1
|
Huang D, Huang W, Liu M, Chen J, Xiao D, Peng Z, He H, Shen H, Jin Q, Chen L, Rao D, Zhao M, Huang J. Progress of mesenchymal stem cell-derived exosomes in targeted delivery of antitumor drugs. Cancer Cell Int 2025; 25:169. [PMID: 40301903 DOI: 10.1186/s12935-025-03795-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 04/17/2025] [Indexed: 05/01/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are currently being used in clinical trials for the treatment of a wide range of diseases and have a wide range of applications in the fields of tissue engineering and regeneration. Exosomes are extracellular vesicles containing a variety of components such as proteins, nucleic acids and lipids, which are widely present in biological fluids and have the functions of participating in intercellular information transfer, immune response and tissue repair, and can also be used as carriers to target and deliver tumors to improve therapeutic effects. Mesenchymal stem cell-derived Exosomes (MSC-Exos), which have the advantages of low immunogenicity and high tumor homing ability, have attracted much attention in targeted drug delivery. Here, we review the current knowledge on the involvement of MSC-Exos in tumor progression and their potential as drug delivery systems in targeted therapies. It also discusses the advantages and prospects of MSC-Exos as a drug carrier and the challenges that still need to be overcome.
Collapse
Affiliation(s)
- Defa Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Wenlong Huang
- Department of General Medicine, First People's Hospital of Zunyi (Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, China
| | - Meijin Liu
- People's Hospital of Ganzhou Economic Development Zone, Ganzhou, 341000, China
| | - Jie Chen
- Department of Laboratory Medicine, the Affiliated Yongchuan Hospital of Chongqing Medical University, Yongchuan, Chongqing, 402177, China
| | - Dewang Xiao
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
| | - Zongbo Peng
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
| | - Haoquan He
- Department of General Practice, Ditian Community health centre, Jinhua jindong, xiaoshun, 321000, China
| | - Haibin Shen
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Qing Jin
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Linli Chen
- Laboratory Medicine, Guizhou Aerospace Hospital, Zunyi, 563100, China
| | - Dingyu Rao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| | - Minghong Zhao
- Laboratory Medicine, Guizhou Aerospace Hospital, Zunyi, 563100, China.
| | - Junyun Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
2
|
Bakr MM, Al Ankily M, Shamel M. The Protective Effects of MSC-Derived Exosomes Against Chemotherapy-Induced Parotid Gland Cytotoxicity. Int J Dent 2025; 2025:5517092. [PMID: 40223864 PMCID: PMC11986938 DOI: 10.1155/ijod/5517092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Fluorouracil (5-FU) is one of the most popular chemotherapeutic agents used in various cancer therapy protocols. Cell-free therapy utilizing exosomes is gaining increased popularity as a safer option due to concerns over potential tumor progression following stem cell therapy. Methods: Parotid glands of albino were treated with a single bone marrow mesenchymal stem cell (BMMSC)-derived exosomes injection (100 μg/kg/dose suspended in 0.2 mL phosphate-buffered saline [PBS]), a single 5-Fu injection (20 mg/kg), and BMMSC-derived exosomes plus 5-FU and compared to control group (daily saline injections). After 30 days, the parotid glands were examined using qualitative histological evaluation, immunohistochemical evaluation using rabbit polyclonal mouse antibody to Ki-67, caspase 3, and iNOS, as well as quantitative real-time polymerase chain reaction (RT-PCR) to evaluate gene expression of TGFβ1, TNF-α, and BCL-2. Results: Histological examination of the parotid gland revealed that BMMSC-derived exosomes restored the glands' architecture and repaired most of the distortion created by 5-FU. Immunohistochemical expression of tumor proliferation and cell death markers were restored to normal levels in the exosome-treated groups that were similar to the control group. Furthermore, BMMSC-derived exosomes reversed the effects of 5-FU on quantitative gene expression levels and showed a significant decrease in TNF-α (p < 0.001) and a significant increase in TGFβ (p < 0.0001) and BCL-2 (p < 0.05) when compared to 5-FU treatment. Conclusion: Within the limitations of the current study, BMMSC-derived exosomes have the potential to counteract the cytotoxic effects of 5-FU on the parotid glands of rats in vivo. Further studies are deemed necessary to simulate clinical scenarios.
Collapse
Affiliation(s)
- Mahmoud M. Bakr
- General Dental Practice, School of Medicine and Dentistry, Griffith University, Gold Coast, Queensland 4215, Australia
| | - Mahmoud Al Ankily
- Faculty of Dentistry, Oral Biology Department, The British University in Egypt, Cairo, Egypt
| | - Mohamed Shamel
- Faculty of Dentistry, Oral Biology Department, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
3
|
Wang X, Xu Z, Wang J, Wu C, Zhang L, Qian C, Luo Y, Gu Y, Wong W, Xiang D. A Mitochondria-Targeted Biomimetic Nanomedicine Capable of Reversing Drug Resistance in Colorectal Cancer Through Mitochondrial Dysfunction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410630. [PMID: 39943694 PMCID: PMC11967798 DOI: 10.1002/advs.202410630] [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: 09/02/2024] [Revised: 01/27/2025] [Indexed: 04/05/2025]
Abstract
Chemoresistance and metastasis are the main obstacles to the clinical success of anticancer treatment and are responsible for most cancer deaths. Developing effective approaches to reverse chemoresistance and inhibit metastasis is essential for efficient chemotherapy. Mitochondria are important sources of cellular energy and are involved in mediating chemoresistance and driving tumor metastasis. Due to the relatively weak DNA repair capacity of mitochondria, targeting mitochondria may reverse chemoresistance and provide a paradigm for metastatic cancer treatment. Herein, exosomes (Exos) modified with integrin ligands and mitochondriotropic molecules are synthesized for encapsulating oxaliplatin (OXA) to construct a sequentially targeted and mitochondrion-dysfunctional nanodrug (OXA@Exo-RD). Subsequent investigations confirm that OXA@Exo-RD targeted cancer cells and mitochondria in sequence, and OXA delivered to mitochondria lacking DNA repair mechanisms reduce the likelihood of deactivation. Furthermore, the OXA@Exo-RD promotes the overproduction of ROS, inhibited ATP generation, and induces mitochondria-mediated apoptosis and mitochondrial dysregulation. Finally, OXA@Exo-RD shows the potential to inhibit the growth and metastasis of HCT116/OXA cells in vitro, which is further validated in subcutaneous and orthotopic CRC models, as well as in CRC metastasis models. Taken together, this dual-targeting nanomedicine induces apoptosis via mitochondrial signaling pathways, providing an attractive strategy for the treatment of drug-resistant CRC.
Collapse
Affiliation(s)
- Xiaohui Wang
- Department of OncologyChongqing University Jiangjin HospitalChongqing402260China
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityHong KongSARChina
| | - Zhiyuan Xu
- Department of Electrical and Electronic EngineeringThe Hong Kong Polytechnic UniversityHong KongSARChina
| | - Jian Wang
- Department of OncologyChongqing University Jiangjin HospitalChongqing402260China
| | - Chunrong Wu
- Department of OncologyChongqing University Jiangjin HospitalChongqing402260China
| | - Lin Zhang
- Department of GastroenterologyChongqing University Jiangjin HospitalChongqing402260China
| | - Chengyuan Qian
- Department of OncologyChongqing University Jiangjin HospitalChongqing402260China
| | - Yang Luo
- Department of Laboratory Medicine, Chongqing General HospitalSchool of MedicineChongqing UniversityChongqing401147China
| | - Yanjuan Gu
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityHong KongSARChina
| | - Wing‐Tak Wong
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityHong KongSARChina
| | - Debing Xiang
- Department of OncologyChongqing University Jiangjin HospitalChongqing402260China
| |
Collapse
|
4
|
Alahmari LA, Ali LS, Fansa HA, Alshaya DS, Al-Salmi FA, El-Hallous EI, Eldesoqui M, Gad Elsaid F, Fayad E, El-Mansy AA, Alsharif G, Khalil DY, Mahmood MB, Khalil RY, Rashwan HM, El-Sawah SG. Antioxidant and Antiapoptotic Effects of Selenium And Nano Selenium-Loaded Exosomes on Hepatic Dysfunction of Type 1 Diabetic Rats. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2025; 343:211-219. [PMID: 39535481 DOI: 10.1002/jez.2881] [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: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Mesenchymal stem cells-derived exosomes (MSCs-EXs) applications have brought a key breakthrough in treating type 1 diabetes mellitus (T1DM) and its diabetic complications. However, various recent strategies aimed to construct prominent engineered EXs with greater precision and higher efficiency for diabetes syndrome were conducted. In this research, we seek to enhance the medicinal potentialities of MSCs-EXs on type 1 diabetic rats' hepatic complications, via loading with either selenium (Se) or nano selenium (NSe) particles. For consecutive 4-weeks, rats were divided into 8 groups as; control, EXs, EXs + Se, EXs + NSe, STZ-diabetic (D), D + EXs, D + EXs + Se, and D + EXs + NSe groups. The three diabetic-treated groups manifested a significant reduction in hepatic contents of oxidative stress (OS) (MDA, NO, and H2O2) inflammatory (IL-6, TNF-α, and TGF-β), and apoptotic (P53, BAX, caspase-3, and Bcl2) markers, with marked elevation in hepatic antioxidant levels (GSH, GPX, SOD, and CAT). Such results were supported by the marked diminish in serum total proteins, liver function enzymes (AST, ALT, and bilirubin), and both serum and liver lipid profile fractions. In addition, hepatic histological examination showed marked improvement in liver architecture of all treated diabetic rats' groups, compared to diabetic untreated rats. Significantly, diabetic rats with EXs loaded with NSe exhibited the most therapeutic superiority.
Collapse
Affiliation(s)
- Layla A Alahmari
- Department of Community Health, College of Applied Medical Sciences, Northern Border University, Arar, Saudi Arabia
| | - Lashin S Ali
- Department of Basic Medical Science, Faculty of Dentistry, Al-Ahliyya Amman University, Amman, Jordan
- Physiology Department, Faculty of Medicine, Mansoura University, Mansours, Egypt
| | - Hoda A Fansa
- Department of Basic Dental Sciences, Faculty of Dentistry, Al-Ahliyya Amman University, Amman, Jordan
- Department of Oral Biology, Faculty of Dentistry, Alexandria University, Egypt
| | - Dalal S Alshaya
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Fawziah A Al-Salmi
- Department of Biology, College of Science, Taif University, Taif, Saudi Arabia
| | - Ehab I El-Hallous
- Department of Biology, College of Science, Taif University, Taif, Saudi Arabia
- Zoology Department, Faculty of Science, Arish University, North Sinai, Egypt
| | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah, Riyadh, Saudi Arabia
| | - Fahmy Gad Elsaid
- Department of Biology, College of Science, King Khalid University, Asir, Abha, Saudi Arabia
| | - Eman Fayad
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Ahmed A El-Mansy
- Department of Basic Medical and Dental Sciences, Faculty of Dentistry, Zarqa University, Zarqa, Jordan
- Department of Medical Histology & Cell Biology, Faculty of Medicine, Mansoura University, Mansours, Egypt
| | - Ghadi Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University of Health Sciences, Jeddah, Saudi Arabia
- Department of Biomedical Research, King Abdullah International Medical Research Center, Jeddah, Saudi Arabia
| | - Dlovan Y Khalil
- Biology Department, Faculty of Science, Sulaimani University, Sulaimaniyah, Iraq
| | - Maryam Bakir Mahmood
- Obstetrics & Gynecology Department, College of Medicine, Slaimani University, Sulaymaniyah, Iraq
| | - Rozhan Yassin Khalil
- Obstetrics & Gynecology Department, College of Medicine, Slaimani University, Sulaymaniyah, Iraq
| | - Hanan M Rashwan
- Zoology Department, Faculty of Science, Arish University, North Sinai, Egypt
| | - Shady G El-Sawah
- Zoology Department, Faculty of Science, Arish University, North Sinai, Egypt
| |
Collapse
|
5
|
Li L, Yao Z, Salimian KJ, Kong J, Zaheer A, Parian A, Gearhart SL, Mao HQ, Selaru FM. Extracellular Vesicles Delivered by a Nanofiber-Hydrogel Composite Enhance Healing In Vivo in a Model of Crohn's Disease Perianal Fistula. Adv Healthc Mater 2025; 14:e2402292. [PMID: 39240055 PMCID: PMC11882933 DOI: 10.1002/adhm.202402292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/24/2024] [Indexed: 09/07/2024]
Abstract
Perianal fistulas represent a common, aggressive, and disabling complication of Crohn's disease (CD). Despite recent drug developments, novel surgical interventions as well as multidisciplinary treatment approaches, the outcome is dismal, with >50% therapy failure rates. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) offer potential therapeutic benefits for treating fistulizing CD, due to the pro-regenerative paracrine signals. However, a significant obstacle to clinical translation of EV-based therapy is the rapid clearance and short half-life of EVs in vivo. Here, an injectable, biodegradable nanofiber-hydrogel composite (NHC) microgel matrix that serves as a carrier to deliver MSC-derived EVs to a rat model of CD perianal fistula (PAF) is reported. It is found that EV-loaded NHC (EV-NHC) yields the best fistula healing when compared to other treatment arms. The MRI assessment reveals that the EV-NHC reduces inflammation at the fistula site and promotes tissue healing. The enhanced therapeutic outcomes are contributed by extended local retention and sustained release of EVs by NHC. In addition, the EV-NHC effectively reduces inflammation at the fistula site and promotes tissue healing and regeneration via macrophage polarization and neo-vascularization. This EV-NHC platform provides an off-the-shelf solution that facilitates its clinical translation.
Collapse
Affiliation(s)
- Ling Li
- Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University; Baltimore, Maryland, USA
| | - Zhicheng Yao
- Institute for NanoBioTechnology, Johns Hopkins University; Baltimore, Maryland, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University; Baltimore, Maryland, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine; Baltimore, Maryland, USA
| | - Kevan J. Salimian
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jiayuan Kong
- Institute for NanoBioTechnology, Johns Hopkins University; Baltimore, Maryland, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University; Baltimore, Maryland, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine; Baltimore, Maryland, USA
| | - Atif Zaheer
- Department of Radiology & Radiological Sciences, School of Medicine, Johns Hopkins University; Baltimore, Maryland, USA
| | - Alyssa Parian
- Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University; Baltimore, Maryland, USA
| | - Susan L. Gearhart
- Division of Colorectal Surgery, Department of Surgery, Johns Hopkins University School of Medicine; Baltimore, Maryland, USA
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University; Baltimore, Maryland, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University; Baltimore, Maryland, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine; Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine; Baltimore, Maryland, USA
| | - Florin M. Selaru
- Division of Gastroenterology and Hepatology, School of Medicine, Johns Hopkins University; Baltimore, Maryland, USA
- Institute for NanoBioTechnology, Johns Hopkins University; Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine; Baltimore, Maryland, USA
- Department of Oncology, Sidney Kimmel Cancer Center, School of Medicine, Johns Hopkins University; Baltimore, Maryland, USA
| |
Collapse
|
6
|
Sharma DK. Recent advancements in nanoparticles for cancer treatment. Med Oncol 2025; 42:72. [PMID: 39928091 DOI: 10.1007/s12032-025-02609-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/13/2025] [Indexed: 02/11/2025]
Abstract
Nanotechnology is a significant factor that has assisted researchers in overcoming medications' permeability and retention effects. This article discusses how different nanoparticles, such as metallic nanoparticles, carbon nanotubes (CNTs), and extracellular vesicles (EVs), are transforming cancer treatments and diagnosis. While CNTs provide photothermal qualities that enable synergistic effects when paired with chemotherapy, EVs provide biocompatibility and immune evasion, enabling effective drug transport. Because of their special optical and magnetic characteristics, metallic nanoparticles are essential for imaging and targeted medication administration. When compared to traditional treatments, these nanoparticles improve bioavailability, decrease systemic toxicity, and increase therapeutic efficacy. Despite increased investigations, the number of licensed nano-drugs has remained relatively high. More investigation is required into targeted drug delivery using nanocarriers to minimize the shielding impact of the protein corona, increase permeability and retention effects, and reduce toxicity to improve clinical translation. This study focuses on novel approaches and state-of-the-art cancer therapies using nanoparticles that target different cancer cells. It also emphasized the advantages of nanoparticle-based cancer therapies over conventional ones, their difficulties, and future promises.
Collapse
Affiliation(s)
- Dinesh Kumar Sharma
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India.
| |
Collapse
|
7
|
Didamoony MA, Soubh AA, Ahmed LA. Cutting-edge insights into liver fibrosis: advanced therapeutic strategies and future perspectives using engineered mesenchymal stem cell-derived exosomes. Drug Deliv Transl Res 2025:10.1007/s13346-024-01784-7. [PMID: 39853531 DOI: 10.1007/s13346-024-01784-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2024] [Indexed: 01/26/2025]
Abstract
Liver fibrosis is still a serious health concern worldwide, and there is increasing interest in mesenchymal stem cells (MSCs) with tremendous potential for treating this disease because of their regenerative and paracrine effects. Recently, many researches have focused on using the released exosomes (EXOs) from stem cells to treat liver fibrosis rather than using parent stem cells themselves. MSC-derived EXOs (MSC-EXOs) have demonstrated favourable outcomes similar to cell treatment in terms of regenerative, immunomodulatory, anti-apoptotic, anti-oxidant, anti-necroptotic, anti-inflammatory and anti-fibrotic actions in several models of liver fibrosis. EXOs are superior to their parent cells in several terms, including lower immunogenicity and risk of tumour formation. However, maintaining the stability and efficacy of EXOs after in vivo transplantation remains a major challenge in their clinical applicability. Therefore, several strategies have been applied in EXOs engineering, such as parental cell modification or modifying EXOs directly to achieve optimum performance of EXOs in treating liver fibrosis. Herein, we discuss the underlying mechanisms of liver fibrosis with an overview of the available therapies, among them EXOs. We also summarise the recent developments in improving the effectiveness of EXOs with the advantages and limitations of these approaches in terms of the upcoming clinical applications.
Collapse
Affiliation(s)
- Manar A Didamoony
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian Russian University, Cairo, 11829, Egypt.
| | - Ayman A Soubh
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza, 12451, Egypt
| | - Lamiaa A Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
8
|
Wang C, Feng Y, Rong X, Yan J, Lv B, Jiang H, Duan L, Jiang J. Mesenchymal stromal cell exosomes for drug delivery of prostate cancer treatments: a review. Stem Cell Res Ther 2025; 16:18. [PMID: 39849570 PMCID: PMC11755940 DOI: 10.1186/s13287-025-04133-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/08/2025] [Indexed: 01/25/2025] Open
Abstract
Interest in prostate cancer as a research topic has gradually increased. As a result, a series of innovative treatment strategies have emerged with an in-depth understanding of the disease. Owing to their unique biological characteristics, mesenchymal stromal cell exosomes (MSC-Exos) have garnered significant attention for their potential to deliver targeted drugs and enable precise prostate cancer treatment. Herein, prostate cancer treatment with MSC-Exos drug-delivery systems is reviewed. This review provides a comprehensive introduction to the advantages of these systems, current research trends and progress, as well as an analysis of current challenges and future research directions. Moreover, this review lays a solid foundation for the continued development and application of MSC-Exos.
Collapse
Affiliation(s)
- Chengran Wang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China
| | - Yanshuo Feng
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China
| | - Xinao Rong
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China
| | - Jingguo Yan
- Department of Intensive Care Unit, Dunhua Hospital, The First Hospital of Jilin University, Dunhua, Jilin Province, People's Republic of China
| | - Baisong Lv
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China
| | - Hongyu Jiang
- Life Spring AKY Pharmaceuticals, Changchun, Jilin Province, People's Republic of China
| | - Lian Duan
- Department of Spine Surgery, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China.
| | - Jinlan Jiang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, Jilin Province, People's Republic of China.
| |
Collapse
|
9
|
He JG, Wu XX, Li S, Yan D, Xiao GP, Mao FG. Exosomes derived from microRNA-540-3p overexpressing mesenchymal stem cells promote immune tolerance via the CD74/nuclear factor-kappaB pathway in cardiac allograft. World J Stem Cells 2024; 16:1022-1046. [PMID: 39734479 PMCID: PMC11669987 DOI: 10.4252/wjsc.v16.i12.1022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 09/16/2024] [Accepted: 11/12/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Heart transplantation is a crucial intervention for severe heart failure, yet the challenge of organ rejection is significant. Bone marrow mesenchymal stem cells (BMSCs) and their exosomes have demonstrated potential in modulating T cells, dendtitic cells (DCs), and cytokines to achieve immunomodulatory effects. DCs, as key antigen-presenting cells, play a critical role in shaping immune responses by influencing T-cell activation and cytokine production. Through this modulation, BMSCs and their exosomes enhance graft tolerance and prolonging survival. AIM To explore the immunomodulatory effects of exosomes derived from BMSCs overexpressing microRNA-540-3p (miR-540-3p) on cardiac allograft tolerance, focusing on how these exosomes modulating DCs and T cells activity through the CD74/nuclear factor-kappaB (NF-κB) pathway. METHODS Rat models were used to assess the impact of miR-540-3p-enhanced exosomes on immune tolerance in cardiac allografts. MiR-540-3p expression was manipulated in BMSCs, and derived exosomes were collected and administered to the rat models post-heart transplantation. The study monitored expression levels of major histocompatibility complex II, CD80, CD86, and CD274 in DCs, and quantified CD4+ and CD8+ T cells, T regulatory cells, and cytokine profiles. RESULTS Exosomes from miR-540-3p-overexpressing BMSCs lead to reduced expression of immune activation markers CD74 and NF-κB p65 in DCs and T cells. Rats treated with these exosomes showed decreased inflammation and improved cardiac function, indicated by lower levels of pro-inflammatory cytokines (interleukin-1β, interferon-γ) and higher levels of anti-inflammatory cytokines (interleukin-10, transforming growth factor β1). Additionally, miR-540-3p skewed the profiles of DCs and T cells towards immune tolerance, increasing the ratio of T regulatory cells and shifting cytokine secretion to favor graft acceptance. CONCLUSION Exosomes derived from BMSCs overexpressing miR-540-3p significantly enhance immune tolerance and prolong cardiac allograft survival by modulating the CD74/NF-κB pathway, which regulates activities of DCs and T cells. These findings highlight a promising therapeutic strategy to improve heart transplantation outcomes and potentially reduce the need for prolonged immunosuppression.
Collapse
Affiliation(s)
- Ji-Gang He
- Department of Cardiovascular Surgery, The First People's Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| | - Xin-Xin Wu
- Yunnan University of Traditional Chinese Medicine, Kunming 650500, Yunnan Province, China
| | - Si Li
- Department of Cardiovascular Surgery, The First People's Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| | - Dan Yan
- Department of Medical Intensive Care Unit, The First People's Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| | - Gao-Peng Xiao
- Department of Anaesthesia, The First People's Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China
| | - Fu-Gang Mao
- Department of Ultrasonic, The First People's Hospital of Yunnan Province, Kunming 650032, Yunnan Province, China.
| |
Collapse
|
10
|
Yadav S, Maity P, Kapat K. The Opportunities and Challenges of Mesenchymal Stem Cells-Derived Exosomes in Theranostics and Regenerative Medicine. Cells 2024; 13:1956. [PMID: 39682706 PMCID: PMC11640604 DOI: 10.3390/cells13231956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Cell-secreted nanovesicles of endosomal origin, called exosomes, are vital for mediating intracellular communication. As local or distal transporters of intracellular cargo, they reflect the unique characteristics of secretory cells and establish cell-specific interactions via characteristic surface proteins and receptors. With the advent of rapid isolation, purification, and identification techniques, exosomes have become an attractive choice for disease diagnosis (exosomal content as biomarkers), cell-free therapy, and tissue regeneration. Mesenchymal stem cell (MSC)-derived exosomes (MSC-exosomes) display angiogenic, immune-modulatory, and other therapeutic effects crucial for cytoprotection, ischemic wound repair, myocardial regeneration, etc. The primary focus of this review is to highlight the widespread application of MSC-exosomes in therapeutics, theranostics, and tissue regeneration. After a brief introduction of exosome properties, biogenesis, isolation, and functions, recent studies on therapeutic and regenerative applications of MSC-exosomes are described, focusing on bone, cartilage, periodontal, cardiovascular, skin, and nerve regeneration. Finally, the review highlights the theranostic potential of exosomes followed by challenges, summary, and outlook.
Collapse
Affiliation(s)
- Sachin Yadav
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, West Bengal, India;
| | - Pritiprasanna Maity
- School of Medicine, University of California Riverside, Riverside, CA 92525, USA
| | - Kausik Kapat
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata 700054, West Bengal, India;
| |
Collapse
|
11
|
Shaban SF, Abdel-Fattah EA, Ali MM, Dessouky AA. The therapeutic efficacy of adipose mesenchymal stem cell-derived microvesicles versus infliximab in a dextran sodium sulfate induced ulcerative colitis rat model. Ultrastruct Pathol 2024; 48:526-549. [PMID: 39545690 DOI: 10.1080/01913123.2024.2426566] [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: 08/18/2024] [Revised: 10/25/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
Ulcerative colitis (UC) is a chronic relapsing intestinal inflammation that is becoming of increasing incidence worldwide and has insufficient treatment. Therefore, finding effective therapies remains a priority. A dextran sodium sulfate colitis model was established to elucidate colonic layers alterations and compare adipose mesenchymal stem cell-derived microvesicles (MSC-MVs) versus infliximab (IFX) efficacy through biochemical, light, and electron microscope studies. Fifty-four rats were allocated to 4 groups: Control (Con), UC, UC+IFX, and UC+MSC-MVs groups. End body weights (BW) and serum malondialdehyde (MDA) levels were recorded. Colitis severity was estimated by disease activity index (DAI). Colonic specimens were processed to evaluate the histological structure, collagen content, surface mucous and goblet cells, CD44, TNF-α, and GFAP immune expression. Morphometric and statistical analyses were performed. The UC group revealed congested, stenosed colons, a significant decline in end BW, and a significant increase in serum MDA and DAI. Furthermore, disturbed histoarchitecture, inflammatory infiltration, depletion of surface mucous and goblet cells, increased collagen, and TNF-α expression and decreased GFAP expression were observed. Alterations were partially attenuated by IFX therapy, whereas MSC-MVs significantly improved all parameters. In conclusion, MSC-MVs were a superior therapeutic option, via attenuating oxidative stress and inflammatory infiltration, in addition to restoring intestinal epithelial integrity and mucosal barrier.
Collapse
Affiliation(s)
- Sahar F Shaban
- Department of Medical Histology and Cell Biology, Medicine, Faculty of medicine, Zagazig University, Zagazig city, Egypt
| | - Eman A Abdel-Fattah
- Department of Medical Histology and Cell Biology, Medicine, Faculty of medicine, Zagazig University, Zagazig city, Egypt
| | - Manar M Ali
- Department of Medical Histology and Cell Biology, Medicine, Faculty of medicine, Zagazig University, Zagazig city, Egypt
| | - Arigue A Dessouky
- Department of Medical Histology and Cell Biology, Medicine, Faculty of medicine, Zagazig University, Zagazig city, Egypt
| |
Collapse
|
12
|
Wu C, Zhai Y, Ji J, Yang X, Ye L, Lu G, Shi X, Zhai G. Advances in tumor stroma-based targeted delivery. Int J Pharm 2024; 664:124580. [PMID: 39142464 DOI: 10.1016/j.ijpharm.2024.124580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/06/2024] [Accepted: 08/10/2024] [Indexed: 08/16/2024]
Abstract
The tumor stroma plays a crucial role in tumor progression, and the interactions between the extracellular matrix, tumor cells, and stromal cells collectively influence tumor progression and the efficacy of therapeutic agents. Currently, utilizing components of the tumor stroma for drug delivery is a noteworthy strategy. A number of targeted drug delivery systems designed based on tumor stromal components are entering clinical trials. Therefore, this paper provides a thorough examination of the function of tumor stroma in the advancement of targeted drug delivery systems. One approach is to use tumor stromal components for targeted drug delivery, which includes certain stromal components possessing inherent targeting capabilities like HA, laminin, along with targeting stromal cells homologously. Another method entails directly focusing on tumor stromal components to reshape the tumor stroma and facilitate drug delivery. These drug delivery systems exhibit great potential in more effective cancer therapy strategies, such as precise targeting, enhanced penetration, improved safety profile, and biocompatibility. Ultimately, the deployment of these drug delivery systems can deepen our comprehension of tumor stroma and the advanced development of corresponding drug delivery systems.
Collapse
Affiliation(s)
- Chunyan Wu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
13
|
Ding K, Kong J, Li L, Selaru FM, Parian A, Mao HQ. Current and emerging therapeutic strategies for perianal fistula in Crohn's disease patients. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 101:159-182. [PMID: 39521599 PMCID: PMC11753511 DOI: 10.1016/bs.apha.2024.10.013] [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] [Indexed: 11/16/2024]
Abstract
The long-term remission rates achieved with current treatment options for Crohn's disease with perianal fistula (CD-PAF)-including antibiotics, biologics, immunomodulators, and Janus kinase inhibitors, often combined with advanced surgical interventions-remain unsatisfactory. This chapter explores several innovative biomaterials-based solutions, such as plugs, adhesives, fillers, and stem cell-based therapies. The key approaches and treatment outcomes of these advanced therapies are examined, focusing on their ability to modulate the immune response, promote tissue healing, and improve patient outcomes. Additionally, the chapter discusses future directions, including the optimization of biomaterial designs, enhancement of delivery and retention of regenerative therapies, and a deeper understanding of the underlying mechanisms of healing.
Collapse
Affiliation(s)
- Kailei Ding
- Institute for NanoBioTechnology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Materials Science and Engineering, Whiting School of Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Translational Tissue Engineering Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jiayuan Kong
- Institute for NanoBioTechnology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Materials Science and Engineering, Whiting School of Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Translational Tissue Engineering Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ling Li
- Division of Gastroenterology and Hepatology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Florin M Selaru
- Institute for NanoBioTechnology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Division of Gastroenterology and Hepatology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Alyssa Parian
- Division of Gastroenterology and Hepatology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Materials Science and Engineering, Whiting School of Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Translational Tissue Engineering Center, The Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
14
|
Shanmugam I, Radhakrishnan S, Santosh S, Ramnath A, Anil M, Devarajan Y, Maheswaran S, Narayanan V, Pitchaimani A. Emerging role and translational potential of small extracellular vesicles in neuroscience. Life Sci 2024; 355:122987. [PMID: 39151884 DOI: 10.1016/j.lfs.2024.122987] [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: 04/26/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Small extracellular vesicles (sEV) are endogenous lipid-bound membrane vesicles secreted by both prokaryotic and eukaryotic cells into the extracellular environment, performs several biological functions such as cell-cell communication, transfer of proteins, mRNA, and ncRNA to target cells in distant sites. Due to their role in molecular pathogenesis and its potential to deliver biological cargo to target cells, it has become a prominent area of interest in recent research in the field of Neuroscience. However, their role in neurological disorders, like neurodegenerative diseases is more complex and still unaddressed. Thus, this review focuses on the role of sEV in neurodegenerative and neurodevelopmental diseases, including their biogenesis, classification, and pathogenesis, with translational advantages and limitations in the area of neurobiology.
Collapse
Affiliation(s)
- Iswarya Shanmugam
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Sivani Radhakrishnan
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Shradha Santosh
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Akansha Ramnath
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Meghna Anil
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Yogesh Devarajan
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Saravanakumar Maheswaran
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Vaibav Narayanan
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India
| | - Arunkumar Pitchaimani
- Precision Nanomedicine and Microfluidic Lab, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology, Vellore. TN, India; School of Biosciences and Technology, Vellore Institute of Technology, Vellore Campus, Tiruvalam Rd, Katpadi, Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
15
|
Kangari P, Salahlou R, Vandghanooni S. Harnessing the Therapeutic Potential of Mesenchymal Stem Cells in Cancer Treatment. Adv Pharm Bull 2024; 14:574-590. [PMID: 39494266 PMCID: PMC11530882 DOI: 10.34172/apb.2024.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 11/05/2024] Open
Abstract
Cancer, as a complicated disease, is considered to be one of the major leading causes of death globally. Although various cancer therapeutic strategies have been established, however, some issues confine the efficacies of the treatments. In recent decades researchers for finding efficient therapeutic solutions have extensively focused on the abilities of stem cells in cancer inhibition. Mesenchymal stem cells (MSCs) are multipotent stromal cells that can the most widely extracted from various sources such as the bone marrow (BM), placenta, umbilical cord (UC), menses blood, Wharton's jelly (WJ), adipose tissue and dental pulp (DP). These cells are capable of differentiating into the osteoblasts, chondrocytes, and adipocytes. Due to the unique characteristics of MSCs such as paracrine effects, immunomodulation, tumor-tropism, and migration, they are considered promising candidates for cancer therapeutics. Currently, MSCs are an excellent living carrier for delivery of therapeutic genes and chemical agents to target tumor sites. Also, exosomes, the most important extracellular vesicle released from MSCs, act as a strong cell-free tool for cancer therapeutics. MSCs can prevent cancer progression by inhibiting several signaling pathways, such as wnt/β-catenin and PI3K/AKT/mTOR. However, there are several challenges associated with the use of MSCs and their exosomes in the field of therapy that need to be considered. This review explores the significance of MSCs in cell-based therapy, focusing on their homing properties and immunomodulatory characteristics. It also examines the potential of using MSCs as carriers for delivery of anticancer agents and their role in modulating the signal transduction pathways of cancer cells.
Collapse
Affiliation(s)
- Parisa Kangari
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Salahlou
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Somayeh Vandghanooni
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Chen M, Lin Y, Guo W, Chen L. BMSC-Derived Exosomes Carrying miR-26a-5p Ameliorate Spinal Cord Injury via Negatively Regulating EZH2 and Activating the BDNF-TrkB-CREB Signaling. Mol Neurobiol 2024; 61:8156-8174. [PMID: 38478142 DOI: 10.1007/s12035-024-04082-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/28/2024] [Indexed: 09/21/2024]
Abstract
BACKGROUND Spinal cord injury (SCI) is a destructive neurological and pathological state that causes major motor, sensory and autonomic dysfunctions. Bone marrow mesenchymal stem cells (BMSCs)-derived exosomes show great therapeutic potential for SCI. Exosomes derived from miR-26a-modified MSCs promote axonal regeneration following SCI. Our study aims to uncover the mechanisms by which BMSC-derived exosomes carrying miR-26a-5p regulate SCI. METHODS BMSCs and BMSC-derived exosomes were isolated and characterized by Oil Red O and alizarin red staining, transmission electron microscopy, flow cytometry, nanoparticle tracking analysis and Western blotting. PC12 cells were treated with lipopolysaccharides (LPS), and SCI was established through laminectomy with contusion injury in rats. Annexin-V staining, CCK-8 and EdU incorporation were applied to determine cell apoptosis, viability, and proliferation. Hematoxylin and Eosin, Nissl and TUNEL staining was used to evaluate SCI injury and apoptosis in the spinal cord. Luciferase and chromatin immunoprecipitation assays were applied to evaluate gene interaction. RESULTS BMSC-derived exosomes facilitated LPS-treated PC12 cell proliferation and inhibited apoptosis by delivering miR-26a-5p. Moreover, BMSC-derived exosomal miR-26a-5p alleviated SCI. Furthermore, miR-26a-5p inhibited EZH2 expression by directly binding to EZH2, and EZH2 inhibited BDNF expression via promoting H3K27me3. Increased phosphorylated CREB enhanced KCC2 transcription and expression by binding to its promoter. Knockdown of miR-26a-5p abrogated BMSC-derived exosome-mediated protection in LPS-treated PC12 cells, but it was reversed by KCC2 overexpression. CONCLUSION BMSC-derived exosomes carrying miR-26a-5p repressed EZH2 expression to promote BDNF and TrkB expression and CREB phosphorylation and subsequently increase KCC2 expression, thus protecting PC12 cells and ameliorating SCI.
Collapse
Affiliation(s)
- Min Chen
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yu Lin
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Wenbin Guo
- Department of Pathology, Fujian Pingtan Comprehensive Experimental Area Hospital, Fuzhou, 350400, China
| | - Lihui Chen
- Laboratory Medicine, Fujian Pingtan Comprehensive Experimental Area Hospital, Fuzhou, 350400, China.
| |
Collapse
|
17
|
Jabermoradi S, Paridari P, Ramawad HA, Gharin P, Roshdi S, Toloui A, Yousefifard M. Stem Cell-Derived Exosomes as a Therapeutic Option for Spinal Cord Injuries; a Systematic Review and Meta-Analysis. ARCHIVES OF ACADEMIC EMERGENCY MEDICINE 2024; 13:e2. [PMID: 39318865 PMCID: PMC11417640 DOI: 10.22037/aaem.v12i1.2261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Introduction Exosomes function as cell signaling carriers and have drawn much attention to the cell-free treatments of regenerative medicine. This meta-analysis aimed to investigate the efficacy of mesenchymal stem cell-derived (MSC-derived) exosomes in animal models of spinal cord injuries (SCI). Method A comprehensive search was conducted in Medline, Embase, Scopus, and Web of Science to attain related articles published by January 31, 2023. The eligible keywords were correlated with the spinal cord injury and MSC-derived exosomes. The evaluated outcomes were locomotion, cavity size, cell apoptosis, inflammation, neuro-regeneration, and microglia activation. A standardized mean difference was calculated for each sample and a pooled effect size was reported. Results 65 papers fully met the inclusion criteria. Treatment with MSC-derived exosomes ultimately improved locomotion and shrunk cavity size (p<0.0001). The administration of MSC-derived exosomes enhanced the expression of beta-tubulin III, NF200, and GAP-43, and increased the number of NeuN-positive and Nissl-positive cells, while reducing the expression of glial fibrillary acidic protein (p<0.0001). The number of apoptotic cells in the treatment group decreased significantly (p<0.0001). Regarding the markers of microglia activation, MSC-derived exosomes increased the number of CD206- and CD68-positive cells (p=0.032 and p<0.0001, respectively). Additionally, MSC-derived exosome administration significantly increased the expression of the anti-inflammatory interleukin (IL)-10 and IL-4 (p<0.001 and p=0.001, respectively) and decreased the expression of the inflammatory IL-1b, IL-6, and TNF-a (p<0.0001). Conclusion MSC-derived exosome treatment resulted in a significantly improved locomotion of SCI animals through ameliorating neuroinflammation, reducing apoptosis, and inducing neuronal regrowth by facilitating a desirable microenvironment.
Collapse
Affiliation(s)
- Sajjad Jabermoradi
- Physiology Research Center, Iran University ofMedical Sciences, Tehran, Iran
- The first and second authors have identical contributions
| | - Parsa Paridari
- Physiology Research Center, Iran University ofMedical Sciences, Tehran, Iran
- The first and second authors have identical contributions
| | - Hamzah Adel Ramawad
- Department of EmergencyMedicine, NYC Health + Hospitals, Coney Island, New York, USA
| | - Pantea Gharin
- Physiology Research Center, Iran University ofMedical Sciences, Tehran, Iran
| | - Shayan Roshdi
- Physiology Research Center, Iran University ofMedical Sciences, Tehran, Iran
| | - Amirmohammad Toloui
- Physiology Research Center, Iran University ofMedical Sciences, Tehran, Iran
| | - Mahmoud Yousefifard
- Physiology Research Center, Iran University ofMedical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Younesi FS, Hinz B. The Myofibroblast Fate of Therapeutic Mesenchymal Stromal Cells: Regeneration, Repair, or Despair? Int J Mol Sci 2024; 25:8712. [PMID: 39201399 PMCID: PMC11354465 DOI: 10.3390/ijms25168712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) can be isolated from various tissues of healthy or patient donors to be retransplanted in cell therapies. Because the number of MSCs obtained from biopsies is typically too low for direct clinical application, MSC expansion in cell culture is required. However, ex vivo amplification often reduces the desired MSC regenerative potential and enhances undesired traits, such as activation into fibrogenic myofibroblasts. Transiently activated myofibroblasts restore tissue integrity after organ injury by producing and contracting extracellular matrix into scar tissue. In contrast, persistent myofibroblasts cause excessive scarring-called fibrosis-that destroys organ function. In this review, we focus on the relevance and molecular mechanisms of myofibroblast activation upon contact with stiff cell culture plastic or recipient scar tissue, such as hypertrophic scars of large skin burns. We discuss cell mechanoperception mechanisms such as integrins and stretch-activated channels, mechanotransduction through the contractile actin cytoskeleton, and conversion of mechanical signals into transcriptional programs via mechanosensitive co-transcription factors, such as YAP, TAZ, and MRTF. We further elaborate how prolonged mechanical stress can create persistent myofibroblast memory by direct mechanotransduction to the nucleus that can evoke lasting epigenetic modifications at the DNA level, such as histone methylation and acetylation. We conclude by projecting how cell culture mechanics can be modulated to generate MSCs, which epigenetically protected against myofibroblast activation and transport desired regeneration potential to the recipient tissue environment in clinical therapies.
Collapse
Affiliation(s)
- Fereshteh Sadat Younesi
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Boris Hinz
- Faculty of Dentistry, University of Toronto, Toronto, ON M5G 1G6, Canada;
- Keenan Research Institute for Biomedical Science, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
19
|
Guinn MT, Fernandez R, Lau S, Loor G. Transcriptomic Signatures in Lung Allografts and Their Therapeutic Implications. Biomedicines 2024; 12:1793. [PMID: 39200257 PMCID: PMC11351513 DOI: 10.3390/biomedicines12081793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/20/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
Ex vivo lung perfusion (EVLP) is a well-established method of lung preservation in clinical transplantation. Transcriptomic analyses of cells and tissues uncover gene expression patterns which reveal granular molecular pathways and cellular programs under various conditions. Coupling EVLP and transcriptomics may provide insights into lung allograft physiology at a molecular level with the potential to develop targeted therapies to enhance or repair the donor lung. This review examines the current landscape of transcriptional analysis of lung allografts in the context of state-of-the-art therapeutics that have been developed to optimize lung allograft function.
Collapse
Affiliation(s)
- Michael Tyler Guinn
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (M.T.G.)
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Ramiro Fernandez
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (M.T.G.)
| | - Sean Lau
- Department of Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Gabriel Loor
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (M.T.G.)
| |
Collapse
|
20
|
Yao H, Sun C, Wang C, Liu J, Li Y, Li L, Zhao B, Liu J. The Double-Edged Sword Property of Mesenchymal Stem Cell-Derived Exosomal microRNAs in Colorectal Cancer. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2024; 35:755-762. [PMID: 39412096 PMCID: PMC11465186 DOI: 10.5152/tjg.2024.23541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 05/19/2024] [Indexed: 10/20/2024]
Abstract
Mesenchymal stem cells (MSCs)-based therapies are promising therapeutic strategies for cancer treatment, because of their strong immunomodulatory and tissue regeneration abilities. In case of colorectal cancer (CRC), MSCs indicate a double-edged sword activity. Some reports declared the inhibitory effects of MSCs on the proliferation, migration, and infiltration of cancer cells to suppress the CRC initiation and development, whereas others showed the tumor-promoter impacts of MSCs on the progression of CRC. Recent investigations have revealed that exosomal microRNAs (Exo-miRs) derived from MSCs (MSCs-Exo-miRs) are attributed to such paradoxical effect. Thus, the current review aimed to seek the role of MSCs-Exo-miRs in CRC progression and their therapeutic potential for the CRC treatment.
Collapse
Affiliation(s)
- Hongliang Yao
- The Second Department of General Surgery, Hengshui People’s Hospital, Hengshui, Hebei Province, China
| | - Caihua Sun
- The Second Department of General Surgery, Hengshui People’s Hospital, Hengshui, Hebei Province, China
| | - Chengjun Wang
- The Second Department of General Surgery, Hengshui People’s Hospital, Hengshui, Hebei Province, China
| | - Jipan Liu
- The Second Department of General Surgery, Hengshui People’s Hospital, Hengshui, Hebei Province, China
| | - Yun Li
- The Second Department of General Surgery, Hengshui People’s Hospital, Hengshui, Hebei Province, China
| | - Li Li
- The Second Department of General Surgery, Hengshui People’s Hospital, Hengshui, Hebei Province, China
| | - Bin Zhao
- The Second Department of General Surgery, Hengshui People’s Hospital, Hengshui, Hebei Province, China
| | - Jia Liu
- The Second Department of General Surgery, Hengshui People’s Hospital, Hengshui, Hebei Province, China
| |
Collapse
|
21
|
Abd-Rabou AA, Kishta MS, Yakout SM, Youssef AM, Abdallah AN, Ahmed HH. Copper/Tin Nanocomposites-Loaded Exosomes Induce Apoptosis and Cell Cycle Arrest at G0/G1 Phase in Skin Cancer Cell Line. Chem Biodivers 2024; 21:e202400486. [PMID: 38860853 DOI: 10.1002/cbdv.202400486] [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: 02/29/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/12/2024]
Abstract
This study aims to explore the efficacy of Copper/Tin (CuS/SnS) nanocomposites loaded into exosomes against skin cancer A431 cell line. CuS/SnS nanocomposites (S1, S2, S3) were synthesized and characterized, then loaded into exosomes (Exo) (S1-Exo, S2-Exo and S3-Exo) and characterized. After that, the loaded samples were investigated in vitro against A431 using cytotoxicity, apoptosis, and cell cycle assays. CuS/SnS nanocomposites were indexed to hexagonal CuS structure and orthorhombic α-SnS phase and showed nano-rode shape. The exosomes loaded with nanocomposites were regular and rounded within the size of 120 nm, with no signs of broken exosomes or leakage of their contents. The cytotoxicity assay indicated the enhanced cytotoxic of S1-Exo versus the free nano-form S1 on A431. Interestingly, S1-Exo recorded 1.109 times more than DOX in its anti-skin cancer capacity. Moreover, S1-Exo recorded 40.2 % for early apoptosis and 22.1 % for late apoptosis. Furthermore, it displayed impact in arresting the cancer cell cycle at G0/G1 phase and reducing G2/M phase. Noteworthy, loaded nanocomposites were safe against normal HSF skin cells. In conclusion, the loaded CuS/SnS nanocomposites into the exosomes could be of great potential as anti-skin cancer candidates through induction of apoptosis and promotion of the cell cycle arrest at G0/G1 phase.
Collapse
Affiliation(s)
- Ahmed A Abd-Rabou
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Cairo, Egypt
- Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Mohamed S Kishta
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Cairo, Egypt
- Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Saad M Yakout
- Inorganic Chemistry Department, Inorganic Chemical Industries and Mineral Resources Research institute, National Research Centre, Dokki, Cairo, Egypt
| | - Ahmed M Youssef
- Inorganic Chemistry Department, Inorganic Chemical Industries and Mineral Resources Research institute, National Research Centre, Dokki, Cairo, Egypt
| | - Ahmed N Abdallah
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Cairo, Egypt
- Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Hanaa H Ahmed
- Hormones Department, Medical Research and Clinical Studies Institute, National Research Centre, Dokki, Cairo, Egypt
- Stem Cell Lab., Center of Excellence for Advanced Sciences, National Research Centre, Dokki, Cairo, 12622, Egypt
| |
Collapse
|
22
|
Kanda P, Gupta A, Dhillon J, Kundapur D, Gottlieb CC. Mesenchymal stem cell based therapies for uveitis: a systematic review of preclinical studies. Eye (Lond) 2024; 38:1845-1854. [PMID: 38600361 PMCID: PMC11226430 DOI: 10.1038/s41433-024-03057-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 02/03/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Abstract
Cell therapy has shown promising results for treating uveitis in preclinical studies. As the field continues to grow towards clinical translation, it is important to review and critically appraise existing studies. Herein, we analysed and critically appraised all preclinical studies using cell therapy or cell derived extracellular vesicles (EVs) for uveitis, and provided insight into mechanisms regulating ocular inflammation. We used PubMed, Medline, and Embase to search for preclinical studies examining stem cell therapy (e.g., mesenchymal stem cells [MSC]) and secreted EVs. All included studies were assessed for quality using the SYstematic Review Center for Laboratory animal Experimentation (SYRCLE) checklist. Sixteen preclinical studies from 2011 to 2022 were analysed and included in this review of which 75% (n = 12) focused only on cell therapy, 18.7% (n = 3) studies focused on EVs, and 6.3% (n = 1) study focused on both cells and EVs. MSCs were the most common type of cells used in preclinical studies (n = 15) and EVs were commonly isolated from MSCs (n = 3). Overall, both MSCs and EVs showed improvements in ocular inflammation (seen on fundoscopy/slit lamp and histology) and electroretinogram outcomes. Overall, MSC and MSC-derived EVs shown great potential as therapeutic agents for treating uveitis. Unfortunately, small sample size, risk of selection/performance bias, and lack of standardized cell harvesting or delivery protocols are some factors which limits clinical translation. Large scaled, randomized preclinical studies are required to understand the full potential of MSCs for treating uveitis.
Collapse
Affiliation(s)
| | - Arnav Gupta
- Department of Medicine, University of Calgary, Calgary, AB, Canada
- College of Public Health, Kent State University, Kent, OH, USA
| | | | | | - Chloe C Gottlieb
- Eye Institute, University of Ottawa, Ottawa, ON, Canada
- Ottawa Hospital Research Institute, The Ottawa Hospital, Ottawa, ON, Canada
| |
Collapse
|
23
|
Taheri M, Tehrani HA, Dehghani S, Alibolandi M, Arefian E, Ramezani M. Nanotechnology and bioengineering approaches to improve the potency of mesenchymal stem cell as an off-the-shelf versatile tumor delivery vehicle. Med Res Rev 2024; 44:1596-1661. [PMID: 38299924 DOI: 10.1002/med.22023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 11/28/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
Targeting actionable mutations in oncogene-driven cancers and the evolution of immuno-oncology are the two prominent revolutions that have influenced cancer treatment paradigms and caused the emergence of precision oncology. However, intertumoral and intratumoral heterogeneity are the main challenges in both fields of precision cancer treatment. In other words, finding a universal marker or pathway in patients suffering from a particular type of cancer is challenging. Therefore, targeting a single hallmark or pathway with a single targeted therapeutic will not be efficient for fighting against tumor heterogeneity. Mesenchymal stem cells (MSCs) possess favorable characteristics for cellular therapy, including their hypoimmune nature, inherent tumor-tropism property, straightforward isolation, and multilineage differentiation potential. MSCs can be loaded with various chemotherapeutics and oncolytic viruses. The combination of these intrinsic features with the possibility of genetic manipulation makes them a versatile tumor delivery vehicle that can be used for in vivo selective tumor delivery of various chemotherapeutic and biological therapeutics. MSCs can be used as biofactory for the local production of chemical or biological anticancer agents at the tumor site. MSC-mediated immunotherapy could facilitate the sustained release of immunotherapeutic agents specifically at the tumor site, and allow for the achievement of therapeutic concentrations without the need for repetitive systemic administration of high therapeutic doses. Despite the enthusiasm evoked by preclinical studies that used MSC in various cancer therapy approaches, the translation of MSCs into clinical applications has faced serious challenges. This manuscript, with a critical viewpoint, reviewed the preclinical and clinical studies that have evaluated MSCs as a selective tumor delivery tool in various cancer therapy approaches, including gene therapy, immunotherapy, and chemotherapy. Then, the novel nanotechnology and bioengineering approaches that can improve the potency of MSC for tumor targeting and overcoming challenges related to their low localization at the tumor sites are discussed.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
24
|
Jin X, Zhang J, Zhang Y, He J, Wang M, Hei Y, Guo S, Xu X, Liu Y. Different origin-derived exosomes and their clinical advantages in cancer therapy. Front Immunol 2024; 15:1401852. [PMID: 38994350 PMCID: PMC11236555 DOI: 10.3389/fimmu.2024.1401852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/13/2024] [Indexed: 07/13/2024] Open
Abstract
Exosomes, as a class of small extracellular vesicles closely related to the biological behavior of various types of tumors, are currently attracting research attention in cancer diagnosis and treatment. Regarding cancer diagnosis, the stability of their membrane structure and their wide distribution in body fluids render exosomes promising biomarkers. It is expected that exosome-based liquid biopsy will become an important tool for tumor diagnosis in the future. For cancer treatment, exosomes, as the "golden communicators" between cells, can be designed to deliver different drugs, aiming to achieve low-toxicity and low-immunogenicity targeted delivery. Signaling pathways related to exosome contents can also be used for safer and more effective immunotherapy against tumors. Exosomes are derived from a wide range of sources, and exhibit different biological characteristics as well as clinical application advantages in different cancer therapies. In this review, we analyzed the main sources of exosomes that have great potential and broad prospects in cancer diagnosis and therapy. Moreover, we compared their therapeutic advantages, providing new ideas for the clinical application of exosomes.
Collapse
Affiliation(s)
- Xiaoyan Jin
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Jing Zhang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
- The Second Affiliated Hospital of Xi‘an Medical University, Xi’an, Shaanxi, China
| | - Yufu Zhang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Jing He
- Laboratory of Obstetrics and Gynecology, The Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Mingming Wang
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Yu Hei
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Shutong Guo
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Xiangrong Xu
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| | - Yusi Liu
- Department of Cell Biology and Genetics, Medical College of Yan’an University, Yan’an, Shaanxi, China
| |
Collapse
|
25
|
Kerkis I, da Silva ÁP, Araldi RP. The impact of interleukin-6 (IL-6) and mesenchymal stem cell-derived IL-6 on neurological conditions. Front Immunol 2024; 15:1400533. [PMID: 39015561 PMCID: PMC11249726 DOI: 10.3389/fimmu.2024.1400533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/04/2024] [Indexed: 07/18/2024] Open
Abstract
Interleukin-6 (IL-6) is a versatile cytokine crucial for immune response modulation, inflammation regulation, and various physiological processes in the body. Its wide-ranging functions underscore its importance in maintaining health. Dysregulated IL-6 is closely associated with many diseases, making it a key research and therapeutic target. Elevated IL-6 levels in the central nervous system worsen neuroinflammation in neurodegenerative diseases by activating microglia and astrocytes and releasing pro-inflammatory cytokines and neurotoxic molecules. Moreover, dysregulated IL-6 weakens the blood-brain barrier, exacerbating neuroinflammation and neuronal damage by allowing peripheral immune cells and inflammatory mediators to enter the brain. Mesenchymal stem cells (MSCs) show promise in modulating neuroinflammation by regulating IL-6 levels. They effectively suppress pro-inflammatory cytokines, including IL-6, while promoting anti-inflammatory factors. This therapeutic approach highlights the importance of targeting IL-6 and other inflammatory mediators to alleviate neuroinflammation and its adverse effects on neurological disorders. This review provides a comprehensive overview of IL-6's involvement in neurological disorders, examining endogenous IL-6 and IL-6 derived from MSCs. We explore IL-6's mechanisms affecting neuronal function, survival, and immune modulation in the central nervous system. Additionally, we discuss the potential of MSC-derived IL-6 in neuroregeneration and neuroprotection. By elucidating IL-6's interplay with neurological pathologies, this review offers insights into novel therapeutic strategies targeting IL-6 signaling pathways for neurological disorders.
Collapse
Affiliation(s)
- Irina Kerkis
- Genetics Laboratory, Center of Development and Innovation, Butantan Institute, São Paulo, Brazil
| | - Álvaro Prieto da Silva
- Genetics Laboratory, Center of Development and Innovation, Butantan Institute, São Paulo, Brazil
| | - Rodrigo Pinheiro Araldi
- BioDecision Analytics Ltda., São Paulo, Brazil
- Post-graduation Program in Structural and Functional Biology, Paulista School of Medicine Escola Paulista de Medicina (EPM), Federal University of São Paulo Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| |
Collapse
|
26
|
Ding Z, Greenberg ZF, Serafim MF, Ali S, Jamieson JC, Traktuev DO, March K, He M. Understanding molecular characteristics of extracellular vesicles derived from different types of mesenchymal stem cells for therapeutic translation. EXTRACELLULAR VESICLE 2024; 3:100034. [PMID: 38957857 PMCID: PMC11218754 DOI: 10.1016/j.vesic.2024.100034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
Mesenchymal stem cells (MSCs) have been studied for decades as candidates for cellular therapy, and their secretome, including secreted extracellular vesicles (EVs), has been identified to contribute significantly to regenerative and reparative functions. Emerging evidence has suggested that MSC-EVs alone, could be used as therapeutics that emulate the biological function of MSCs. However, just as with MSCs, MSC-EVs have been shown to vary in composition, depending on the tissue source of the MSCs as well as the protocols employed in culturing the MSCs and obtaining the EVs. Therefore, the importance of careful choice of cell sources and culture environments is receiving increasing attention. Many factors contribute to the therapeutic potential of MSC-EVs, including the source tissue, isolation technique, and culturing conditions. This review illustrates the molecular landscape of EVs derived from different types of MSC cells along with culture strategies. A thorough analysis of publicly available omic datasets was performed to advance the precision understanding of MSC-EVs with unique tissue source-dependent molecular characteristics. The tissue-specific protein and miRNA-driven Reactome ontology analysis was used to reveal distinct patterns of top Reactome ontology pathways across adipose, bone marrow, and umbilical MSC-EVs. Moreover, a meta-analysis assisted by an AI technique was used to analyze the published literature, providing insights into the therapeutic translation of MSC-EVs based on their source tissues.
Collapse
Affiliation(s)
- Zuo Ding
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Zachary F. Greenberg
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Maria Fernanda Serafim
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Samantha Ali
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Julia C. Jamieson
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| | - Dmitry O. Traktuev
- UF Center for Regenerative Medicine, Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Keith March
- UF Center for Regenerative Medicine, Division of Cardiovascular Medicine, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Mei He
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
27
|
Moghassemi S, Dadashzadeh A, Sousa MJ, Vlieghe H, Yang J, León-Félix CM, Amorim CA. Extracellular vesicles in nanomedicine and regenerative medicine: A review over the last decade. Bioact Mater 2024; 36:126-156. [PMID: 38450204 PMCID: PMC10915394 DOI: 10.1016/j.bioactmat.2024.02.021] [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: 12/01/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
Small extracellular vesicles (sEVs) are known to be secreted by a vast majority of cells. These sEVs, specifically exosomes, induce specific cell-to-cell interactions and can activate signaling pathways in recipient cells through fusion or interaction. These nanovesicles possess several desirable properties, making them ideal for regenerative medicine and nanomedicine applications. These properties include exceptional stability, biocompatibility, wide biodistribution, and minimal immunogenicity. However, the practical utilization of sEVs, particularly in clinical settings and at a large scale, is hindered by the expensive procedures required for their isolation, limited circulation lifetime, and suboptimal targeting capacity. Despite these challenges, sEVs have demonstrated a remarkable ability to accommodate various cargoes and have found extensive applications in the biomedical sciences. To overcome the limitations of sEVs and broaden their potential applications, researchers should strive to deepen their understanding of current isolation, loading, and characterization techniques. Additionally, acquiring fundamental knowledge about sEVs origins and employing state-of-the-art methodologies in nanomedicine and regenerative medicine can expand the sEVs research scope. This review provides a comprehensive overview of state-of-the-art exosome-based strategies in diverse nanomedicine domains, encompassing cancer therapy, immunotherapy, and biomarker applications. Furthermore, we emphasize the immense potential of exosomes in regenerative medicine.
Collapse
Affiliation(s)
- Saeid Moghassemi
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Arezoo Dadashzadeh
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Maria João Sousa
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Hanne Vlieghe
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jie Yang
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Cecibel María León-Félix
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christiani A. Amorim
- Pôle de Recherche en Physiopathologie de La Reproduction, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
28
|
Jalilivand S, Izadirad M, Vazifeh Shiran N, Gharehbaghian A, Naserian S. The effect of bone marrow mesenchymal stromal cell exosomes on acute myeloid leukemia's biological functions: a focus on the potential role of LncRNAs. Clin Exp Med 2024; 24:108. [PMID: 38777995 PMCID: PMC11111499 DOI: 10.1007/s10238-024-01364-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Acute myeloid leukemia represents a group of malignant blood disorders that originate from clonal over-proliferation and the differentiation failure of hematopoietic precursors, resulting in the accumulation of blasts in the bone marrow. Mesenchymal stromal cells (MSCs) have been shown to exert diverse effects on tumor cells through direct and indirect interaction. Exosomes, as one of the means of indirect intercellular communication, are released from different types of cells, including MSCs, and their various contents, such as lncRNAs, enable them to exert significant impacts on target cells. Our study aims to investigate the effects of BM-MSC exosomes on the cellular and molecular characterization of HL-60 AML cells, particularly detecting the alterations in the expression of lncRNAs involved in AML leukemogenesis, cell growth, drug resistance, and poor prognosis. BM-MSCs were cultured with serum-free culture media to isolate exosomes from their supernatants. The validation of exosomes was performed in three stages: morphological analysis using TEM, size evaluation using DLS, and CD marker identification using flow cytometry. Subsequently, the HL-60 AML cells were treated with isolated BM-MSC exosomes to determine the impact of their contents on leukemic cells. Cell metabolic activity was evaluated by the MTT assay, while cell cycle progression, apoptosis, ROS levels, and proliferation were assessed by flow cytometry. Furthermore, RT-qPCR was conducted to determine the expression levels of lncRNAs and apoptosis-, ROS-, and cell cycle-related genes. MTT assay and flow cytometry analysis revealed that BM-MSC exosomes considerably suppressed cell metabolic activity, proliferation, and cell cycle progression. Also, these exosomes could effectively increase apoptosis and ROS levels in HL-60 cells. The expression levels of p53, p21, BAX, and FOXO4 were increased, while the BCL2 and c-Myc levels decreased. MALAT1, HOTAIR, and H19 expression levels were also significantly decreased in treated HL-60 cells compared to their untreated counterparts. BM-MSC exosomes suppress cell cycle progression, proliferation, and metabolic activity while simultaneously elevating the ROS index and apoptosis ratio in HL-60 cells, likely by reducing the expression levels of MALAT1, HOTAIR, and H19. These findings suggest that BM-MSC exosomes might serve as potential supportive therapies for leukemia.
Collapse
Affiliation(s)
- Sahar Jalilivand
- Department of Laboratory Hematology and Blood Bank, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Izadirad
- Department of Laboratory Hematology and Blood Bank, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nader Vazifeh Shiran
- Department of Laboratory Hematology and Blood Bank, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Gharehbaghian
- Department of Laboratory Hematology and Blood Bank, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
29
|
Tahmasebi F, Asl ER, Vahidinia Z, Barati S. Stem Cell-Derived Exosomal MicroRNAs as Novel Potential Approach for Multiple Sclerosis Treatment. Cell Mol Neurobiol 2024; 44:44. [PMID: 38713302 PMCID: PMC11076329 DOI: 10.1007/s10571-024-01478-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 04/09/2024] [Indexed: 05/08/2024]
Abstract
Multiple Sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) characterized by inflammation and demyelination of CNS neurons. Up to now, there are many therapeutic strategies for MS but they are only being able to reduce progression of diseases and have not got any effect on repair and remyelination. Stem cell therapy is an appropriate method for regeneration but has limitations and problems. So recently, researches were used of exosomes that facilitate intercellular communication and transfer cell-to-cell biological information. MicroRNAs (miRNAs) are a class of short non-coding RNAs that we can used to their dysregulation in order to diseases diagnosis. The miRNAs of microvesicles obtained stem cells may change the fate of transplanted cells based on received signals of injured regions. The miRNAs existing in MSCs may be displayed the cell type and their biological activities. Current studies show also that the miRNAs create communication between stem cells and tissue-injured cells. In the present review, firstly we discuss the role of miRNAs dysregulation in MS patients and miRNAs expression by stem cells. Finally, in this study was confirmed the relationship of microRNAs involved in MS and miRNAs expressed by stem cells and interaction between them in order to find appropriate treatment methods in future for limit to disability progression.
Collapse
Affiliation(s)
- Fatemeh Tahmasebi
- Department of Anatomy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elmira Roshani Asl
- Department of Biochemistry, Saveh University of Medical Sciences, Saveh, Iran
| | - Zeinab Vahidinia
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran.
| |
Collapse
|
30
|
Tang J, Wang X, Lin X, Wu C. Mesenchymal stem cell-derived extracellular vesicles: a regulator and carrier for targeting bone-related diseases. Cell Death Discov 2024; 10:212. [PMID: 38697996 PMCID: PMC11066013 DOI: 10.1038/s41420-024-01973-w] [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: 02/05/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
The escalating threat of bone-related diseases poses a significant challenge to human health. Mesenchymal stem cell (MSC)-derived extracellular vesicles (MSC-EVs), as inherent cell-secreted natural products, have emerged as promising treatments for bone-related diseases. Leveraging outstanding features such as high biocompatibility, low immunogenicity, superior biological barrier penetration, and extended circulating half-life, MSC-EVs serve as potent carriers for microRNAs (miRNAs), long no-code RNAs (lncRNAs), and other biomolecules. These cargo molecules play pivotal roles in orchestrating bone metabolism and vascularity through diverse mechanisms, thereby contributing to the amelioration of bone diseases. Additionally, engineering modifications enhance the bone-targeting ability of MSC-EVs, mitigating systemic side effects and bolstering their clinical translational potential. This review comprehensively explores the mechanisms through which MSC-EVs regulate bone-related disease progression. It delves into the therapeutic potential of MSC-EVs as adept drug carriers, augmented by engineered modification strategies tailored for osteoarthritis (OA), rheumatoid arthritis (RA), osteoporosis, and osteosarcoma. In conclusion, the exceptional promise exhibited by MSC-EVs positions them as an excellent solution with considerable translational applications in clinical orthopedics.
Collapse
Affiliation(s)
- Jiandong Tang
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China
| | - Xiangyu Wang
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China
| | - Xu Lin
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China
| | - Chao Wu
- Orthopaedics Center, Zigong Fourth People's Hospital, Tan mu lin Street 19#, Zigong, 643099, Sichuan Province, China.
| |
Collapse
|
31
|
Lu W, Guo Y, Liu H, Zhang T, Zhang M, Li X, Li Z, Shi M, Jiang Z, Zhao Z, Yang S, Li Z. The Inhibition of Fibrosis and Inflammation in Obstructive Kidney Injury via the miR-122-5p/SOX2 Axis Using USC-Exos. Biomater Res 2024; 28:0013. [PMID: 38617751 PMCID: PMC11014086 DOI: 10.34133/bmr.0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 02/14/2024] [Indexed: 04/16/2024] Open
Abstract
Background: Fibrosis and inflammation due to ureteropelvic junction obstruction substantially contributes to poor renal function. Urine-derived stem-cell-derived exosomes (USC-Exos) have therapeutic effects through paracrine. Methods: In vitro, the effects of USC-Exos on the biological functions of HK-2 and human umbilical vein endothelial cells were tested. Cell inflammation and fibrosis were induced by transforming growth factor-β1 and interleukin-1β, and their anti-inflammatory and antifibrotic effects were observed after exogenous addition of USC-Exos. Through high-throughput sequencing of microRNA in USC-Exos, the pathways and key microRNAs were selected. Then, the antifibrotic and anti-inflammatory effects of exosomal miR-122-5p and target genes were verified. The role of the miR-122-5p/SOX2 axis in anti-inflammatory and antifibrotic effects was verified. In vivo, a rabbit model of partial unilateral ureteral obstruction (PUUO) was established. Magnetic resonance imaging recorded the volume of the renal pelvis after modeling, and renal tissue was pathologically analyzed. Results: We examined the role of USC-Exos and their miR-122-5p content in obstructive kidney injury. These Exos exhibit antifibrotic and anti-inflammatory activities. SOX2 is the hub gene in PUUO and negatively related to renal function. We confirmed the binding relationship between miR-122-5p and SOX2. The anti-inflammatory and antifibrotic effects of miR-122-5p were inhibited, indicating that miR-122-5p has anti-inflammatory and antifibrotic effects by inhibiting SOX2 expression. In vivo, the PUUO group showed typical obstructive kidney injury after modeling. After USC-Exo treatment, the shape of the renal pelvis shown a remarkable improvement, and inflammation and fibrosis decreased. Conclusions: We confirmed that miR-122-5p from USC-Exos targeting SOX2 is a new molecular target for postoperative recovery treatment of obstructive kidney injury.
Collapse
Affiliation(s)
- Wenjun Lu
- Department of Pediatric Surgery,
The Sixth Hospital Affiliated to Harbin Medical University, Harbin Medical University, No.998 Aiying Street, Harbin 150027, Heilongjiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province,
School of Life Sciences, Westlake University,Hangzhou 310024, Zhejiang, China
- Center for Infectious Disease Research,
Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, Zhejiang, China
- Laboratory of Systems Immunology,
Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, Zhejiang, China
| | - Yujun Guo
- Department of Pediatric Surgery,
The Sixth Hospital Affiliated to Harbin Medical University, Harbin Medical University, No.998 Aiying Street, Harbin 150027, Heilongjiang, China
| | - Hengchen Liu
- Department of General Surgery,
The Second Hospital Affiliated to Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou 310022, Zhejiang, China
| | - Tingting Zhang
- Department of Pediatric Surgery,
The Sixth Hospital Affiliated to Harbin Medical University, Harbin Medical University, No.998 Aiying Street, Harbin 150027, Heilongjiang, China
| | - Mingzhao Zhang
- Department of General Surgery,
The Second Hospital Affiliated to Anhui Medical University, No. 678 Furong Road, Hefei 230031, Anhui, China
| | - Xiangqi Li
- Department of Pediatric Surgery,
The Sixth Hospital Affiliated to Harbin Medical University, Harbin Medical University, No.998 Aiying Street, Harbin 150027, Heilongjiang, China
| | - Zhou Li
- Department of Pediatric Surgery,
The Sixth Hospital Affiliated to Harbin Medical University, Harbin Medical University, No.998 Aiying Street, Harbin 150027, Heilongjiang, China
| | - Manyu Shi
- Department of Pediatric Surgery,
The Sixth Hospital Affiliated to Harbin Medical University, Harbin Medical University, No.998 Aiying Street, Harbin 150027, Heilongjiang, China
| | - Zhitao Jiang
- Department of Pediatric Surgery,
The Sixth Hospital Affiliated to Harbin Medical University, Harbin Medical University, No.998 Aiying Street, Harbin 150027, Heilongjiang, China
| | - Zheng Zhao
- Department of Pediatric Surgery,
The Sixth Hospital Affiliated to Harbin Medical University, Harbin Medical University, No.998 Aiying Street, Harbin 150027, Heilongjiang, China
| | - Shulong Yang
- Department of Pediatric Surgery,
The Sixth Hospital Affiliated to Harbin Medical University, Harbin Medical University, No.998 Aiying Street, Harbin 150027, Heilongjiang, China
| | - Zhaozhu Li
- Department of Pediatric Surgery,
The Sixth Hospital Affiliated to Harbin Medical University, Harbin Medical University, No.998 Aiying Street, Harbin 150027, Heilongjiang, China
| |
Collapse
|
32
|
Mirsanei Z, Jamshidi-Adegani F, Vakilian S, Ahangari F, Soufihasanabad S, Al-Riyami K, Soudi S, Ghaffari Khaligh S, Al-Hashmi S, Hashemi SM. Synergistic effects of mesenchymal stem cell-derived extracellular vesicles and dexamethasone on macrophage polarization under inflammatory conditions. Inflammopharmacology 2024; 32:1317-1332. [PMID: 38512654 DOI: 10.1007/s10787-024-01438-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/22/2023] [Indexed: 03/23/2024]
Abstract
The undesirable inflammation and the excessive M1 macrophage activity may lead to inflammatory diseases. Corticosteroids and stem cell therapy are used in clinical practice to promote anti-inflammatory responses. However, this protocol has limitations and is associated with numerous side effects. In this study, the synergistic anti-inflammatory effects of dexamethasone (Dex) and mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) were evaluated to enhance the polarization of M1 inflammatory macrophages into the anti-inflammatory (M2) phenotype. Hence, we designed different combinations of Dex and EVs using three methods, including EVs isolated from Dex-preconditioned MSCs (Pre-Dex-EVs), EVs loaded with Dex (L-Dex-EVs), and EVs and Dex co-administration (Dex + EVs). All designed EVs had a significant effect on reducing the expression of M1-related genes (iNOS, Stat1, and IRF5), cytokines (IL6 and TNF-a), and CD markers (CD86) in lipopolysaccharide-stimulated macrophages. On the other hand, these combinations promoted the expression of alternative-activated M2-related genes (Arg-1, Stat6, and IRF4), cytokine (IL10), and CD markers (CD206).The combination of Dex and MSC-EVs enhances the effectiveness of both and synergistically promotes the conversion of inflammatory macrophages into an anti-inflammatory state.
Collapse
Affiliation(s)
- Zahra Mirsanei
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Jamshidi-Adegani
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, P. O. Box: 33, PC, 616, Nizwa, Oman
| | - Saeid Vakilian
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, P. O. Box: 33, PC, 616, Nizwa, Oman
| | - Fateme Ahangari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Soufihasanabad
- Department of Biology, School of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Khamis Al-Riyami
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, P. O. Box: 33, PC, 616, Nizwa, Oman
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | | | - Sulaiman Al-Hashmi
- Laboratory for Stem Cell and Regenerative Medicine, Natural and Medical Sciences Research Center, University of Nizwa, P. O. Box: 33, PC, 616, Nizwa, Oman.
| | - Seyed Mahmoud Hashemi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Hyun J, Eom J, Im J, Kim YJ, Seo I, Kim SW, Im GB, Kim YH, Lee DH, Park HS, Yun DW, Kim DI, Yoon JK, Um SH, Yang DH, Bhang SH. Fibroblast function recovery through rejuvenation effect of nanovesicles extracted from human adipose-derived stem cells irradiated with red light. J Control Release 2024; 368:453-465. [PMID: 38447812 DOI: 10.1016/j.jconrel.2024.02.047] [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: 11/03/2023] [Revised: 02/07/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
Fibroblasts (hDFs) are widely employed for skin regeneration and the treatment of various skin disorders, yet research were rarely investigated about restoration of diminished therapeutic efficacy due to cell senescence. The application of stem cell and stem cell-derived materials, exosomes, were drawn attention for the restoration functionality of fibroblasts, but still have limitation for unintended side effect or low yield. To advance, stem cell-derived nanovesicle (NV) have developed for effective therapeutic reagents with high yield and low risk. In this study, we have developed a method using red light irradiated human adipose-derived stem cells (hADSCs) derived NV (R-NVs) for enhancing the therapeutic efficacy and rejuvenating hDFs. Through red light irradiation, we were able to significantly increase the content of stemness factors and angiogenic biomolecules in R-NVs. Treatment with these R-NVs was found to enhance the migration ability and leading to rejuvenation of old hDFs to levels similar to those of young hDFs. In subsequent in vivo experiments, the treatment of old hDFs with R-NVs demonstrated a superior skin wound healing effect, surpassing that of young hDFs. In summary, this study successfully induced rejuvenation and leading to increased therapeutic efficacy to R-NVs treated old hDFs previously considered as biowaste.
Collapse
Affiliation(s)
- Jiyu Hyun
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jiin Eom
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jisoo Im
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Yu-Jin Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Inwoo Seo
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sung-Won Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gwang-Bum Im
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Surgery, Harvard Medical School, Boston, MA, 02115 USA
| | - Yeong Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dong-Hyun Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyun Su Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dae Won Yun
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dong-Ik Kim
- Division of Vascular Surgery, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul 06351, South Korea
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong 4726, Republic of Korea
| | - Soong Ho Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dae Hyeok Yang
- Department of Medical Life Sciences, College of Medicine, Institute of Cell and Tissue Engineering, The Catholic University of Korea, Seoul, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| |
Collapse
|
34
|
Tilotta V, Vadalà G, Ambrosio L, Di Giacomo G, Cicione C, Russo F, Darinskas A, Papalia R, Denaro V. Wharton's Jelly mesenchymal stromal cell-derived extracellular vesicles promote nucleus pulposus cell anabolism in an in vitro 3D alginate-bead culture model. JOR Spine 2024; 7:e1274. [PMID: 38222813 PMCID: PMC10782051 DOI: 10.1002/jsp2.1274] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/29/2023] [Accepted: 07/02/2023] [Indexed: 01/16/2024] Open
Abstract
Background Intradiscal transplantation of mesenchymal stromal cells (MSCs) has emerged as a promising therapy for intervertebral disc degeneration (IDD). However, the hostile microenvironment of the intervertebral disc (IVD) may compromise the survival of implanted cells. Interestingly, studies reported that paracrine factors, such as extracellular vesicles (EVs) released by MSCs, may regenerate the IVD. The aim of this study was to investigate the therapeutic effects of Wharton's Jelly MSC (WJ-MSC)-derived EVs on human nucleus pulposus cells (hNPCs) using an in vitro 3D alginate-bead culture model. Methods After EV isolation and characterization, hNPCs isolated from surgical specimens were encapsulated in alginate beads and treated with 10, 50, and 100 μg/mL WJ-MSC-EVs. Cell proliferation and viability were assessed by flow cytometry and live/dead staining. Nitrite and glycosaminoglycan (GAG) content was evaluated through Griess and 1,9-dimethylmethylene blue assays. hNPCs in alginate beads were paraffin-embedded and stained for histological analysis (hematoxylin-eosin and Alcian blue) to assess extracellular matrix (ECM) composition. Gene expression levels of catabolic (MMP1, MMP13, ADAMTS5, IL6, NOS2), anabolic (ACAN), and hNPC marker (SOX9, KRT19) genes were analyzed through qPCR. Collagen type I and type II content was assessed with Western blot analysis. Results Treatment with WJ-MSC-EVs resulted in an increase in cell content and a decrease in cell death in degenerated hNPCs. Nitrite production was drastically reduced by EV treatment compared to the control. Furthermore, proteoglycan content was enhanced and confirmed by Alcian blue histological staining. EV stimulation attenuated ECM degradation and inflammation by suppressing catabolic and inflammatory gene expression levels. Additionally, NPC phenotypic marker genes were also maintained by the EV treatment. Conclusions WJ-MSC-derived EVs ameliorated hNPC growth and viability, and attenuated ECM degradation and oxidative stress, offering new opportunities for IVD regeneration as an attractive alternative strategy to cell therapy, which may be jeopardized by the harsh microenvironment of the IVD.
Collapse
Affiliation(s)
- Veronica Tilotta
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and SurgeryUniversità Campus Bio‐Medico di RomaRomeItaly
| | - Gianluca Vadalà
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and SurgeryUniversità Campus Bio‐Medico di RomaRomeItaly
- Operative Research Unit of Orthopaedic and Trauma SurgeryFondazione Policlinico Universitario Campus Bio‐MedicoRomeItaly
| | - Luca Ambrosio
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and SurgeryUniversità Campus Bio‐Medico di RomaRomeItaly
- Operative Research Unit of Orthopaedic and Trauma SurgeryFondazione Policlinico Universitario Campus Bio‐MedicoRomeItaly
| | - Giuseppina Di Giacomo
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and SurgeryUniversità Campus Bio‐Medico di RomaRomeItaly
| | - Claudia Cicione
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and SurgeryUniversità Campus Bio‐Medico di RomaRomeItaly
| | - Fabrizio Russo
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and SurgeryUniversità Campus Bio‐Medico di RomaRomeItaly
- Operative Research Unit of Orthopaedic and Trauma SurgeryFondazione Policlinico Universitario Campus Bio‐MedicoRomeItaly
| | - Adas Darinskas
- Laboratory of Immunology, National Cancer InstituteVilniusLithuania
- JSC Innovita Research, Tissue BankVilniusLithuania
| | - Rocco Papalia
- Laboratory for Regenerative Orthopaedics, Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and SurgeryUniversità Campus Bio‐Medico di RomaRomeItaly
- Operative Research Unit of Orthopaedic and Trauma SurgeryFondazione Policlinico Universitario Campus Bio‐MedicoRomeItaly
| | - Vincenzo Denaro
- Operative Research Unit of Orthopaedic and Trauma SurgeryFondazione Policlinico Universitario Campus Bio‐MedicoRomeItaly
| |
Collapse
|
35
|
Tashima T. Mesenchymal Stem Cell (MSC)-Based Drug Delivery into the Brain across the Blood-Brain Barrier. Pharmaceutics 2024; 16:289. [PMID: 38399342 PMCID: PMC10891589 DOI: 10.3390/pharmaceutics16020289] [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: 01/17/2024] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
At present, stem cell-based therapies using induced pluripotent stem cells (iPSCs) or mesenchymal stem cells (MSCs) are being used to explore the potential for regenerative medicine in the treatment of various diseases, owing to their ability for multilineage differentiation. Interestingly, MSCs are employed not only in regenerative medicine, but also as carriers for drug delivery, homing to target sites in injured or damaged tissues including the brain by crossing the blood-brain barrier (BBB). In drug research and development, membrane impermeability is a serious problem. The development of central nervous system drugs for the treatment of neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease, remains difficult due to impermeability in capillary endothelial cells at the BBB, in addition to their complicated pathogenesis and pathology. Thus, intravenously or intraarterially administered MSC-mediated drug delivery in a non-invasive way is a solution to this transendothelial problem at the BBB. Substances delivered by MSCs are divided into artificially included materials in advance, such as low molecular weight compounds including doxorubicin, and expected protein expression products of genetic modification, such as interleukins. After internalizing into the brain through the fenestration between the capillary endothelial cells, MSCs release their cargos to the injured brain cells. In this review, I introduce the potential and advantages of drug delivery into the brain across the BBB using MSCs as a carrier that moves into the brain as if they acted of their own will.
Collapse
Affiliation(s)
- Toshihiko Tashima
- Tashima Laboratories of Arts and Sciences, 1239-5 Toriyama-cho, Kohoku-ku, Yokohama 222-0035, Japan
| |
Collapse
|
36
|
dos Santos CC, Lopes-Pacheco M, English K, Rolandsson Enes S, Krasnodembskaya A, Rocco PRM. The MSC-EV-microRNAome: A Perspective on Therapeutic Mechanisms of Action in Sepsis and ARDS. Cells 2024; 13:122. [PMID: 38247814 PMCID: PMC10813908 DOI: 10.3390/cells13020122] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) and MSC-derived extracellular vesicles (EVs) have emerged as innovative therapeutic agents for the treatment of sepsis and acute respiratory distress syndrome (ARDS). Although their potential remains undisputed in pre-clinical models, this has yet to be translated to the clinic. In this review, we focused on the role of microRNAs contained in MSC-derived EVs, the EV microRNAome, and their potential contribution to therapeutic mechanisms of action. The evidence that miRNA transfer in MSC-derived EVs has a role in the overall therapeutic effects is compelling. However, several questions remain regarding how to reconcile the stochiometric issue of the low copy numbers of the miRNAs present in the EV particles, how different miRNAs delivered simultaneously interact with their targets within recipient cells, and the best miRNA or combination of miRNAs to use as therapy, potency markers, and biomarkers of efficacy in the clinic. Here, we offer a molecular genetics and systems biology perspective on the function of EV microRNAs, their contribution to mechanisms of action, and their therapeutic potential.
Collapse
Affiliation(s)
- Claudia C. dos Santos
- Institute of Medical Sciences and Interdepartmental Division of Critical Care, Department of Medicine, University of Toronto, Toronto, ON M5B 1T8, Canada
- Keenan Center for Biomedical Research, Unity Health Toronto, St. Michael’s Hospital, Toronto, ON M5B 1T8, Canada
| | - Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, 1749-016 Lisbon, Portugal;
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Karen English
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, W23 F2H6 Maynooth, Ireland;
- Department of Biology, Maynooth University, W23 F2H6 Maynooth, Ireland
| | - Sara Rolandsson Enes
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, 22184 Lund, Sweden;
| | - Anna Krasnodembskaya
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University of Belfast, Belfast BT9 7BL, UK;
| | - Patricia R. M. Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro 21941-599, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSaúde, Research Support Foundation of the State of Rio de Janeiro, Rio de Janeiro 20020-000, Brazil
| |
Collapse
|
37
|
Uysal O, Erybeh H, Canbek M, Ekenel EQ, Gunes S, Büyükköroğlu G, Semerci Sevimli T, Cemrek F, Sariboyaci AE. Stem Cell-Based or Cell-Free Gene Therapy in Chondrocyte Regeneration: Synovial Fluid-Derived Mesenchymal Stem Cell Exosomes. Curr Mol Med 2024; 24:906-919. [PMID: 37859306 PMCID: PMC11327740 DOI: 10.2174/0115665240266016231014081916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023]
Abstract
BACKGROUND Cartilage injuries are currently the most prevalent joint disease. Previous studies have emphasized the use of stem cells as the effective treatment for regenerating cartilage damage. OBJECTIVE In this study, considering the difficulties of the cellular therapy method, it was hypothesized that human synovial fluid-derived mesenchymal stem cell (hSFMSC) exosomes as a SC source could be used to treat these injuries as a safer and cell-free therapeutic alternative procedure due to its direct relevance to cartilage regeneration. Moreover, this study aimed to determine the miRNA and target genes required for the formation of SC treatment combined with gene therapy in order to reveal the mechanism of cartilage regeneration and increase its effectiveness. METHODS MSCs were characterized by flow cytometry, and immunocytochemical and differentiation analyses were done. To characterize functionally isolated exosomes, in vitro uptake analysis was performed. RT-qPCR was used to examine in terms of the advantages of cellular and cell-free therapy, mature human chondroblasts derived by differentiation from hSF-MSCs and human chondrocyte profiles were compared in order to demonstrate the above profile of hSF-MSCs and exosomes isolated from them, and the effectiveness of SC therapy in repairing cartilage damage. RESULTS According to our findings, the expression level of hsa-miR-155-5p was found to be considerably higher in chondrocytes differentiated from human synovial fluid MSCs than in mature human chondrocytes. These findings were also supported by the TGF-signalling pathway and chondrogenesis marker genes. CONCLUSION It was concluded that hSF-MSCs and exosomes can be used in the treatment of cartilage damage, and hsa-miR-155-5p can be used as a target miRNA in a new gene therapy approach because it increases the therapeutic effect on cartilage damage.
Collapse
Affiliation(s)
- Onur Uysal
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Eskisehir, Turkiye
| | - Haya Erybeh
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Molecular Biology, Science Faculty, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Mediha Canbek
- Department of Molecular Biology, Science Faculty, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Emilia Qomi Ekenel
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Sibel Gunes
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Eskisehir, Turkiye
| | - Gülay Büyükköroğlu
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Anadolu University, Eskisehir, Turkiye
| | - Tugba Semerci Sevimli
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Fatih Cemrek
- Department of Statistics, Faculty of Science and Letters, Eskisehir Osmangazi University, Eskisehir, Turkiye
| | - Ayla Eker Sariboyaci
- Cellular Therapy and Stem Cell Production Application and Research Centre, ESTEM, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Stem Cell, Institute of Health Sciences, Eskisehir Osmangazi University, Eskisehir, Turkiye
- Department of Medical Laboratory Techniques, Vocational School of Health Services, Eskisehir, Turkiye
| |
Collapse
|
38
|
Xining Z, Sai L. The Evolving Function of Vasculature and Pro-angiogenic Therapy in Fat Grafting. Cell Transplant 2024; 33:9636897241264976. [PMID: 39056562 PMCID: PMC11282510 DOI: 10.1177/09636897241264976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 07/28/2024] Open
Abstract
Autologous fat grating is a widely-accepted method to correct soft tissue deficiency. Although fat transplantation shows excellent biocompatibility and simple applicability, the relatively low retention rate caused by fat necrosis is still a challenge. The vasculature is integral after fat grafting, serving multiple crucial functions. Rapid and effective angiogenesis within grafts is essential for supplying oxygen necessary for adipocytes' survival. It facilitates the influx of inflammatory cells to remove necrotic adipocytes and aids in the delivery of regenerative cells for adipose tissue regeneration in fat grafts. The vasculature also provides a niche for interaction between adipose progenitor cells and vascular progenitor cells, enhancing angiogenesis and adipogenesis in grafts. Various methods, such as enriching grafts with diverse pro-angiogenic cells or utilizing cell-free approaches, have been employed to enhance angiogenesis. Beige and dedifferentiated adipocytes in grafts could increase vessel density. This review aims to outline the function of vasculature in fat grafting and discuss different cell or cell-free approaches that can enhance angiogenesis following fat grafting.
Collapse
Affiliation(s)
- Zhang Xining
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Luo Sai
- The Plastic and Aesthetic Center, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
39
|
Arte PA, Tungare K, Bhori M, Jobby R, Aich J. Treatment of type 2 diabetes mellitus with stem cells and antidiabetic drugs: a dualistic and future-focused approach. Hum Cell 2024; 37:54-84. [PMID: 38038863 DOI: 10.1007/s13577-023-01007-0] [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: 05/06/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023]
Abstract
Type 2 Diabetes Mellitus (T2DM) accounts for more than 90% of total diabetes mellitus cases all over the world. Obesity and lack of balance between energy intake and energy expenditure are closely linked to T2DM. Initial pharmaceutical treatment and lifestyle interventions can at times lead to remission but usually help alleviate it to a certain extent and the condition remains, thus, recurrent with the patient being permanently pharmaco-dependent. Mesenchymal stromal cells (MSCs) are multipotent, self-renewing cells with the ability to secrete a variety of biological factors that can help restore and repair injured tissues. MSC-derived exosomes possess these properties of the original stem cells and are potentially able to confer superior effects due to advanced cell-to-cell signaling and the presence of stem cell-specific miRNAs. On the other hand, the repository of antidiabetic agents is constantly updated with novel T2DM disease-modifying drugs, with higher efficacy and increasingly convenient delivery protocols. Delving deeply, this review details the latest progress and ongoing studies related to the amalgamation of stem cells and antidiabetic drugs, establishing how this harmonized approach can exert superior effects in the management and potential reversal of T2DM.
Collapse
Affiliation(s)
- Priyamvada Amol Arte
- School of Biotechnology and Bioinformatics, DY Patil Deemed to Be University, CBD Belapur, Navi Mumbai, Maharashtra, 400614, India.
- Anatek Services PVT LTD, Sai Chamber, 10, Near Santacruz Railway Bridge, Sen Nagar, Santacruz East, Mumbai, Maharashtra, 400055, India.
| | - Kanchanlata Tungare
- School of Biotechnology and Bioinformatics, DY Patil Deemed to Be University, CBD Belapur, Navi Mumbai, Maharashtra, 400614, India
| | - Mustansir Bhori
- Inveniolife Technology PVT LTD, Office No.118, Grow More Tower, Plot No.5, Sector 2, Kharghar, Navi Mumbai, Maharashtra, 410210, India
| | - Renitta Jobby
- Amity Institute of Biotechnology, Amity University Maharashtra, Mumbai-Pune Expressway, Bhatan, Panvel, Navi Mumbai, Maharashtra, 410206, India
- Amity Centre of Excellence in Astrobiology, Amity University Maharashtra, Mumbai-Pune Expressway, Bhatan, Panvel, Navi Mumbai, Maharashtra, 410206, India
| | - Jyotirmoi Aich
- School of Biotechnology and Bioinformatics, DY Patil Deemed to Be University, CBD Belapur, Navi Mumbai, Maharashtra, 400614, India
| |
Collapse
|
40
|
Ghosh M, Pearse DD. Schwann Cell-Derived Exosomal Vesicles: A Promising Therapy for the Injured Spinal Cord. Int J Mol Sci 2023; 24:17317. [PMID: 38139147 PMCID: PMC10743801 DOI: 10.3390/ijms242417317] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/02/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Exosomes are nanoscale-sized membrane vesicles released by cells into their extracellular milieu. Within these nanovesicles reside a multitude of bioactive molecules, which orchestrate essential biological processes, including cell differentiation, proliferation, and survival, in the recipient cells. These bioactive properties of exosomes render them a promising choice for therapeutic use in the realm of tissue regeneration and repair. Exosomes possess notable positive attributes, including a high bioavailability, inherent safety, and stability, as well as the capacity to be functionalized so that drugs or biological agents can be encapsulated within them or to have their surface modified with ligands and receptors to imbue them with selective cell or tissue targeting. Remarkably, their small size and capacity for receptor-mediated transcytosis enable exosomes to cross the blood-brain barrier (BBB) and access the central nervous system (CNS). Unlike cell-based therapies, exosomes present fewer ethical constraints in their collection and direct use as a therapeutic approach in the human body. These advantageous qualities underscore the vast potential of exosomes as a treatment option for neurological injuries and diseases, setting them apart from other cell-based biological agents. Considering the therapeutic potential of exosomes, the current review seeks to specifically examine an area of investigation that encompasses the development of Schwann cell (SC)-derived exosomal vesicles (SCEVs) as an approach to spinal cord injury (SCI) protection and repair. SCs, the myelinating glia of the peripheral nervous system, have a long history of demonstrated benefit in repair of the injured spinal cord and peripheral nerves when transplanted, including their recent advancement to clinical investigations for feasibility and safety in humans. This review delves into the potential of utilizing SCEVs as a therapy for SCI, explores promising engineering strategies to customize SCEVs for specific actions, and examines how SCEVs may offer unique clinical advantages over SC transplantation for repair of the injured spinal cord.
Collapse
Affiliation(s)
- Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
41
|
Yang F, Li Z, Cai Z, He Y, Ke C, Wang J, Lin M, Li L. Pluronic F-127 Hydrogel Loaded with Human Adipose-Derived Stem Cell-Derived Exosomes Improve Fat Graft Survival via HIF-1α-Mediated Enhancement of Angiogenesis. Int J Nanomedicine 2023; 18:6781-6796. [PMID: 38026529 PMCID: PMC10658963 DOI: 10.2147/ijn.s435106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/09/2023] [Indexed: 12/01/2023] Open
Abstract
Purpose Autologous fat grafting is playing an increasingly important role in plastic surgery. However, high absorption and low survival of autologous fat grafts limit their clinical application. This study aimed to investigate whether human adipose-derived stem cell-derived exosomes (hASC-Exos) encapsulated in a PF-127 hydrogel can improve the survival of autologous fat grafts and to elucidate the underlying mechanisms. Patients and Methods Exosomes were isolated from hASCs and identified using transmission electron microscopy, nanoparticle tracking analysis and Western blotting. We performed functional assays in vitro to assess the effect of hASC-Exos on proliferation, migration, and tube formation as well as their regulatory role in the HIF-1α/VEGF signaling pathway. hASC-Exos encapsulated in the PF-127 hydrogel were used as an in vivo autologous fat graft model. The effects of the PF-127 hydrogel/hASC-Exos and the role of the HIF-1α/VEGF signaling pathway in promoting angiogenesis in an autologous fat grafting model were assessed. Results hASC-Exos were taken up by human umbilical vein endothelial cells and enhanced their proliferation, migration, and tubule formation in vitro. The effects of hASC-Exos on promoting angiogenesis were mediated by the HIF-1α/VEGF signaling pathway. Moreover, we fabricated a PF-127 hydrogel for the sustained release of hASC-Exos, and in vivo results showed that hASC-Exos encapsulated in PF-127 hydrogel improved the survival of autologous fat grafts. Conclusion Our findings indicated that hASC-Exos encapsulated in PF-127 hydrogel serve as a key regulator of angiogenesis by activating the HIF-1α/VEGF signaling pathway and provide a promising strategy for autologous fat grafting treatment.
Collapse
Affiliation(s)
- Fangfang Yang
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Zihao Li
- Department of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Zhongming Cai
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Yucang He
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Chen Ke
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Jingping Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Ming Lin
- Department of Obstetrics and Gynecology, Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| | - Liqun Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
42
|
Wang YH, Chen EQ. Mesenchymal Stem Cell Therapy in Acute Liver Failure. Gut Liver 2023; 17:674-683. [PMID: 36843422 PMCID: PMC10502502 DOI: 10.5009/gnl220417] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/04/2022] [Accepted: 11/18/2022] [Indexed: 02/28/2023] Open
Abstract
Acute liver failure (ALF) is a severe liver disease syndrome with rapid deterioration and high mortality. Liver transplantation is the most effective treatment, but the lack of donor livers and the high cost of transplantation limit its broad application. In recent years, there has been no breakthrough in the treatment of ALF, and the application of stem cells in the treatment of ALF is a crucial research field. Mesenchymal stem cells (MSCs) are widely used in disease treatment research due to their abundant sources, low immunogenicity, and no ethical restrictions. Although MSCs are effective for treating ALF, the application of MSCs to ALF needs to be further studied and optimized. In this review, we discuss the potential mechanisms of MSCs therapy for ALF, summarize some methods to enhance the efficacy of MSCs, and explore optimal approaches for MSC transplantation.
Collapse
Affiliation(s)
- Yong-Hong Wang
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - En-Qiang Chen
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
43
|
Mori T, Giovannelli L, Bilia AR, Margheri F. Exosomes: Potential Next-Generation Nanocarriers for the Therapy of Inflammatory Diseases. Pharmaceutics 2023; 15:2276. [PMID: 37765245 PMCID: PMC10537720 DOI: 10.3390/pharmaceutics15092276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/28/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Inflammatory diseases are common pathological processes caused by various acute and chronic factors, and some of them are autoimmune diseases. Exosomes are fundamental extracellular vesicles secreted by almost all cells, which contain a series of constituents, i.e., cytoskeletal and cytosolic proteins (actin, tubulin, and histones), nucleic acids (mRNA, miRNA, and DNA), lipids (diacylglycerophosphates, cholesterol, sphingomyelin, and ceramide), and other bioactive components (cytokines, signal transduction proteins, enzymes, antigen presentation and membrane transport/fusion molecules, and adhesion molecules). This review will be a synopsis of the knowledge on the contribution of exosomes from different cell sources as possible therapeutic agents against inflammation, focusing on several inflammatory diseases, neurological diseases, rheumatoid arthritis and osteoarthritis, intestinal bowel disease, asthma, and liver and kidney injuries. Current knowledge indicates that the role of exosomes in the therapy of inflammation and in inflammatory diseases could be distinctive. The main limitations to their clinical translation are still production, isolation, and storage. Additionally, there is an urgent need to personalize the treatments in terms of the selection of exosomes; their dosages and routes of administration; and a deeper knowledge about their biodistribution, type and incidence of adverse events, and long-term effects of exosomes. In conclusion, exosomes can be a very promising next-generation therapeutic option, superior to synthetic nanocarriers and cell therapy, and can represent a new strategy of effective, safe, versatile, and selective delivery systems in the future.
Collapse
Affiliation(s)
- Tosca Mori
- Department of Chemistry “Ugo Schiff” (DICUS), University of Florence, Via Ugo Schiff 6, Sesto Fiorentino, 50019 Florence, Italy;
| | - Lisa Giovannelli
- Department of Neurosciences (Department of Neurosciences, Psychology, Drug Research and Child Health), University of Florence, 50139 Florence, Italy
| | - Anna Rita Bilia
- Department of Chemistry “Ugo Schiff” (DICUS), University of Florence, Via Ugo Schiff 6, Sesto Fiorentino, 50019 Florence, Italy;
| | - Francesca Margheri
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50121 Florence, Italy;
| |
Collapse
|
44
|
Xiang H, Xu S, Zhang W, Xue X, Li Y, Lv Y, Chen J, Miao X. Dissolving microneedles for alopecia treatment. Colloids Surf B Biointerfaces 2023; 229:113475. [PMID: 37536169 DOI: 10.1016/j.colsurfb.2023.113475] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/20/2023] [Accepted: 07/22/2023] [Indexed: 08/05/2023]
Abstract
Alopecia is a treatable benign disease, however, approximately 15-30% of women and 50% of men suffer from alopecia, which greatly affects patient's self-esteem and quality of life. Currently, commercial products for alopecia treatment include topical minoxidil solution, oral finasteride tablets and oral baricitinib tablets. However, the barrier of stratum corneum, systemic adverse effects and poor cure rate limit the application of commercial products. Therefore, researchers investigated the mechanism of alopecia, and developed new drugs that could target lactate dehydrogenase-related pathways, remove excessive reactive oxygen in hair follicles, and reduce the escape of hair follicle stem cells, thus injecting new strength into the treatment of alopecia. Moreover, starting from improving drug stratum corneum penetration and reducing side effects, researchers have developed hair loss treatment strategies based on dissolved microneedles (MNs), such as drug powders/microparticles, nanoparticles, biomimetic cell membranes, phototherapy and magnetically responsive soluble microneedles, which show exciting alopecia treatment effects. However, there are still some challenges in the practical application of the current alopecia treatment strategy with soluble microneedles, and further studies are needed to accelerate its clinical translation.
Collapse
Affiliation(s)
- Hong Xiang
- Marine College, Shandong University, Weihai 264209, China
| | - Sai Xu
- Marine College, Shandong University, Weihai 264209, China
| | - Weiwei Zhang
- Drug Research and Development Center, Shandong Drug and Food Vocational College, Weihai 264209, China
| | - Xinyue Xue
- Marine College, Shandong University, Weihai 264209, China
| | - Yixuan Li
- Marine College, Shandong University, Weihai 264209, China
| | - Yanyu Lv
- Drug Research and Development Center, Shandong Drug and Food Vocational College, Weihai 264209, China
| | - Jing Chen
- Marine College, Shandong University, Weihai 264209, China
| | - Xiaoqing Miao
- Marine College, Shandong University, Weihai 264209, China.
| |
Collapse
|
45
|
Pavelić K, Pavelić SK, Bulog A, Agaj A, Rojnić B, Čolić M, Trivanović D. Nanoparticles in Medicine: Current Status in Cancer Treatment. Int J Mol Sci 2023; 24:12827. [PMID: 37629007 PMCID: PMC10454499 DOI: 10.3390/ijms241612827] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Cancer is still a leading cause of deaths worldwide, especially due to those cases diagnosed at late stages with metastases that are still considered untreatable and are managed in such a way that a lengthy chronic state is achieved. Nanotechnology has been acknowledged as one possible solution to improve existing cancer treatments, but also as an innovative approach to developing new therapeutic solutions that will lower systemic toxicity and increase targeted action on tumors and metastatic tumor cells. In particular, the nanoparticles studied in the context of cancer treatment include organic and inorganic particles whose role may often be expanded into diagnostic applications. Some of the best studied nanoparticles include metallic gold and silver nanoparticles, quantum dots, polymeric nanoparticles, carbon nanotubes and graphene, with diverse mechanisms of action such as, for example, the increased induction of reactive oxygen species, increased cellular uptake and functionalization properties for improved targeted delivery. Recently, novel nanoparticles for improved cancer cell targeting also include nanobubbles, which have already demonstrated increased localization of anticancer molecules in tumor tissues. In this review, we will accordingly present and discuss state-of-the-art nanoparticles and nano-formulations for cancer treatment and limitations for their application in a clinical setting.
Collapse
Affiliation(s)
- Krešimir Pavelić
- Faculty of Medicine, Juraj Dobrila University of Pula, Zagrebačka 30, 52100 Pula, Croatia
| | - Sandra Kraljević Pavelić
- Faculty of Health Studies, University of Rijeka, Ulica Viktora Cara Emina 5, 51000 Rijeka, Croatia
| | - Aleksandar Bulog
- Teaching Institute for Public Health of Primorsko-Goranska County, Krešimirova Ulica 52, 51000 Rijeka, Croatia
- Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Andrea Agaj
- Faculty of Medicine, Juraj Dobrila University of Pula, Zagrebačka 30, 52100 Pula, Croatia
| | - Barbara Rojnić
- Faculty of Medicine, Juraj Dobrila University of Pula, Zagrebačka 30, 52100 Pula, Croatia
| | - Miroslav Čolić
- Clear Water Technology Inc., 13008 S Western Avenue, Gardena, CA 90429, USA;
| | - Dragan Trivanović
- Faculty of Medicine, Juraj Dobrila University of Pula, Zagrebačka 30, 52100 Pula, Croatia
- Department of Oncology and Hematology, General Hospital Pula, Santorijeva 24a, 52200 Pula, Croatia
| |
Collapse
|
46
|
Abudurexiti M, Zhao Y, Wang X, Han L, Liu T, Wang C, Yuan Z. Bio-Inspired Nanocarriers Derived from Stem Cells and Their Extracellular Vesicles for Targeted Drug Delivery. Pharmaceutics 2023; 15:2011. [PMID: 37514197 PMCID: PMC10386614 DOI: 10.3390/pharmaceutics15072011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
With their seemingly limitless capacity for self-improvement, stem cells have a wide range of potential uses in the medical field. Stem-cell-secreted extracellular vesicles (EVs), as paracrine components of stem cells, are natural nanoscale particles that transport a variety of biological molecules and facilitate cell-to-cell communication which have been also widely used for targeted drug delivery. These nanocarriers exhibit inherent advantages, such as strong cell or tissue targeting and low immunogenicity, which synthetic nanocarriers lack. However, despite the tremendous therapeutic potential of stem cells and EVs, their further clinical application is still limited by low yield and a lack of standardized isolation and purification protocols. In recent years, inspired by the concept of biomimetics, a new approach to biomimetic nanocarriers for drug delivery has been developed through combining nanotechnology and bioengineering. This article reviews the application of biomimetic nanocarriers derived from stem cells and their EVs in targeted drug delivery and discusses their advantages and challenges in order to stimulate future research.
Collapse
Affiliation(s)
- Munire Abudurexiti
- College of Pharmacy, Southwest Minzu University, Chendu 610041, China; (M.A.); (X.W.); (L.H.)
| | - Yue Zhao
- Department of Pharmacy, Sichuan Tianfu New Area People’s Hospital, Chengdu 610213, China;
| | - Xiaoling Wang
- College of Pharmacy, Southwest Minzu University, Chendu 610041, China; (M.A.); (X.W.); (L.H.)
| | - Lu Han
- College of Pharmacy, Southwest Minzu University, Chendu 610041, China; (M.A.); (X.W.); (L.H.)
| | - Tianqing Liu
- NICM Health Research Institute, Western Sydney University, Westmead 2145, Australia;
| | - Chengwei Wang
- Division of Internal Medicine, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhixiang Yuan
- College of Pharmacy, Southwest Minzu University, Chendu 610041, China; (M.A.); (X.W.); (L.H.)
| |
Collapse
|
47
|
Lee BW, Kwok SK. Mesenchymal Stem/Stromal Cell-Based Therapies in Systemic Rheumatic Disease: From Challenges to New Approaches for Overcoming Restrictions. Int J Mol Sci 2023; 24:10161. [PMID: 37373308 PMCID: PMC10299481 DOI: 10.3390/ijms241210161] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/10/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Systemic rheumatic diseases, such as rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis, are chronic autoimmune diseases affecting multiple organs and tissues. Despite recent advances in treatment, patients still experience significant morbidity and disability. Mesenchymal stem/stromal cell (MSC)-based therapy is promising for treating systemic rheumatic diseases due to the regenerative and immunomodulatory properties of MSCs. However, several challenges need to be overcome to use MSCs in clinical practice effectively. These challenges include MSC sourcing, characterization, standardization, safety, and efficacy issues. In this review, we provide an overview of the current state of MSC-based therapies in systemic rheumatic diseases, highlighting the challenges and limitations associated with their use. We also discuss emerging strategies and novel approaches that can help overcome the limitations. Finally, we provide insights into the future directions of MSC-based therapies for systemic rheumatic diseases and their potential clinical applications.
Collapse
Affiliation(s)
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| |
Collapse
|
48
|
Kim IY, Kim HY, Song HW, Park JO, Choi YH, Choi E. Functional enhancement of exosomes derived from NK cells by IL-15 and IL-21 synergy against hepatocellular carcinoma cells: The cytotoxicity and apoptosis in vitro study. Heliyon 2023; 9:e16962. [PMID: 37484408 PMCID: PMC10361042 DOI: 10.1016/j.heliyon.2023.e16962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 07/25/2023] Open
Abstract
Exosomes are released by various cells, including natural killer (NK) cells and transport signaling molecules for the intercellular communication. Hepatocellular carcinoma (HCC), also known as primary liver cancer, is often inoperable and difficult to accurate diagnosis. Notably, the prognosis and underlying mechanisms of HCC are not fully understood. Exosomes-derived NK cells (NK-exos) express unique cytotoxic proteins with a killing ability in tumors and can easily penetrate tumor tissues to improve their targeting ability. NK cell functions, inducing cellular cytotoxicity are modulated by cytokines such as interleukin (IL)-15 and IL-21. However, the mechanisms and effects of cytokines-stimulated NK-exos for the treatment of liver cancer, including HCC, are not well known. In this study, we aimed to investigate the synergistic anti-tumor effects of NK-exos stimulated with IL-15 and IL-21 (NK-exosIL-15/21) in Hep3B cells. Our findings revealed that NK-exosIL-15/21 expressed cytotoxic proteins (perforin and granzyme B) and contained typical exosome markers (CD9 and CD63) within the size range of 100-150 nm. Moreover, we demonstrated that NK-exosIL-15/21 induced the enhancement of cytotoxicity and apoptotic activity in Hep3B cells by activating the specific pro-apoptotic proteins (Bax, cleaved caspase 3, cleaved PARP, perforin, and granzyme B) and inhibiting the anti-apoptotic protein (Bcl-2). In summary, our results suggest that NK-exosIL-15/21 regulate strong anti-tumor effects of HCC cells, by increasing the cytotoxicity and apoptosis through the activation of specific cytotoxic molecules.
Collapse
Affiliation(s)
- In-Young Kim
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - Ho Yong Kim
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - Hyeong-woo Song
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - Jong-Oh Park
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - You Hee Choi
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
| | - Eunpyo Choi
- Korea Institute of Medical Microrobotics, 43-26 Cheomdangwagi-ro, Buk-gu, Gwangju 61011, Republic of Korea
- School of Mechanical Engineering, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea
| |
Collapse
|
49
|
Wang H, Wang J, Liu T, Leng Y, Yang W. Stem cell-derived exosomal MicroRNAs: Potential therapies in diabetic kidney disease. Biomed Pharmacother 2023; 164:114961. [PMID: 37257230 DOI: 10.1016/j.biopha.2023.114961] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/23/2023] [Accepted: 05/27/2023] [Indexed: 06/02/2023] Open
Abstract
The diabetic kidney disease (DKD) is chronic kidney disease caused by diabetes and one of the most common comorbidities. It is often more difficult to treat end-stage renal disease once it develops because of its complex metabolic disorders, so early prevention and treatment are important. However, currently available DKD therapies are not ideal, and novel therapeutic strategies are urgently needed. The potential of stem cell therapies partly depends on their ability to secrete exosomes. More and more studies have shown that stem cell-derived exosomes take part in the DKD pathophysiological process, which may offer an effective therapy for DKD treatment. Herein, we mainly review potential therapies of stem cell-derived exosomes mainly stem cell-derived exosomal microRNAs in DKD, including their protective effects on mesangial cells, podocytes and renal tubular epithelial cells. Using this secretome as possible therapeutic drugs without potential carcinogenicity should be the focus of further research.
Collapse
Affiliation(s)
- Han Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Jiajia Wang
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Tiejun Liu
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yan Leng
- Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130021, China
| | - Weipeng Yang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
50
|
Li J, Huang Y, Sun H, Yang L. Mechanism of mesenchymal stem cells and exosomes in the treatment of age-related diseases. Front Immunol 2023; 14:1181308. [PMID: 37275920 PMCID: PMC10232739 DOI: 10.3389/fimmu.2023.1181308] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/08/2023] [Indexed: 06/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) from multiple tissues have the capability of multidirectional differentiation and self-renewal. Many reports indicated that MSCs exert curative effects on a variety of age-related diseases through regeneration and repair of aging cells and organs. However, as research has progressed, it has become clear that it is the MSCs derived exosomes (MSC-Exos) that may have a real role to play, and that they can be modified to achieve better therapeutic results, making them even more advantageous than MSCs for treating disease. This review generalizes the biological characteristics of MSCs and exosomes and their mechanisms in treating age-related diseases, for example, MSCs and their exosomes can treat age-related diseases through mechanisms such as oxidative stress (OS), Wnt/β-catenin signaling pathway, mitogen-activated protein kinases (MAPK) signaling pathway, and so on. In addition, current in vivo and in vitro trials are described, and ongoing clinical trials are discussed, as well as the prospects and challenges for the future use of exosomes in disease treatment. This review will provide references for using exosomes to treat age-related diseases.
Collapse
Affiliation(s)
- Jia Li
- Departments of Geriatrics, The First Hospital of China Medical University, Shenyang, China
| | - Yuling Huang
- Departments of Geriatrics, The First Hospital of China Medical University, Shenyang, China
| | - Haiyan Sun
- Department of Endodontics, School of Stomatology, China Medical University, Shenyang, China
| | - Lina Yang
- Departments of Geriatrics, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|