencia, Spain
| |
Collapse
|
39
|
Almeria C, Kreß S, Weber V, Egger D, Kasper C. Heterogeneity of mesenchymal stem cell-derived extracellular vesicles is highly impacted by the tissue/cell source and culture conditions. Cell Biosci 2022; 12:51. [PMID: 35501833 PMCID: PMC9063275 DOI: 10.1186/s13578-022-00786-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/10/2022] [Indexed: 12/19/2022] Open
Abstract
AbstractExtracellular vesicles (EVs) are cell-derived membrane structures exerting major effects in physiological as well as pathological processes by functioning as vehicles for the delivery of biomolecules to their target cells. An increasing number of effects previously attributed to cell-based therapies have been recognized to be actually mediated by EVs derived from the respective cells, suggesting the administration of purified EVs instead of living cells for cell-based therapies. In this review, we focus on the heterogeneity of EVs derived from mesenchymal stem/stromal cells (MSC) and summarize upstream process parameters that crucially affect the resulting therapeutic properties and biological functions. Hereby, we discuss the effects of the cell source, medium composition, 3D culture, bioreactor culture and hypoxia. Furthermore, aspects of the isolation and storage strategies influences EVs are described. Conclusively, optimization of upstream process parameters should focus on controlling MSC-derived EV heterogeneity for specific therapeutic applications.
Graphical Abstract
Collapse
|
40
|
Vanhie JJ, Kim W, Ek Orloff L, Ngu M, Collao N, De Lisio M. The role of exercise-and high fat diet-induced bone marrow extracellular vesicles in stress hematopoiesis. Front Physiol 2022; 13:1054463. [PMID: 36505084 PMCID: PMC9728614 DOI: 10.3389/fphys.2022.1054463] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
Exercise and obesity regulate hematopoiesis, in part through alterations in cellular and soluble components of the bone marrow niche. Extracellular vesicles (EVs) are components of the bone marrow niche that regulate hematopoiesis; however, the role of exercise training or obesity induced EVs in regulating hematopoiesis remains unknown. To address this gap, donor EVs were isolated from control diet-fed, sedentary mice (CON-SED), control diet-fed exercise trained mice (CON-EX), high fat diet-fed, sedentary mice (HFD-SED), and high fat diet-fed, exercise trained mice (HFD-EX) and injected into recipient mice undergoing stress hematopoiesis. Hematopoietic and niche cell populations were quantified, and EV miRNA cargo was evaluated. EV content did not differ between the four groups. Mice receiving HFD-EX EVs had fewer hematopoietic stem cells (HSCs) (p < 0.01), long-term HSC (p < 0.05), multipotent progenitors (p < 0.01), common myeloid progenitors (p<0.01), common lymphoid progenitors (p < 0.01), and granulocyte-macrophage progenitors (p < 0.05), compared to mice receiving HFD-SED EVs. Similarly, mice receiving EX EVs had fewer osteoprogenitor cells compared to SED (p < 0.05) but enhanced mesenchymal stromal cell (MSC) osteogenic differentiation in vitro (p < 0.05) compared to SED EVs. HFD EVs enhanced mesenchymal stromal cell (MSC) adipogenesis in vitro (p < 0.01) compared to CON EVs. HFD-EX EVs had lower microRNA-193 and microRNA-331-5p content, microRNAs implicated in inhibiting osteogenesis and leukemic cell expansion respectively, compared to HFD-SED EVs. The results identify alterations in EV cargo as a novel mechanism by which exercise training alters stress hematopoiesis and the bone marrow niche.
Collapse
Affiliation(s)
- James J. Vanhie
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Wooseok Kim
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Lisa Ek Orloff
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Matthew Ngu
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Nicolas Collao
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada
| | - Michael De Lisio
- School of Human Kinetics, Faculty of Health Sciences, Ottawa, ON, Canada,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada,*Correspondence: Michael De Lisio,
| |
Collapse
|
41
|
Liu Z, Wang S, Huo N, Yang S, Shi Q, Xu J. Extracellular vesicles: A potential future strategy for dental and maxillofacial tissue repair and regeneration. Front Physiol 2022; 13:1012241. [PMID: 36479350 PMCID: PMC9719951 DOI: 10.3389/fphys.2022.1012241] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/09/2022] [Indexed: 06/18/2024] Open
Abstract
Extracellular vesicles (EVs), nano-sized bilayer membrane structures containing lipids, proteins and nucleic acids, play key roles in intercellular communication. Compared to stem cells, EVs have lower tumorigenicity and immunogenicity, are easier to manage and cause fewer ethic problems. In recent years, EVs have emerged as a potential solution for tissue regeneration in stomatology through cell-free therapies. The present review focuses on the role of EVs in dental and maxillofacial tissue repair and regeneration, including in dental and periodontal tissue, maxilla and mandible bone, temporomandibular joint cartilage, peripheral nerve and soft tissue. We also make a brief overview on the mechanism of EVs performing functions. However, limitations and challenges in clinical application of EVs still exist and should be addressed in future researches.
Collapse
Affiliation(s)
- Ziwei Liu
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Orthopedic Laboratory of PLA General Hospital, Beijing, China
| | - Situo Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Orthopedic Laboratory of PLA General Hospital, Beijing, China
| | - Na Huo
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shuo Yang
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Quan Shi
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Juan Xu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
42
|
Liu C, Li Y, Han G. Advances of Mesenchymal Stem Cells Released Extracellular Vesicles in Periodontal Bone Remodeling. DNA Cell Biol 2022; 41:935-950. [PMID: 36315196 DOI: 10.1089/dna.2022.0359] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles that include exosomes, microvesicles, and apoptotic bodies; they interact with target cell surface receptors and transport contents, including mRNA, proteins, and enzymes into the cytoplasm of target cells to function. The biological fingerprints of EVs practically mirror those of the parental cells they originated from. In the bone remodeling microenvironment, EVs could act on osteoblasts to regulate the bone formation, promote osteoclast differentiation, and regulate bone resorption. Therefore, there have been many attempts wherein EVs were used to achieve targeted therapy in bone-related diseases. Periodontitis, a common bacterial infectious disease, could cause severe alveolar bone resorption, resulting in tooth loss, whereas research on periodontal bone regeneration is also an urgent question. Therefore, EVs-related studies are important for periodontal bone remodeling. In this review, we summarize the current knowledge of mesenchymal stem cell-EVs involved in periodontal bone remodeling and explore the functional gene expression through a comparative analysis of transcriptomic content.
Collapse
Affiliation(s)
- Chaoran Liu
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Yanan Li
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| | - Guanghong Han
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, People's Republic of China
| |
Collapse
|
43
|
Tang H, He Y, Liang Z, Li J, Dong Z, Liao Y. The therapeutic effect of adipose-derived stem cells on soft tissue injury after radiotherapy and their value for breast reconstruction. Stem Cell Res Ther 2022; 13:493. [PMID: 36195925 PMCID: PMC9531407 DOI: 10.1186/s13287-022-02952-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/08/2022] [Indexed: 12/24/2022] Open
Abstract
Background Postmastectomy radiotherapy is considered to be a necessary treatment in the therapy of breast cancer, while it will cause soft tissue damage and complications, which are closely related to the success rate and effectiveness of breast reconstruction. After radiotherapy, cutaneous tissue becomes thin and brittle, and its compliance decreases. Component fat grafting and adipose-derived stem cell therapy are considered to have great potential in treating radiation damage and improving skin compliance after radiotherapy. Main body In this paper, the basic types and pathological mechanisms of skin and soft tissue damage to breast skin caused by radiation therapy are described. The 2015–2021 studies related to stem cell therapy in PubMed were also reviewed. Studies suggest that adipose-derived stem cells exert their biological effects mainly through cargoes carried in extracellular vesicles and soluble secreted factors. Compared to traditional fat graft breast reconstruction, ADSC therapy amplifies the effects of stem cells in it. In order to obtain a more purposeful therapeutic effect, proper stem cell pretreatment may achieve more ideal and safe results. Conclusion Recent research works about ADSCs and other MSCs mainly focus on curative effects in the acute phase of radiation injury, and there is little research about treatment of chronic phase complications. The efficacy of stem cell therapy on alleviating skin fibrosis and its underlying mechanism require further research.
Collapse
Affiliation(s)
- Haojing Tang
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Yufei He
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Zhuokai Liang
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Jian Li
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China
| | - Ziqing Dong
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China.
| | - Yunjun Liao
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
44
|
Pozzobon M, D’Agostino S, Roubelakis MG, Cargnoni A, Gramignoli R, Wolbank S, Gindraux F, Bollini S, Kerdjoudj H, Fenelon M, Di Pietro R, Basile M, Borutinskaitė V, Piva R, Schoeberlein A, Eissner G, Giebel B, Ponsaerts P. General consensus on multimodal functions and validation analysis of perinatal derivatives for regenerative medicine applications. Front Bioeng Biotechnol 2022; 10:961987. [PMID: 36263355 PMCID: PMC9574482 DOI: 10.3389/fbioe.2022.961987] [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: 06/05/2022] [Accepted: 07/01/2022] [Indexed: 11/26/2022] Open
Abstract
Perinatal tissues, such as placenta and umbilical cord contain a variety of somatic stem cell types, spanning from the largely used hematopoietic stem and progenitor cells to the most recently described broadly multipotent epithelial and stromal cells. As perinatal derivatives (PnD), several of these cell types and related products provide an interesting regenerative potential for a variety of diseases. Within COST SPRINT Action, we continue our review series, revising and summarizing the modalities of action and proposed medical approaches using PnD products: cells, secretome, extracellular vesicles, and decellularized tissues. Focusing on the brain, bone, skeletal muscle, heart, intestinal, liver, and lung pathologies, we discuss the importance of potency testing in validating PnD therapeutics, and critically evaluate the concept of PnD application in the field of tissue regeneration. Hereby we aim to shed light on the actual therapeutic properties of PnD, with an open eye for future clinical application. This review is part of a quadrinomial series on functional/potency assays for validation of PnD, spanning biological functions, such as immunomodulation, anti-microbial/anti-cancer, anti-inflammation, wound healing, angiogenesis, and regeneration.
Collapse
Affiliation(s)
- Michela Pozzobon
- Department of Women’s and Children’s Health, University of Padova, Padova, Italy
| | - Stefania D’Agostino
- Department of Women’s and Children’s Health, University of Padova, Padova, Italy
| | - Maria G. Roubelakis
- Laboratory of Biology, Medical School of Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Cargnoni
- Centro di Ricerca E. Menni, Fondazione Poliambulanza Istituto Ospedaliero, Brescia, Italy
| | - Roberto Gramignoli
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Susanne Wolbank
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, The Research Center in Cooperation with AUVA Trauma Research Center, Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Florelle Gindraux
- Service de Chirurgie Orthopédique, Traumatologique et plastique, CHU Besançon, Laboratoire de Nanomédecine, Imagerie, Thérapeutique EA 4662, University Bourgogne Franche-Comté, Besançon, France
| | - Sveva Bollini
- Department of Experimental Medicine (DIMES), School of Medical and Pharmaceutical Sciences, University of Genova, Genova, Italy
| | - Halima Kerdjoudj
- University of Reims Champagne Ardenne, EA 4691 BIOS “Biomatériaux et Inflammation en Site Osseux”, UFR d’Odontologie, Reims, France
| | | | - Roberta Di Pietro
- Department of Medicine and Ageing Sciences, Section of Biomorphology, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Mariangela Basile
- Department of Medicine and Ageing Sciences, Section of Biomorphology, G. d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Veronika Borutinskaitė
- Department of Molecular Cell Biology, Institute of Biochemistry, Vilnius University, Vilnius, Lithuania
| | - Roberta Piva
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Andreina Schoeberlein
- Department of Obstetrics and Feto-maternal Medicine, Inselspital, Bern University Hospital, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Guenther Eissner
- Systems Biology Ireland, School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
45
|
Abstract
Lifestyle factors are modifiable behavioral factors that have a significant impact on health and longevity. Diet-induced obesity and physical activity/exercise are two prevalent lifestyle factors that have strong relationships to overall health. The mechanisms linking obesity to negative health outcomes and the mechanisms linking increased participation in physical activity/exercise to positive health outcomes are beginning to be elucidated. Chronic inflammation, due in part to overproduction of myeloid cells from hematopoietic stem cells (HSCs) in the bone marrow, is an established mechanism responsible for the negative health effects of obesity. Recent work has shown that exercise training can reverse the aberrant myelopoiesis present in obesity in part by restoring the bone marrow microenvironment. Specifically, exercise training reduces marrow adipose tissue, increases HSC retention factor expression, and reduces pro-inflammatory cytokine levels in the bone marrow. Other, novel mechanistic factors responsible for these exercise-induced effects, including intercellular communication using extracellular vesicles (EVs), is beginning to be explored. This review will summarize the recent literature describing the effects of exercise on hematopoiesis in individuals with obesity and introduce the potential contribution of EVs to this process.
Collapse
|
46
|
Sun G, Gu Q, Zheng J, Cheng H, Cheng T. Emerging roles of extracellular vesicles in normal and malignant hematopoiesis. J Clin Invest 2022; 132:160840. [PMID: 36106632 PMCID: PMC9479752 DOI: 10.1172/jci160840] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hematopoietic stem cells, regulated by their microenvironment (or “niche”), sustain the production of mature blood and immune cells. Leukemia cells remodel the microenvironment to enhance their survival, which is accompanied by the loss of support for normal hematopoiesis in hematologic malignancies. Extracellular vesicles (EVs) mediate intercellular communication in physiological and pathological conditions, and deciphering their functions in cell-cell interactions in the ecosystem can highlight potential therapeutic targets. In this Review, we illustrate the utility of EVs derived from various cell types, focusing on the biological molecules they contain and the behavioral alterations they can induce in recipient cells. We also discuss the potential for clinical application in hematologic malignancies, including EV-based therapeutic regimens, drug delivery via EVs, and the use of EVs (or their cargoes) as biomarkers.
Collapse
Affiliation(s)
- Guohuan Sun
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Quan Gu
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Junke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Cheng
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
- Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
- Department of Stem Cell and Regenerative Medicine, Peking Union Medical College, Tianjin, China
| |
Collapse
|
47
|
Lin Z, Wu Y, Xu Y, Li G, Li Z, Liu T. Mesenchymal stem cell-derived exosomes in cancer therapy resistance: recent advances and therapeutic potential. Mol Cancer 2022; 21:179. [PMID: 36100944 PMCID: PMC9468526 DOI: 10.1186/s12943-022-01650-5] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 08/30/2022] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stromal cells that can be obtained from various human tissues and organs. They can differentiate into a wide range of cell types, including osteoblasts, adipocytes and chondrocytes, thus exhibiting great potential in regenerative medicine. Numerous studies have indicated that MSCs play critical roles in cancer biology. The crosstalk between tumour cells and MSCs has been found to regulate many tumour behaviours, such as proliferation, metastasis and epithelial-mesenchymal transition (EMT). Multiple lines of evidence have demonstrated that MSCs can secrete exosomes that can modulate the tumour microenvironment and play important roles in tumour development. Notably, very recent works have shown that mesenchymal stem cell-derived exosomes (MSC-derived exosomes) are critically involved in cancer resistance to chemotherapy agents, targeted-therapy drugs, radiotherapy and immunotherapy. In this review, we systematically summarized the emerging roles and detailed molecular mechanisms of MSC-derived exosomes in mediating cancer therapy resistance, thus providing novel insights into the clinical applications of MSC-derived exosomes in cancer management.
Collapse
|
48
|
Determination of the Loading Capacity and Recovery of Extracellular Vesicles Derived from Human Embryonic Kidney Cells and Urine Matrices on Capillary-Channeled Polymer (C-CP) Fiber Columns. SEPARATIONS 2022. [DOI: 10.3390/separations9090251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles (EVs) are 50–1000 nm membranous vesicles secreted from all cells that play important roles in many biological processes. Exosomes, a smaller-sized subset of EVs, have become of increasing interest in fundamental biochemistry and clinical fields due to their rich biological cargos and their roles in processes such as cell-signaling, maintaining homeostasis, and regulating cellular functions. To be implemented effectively in fundamental biochemistry and clinical diagnostics fields of study, and for their proposed use as vectors in gene therapies, there is a need for new methods for the isolation of large concentrations of high-purity exosomes from complex matrices in a timely manner. To address current limitations regarding recovery and purity, described here is a frontal throughput and recovery analysis of exosomes derived from human embryonic kidney (HEK) cell cultures and human urine specimens using capillary-channeled polymer (C-CP) fiber stationary phases via high performance liquid chromatography (HPLC). Using the C-CP fiber HPLC method for EV isolations, the challenge of recovering purified EVs from small sample volumes imparted by the traditional techniques was overcome while introducing significant benefits in processing, affordability (~5 $ per column), loading (~1012 particles), and recovery (1011–1012 particles) from whole specimens without further processing requirements.
Collapse
|
49
|
Srivastava J, Katiyar S, Chaturvedi CP, Nityanand S. Extracellular vesicles from bone marrow mesenchymal stromal cells of severe aplastic anemia patients attenuate hematopoietic functions of CD34 + hematopoietic stem and progenitor cells. Cell Biol Int 2022; 46:1970-1976. [PMID: 35998254 DOI: 10.1002/cbin.11885] [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: 10/05/2021] [Revised: 05/30/2022] [Accepted: 07/25/2022] [Indexed: 11/07/2022]
Abstract
Mesenchymal stromal cells (MSC) regulate hematopoiesis in the bone marrow (BM) niche and extracellular vesicles (EVs) released by BM-MSC are important mediators of the cross-talk between BM-MSC and hematopoietic stem and progenitor cells (HSPC). We have previously demonstrated that BM-MSC of severe aplastic anemia (SAA) patients have an altered expression of hematopoiesis regulatory molecules. In the present study, we observed that CD34+ HSPC when cocultured with BM-MSC EVs from aplastic anemia patients exhibited a significant reduction in colony-forming units (p = .001), cell proliferation (p = .002), and increased apoptosis (p > .001) when compared to coculture with BM-MSC EVs from controls. Collectively, our results highlight that EVs derived from the BM-MSC of SAA patients impair the hematopoiesis supporting function of HSPC.
Collapse
Affiliation(s)
- Jyotika Srivastava
- Department of Hematology and Stem Cell Research Centre, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Shobhita Katiyar
- Department of Hematology and Stem Cell Research Centre, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Chandra P Chaturvedi
- Department of Hematology and Stem Cell Research Centre, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Soniya Nityanand
- Department of Hematology and Stem Cell Research Centre, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| |
Collapse
|
50
|
Human Mesenchymal Stromal Cells Do Not Cause Radioprotection of Head-and-Neck Squamous Cell Carcinoma. Int J Mol Sci 2022; 23:ijms23147689. [PMID: 35887032 PMCID: PMC9323822 DOI: 10.3390/ijms23147689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/03/2022] [Accepted: 07/09/2022] [Indexed: 02/01/2023] Open
Abstract
Radiotherapy of head-and-neck squamous cell carcinoma (HNSCC) can cause considerable normal tissue injuries, and mesenchymal stromal cells (MSCs) have been shown to aid regeneration of irradiation-damaged normal tissues. However, utilization of MSC-based treatments for HNSCC patients undergoing radiotherapy is hampered by concerns regarding potential radioprotective effects. We therefore investigated the influence of MSCs on the radiosensitivity of HNSCCs. Several human papillomavirus (HPV)-negative and HPV-positive HNSCCs were co-cultured with human bone marrow-derived MSCs using two-dimensional and three-dimensional assays. Clonogenic survival, proliferation, and viability of HNSCCs after radiotherapy were assessed depending on MSC co-culture. Flow cytometry analyses were conducted to examine the influence of MSCs on irradiation-induced cell cycle distribution and apoptosis induction in HNSCCs. Immunofluorescence stainings of γH2AX were conducted to determine the levels of residual irradiation-induced DNA double-strand breaks. Levels of connective tissue growth factor (CTGF), a multifunctional pro-tumorigenic cytokine, were analyzed using enzyme-linked immunosorbent assays. Neither direct MSC co-culture nor MSC-conditioned medium exerted radioprotective effects on HNSCCs as determined by clonogenic survival, proliferation, and viability assays. Consistently, three-dimensional microwell arrays revealed no radioprotective effects of MSCs. Irradiation resulted in a G2/M arrest of HNSCCs at 96 h independently of MSC co-culture. HNSCCs’ apoptosis rates were increased by irradiation irrespective of MSCs. Numbers of residual γH2AX foci after irradiation with 2 or 8 Gy were comparable between mono- and co-cultures. MSC mono-cultures and HNSCC-MSC co-cultures exhibited comparable CTGF levels. We did not detect radioprotective effects of human MSCs on HNSCCs. Our results suggest that the usage of MSC-based therapies for radiotherapy-related toxicities in HNSCC patients may be safe in the context of absent radioprotection.
Collapse
|