1
|
Fayazi M, Rostami M, Amiri Moghaddam M, Nasiri K, Tadayonfard A, Roudsari MB, Ahmad HM, Parhizgar Z, Majbouri Yazdi A. A state-of-the-art review of the recent advances in drug delivery systems for different therapeutic agents in periodontitis. J Drug Target 2025; 33:612-647. [PMID: 39698877 DOI: 10.1080/1061186x.2024.2445051] [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: 10/07/2024] [Revised: 12/08/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
Periodontitis (PD) is a chronic gum illness that may be hard to cure for a number of reasons, including the fact that no one knows what causes it, the side effects of anti-microbial treatment, and how various kinds of bacteria interact with one another. As a result, novel therapeutic approaches for PD treatment must be developed. Additionally, supplementary antibacterial regimens, including local and systemic medication administration of chemical agents, are necessary for deep pockets to assist with mechanical debridement of tooth surfaces. As our knowledge of periodontal disease and drug delivery systems (DDSs) grows, new targeted delivery systems like extracellular vesicles, lipid-based nanoparticles (NPs), metallic NPs, and polymer NPs have been developed. These systems aim to improve the targeting and precision of PD treatments while reducing the systemic side effects of antibiotics. Nanozymes, photodermal therapy, antibacterial metallic NPs, and traditional PD therapies have all been reviewed in this research. Medicinal herbs, antibiotics, photothermal therapy, nanozymes, antibacterial metallic NPs, and conventional therapies for PD have all been examined in this research. After that, we reviewed the key features of many innovative DDSs and how they worked for PD therapy. Finally, we have discussed the advantages and disadvantages of these DDSs.
Collapse
Affiliation(s)
- Mehrnaz Fayazi
- School of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Mitra Rostami
- School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Kamyar Nasiri
- Department of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Azadeh Tadayonfard
- Department of Prosthodontics, Dental Faculty, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Behnam Roudsari
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Zahra Parhizgar
- Resident of Periodontology, Department of Periodontics, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|
2
|
Dey S, Dinakar YH, R S, Jain V, Jain R. Navigating the therapeutic landscape for breast cancer: targeting breast cancer stem cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2387-2406. [PMID: 39441235 DOI: 10.1007/s00210-024-03542-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024]
Abstract
Breast cancer is a common and deadly malignancy that affects women globally, and breast cancer stem cells (BCSCs) play an important role in tumorigenesis, development, metastasis, and recurrence. Traditional therapies often fail to eliminate BCSCs, leading to treatment resistance and relapse. This review explores the therapeutic strategies which are designed to target BCSCs, including inhibition of key signaling pathway and targeting receptor. This paper also explores the approaches to targeting BCSCs including chemotherapy, phytomedicines, and nanotechnology. Nanotechnology has gained a lot of importance in cancer therapy because of its ability to deliver therapeutic agents with more precision and minimal side effects. Various chemotherapeutic drugs, siRNAs, or gene editing tools are delivered efficiently with the use of nanocarriers which target pathways, receptors, and proteins associated with BCSCs. Over the past few years, stimuli-responsive and receptor-targeted nanocarriers have been explored for better therapeutic effects. In recent times, strategies such as chimeric antigen receptor (CAR) T-cell therapy, ablation therapy, and cell-free therapies are explored for targeting these stem cells. This review provides a recent developmental overview of strategies to attack BCSCs from conventional chemotherapeutic agents to nanotechnological platforms such as polymeric, lipidic, and metal-based nanoparticles and advanced technologies like CAR T cell therapies.
Collapse
Affiliation(s)
- Soudeep Dey
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Yirivinti Hayagreeva Dinakar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Soundarya R
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India.
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, 570015, India.
| |
Collapse
|
3
|
Chen Q, Yu T, Gong J, Shan H. Advanced Nanomedicine Delivery Systems for Cardiovascular Diseases: Viral and Non-Viral Strategies in Targeted Therapy. Molecules 2025; 30:962. [PMID: 40005272 PMCID: PMC11858567 DOI: 10.3390/molecules30040962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/08/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiovascular diseases (CVDs) represent a leading global health crisis, significantly impairing patients' quality of life and posing substantial risks to their survival. Conventional therapies for CVDs often grapple with challenges such as inadequate targeting precision, suboptimal therapeutic efficacy, and potential adverse side effects. To address these shortcomings, researchers are intensively developing advanced drug delivery systems characterized by high specificity and selectivity, excellent biodegradability, superior biocompatibility, and minimal toxicity. These innovative systems enable the precise delivery of pharmaceuticals with high drug-loading capacities, minimal leakage, and expansive specific surface areas, thereby enhancing therapeutic outcomes. In this review, we summarize and classify various drug delivery materials targeting CVDs and application values. We also evaluate the feasibility and efficacy of viral and non-viral vectors for the treatment of CVDs, the existing limitations and application prospects are also discussed. We hope that this review will provide new perspectives for the future development of drug delivery systems for the treatment of CVDs, ultimately contributing to improved patient care and outcomes.
Collapse
Affiliation(s)
| | - Tong Yu
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China; (Q.C.); (J.G.)
| | | | - Hongli Shan
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China; (Q.C.); (J.G.)
| |
Collapse
|
4
|
Dong Z, Fu Y, Cai Z, Dai H, He Y. Recent advances in adipose-derived mesenchymal stem cell-derived exosomes for regulating macrophage polarization. Front Immunol 2025; 16:1525466. [PMID: 39963133 PMCID: PMC11830602 DOI: 10.3389/fimmu.2025.1525466] [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: 11/09/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025] Open
Abstract
Adipose-derived mesenchymal stem cells (ADSCs) exhibit superior immunomodulatory properties and have broad therapeutic applications. They induce macrophage M2 polarization for anti-inflammatory responses. Exosomes derived from ADSCs (ADSC-EXOs) exhibit biological functions similar to those of ADSCs but can circumvent the limitations associated with cellular injection therapies. Potent anti-inflammatory substances contained in exosomes include the glycoprotein MFGE8, the cytokines such as prostaglandin E2, IL-6, and IGF, as well as non-coding nucleotides (miR-451a, miR-23, miR-30d-5p, let-7, lncRNA DLEU2, circRps5, Circ-Ptpn4, and mmu_ circ_0001359). The anti-inflammatory and immunomodulatory properties of these exosomes provide new perspectives for therapeutic approaches for graft inflammation, bone healing, acute lung injury, kidney stones, myocardial infarction, and diabetes-related diseases. This review summarizes the contents and functions of ADSC-EXOs, outlines their properties and the characteristics of macrophage phenotypes, and emphasizes their impact on macrophage polarization and their contribution to immune-related diseases.
Collapse
Affiliation(s)
- Zhewei Dong
- Renji College, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yingli Fu
- Department of Plastic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhongming Cai
- Department of Breast Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hao Dai
- Department of Plastic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yucang He
- Department of Plastic Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
5
|
Rached RD, Helfenstein T, Kim AH, Da Costa TR, Araújo RL. Mesenchymal Stem Cell Therapy for Superior Gluteal Nerve Injury Post-hip Arthroplasty: A Case Report. Cureus 2025; 17:e79088. [PMID: 40109801 PMCID: PMC11920847 DOI: 10.7759/cureus.79088] [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] [Accepted: 02/15/2025] [Indexed: 03/22/2025] Open
Abstract
Peripheral nerve injuries, such as axonal injury of the superior gluteal nerve, are rare but debilitating complications that can occur after hip arthroplasty. This case report describes the use of adipose-derived mesenchymal stem cells (MSCs) to treat an axonal injury of the superior gluteal nerve in a 71-year-old patient. After conventional rehabilitation failed, MSC infiltration was chosen and performed with ultrasound (US) guidance. Two months later, the patient showed normalization of electromyography (EMG), indicating full nerve recovery, along with significant improvement in neuropathic pain. The patient also demonstrated a 55% increase in maximum torque and a 9% increase in power during right hip extension in isokinetic evaluation, resulting in improvement of muscle strength and functionality. This case highlights the potential of MSCs in promoting nerve regeneration, suggesting that this approach may accelerate nerve recovery and improve short-term clinical outcomes. Although the results are promising, further studies are needed to confirm the efficacy and safety of this treatment in a larger population. This integrated model of cell therapy and physical rehabilitation represents a significant advance in recovering from complex nerve injuries.
Collapse
Affiliation(s)
- Roberto D Rached
- Physical Medicine and Rehabilitation, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRA
| | - Thomas Helfenstein
- Physical Medicine and Rehabilitation, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRA
| | - Angela H Kim
- Physical Medicine and Rehabilitation, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRA
| | - Thadeu R Da Costa
- Physical Medicine and Rehabilitation, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRA
| | - Ricardo L Araújo
- Physical Medicine and Rehabilitation, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, BRA
| |
Collapse
|
6
|
Wu KC, Yang HI, Chang YH, Chiang RYS, Ding DC. Extracellular Vesicles Derived from Human Umbilical Mesenchymal Stem Cells Transfected with miR-7704 Improved Damaged Cartilage and Reduced Matrix Metallopeptidase 13. Cells 2025; 14:82. [PMID: 39851510 PMCID: PMC11763736 DOI: 10.3390/cells14020082] [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: 12/24/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
We aimed to explore the therapeutic efficacy of miR-7704-modified extracellular vesicles (EVs) derived from human umbilical cord mesenchymal stem cells (HUCMSCs) for osteoarthritis (OA) treatment. In vitro experiments demonstrated the successful transfection of miR-7704 into HUCMSCs and the isolation of EVs from these cells. In vivo experiments used an OA mouse model to assess the effects of the injection of miR-7704-modified EVs intra-articularly. Walking capacity (rotarod test), cartilage morphology, histological scores, and the expression of type II collagen, aggrecan, interleukin-1 beta, and matrix metalloproteinase 13 (MMP13) in the cartilage were evaluated. The EVs were characterized to confirm their suitability for therapeutic use. IL-1beta-treated chondrocytes increased type II collagen and decreased MMP13 after treatment with miR-7704-overexpressed EVs. In vivo experiments revealed that an intra-articular injection of miR-7704-overexpressed EVs significantly improved walking capacity, preserved cartilage morphology, and resulted in higher histological scores compared to in the controls. Furthermore, the decreased expression of MMP13 in the cartilage post treatment suggests a potential mechanism for the observed therapeutic effects. Therefore, miR-7704-overexpressed EVs derived from HUCMSCs showed potential as an innovative therapeutic strategy for treating OA. Further investigations should focus on optimizing dosage, understanding mechanisms, ensuring safety and efficacy, developing advanced delivery systems, and conducting early-phase clinical trials to establish the therapeutic potential of HUCMSC-derived EVs for OA management.
Collapse
Affiliation(s)
- Kun-Chi Wu
- Department of Orthopedics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Hui-I Yang
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan;
| | - Yu-Hsun Chang
- Department of Pediatrics, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Raymond Yuh-Shyan Chiang
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
| | - Dah-Ching Ding
- Department of Obstetrics and Gynecology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien 970, Taiwan;
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
7
|
Deng Z, Iwasaki K, Peng Y, Honda Y. Mesenchymal Stem Cell Extract Promotes Skin Wound Healing. Int J Mol Sci 2024; 25:13745. [PMID: 39769505 PMCID: PMC11679360 DOI: 10.3390/ijms252413745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Recently, it has been reported that mesenchymal stem cell (MSC)-derived humoral factors promote skin wound healing. As these humoral factors are transiently stored in cytoplasm, we collected them as part of the cell extracts from MSCs (MSC-ext). This study aimed to investigate the effects of MSC-ext on skin wound healing. We examined the effects of MSC-ext on cell proliferation and migration. Additionally, the effect of MSC-ext on skin wound healing was evaluated using a mouse skin defect model. The MSC-ext enhanced the proliferation of dermal fibroblasts, epithelial cells, and endothelial cells. It also increased the number of migrating fibroblasts and epithelial cells. The skin defects treated with MSC-ext demonstrated rapid wound closure compared to those treated with phosphate-buffered saline. The MSC-ext group exhibited a thicker dermis, larger Picrosirius red-positive areas, and a higher number of Ki67-positive cells. Our results indicate that MSC-ext promotes the proliferation and/or migration of fibroblasts, epithelial cells, and endothelial cells, and enhances skin wound healing. This suggests the therapeutic potential of MSC-ext in treating skin defects as a novel cell-free treatment modality.
Collapse
Affiliation(s)
- Zi Deng
- Department of Oral Anatomy, Osaka Dental University, Osaka 573-1121, Japan; (Z.D.); (Y.H.)
| | - Kengo Iwasaki
- Advanced Medicine Research Center, Translational Research Institute for Medical Innovation (TRIMI), Osaka Dental University, Osaka 573-1121, Japan
| | - Yihao Peng
- Department of Periodontology, Osaka Dental University, Osaka 573-1121, Japan;
| | - Yoshitomo Honda
- Department of Oral Anatomy, Osaka Dental University, Osaka 573-1121, Japan; (Z.D.); (Y.H.)
| |
Collapse
|
8
|
Qu Q, Liu L, Wang L, Cui Y, Liu C, Jing X, Xu X. Exosomes derived from hypoxic mesenchymal stem cells restore ovarian function by enhancing angiogenesis. Stem Cell Res Ther 2024; 15:496. [PMID: 39709481 DOI: 10.1186/s13287-024-04111-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND hucMSC-exosomes can be engineered to strengthen their therapeutic potential, and the present study aimed to explore whether hypoxic preconditioning can enhance the angiogenic potential of hucMSC-exosomes in an experimental model of POF. METHODS Primary hucMSCs and ROMECs were isolated from fresh tissue samples and assessed through a series of experiments. Exosomes were isolated from hucMSCs under normoxic or hypoxic conditions (norm-Exos and hypo-Exos, respectively) and then characterized using classic experimental methods. Based on a series of angiogenesis-related assays, we found that hypo-Exos significantly promoted ROMEC proliferation, migration, and tube formation and increased angiogenesis-promoting molecules in vitro. Histology, immunohistochemistry, and immunofluorescence experiments in a rat model of POF demonstrated that hypoxia pretreatment strengthens the therapeutic angiogenic effect of hucMSC-exosomes in vivo. Subsequently, high-throughput miRNA sequencing, qRT‑PCR analysis, and western blotting were employed to identify the potential molecular mechanism. RESULTS We found that hypo-Exos enhance endothelial function and angiogenesis via the transfer of miR-205-5p in vitro and in vivo. Finally, based on the results of bioinformatics analysis, dual luciferase reporter assays, and gain- and loss-of-function studies, we found evidence indicating that exosomal miR-205-5p enhances angiogenesis by targeting the PTEN/PI3K/AKT/mTOR signalling pathway. These results indicated for the first time that exosomes derived from hypoxia-conditioned hucMSCs strongly enhance angiogenesis via the transfer of miR-205-5p by targeting the PTEN/PI3K/AKT/mTOR signalling pathway. CONCLUSIONS Our findings provide a theoretical basis and demonstrate the potential application of a novel cell-free approach with stem cell-derived products in the treatment of POF.
Collapse
Affiliation(s)
- Qingxi Qu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Linghong Liu
- Research Center of Stem Cell and Regenerative Medicine, Shandong University, Jinan, 250012, P.R. China.
- Laboratory of Cryomedicine, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China.
| | - Limei Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Yuqian Cui
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Chunxiao Liu
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Xuanxuan Jing
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| | - Xiaoxuan Xu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, 250012, P.R. China
| |
Collapse
|
9
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Extracellular Vesicles in Viral Liver Diseases. Viruses 2024; 16:1785. [PMID: 39599900 PMCID: PMC11598962 DOI: 10.3390/v16111785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/12/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Extracellular vesicles (EVs) are bilayer vesicles released by cells in the microenvironment of the liver including parenchymal and non-parenchymal cells. They are the third important mechanism in the communications between cells, besides the secretion of cytokines and chemokines and the direct cell-to-cell contact. The aim of this review is to discuss the important role of EVs in viral liver disease, as there is increasing evidence that the transportation of viral proteins, all types of RNA, and viral particles including complete virions is implicated in the pathogenesis of both viral cirrhosis and viral-related hepatocellular carcinoma. The biogenesis of EVs is discussed and their role in the pathogenesis of viral liver diseases is presented. Their use as diagnostic and prognostic biomarkers is also analyzed. Most importantly, the significance of possible novel treatment strategies for liver fibrosis and hepatocellular carcinoma is presented, although available data are based on experimental evidence and clinical trials have not been reported.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Ioannis Tsomidis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece;
| |
Collapse
|
10
|
Zhou X, He S, He J, Xiong Y, Hu Z, Xian H, Guo G, Tan S, Ouyang D, Liu R, Gao Z, Zhu X, Abulimiti A, Zheng S, Hu D. HUC-MSC-derived exosomes repaired the damage induced by hydroquinone to 16HBE cells via miR-221/PTEN pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117120. [PMID: 39357375 DOI: 10.1016/j.ecoenv.2024.117120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 09/13/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Mesenchymal stem cell - originated exosomes (MSC-exo) are promising non-cellular treatment agents for various diseases. The present study aimed to explore whether human umbilical cord MSC - originated exosomes (HUC-MSC-exo) have the function of protecting human cells (16HBE) against the damage caused by HQ and the related mechanism. HUC-MSC-exo was isolated with differential gradient ultracentrifugation method and characterized by using transmission electron microscope (TEM). 16HBE cells were used as the tool cells and co-cultured with HUC-MSC-exo. Confocal laser scanning microscope was employed to confirm the ingestion of HUC-MSC-exo by 16HBE. Cell proliferation, migration, oxidative stress, DNA and chromosome damages of 16HBE were analyzed under HQ stress, and the role of miR-221/PTEN axis was investigated. Our data showed that under HQ stress, different groups of cells exhibited significantly decreased proliferation and migration abilities, and significant oxidative stress, DNA and chromosome damage effects. HUC-MSC-exo could alleviate the cytotoxic, oxidative stress and genotoxic damage effects of HQ on 16HBE cells. Mechanistically, HQ exposure up-regulated the level of miR-221 and down-regulated PTEN, while HUC-MSC-exo could significantly reduce the level of miR-221 and promote PTEN expression, which was involved in alleviating the toxic effects of HQ on 16HBE cells. Our data indicates that HUC-MSC-exo can alleviate the oxidative stress, cytotoxic and genotoxic effects of HQ on 16HBE cells via miR-221/PTEN pathway, and it may be a promising agent for protecting against the toxicity of HQ.
Collapse
Affiliation(s)
- Xiaotao Zhou
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China; Public Health Service Centre of Baoan District, Shenzhen City 518000, PR China
| | - Shanshan He
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China
| | - Jiayi He
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China
| | - Yiren Xiong
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China
| | - Zuqing Hu
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China
| | - Hongyi Xian
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China
| | - Guoqiang Guo
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China; Public Health Service Centre of Baoan District, Shenzhen City 518000, PR China
| | - Suqin Tan
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China
| | - Di Ouyang
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China
| | - Renyi Liu
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China
| | - Zhenjie Gao
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China
| | - Xiaoqi Zhu
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China
| | - Abudumijiti Abulimiti
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China
| | - Sujin Zheng
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China
| | - Dalin Hu
- Department of Environmental Health, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, 1838 Guangzhou Road North, Guangzhou 510515, PR China.
| |
Collapse
|
11
|
Shi L, Zeng H, An Z, Chen W, Shan Y, Ji C, Qian H. Extracellular vesicles: Illuminating renal pathophysiology and therapeutic frontiers. Eur J Pharmacol 2024; 978:176720. [PMID: 38880217 DOI: 10.1016/j.ejphar.2024.176720] [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: 03/09/2024] [Revised: 05/21/2024] [Accepted: 06/05/2024] [Indexed: 06/18/2024]
Abstract
Extracellular vesicles (EVs) are minute sacs released by cells into the extracellular milieu, harboring an array of biomolecules including proteins, nucleic acids, and lipids. Notably, a large number of studies have demonstrated the important involvement of EVs in both physiological and pathological aspects of renal function. EVs can facilitate communication between different renal cells, but it is important to recognize their dual role: they can either transmit beneficial information or lead to renal damage and worsening of existing conditions. The composition of EVs in the context of the kidneys offers valuable insights into the intricate mechanisms underlying specific renal functions or disease states. In addition, mesenchymal stem cell-derived EVs have the potential to alleviate acute and chronic kidney diseases. More importantly, the innate nanoparticle properties of EVs, coupled with their engineering potential, make them effective tools for drug delivery and therapeutic intervention. In this review, we focus on the intricate biological functions of EVs in the kidney. In addition, we explore the emerging role of EVs as diagnostic tools and innovative therapeutic agents in a range of renal diseases.
Collapse
Affiliation(s)
- Linru Shi
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Houcheng Zeng
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Zhongwu An
- Department of Laboratory, Lianyungang Oriental Hospital, Lianyungang, 222042, Jiangsu, China
| | - Wenya Chen
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yunjie Shan
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Cheng Ji
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
12
|
Zhang X, Liu H, Xiu X, Cheng J, Li T, Wang P, Men L, Qiu J, Jin Y, Zhao J. Exosomal GDNF from Bone Marrow Mesenchymal Stem Cells Moderates Neuropathic Pain in a Rat Model of Chronic Constriction Injury. Neuromolecular Med 2024; 26:34. [PMID: 39167282 DOI: 10.1007/s12017-024-08800-6] [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: 06/21/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Both of exosomes derived from mesenchymal stem cells (MSCs) and glial cell line-derived neurotrophic factor (GDNF) show potential for the treatment of neuropathic pain. Here, the analgesic effects of exosomes derived from bone marrow MSCs (BMSCs) were investigated. BMSCs-derived exosomes were isolated and characterized. Chronic constriction injury (CCI) was constructed to induce neuropathic pain in rats, which were then treated with exosomes. Pain behaviors were evaluated by measuring paw withdrawal thresholds and latency. The changes of key proteins, including cytokines, were explored using Western blot and ELISA. Administration of BMSCs-derived exosomes alleviated neuropathic pain, as demonstrated by the decrease of thermal hyperalgesia and mechanical allodynia, as well as the reduced secretion of pro-inflammatory cytokines in CCI rats. These effects were comparable to the treatment of GDNF alone. Mechanically, the exosomes suppressed the CCI-induced activation of TLR2/MyD88/NF-κB signaling pathway, while GDNF knockdown impaired their analgesic effects on CCI rat. BMSCs-derived exosomes may alleviate CCI-induced neuropathic pain and inflammation in rats by transporting GDNF.
Collapse
Affiliation(s)
- Xuelei Zhang
- Graduate School, Hebei University of Chinese Medicine, No. 3 Xingyuan Road, Luquan District, Shijiazhuang, 050200, Hebei, China.
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China.
- Hebei Key Laboratory of Intergraded Traditional and Western Medicine in Osteoarthrosis Research (Preparing), Cangzhou, 061000, Hebei, China.
| | - Huan Liu
- Graduate School, Hebei University of Chinese Medicine, No. 3 Xingyuan Road, Luquan District, Shijiazhuang, 050200, Hebei, China
| | - Xiaolei Xiu
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China
| | - Jibo Cheng
- Chengde Medical University, Anyuan Road, Chengde, 067000, Hebei, China
| | - Tong Li
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China
| | - Ping Wang
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China
| | - Lili Men
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China
| | - Junru Qiu
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China
| | - Yanyan Jin
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China
| | - Jianyong Zhao
- Department of Hand Microsurgery, The Hebei Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine, Cangzhou, 061000, Hebei, China.
| |
Collapse
|
13
|
Brooks B, D'Egidio F, Borlongan MC, Borlongan MC, Lee JY. Stem cell grafts enhance endogenous extracellular vesicle expression in the stroke brain. Brain Res Bull 2024; 214:110999. [PMID: 38851436 DOI: 10.1016/j.brainresbull.2024.110999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Endogenous brain repair occurs following an ischemic stroke but is transient, thus unable to fully mount a neuroprotective response against the evolving secondary cell death. Finding a treatment strategy that may render robust and long-lasting therapeutic effects stands as a clinically relevant therapy for stroke. Extracellular vesicles appear to be upregulated after stroke, which may represent a candidate target for neuroprotection. In this study, we probed whether transplanted stem cells could enhance the expression of extracellular vesicles to afford stable tissue remodeling in the ischemic stroke brain. Aged rats were initially exposed to the established ischemic stroke model of middle cerebral artery occlusion then received intravenous delivery of either bone marrow-derived mesenchymal stem cell transplantation or vehicle. A year later, the animals were assayed for brain damage, inflammation, and extracellular vesicle expression. Our findings revealed that while core infarction was not reduced, the stroke animals transplanted with stem cells displayed a significant reduction in peri-infarct cell loss that coincided with downregulated Iba1-labeled inflammatory cells and upregulated CD63-positive extracellular vesicles that appeared to be co-localized with GFAP-positive astrocytes. Interestingly, grafted stem cells were not detected at one year post-transplantation period, suggesting that the extracellular vesicles likely originated within the host brain. That long-lasting functional benefits persisted in the absence of surviving transplanted stem cells, but with upregulation of endogenous extracellular vesicles, advances the concept that transplantation of stem cells acutely after stroke propels host extracellular vesicles to the ischemic brain, altogether promoting chronic brain remodeling.
Collapse
Affiliation(s)
- Beverly Brooks
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, United States
| | - Francesco D'Egidio
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, United States
| | - Maximillian C Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, United States
| | - Mia C Borlongan
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, United States
| | - Jea-Young Lee
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, United States.
| |
Collapse
|
14
|
Jiang Y, Yusoff NM, Du J, Moses EJ, Lin JT. Current perspectives on mesenchymal stem cells as a potential therapeutic strategy for non-alcoholic fatty liver disease. World J Stem Cells 2024; 16:760-772. [PMID: 39086561 PMCID: PMC11287429 DOI: 10.4252/wjsc.v16.i7.760] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 05/18/2024] [Accepted: 06/14/2024] [Indexed: 07/25/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as a significant health challenge, characterized by its widespread prevalence, intricate natural progression and multifaceted pathogenesis. Although NAFLD initially presents as benign fat accumulation, it may progress to steatosis, non-alcoholic steatohepatitis, cirrhosis, and hepatocellular carcinoma. Mesenchymal stem cells (MSCs) are recognized for their intrinsic self-renewal, superior biocompatibility, and minimal immunogenicity, positioning them as a therapeutic innovation for liver diseases. Therefore, this review aims to elucidate the potential roles of MSCs in alleviating the progression of NAFLD by alteration of underlying molecular pathways, including glycolipid metabolism, inflammation, oxidative stress, endoplasmic reticulum stress, and fibrosis. The insights are expected to provide further understanding of the potential of MSCs in NAFLD therapeutics, and support the development of MSC-based therapy in the treatment of NAFLD.
Collapse
Affiliation(s)
- Yan Jiang
- School of Nursing, Xinxiang Medical University, Xinxiang 453000, Henan Province, China
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas 13200, Pulau Pinang, Malaysia
| | - Narazah Mohd Yusoff
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas 13200, Pulau Pinang, Malaysia
| | - Jiang Du
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
| | - Emmanuel Jairaj Moses
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas 13200, Pulau Pinang, Malaysia
| | - Jun-Tang Lin
- Stem Cells and Biotherapy Engineering Research Center of Henan, National Joint Engineering Laboratory of Stem Cells and Biotherapy, School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, Henan Province, China
- Henan Joint International Research Laboratory of Stem Cell Medicine, School of Medical Engineering, Xinxiang Medical University, Xinxiang 453000, Henan Province, China.
| |
Collapse
|
15
|
Lv H, Feng Z, Chen X, Zhang Z, Zhou T, Wei J, Feng L, Tao Y, Chen F, Lu S. Global scientific trends on exosomes therapy for osteoporosis from 2004 to 2023: A bibliometric and visualized analysis. Medicine (Baltimore) 2024; 103:e38835. [PMID: 38996093 PMCID: PMC11245275 DOI: 10.1097/md.0000000000038835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 06/14/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Exosomes have emerged as pivotal mediators in modulating physiological and pathological processes implicated in osteoporosis (OP) through their distinctive mode of intracellular communication. The use of exosomes has evoked considerable interest, catalyzing a surge in research endeavors on a global scale. This study endeavors to scrutinize contemporary landscapes and burgeoning trends in this realm. METHODS The Web of Science Core Collection was used to retrieve publications on exosomes therapy for OP within the time frame of January 1, 2004 to December 31, 2023. The bibliometric methodology was applied to study and index the collected data. VOSviewer and citespace software were used to conduct visualization, co-authorship, co-occurrence, and publication trend analyses of exosome therapy in OP. RESULTS A total of 610 publications (443 articles and 167 reviews) from 51 countries and 911 institutions were included in this study. Shanghai Jiao Tong University, Central South University, Sichuan University, and Zhejiang University are leading research institutions in this field. Stem Cell Research Therapy published the highest number of articles and has emerged as the most cited journal. Of the 4077 scholars who participated in the study, Xie, Hui, Zhang, Yan, Tan, and Yi-Juan had the largest number of articles. Furthermore, according to the cluster analysis of external keywords, future research hotspots can be categorized into 3 directions: research status of exosomes for the treatment of OP, treatment of OP through exosome-regulated signaling pathways, and exosomes as targeted drug delivery systems. CONCLUSION This study suggests that the number of future publications on exosome therapy for OP will increase, with a focus on fundamental investigations into drug-loading capacities and molecular mechanisms. In summary, this study presents the first systematic bibliometric analysis of exosome therapy publications in OP, providing an objective and comprehensive overview of the field and a valuable reference for researchers in this domain.
Collapse
Affiliation(s)
- He Lv
- Faculty of Postgraduate, Guangxi University of Chinese Medicine, Nanning, China
| | - Zhe Feng
- Joint & Sports Medicine Surgery Division, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Xingyu Chen
- Faculty of Postgraduate, Guangxi University of Chinese Medicine, Nanning, China
| | - Zhenyu Zhang
- Gynecology Department, Guangdong Medical University Shunde Women and Children’s Hospital, Foshan, China
| | - Tianhao Zhou
- Faculty of Postgraduate, Guangxi University of Chinese Medicine, Nanning, China
| | - Jihu Wei
- Faculty of Postgraduate, Guangxi University of Chinese Medicine, Nanning, China
| | - Lin Feng
- Faculty of Postgraduate, Guangxi University of Chinese Medicine, Nanning, China
| | - Yizi Tao
- Faculty of Postgraduate, Guangxi University of Chinese Medicine, Nanning, China
| | - Feng Chen
- Spine Surgery Division, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Shijin Lu
- Centre for Translational Medical Research in Integrative Chinese and Western Medicine, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
16
|
Souza ILM, Suzukawa AA, Josino R, Marcon BH, Robert AW, Shigunov P, Correa A, Stimamiglio MA. Cellular In Vitro Responses Induced by Human Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles Obtained from Suspension Culture. Int J Mol Sci 2024; 25:7605. [PMID: 39062847 PMCID: PMC11277484 DOI: 10.3390/ijms25147605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) and their extracellular vesicles (MSC-EVs) have been described to have important roles in tissue regeneration, including tissue repair, control of inflammation, enhancing angiogenesis, and regulating extracellular matrix remodeling. MSC-EVs have many advantages for use in regeneration therapies such as facility for dosage, histocompatibility, and low immunogenicity, thus possessing a lower possibility of rejection. In this work, we address the potential activity of MSC-EVs isolated from adipose-derived MSCs (ADMSC-EVs) cultured on cross-linked dextran microcarriers, applied to test the scalability and reproducibility of EV production. Isolated ADMSC-EVs were added into cultured human dermal fibroblasts (NHDF-1), keratinocytes (HaCat), endothelial cells (HUVEC), and THP-1 cell-derived macrophages to evaluate cellular responses (i.e., cell proliferation, cell migration, angiogenesis induction, and macrophage phenotype-switching). ADMSC viability and phenotype were assessed during cell culture and isolated ADMSC-EVs were monitored by nanotracking particle analysis, electron microscopy, and immunophenotyping. We observed an enhancement of HaCat proliferation; NHDF-1 and HaCat migration; endothelial tube formation on HUVEC; and the expression of inflammatory cytokines in THP-1-derived macrophages. The increased expression of TGF-β and IL-1β was observed in M1 macrophages treated with higher doses of ADMSC-EVs. Hence, EVs from microcarrier-cultivated ADMSCs are shown to modulate cell behavior, being able to induce skin tissue related cells to migrate and proliferate as well as stimulate angiogenesis and cause balance between pro- and anti-inflammatory responses in macrophages. Based on these findings, we suggest that the isolation of EVs from ADMSC suspension cultures makes it possible to induce in vitro cellular responses of interest and obtain sufficient particle numbers for the development of in vivo concept tests for tissue regeneration studies.
Collapse
Affiliation(s)
- Ingrid L. M. Souza
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| | - Andreia A. Suzukawa
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| | - Raphaella Josino
- Albert Einstein Israelite Hospital, São Paulo 05652-900, SP, Brazil
| | - Bruna H. Marcon
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
- Confocal and Electronic Microscopy Facility (RPT07C), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil
| | - Anny W. Robert
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
- Confocal and Electronic Microscopy Facility (RPT07C), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil
| | - Patrícia Shigunov
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| | - Alejandro Correa
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| | - Marco A. Stimamiglio
- Laboratory of Basic Biology of Stem Cells (Labcet), Carlos Chagas Institute, Fiocruz, Curitiba 81350-010, PR, Brazil (A.A.S.); (B.H.M.); (A.W.R.); (P.S.)
| |
Collapse
|
17
|
Pinto H, Sánchez-Vizcaíno Mengual E. Exosomes in the Real World of Medical Aesthetics: A Review. Aesthetic Plast Surg 2024; 48:2513-2527. [PMID: 38315231 DOI: 10.1007/s00266-023-03844-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 12/30/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND Exosomes are cell-derived nanovesicles that transport proteins, nucleic acids, and lipids and play a significant role in almost every physiological process in the human body. They have generated great interest, especially in the field of tissue regeneration. Studies in the last decade support their great regenerating and rejuvenating potential. However, the lack of standardized procedures, limited knowledge regarding their action mechanism, and little clinical evidence impair their implementation and approval in the medical setting. This review aimed to identify published studies and clinical trials using exosomes in human patients for clinical treatments in aesthetic medicine. MATERIALS AND METHODS A systematic search was conducted in the PubMed database using the search term "exosomes" and 25 terms related to aesthetic medicine treatments in human patients. Additionally, a search was conducted in the ClinicalTrials.gov database for interventional clinical trials using exosomes for aesthetic treatments in adults 18 to ≥ 65 years of age. RESULTS Nine articles were selected after debugging the initial list of published articles in which exosomes were related to Aesthetic Medicine (633 articles). Nine studies were identified from the initial search on ClinicalTrial.gov (104 trials with exosomes). CONCLUSIONS There is no doubt about the scientific basis of exosome regenerative potential and the growing interest in exosomes in Aesthetic Medicine. However, companies must spend more on research to develop standardized and reliable procedures to obtain exosomes for their approval and application in clinical practice. LEVEL OF EVIDENCE III This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 . This review highlights the large amount of published research on exosomes related to aesthetic medicine and, at the same time, the lack of products approved by regulatory agencies. Several issues have been suggested to elucidate a response, such as the need for standardized protocols and more knowledge to ensure safe treatments. It also highlights the few clinical trials conducted to evaluate exosome properties in aesthetic medicine treatments.
Collapse
Affiliation(s)
- Hernán Pinto
- Instituto de Investigaciones Biomédicas i2e3, Santa Coloma de Gramenet, Spain
| | | |
Collapse
|
18
|
Haider KH. Priming mesenchymal stem cells to develop "super stem cells". World J Stem Cells 2024; 16:623-640. [PMID: 38948094 PMCID: PMC11212549 DOI: 10.4252/wjsc.v16.i6.623] [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: 04/02/2024] [Revised: 05/04/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
The stem cell pre-treatment approaches at cellular and sub-cellular levels encompass physical manipulation of stem cells to growth factor treatment, genetic manipulation, and chemical and pharmacological treatment, each strategy having advantages and limitations. Most of these pre-treatment protocols are non-combinative. This editorial is a continuum of Li et al's published article and Wan et al's editorial focusing on the significance of pre-treatment strategies to enhance their stemness, immunoregulatory, and immunosuppressive properties. They have elaborated on the intricacies of the combinative pre-treatment protocol using pro-inflammatory cytokines and hypoxia. Applying a well-defined multi-pronged combinatorial strategy of mesenchymal stem cells (MSCs), pre-treatment based on the mechanistic understanding is expected to develop "Super MSCs", which will create a transformative shift in MSC-based therapies in clinical settings, potentially revolutionizing the field. Once optimized, the standardized protocols may be used with slight modifications to pre-treat different stem cells to develop "super stem cells" with augmented stemness, functionality, and reparability for diverse clinical applications with better outcomes.
Collapse
Affiliation(s)
- Khawaja Husnain Haider
- Department of Basic Sciences, Sulaiman AlRajhi University, AlQaseem 52736, Saudi Arabia.
| |
Collapse
|
19
|
Li J, Liu Y, Zhang R, Yang Q, Xiong W, He Y, Ye Q. Insights into the role of mesenchymal stem cells in cutaneous medical aesthetics: from basics to clinics. Stem Cell Res Ther 2024; 15:169. [PMID: 38886773 PMCID: PMC11184751 DOI: 10.1186/s13287-024-03774-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
With the development of the economy and the increasing prevalence of skin problems, cutaneous medical aesthetics are gaining more and more attention. Skin disorders like poor wound healing, aging, and pigmentation have an impact not only on appearance but also on patients with physical and psychological issues, and even impose a significant financial burden on families and society. However, due to the complexities of its occurrence, present treatment options cannot produce optimal outcomes, indicating a dire need for new and effective treatments. Mesenchymal stem cells (MSCs) and their secretomics treatment is a new regenerative medicine therapy that promotes and regulates endogenous stem cell populations and/or replenishes cell pools to achieve tissue homeostasis and regeneration. It has demonstrated remarkable advantages in several skin-related in vivo and in vitro investigations, aiding in the improvement of skin conditions and the promotion of skin aesthetics. As a result, this review gives a complete description of recent scientific breakthroughs in MSCs for skin aesthetics and the limitations of their clinical applications, aiming to provide new ideas for future research and clinical transformation.
Collapse
Affiliation(s)
- Junyi Li
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ye Liu
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Rui Zhang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qianyu Yang
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wei Xiong
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, 430030, China.
| | - Qingsong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
20
|
Alshahrani MY, Jasim SA, Altalbawy FMA, Bansal P, Kaur H, Al-Hamdani MM, Deorari M, Abosaoda MK, Hamzah HF, A Mohammed B. A comprehensive insight into the immunomodulatory role of MSCs-derived exosomes (MSC-Exos) through modulating pattern-recognition receptors (PRRs). Cell Biochem Funct 2024; 42:e4029. [PMID: 38773914 DOI: 10.1002/cbf.4029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/16/2024] [Accepted: 04/25/2024] [Indexed: 05/24/2024]
Abstract
Mesenchymal stem cell-derived exosomes (MSC-Exos) are emerging as remarkable agents in the field of immunomodulation with vast potential for diagnosing and treating various diseases, including cancer and autoimmune disorders. These tiny vesicles are laden with a diverse cargo encompassing proteins, nucleic acids, lipids, and bioactive molecules, offering a wealth of biomarkers and therapeutic options. MSC-Exos exhibit their immunomodulatory prowess by skillfully regulating pattern-recognition receptors (PRRs). They conduct a symphony of immunological responses, modulating B-cell activities, polarizing macrophages toward anti-inflammatory phenotypes, and fine-tuning T-cell activity. These interactions have profound implications for precision medicine, cancer immunotherapy, autoimmune disease management, biomarker discovery, and regulatory approvals. MSC-Exos promises to usher in a new era of tailored therapies, personalized diagnostics, and more effective treatments for various medical conditions. As research advances, their transformative potential in healthcare becomes increasingly evident.
Collapse
Affiliation(s)
- Mohammad Y Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | | | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh, India
- Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand, India
| | | | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Munther Kadhim Abosaoda
- College of Pharmacy, The Islamic University, Najaf, Iraq
- College of Pharmacy, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Pharmacy, The Islamic University of Babylon, Al Diwaniyah, Iraq
| | - Hamza Fadhel Hamzah
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | - Bahira A Mohammed
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| |
Collapse
|
21
|
Mushtaq M, Zineldeen DH, Mateen MA, Haider KH. Mesenchymal stem cells' "garbage bags" at work: Treating radial nerve injury with mesenchymal stem cell-derived exosomes. World J Stem Cells 2024; 16:467-478. [PMID: 38817330 PMCID: PMC11135253 DOI: 10.4252/wjsc.v16.i5.467] [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: 02/20/2024] [Revised: 04/03/2024] [Accepted: 04/25/2024] [Indexed: 05/24/2024] Open
Abstract
Unlike central nervous system injuries, peripheral nerve injuries (PNIs) are often characterized by more or less successful axonal regeneration. However, structural and functional recovery is a senile process involving multifaceted cellular and molecular processes. The contemporary treatment options are limited, with surgical intervention as the gold-standard method; however, each treatment option has its associated limitations, especially when the injury is severe with a large gap. Recent advancements in cell-based therapy and cell-free therapy approaches using stem cell-derived soluble and insoluble components of the cell secretome are fast-emerging therapeutic approaches to treating acute and chronic PNI. The recent pilot study is a leap forward in the field, which is expected to pave the way for more enormous, systematic, and well-designed clinical trials to assess the therapeutic efficacy of mesenchymal stem cell-derived exosomes as a bio-drug either alone or as part of a combinatorial approach, in an attempt synergize the best of novel treatment approaches to address the complexity of the neural repair and regeneration.
Collapse
Affiliation(s)
- Mazhar Mushtaq
- Department of Basic Sciences, Sulaiman AlRajhi University, Albukairiyah 52736, AlQaseem, Saudi Arabia
| | - Doaa Hussein Zineldeen
- Department of Basic Sciences, Sulaiman AlRajhi University, Albukairiyah 52736, AlQaseem, Saudi Arabia
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Tanta University, Tanta 6632110, Egypt
| | - Muhammad Abdul Mateen
- Department of Basic Sciences, Sulaiman AlRajhi University, Albukairiyah 52736, AlQaseem, Saudi Arabia
| | - Khawaja Husnain Haider
- Department of Basic Sciences, Sulaiman AlRajhi University, Albukairiyah 52736, AlQaseem, Saudi Arabia.
| |
Collapse
|
22
|
Krishnan I, Chan AML, Law JX, Ng MH, Jayapalan JJ, Lokanathan Y. Proteomic Analysis of Umbilical Cord Mesenchymal Stem Cell-Derived Extracellular Vesicles: A Systematic Review. Int J Mol Sci 2024; 25:5340. [PMID: 38791378 PMCID: PMC11121203 DOI: 10.3390/ijms25105340] [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: 03/26/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Numerous challenges remain within conventional cell-based therapy despite the growing trend of stem cells used to treat various life-debilitating diseases. These limitations include batch-to-batch heterogeneity, induced alloreactivity, cell survival and integration, poor scalability, and high cost of treatment, thus hindering successful translation from lab to bedside. However, recent pioneering technology has enabled the isolation and enrichment of small extracellular vesicles (EVs), canonically known as exosomes. EVs are described as a membrane-enclosed cargo of functional biomolecules not limited to lipids, nucleic acid, and proteins. Interestingly, studies have correlated the biological role of MSC-EVs to the paracrine activity of MSCs. This key evidence has led to rigorous studies on MSC-EVs as an acellular alternative. Using EVs as a therapy was proposed as a model leading to improvements through increased safety; enhanced bioavailability due to size and permeability; reduced heterogeneity by selective and quantifiable properties; and prolonged shelf-life via long-term freezing or lyophilization. Yet, the identity and potency of EVs are still relatively unknown due to various methods of preparation and to qualify the final product. This is reflected by the absence of regulatory strategies overseeing manufacturing, quality control, clinical implementation, and product registration. In this review, the authors review the various production processes and the proteomic profile of MSC-EVs.
Collapse
Affiliation(s)
- Illayaraja Krishnan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| | - Alvin Man Lung Chan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| | - Jia Xian Law
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| | | | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia; (I.K.); (A.M.L.C.); (J.X.L.); (M.H.N.)
| |
Collapse
|
23
|
Yan C, Wang X, Wang Q, Li H, Song H, Zhou J, Peng Z, Yin W, Fan X, Yang K, Zhou B, Liang Y, Jiang Z, Shi Y, Zhang S, He S, Li R, Xie J. A Novel Conductive Polypyrrole-Chitosan Hydrogel Containing Human Endometrial Mesenchymal Stem Cell-Derived Exosomes Facilitated Sustained Release for Cardiac Repair. Adv Healthc Mater 2024; 13:e2304207. [PMID: 38175149 PMCID: PMC11468178 DOI: 10.1002/adhm.202304207] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Indexed: 01/05/2024]
Abstract
Myocardial infarction (MI) results in cardiomyocyte necrosis and conductive system damage, leading to sudden cardiac death and heart failure. Studies have shown that conductive biomaterials can restore cardiac conduction, but cannot facilitate tissue regeneration. This study aims to add regenerative capabilities to the conductive biomaterial by incorporating human endometrial mesenchymal stem cell (hEMSC)-derived exosomes (hEMSC-Exo) into poly-pyrrole-chitosan (PPY-CHI), to yield an injectable hydrogel that can effectively treat MI. In vitro, PPY-CHI/hEMSC-Exo, compared to untreated controls, PPY-CHI, or hEMSC-Exo alone, alleviates H2O2-induced apoptosis and promotes tubule formation, while in vivo, PPY-CHI/hEMSC-Exo improves post-MI cardiac functioning, along with counteracting against ventricular remodeling and fibrosis. All these activities are facilitated via increased epidermal growth factor (EGF)/phosphoinositide 3-kinase (PI3K)/AKT signaling. Furthermore, the conductive properties of PPY-CHI/hEMSC-Exo are able to resynchronize cardiac electrical transmission to alleviate arrythmia. Overall, PPY-CHI/hEMSC-Exo synergistically combines the cardiac regenerative capabilities of hEMSC-Exo with the conductive properties of PPY-CHI to improve cardiac functioning, via promoting angiogenesis and inhibiting apoptosis, as well as resynchronizing electrical conduction, to ultimately enable more effective MI treatment. Therefore, incorporating exosomes into a conductive hydrogel provides dual benefits in terms of maintaining conductivity, along with facilitating long-term exosome release and sustained application of their beneficial effects.
Collapse
Affiliation(s)
- Changping Yan
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
- Department of GynecologyAffiliated Cancer Hospital of Shanxi Medical UniversityTaiyuan030013China
| | - Xinzhu Wang
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Qi Wang
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Haiyan Li
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Huifang Song
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
- Department of AnatomyShanxi Medical UniversityTaiyuan030001China
| | - Jingli Zhou
- Shanxi Provincial People's HospitalAffiliated Hospital of Shanxi Medical UniversityTaiyuan030012China
| | - Zexu Peng
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Wenjuan Yin
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Xuemei Fan
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Kun Yang
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Bingrui Zhou
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Yuxiang Liang
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Zengyu Jiang
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Yuwei Shi
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
- NHC Key Laboratory of PneumoconiosisShanxi Province Key Laboratory of RespiratoryDepartment of Pulmonary and Critical Care MedicineThe First Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Sanyuan Zhang
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Sheng He
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| | - Ren‐Ke Li
- Toronto General Hospital Research InstituteDivision of Cardiovascular SurgeryUniversity Health NetworkUniversity of TorontoTorontoONM5G 2C4Canada
| | - Jun Xie
- The First Hospital of Shanxi Medical UniversityDepartment of Biochemistry and Molecular BiologyShanxi Key Laboratory of Birth Defect and Cell RegenerationMOE Key Laboratory of Coal Environmental Pathogenicity and PreventionShanxi Medical UniversityTaiyuan030001China
| |
Collapse
|
24
|
Tian X, Wu L, Li X, Zheng W, Zuo H, Song H. Exosomes derived from bone marrow mesenchymal stem cells alleviate biliary ischemia reperfusion injury in fatty liver transplantation by inhibiting ferroptosis. Mol Cell Biochem 2024; 479:881-894. [PMID: 37243945 PMCID: PMC11016128 DOI: 10.1007/s11010-023-04770-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 05/15/2023] [Indexed: 05/29/2023]
Abstract
Fatty liver grafts are susceptible to ischemia reperfusion injury (IRI), increasing the risk of biliary complications after liver transplantation (LT). Ferroptosis, a newly recognized programmed cell death, is expected to be a novel therapeutic target for IRI. We investigated whether exosomes derived from heme oxygenase 1-modified bone marrow mesenchymal stem cells (HExos) relieve ferroptosis and protect biliary tracts from IRI in a rat fatty liver transplantation model. Rats were fed with a methionine choline deficient (MCD) diet for 2 weeks to induce severe hepatic steatosis. Steatotic grafts were implanted and HExos were administered after liver transplantation. A series of functional assays and pathological analysis were performed to assess ferroptosis and biliary IRI. The HExos attenuated IRI following liver transplantation, as demonstrated by less ferroptosis, improved liver function, less Kupffer and T cell activation, and less long-term biliary fibrosis. MicroRNA (miR)-204-5p delivered by HExos negatively regulated ferroptosis by targeting a key pro-ferroptosis enzyme, ACSL4. Ferroptosis contributes to biliary IRI in fatty liver transplantation. HExos protect steatotic grafts by inhibiting ferroptosis, and may become a promising strategy to prevent biliary IRI and expand the donor pool.
Collapse
Affiliation(s)
- Xuan Tian
- School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Longlong Wu
- School of Medicine, Nankai University, Tianjin, People's Republic of China
| | - Xiang Li
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Weiping Zheng
- Department of Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China
- NHC Key Laboratory of Critical Care Medicine, Tianjin, 300192, People's Republic of China
| | - Huaiwen Zuo
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Hongli Song
- Department of Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, No. 24 Fukang Road, Nankai District, Tianjin, 300192, People's Republic of China.
- Tianjin Key Laboratory of Organ Transplantation, Tianjin, People's Republic of China.
| |
Collapse
|
25
|
Kao YH, Chang CY, Lin YC, Chen PH, Lee PH, Chang HR, Chang WY, Chang YC, Wun SF, Sun CK. Mesenchymal Stem Cell-Derived Exosomes Mitigate Acute Murine Liver Injury via Ets-1 and Heme Oxygenase-1 Up-regulation. Curr Stem Cell Res Ther 2024; 19:906-918. [PMID: 37723631 DOI: 10.2174/1574888x19666230918102826] [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: 06/14/2023] [Revised: 07/21/2023] [Accepted: 08/11/2023] [Indexed: 09/20/2023]
Abstract
BACKGROUND Mesenchymal stem cells (MSCs)-derived exosomes have been previously demonstrated to promote tissue regeneration in various animal disease models. This study investigated the protective effect of exosome treatment in carbon tetrachloride (CCl4)-induced acute liver injury and delineated possible underlying mechanism. METHODS Exosomes collected from conditioned media of previously characterized human umbilical cord-derived MSCs were intravenously administered into male CD-1 mice with CCl4-induced acute liver injury. Biochemical, histological and molecular parameters were used to evaluate the severity of liver injury. A rat hepatocyte cell line, Clone-9, was used to validate the molecular changes by exosome treatment. RESULTS Exosome treatment significantly suppressed plasma levels of AST, ALT, and pro-inflammatory cytokines, including IL-6 and TNF-α, in the mice with CCl4-induced acute liver injury. Histological morphometry revealed a significant reduction in the necropoptic area in the injured livers following exosome therapy. Consistently, western blot analysis indicated marked elevations in hepatic expression of PCNA, c-Met, Ets-1, and HO-1 proteins after exosome treatment. Besides, the phosphorylation level of signaling mediator JNK was significantly increased, and that of p38 was restored by exosome therapy. Immunohistochemistry double staining confirmed nuclear Ets-1 expression and cytoplasmic localization of c-Met and HO-1 proteins. In vitro studies demonstrated that exosome treatment increased the proliferation of Clone-9 hepatocytes and protected them from CCl4-induced cytotoxicity. Kinase inhibition experiment indicated that the exosome-driven hepatoprotection might be mediated through the JNK pathway. CONCLUSION Exosome therapy activates the JNK signaling activation pathway as well as up-regulates Ets-1 and HO-1 expression, thereby protecting hepatocytes against hepatotoxin-induced cell death.
Collapse
Affiliation(s)
- Ying-Hsien Kao
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Chih-Yang Chang
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, E-Da Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Yu-Chun Lin
- Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, 52445, Taiwan
| | - Po-Han Chen
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Po-Huang Lee
- Department of Surgery, E-Da Hospital, I-Shou University, Kaohsiung, 52445, Taiwan
- Committee for Integration and Promotion of Advanced Medicine and Biotechnology, E-Da Healthcare Group, Kaohsiung, 82445, Taiwan
| | - Huoy-Rou Chang
- Departments of Biomedical Engineering, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Wen-Yu Chang
- Department of Dermatology, EDa Cancer Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
- The School of Medicine for International Students, College of Medicine, IShou University, Kaohsiung, 82445, Taiwan
| | - Yo-Chen Chang
- Department of Ophthalmology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Shen-Fa Wun
- Departments of Biomedical Engineering, I-Shou University, Kaohsiung, 82445, Taiwan
| | - Cheuk-Kwan Sun
- Department of Medical Research, E-Da Hospital, I-Shou University, Kaohsiung, 82445, Taiwan
- The School of Medicine for International Students, College of Medicine, IShou University, Kaohsiung, 82445, Taiwan
| |
Collapse
|
26
|
Hormozi A, Hasanzadeh S, Ebrahimi F, Daei N, Hajimortezayi Z, Mehdizadeh A, Zamani M. Treatment with Exosomes Derived from Mesenchymal Stem Cells: A New Window of Healing Science in Regenerative Medicine. Curr Stem Cell Res Ther 2024; 19:879-893. [PMID: 37622719 DOI: 10.2174/1574888x18666230824165014] [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: 03/11/2023] [Revised: 07/07/2023] [Accepted: 07/12/2023] [Indexed: 08/26/2023]
Abstract
Many studies have been conducted on the potential applications of mesenchymal stem cells (MSCs) over recent years due to their growing importance in regenerative medicine. Exosomes are considered cargos capable of transporting proteins, peptides, lipids, mRNAs, and growth factors. MSCsderived exosomes are also involved in the prevention or treatment of a variety of diseases, including cardiovascular diseases, neurological diseases, skin disorders, lung diseases, osteoarthritis, damaged tissue repair, and other diseases. This review attempted to summarize the importance of employing MSCs in regenerative medicine by gathering and evaluating information from current literature. The role of MSCs and the potential applications of MSCs-derived exosomes have also been discussed.
Collapse
Affiliation(s)
- Arezoo Hormozi
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Sajedeh Hasanzadeh
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Faezeh Ebrahimi
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Narges Daei
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Zahra Hajimortezayi
- Student Research Committee, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Zamani
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
27
|
Al-Jipouri A, Eritja À, Bozic M. Unraveling the Multifaceted Roles of Extracellular Vesicles: Insights into Biology, Pharmacology, and Pharmaceutical Applications for Drug Delivery. Int J Mol Sci 2023; 25:485. [PMID: 38203656 PMCID: PMC10779093 DOI: 10.3390/ijms25010485] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles released from various cell types that have emerged as powerful new therapeutic option for a variety of diseases. EVs are involved in the transmission of biological signals between cells and in the regulation of a variety of biological processes, highlighting them as potential novel targets/platforms for therapeutics intervention and/or delivery. Therefore, it is necessary to investigate new aspects of EVs' biogenesis, biodistribution, metabolism, and excretion as well as safety/compatibility of both unmodified and engineered EVs upon administration in different pharmaceutical dosage forms and delivery systems. In this review, we summarize the current knowledge of essential physiological and pathological roles of EVs in different organs and organ systems. We provide an overview regarding application of EVs as therapeutic targets, therapeutics, and drug delivery platforms. We also explore various approaches implemented over the years to improve the dosage of specific EV products for different administration routes.
Collapse
Affiliation(s)
- Ali Al-Jipouri
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
| | - Àuria Eritja
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| | - Milica Bozic
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| |
Collapse
|
28
|
Bicer M. Exploring therapeutic avenues: mesenchymal stem/stromal cells and exosomes in confronting enigmatic biofilm-producing fungi. Arch Microbiol 2023; 206:11. [PMID: 38063945 DOI: 10.1007/s00203-023-03744-0] [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: 10/04/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 12/18/2023]
Abstract
Fungal infections concomitant with biofilms can demonstrate an elevated capacity to withstand substantially higher concentrations of antifungal agents, contrasted with infectious diseases caused by planktonic cells. This inherent resilience intrinsic to biofilm-associated infections engenders a formidable impediment to effective therapeutic interventions. The different mechanisms that are associated with the intrinsic resistance of Candida species encompass drug sequestration by the matrix, drug efflux pumps, stress response cell density, and the presence of persister cells. These persisters, a subset of fungi capable of surviving hostile conditions, pose a remarkable challenge in clinical settings in virtue of their resistance to conventional antifungal therapies. Hence, an exigent imperative has arisen for the development of novel antifungal therapeutics with specific targeting capabilities focused on these pathogenic persisters. On a global scale, fungal persistence and their resistance within biofilms generate an urgent clinical need for investigating recently introduced therapeutic strategies. This review delves into the unique characteristics of Mesenchymal stem/stromal cells (MSCs) and their secreted exosomes, which notably exhibit immunomodulatory and regenerative properties. By comprehensively assessing the current literature and ongoing research in this field, this review sheds light on the plausible mechanisms by which MSCs and their exosomes can be harnessed to selectively target fungal persisters. Additionally, prospective approaches in the use of cell-based therapeutic modalities are examined, emphasizing the importance of further research to overcome the enigmatic fungal persistence.
Collapse
Affiliation(s)
- Mesude Bicer
- Department of Bioengineering, Faculty of Life and Natural Sciences, Abdullah Gul University, Kayseri, 38080, Turkey.
| |
Collapse
|
29
|
Ling MTM, Govindaraju K, Lokanathan Y, Abidin AZ, Ibrahim B. Mesenchymal stem cell-derived extracellular vesicles for metabolic syndrome therapy: Assessing efficacy with nuclear magnetic resonance spectroscopy. Cell Biochem Funct 2023; 41:1044-1059. [PMID: 37933415 DOI: 10.1002/cbf.3881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 09/27/2023] [Accepted: 10/21/2023] [Indexed: 11/08/2023]
Abstract
Metabolic syndrome (MetS) represents a cluster of metabolic abnormalities. The prevalence of MetS has surged, transforming it into a pressing public health concern that could potentially affect around 20%-25% of the global population. As MetS continues its ascent, diverse interventions, pharmacological, nonpharmacological and combined have been deployed. Yet, a comprehensive remedy that fully eradicates MetS symptoms remains elusive, compounded by the risks of polypharmacy's emergence. Acknowledging the imperative to grasp MetS's intricate pathologies, deeper insights for future research and therapy optimisation become paramount. Conventional treatments often target specific syndrome elements. However, a novel approach emerges in mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) therapy, promising a holistic shift. MSC-EVs, tiny membranous vesicles secreted by mesenchymal stem cells, have garnered immense attention for their multifaceted bioactivity and regenerative potential. Their ability to modulate inflammation, enhance tissue repair and regulate metabolic pathways has prompted researchers to explore their therapeutic application in MetS. This review primarily aims to provide an overview of how MSC-EVs therapy can improve metabolic parameters in subjects with MetS disease and also introduce the usefulness of NMR spectroscopy in assessing the efficacy of MSC-EVs therapy for treating MetS.
Collapse
Affiliation(s)
- Magdalene Tan Mei Ling
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Kayatri Govindaraju
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Yogeswaran Lokanathan
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Asmaa' Zainal Abidin
- Department of Chemistry and Biology, Centre for Defense Foundation Studies, Universiti Pertahanan Nasional Malaysia, Kuala Lumpur, Malaysia
| | - Baharudin Ibrahim
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
30
|
Waseem A, Saudamini, Haque R, Janowski M, Raza SS. Mesenchymal stem cell-derived exosomes: Shaping the next era of stroke treatment. NEUROPROTECTION 2023; 1:99-116. [PMID: 38283953 PMCID: PMC10811806 DOI: 10.1002/nep3.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/05/2023] [Accepted: 11/10/2023] [Indexed: 01/30/2024]
Abstract
Exosome-based treatments are gaining traction as a viable approach to addressing the various issues faced by an ischemic stroke. These extracellular vesicles, mainly produced by Mesenchymal Stem Cells (MSCs), exhibit many properties with substantial therapeutic potential. Exosomes are particularly appealing for stroke therapy because of their low immunogenicity, effective cargo transport, and ability to cross the blood-brain barrier. Their diverse effects include neuroprotection, angiogenesis stimulation, inflammatory response modulation, and cell death pathway attenuation, synergistically promoting neuronal survival, tissue regeneration, and functional recovery. Exosomes also show potential as diagnostic indicators for early stroke identification and customized treatment options. Despite these promising qualities, current exosome-based therapeutics have some limitations. The heterogeneity of exosome release among cell types, difficulty in standardization and isolation techniques, and complications linked to dosage and targeted administration necessitates extensive investigation. It is critical to thoroughly understand exosomal processes and their complicated interactions within the cellular milieu. To improve the practicality and efficacy of exosome-based medicines, research efforts must focus on improving production processes, developing robust evaluation criteria, and developing large-scale isolation techniques. Altogether, exosomes' multifunctional properties offer a new route for transforming stroke treatment and significantly improving patient outcomes.
Collapse
Affiliation(s)
- Arshi Waseem
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College HospitalEra University, SarfarazganjLucknowIndia
| | - Saudamini
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College HospitalEra University, SarfarazganjLucknowIndia
- Department of BiotechnologyCentral University of South BiharGayaIndia
| | - Rizwanul Haque
- Department of BiotechnologyCentral University of South BiharGayaIndia
| | - Miroslaw Janowski
- Center for Advanced Imaging Research, Department of Diagnostic Radiology and Nuclear MedicineUniversity of MarylandBaltimoreMarylandUSA
| | - Syed S. Raza
- Laboratory for Stem Cell & Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College HospitalEra University, SarfarazganjLucknowIndia
- Department of Stem Cell Biology and Regenerative Medicine, Era's Lucknow Medical College HospitalEra University, SarfarazganjLucknowIndia
| |
Collapse
|
31
|
Mohamed AS, Abdel-Fattah DS, Abdel-Aleem GA, El-Sheikh TF, Elbatch MM. Biochemical study of the effect of mesenchymal stem cells-derived exosome versus L-Dopa in experimentally induced Parkinson's disease in rats. Mol Cell Biochem 2023; 478:2795-2811. [PMID: 36966421 PMCID: PMC10627934 DOI: 10.1007/s11010-023-04700-8] [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: 01/01/2023] [Accepted: 03/02/2023] [Indexed: 03/27/2023]
Abstract
Parkinson's disease (PD) is a chronic and ongoing neurological condition. Unfortunately, as the dopaminergic terminals continue to deteriorate, the effectiveness of anti-Parkinson therapy decreases. This study aimed to examine the effects of BM-MSCs-derived exosomes in rats induced with Parkinson's disease. The goal was to determine their potential for neurogenic repair and functional restoration. Forty male albino rats were divided into four groups: control (group I), PD (group II), PD-L-Dopa (group III), and PD-exosome (group IV). Motor tests, histopathological examinations, and immunohistochemistry for tyrosine hydroxylase were performed on brain tissue. The levels of α-synuclein, DJ-1, PARKIN, circRNA.2837, and microRNA-34b were measured in brain homogenates. Rotenone induced motor deficits and neuronal alterations. Groups (III) and (IV) showed improvement in motor function, histopathology, α-synuclein, PARKIN, and DJ-1 compared to group (II). Group (IV) showed improvement in microRNA-34b and circRNA.2837 compared to groups (III) and (II). MSC-derived exosomes showed a greater suppression of neurodegenerative disease (ND) compared to L-Dopa in Parkinson's patients.
Collapse
Affiliation(s)
- Asmaa S Mohamed
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, El-Geish Street, Tanta, El Gharbia, Egypt.
| | - Dina S Abdel-Fattah
- Medical Biochemistry Department, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Ghada A Abdel-Aleem
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, El-Geish Street, Tanta, El Gharbia, Egypt
| | - Thanaa F El-Sheikh
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, El-Geish Street, Tanta, El Gharbia, Egypt
| | - Manal M Elbatch
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, El-Geish Street, Tanta, El Gharbia, Egypt
| |
Collapse
|
32
|
Chou Y, Alfarafisa NM, Ikezawa M, Khairani AF. Progress in the Development of Stem Cell-Derived Cell-Free Therapies for Skin Aging. Clin Cosmet Investig Dermatol 2023; 16:3383-3406. [PMID: 38021432 PMCID: PMC10676866 DOI: 10.2147/ccid.s434439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023]
Abstract
Introduction The skin is a vital organ as the body's largest barrier, but its function declines with aging. Therefore, research into effective regeneration treatments must continue to advance. Stem cell transplantation, a cell-based therapy, has become a popular skin-aging treatment, although it comes with drawbacks like host immune reactions. Stem cell-derived cell-free therapies have emerged as an alternative, backed by promising preclinical findings. Stem cell secretomes and extracellular vesicles (EVs) are the key components in cell-free therapy from stem cells. However, comprehensive reviews on the mechanisms of such treatments for skin aging are still limited. Purpose This review discusses stem cell-derived cell-free therapy's potential mechanisms of action related to skin aging prevention by identifying specific molecular targets suitable for the interventions. Methods A search identified 27 relevant in vitro studies on stem cell-derived cell-free therapy interventions in skin aging model cells without restricting publication years using PubMed, Scopus, and Google Scholar. Results Stem cell-derived cell-free therapy can prevent skin aging through various mechanisms, such as (1) involvement of multiple regenerative pathways [NFkb, AP-1, MAPK, P-AKT, NRF2, SIRT-1]; (2) oxidative stress regulation [by reducing oxidants (HO-1, NQO1) and enhancing antioxidants (SOD1, CAT, GP, FRAP)]; (3) preventing ECM degradation [by increasing elastin, collagen, HA, TIMP, and reducing MMP]; (4) regulating cell activity [by reducing cell senescence (SA-β-gal), apoptosis, and cell cycle arrest (P53, P12, P16); and enhancing autophagy, cell migration, and cell proliferation (Ki67)] (5) Regulating the inflammatory pathway [by reducing IL-6, IL-1, TNF-⍺, and increasing TGF-β]. Several clinical trials have also revealed improvements in wrinkles, elasticity, hydration, pores, and pigmentation. Conclusion Stem cell-derived cell-free therapy is a potential novel treatment for skin aging by cell rejuvenation through various molecular mechanisms.
Collapse
Affiliation(s)
- Yoan Chou
- Graduate School of Master Program in Anti Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| | - Nayla Majeda Alfarafisa
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| | - Maiko Ikezawa
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Astrid Feinisa Khairani
- Graduate School of Master Program in Anti Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| |
Collapse
|
33
|
Li Y, Liu C, Han G. Research progress of odontogenic extracellular vesicles in regeneration of dental pulp. Oral Dis 2023; 29:2565-2577. [PMID: 36415913 DOI: 10.1111/odi.14451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/26/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022]
Abstract
It is well understood that maintaining viable pulp is critical for tooth retention. This review focused on cell-free therapy based on extracellular vesicles (EVs), a novel minimally invasive treatment strategy for endodontic restoration. This study was conducted by searching mainstream electronic databases such as Web of Science and PubMed for relevant studies on the therapeutic role of odontogenic EVs in pulp healing published in the last five years. We selected 89 relevant articles and discovered that dental stem cells (DSCs) derived EVs (DSC-EVs) have become a research hotspot in oral regenerative medicine, with significant advantages over cell transplantation in terms of low immunogenicity, ease of isolation, preservation, and management. Here, we introduce in detail the therapeutic effects of DSC-EVs for pulp restoration from three perspectives: excellent odontogenic properties, clinical applications, and possible molecular mechanisms. This article contributes a new viewpoint to the field of regenerative endodontics.
Collapse
Affiliation(s)
- Yanan Li
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Chaoran Liu
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Guanghong Han
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
34
|
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
|
35
|
Liu X, Wu C, Zhang Y, Chen S, Ding J, Chen Z, Wu K, Wu X, Zhou T, Zeng M, Wei D, Sun J, Fan H, Zhou L. Hyaluronan-based hydrogel integrating exosomes for traumatic brain injury repair by promoting angiogenesis and neurogenesis. Carbohydr Polym 2023; 306:120578. [PMID: 36746568 DOI: 10.1016/j.carbpol.2023.120578] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/19/2023]
Abstract
With wide clinical demands, therapies for traumatic brain injury (TBI) are far from satisfactory. Combining the merits of stem cells but avoiding the risk of immunologic rejection, bone marrow mesenchymal stem cell-derived exosomes (BME) attract increasing interests and have been proved effective for TBI repair by intravenous or in situ injection. However, difficulties in sustained delivery or aggregation in lesion sites remain obstacle to using BME for TBI. Inspired by that hydrogels are promising to bridge the destroyed neural gap and provide neural niches, we raised a novel strategy of incorporating BME into hyaluronan-collagen hydrogel (DHC-BME) to achieve both mimicking of brain matrix and steady release of exosomes, and thus realizing TBI repair. External characterizations proved that the BME and DHC synergistically promoted neural stem cells (NSCs) differentiation into neurons and oligodendrocytes while inhibited astrocytes differentiation. DHC-BME induced angiogenesis and neurogenesis, from endogenous NSC recruitment to neuronal differentiation and vascularization to synergistically promote axonal regeneration, remyelination, synapse formation and even brain structural remodeling, and lastly, neurological functional recovery of TBI.
Collapse
Affiliation(s)
- Xiaoyin Liu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chengheng Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China; Institute of Regulatory Science for Medical Devices, Sichuan University, Chengdu 610065, Sichuan, China
| | - Yusheng Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Suping Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Jie Ding
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Zhihong Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Kai Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Xiaoyang Wu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Ting Zhou
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Mingze Zeng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Dan Wei
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Jing Sun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China
| | - Hongsong Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, Sichuan, China.
| | - Liangxue Zhou
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
36
|
Yang T, Li C, Li Y, Cai G, Wang G, He L, He C. MicroRNA-146a-5p alleviates the pathogenesis of osteoarthritis by inhibiting SDF-1/CXCR4-induced chondrocyte autophagy. Int Immunopharmacol 2023; 117:109938. [PMID: 36863142 DOI: 10.1016/j.intimp.2023.109938] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 02/13/2023] [Accepted: 02/23/2023] [Indexed: 03/04/2023]
Abstract
BACKGROUND SDF-1/CXCR4 signaling promotes osteoarthritis (OA) development. CXCR4 is a potential target of miR-146a-5p. This study investigated the therapeutic role and the underlying mechanism of miR-146a-5p in OA. METHODS Human primary chondrocytes C28/I2 were stimulated with SDF-1. Cell viability and LDH release were examined. Chondrocyte autophagy was assessed using Western blot analysis, ptfLC3 transfection, and transmission electron microscopy. MiR-146a-5p mimics were transfected into C28/I2 cells to investigate the role of miR-146a-5p in SDF-1/CXCR4-induced autophagy of chondrocytes. An SDF-1-induced rabbit OA model was established to investigate the therapeutic role of miR-146a-5p in OA. Histological staining was performed to observe the morphology of osteochondral tissue. RESULTS SDF-1/CXCR4 signaling promoted autophagy in C28/I2 cells, as demonstrated by increased LC3-II protein expression and autophagic flux induced by SDF-1. SDF-1 treatment significantly inhibited cell proliferation while promoting necrosis and autophagosome formation in C28/I2 cells. In the presence of SDF-1, miR-146a-5p overexpression in C28/I2 cells suppressed CXCR4 mRNA expression, LC3-II and Beclin-1 protein expression, LDH release, and autophagic flux. In addition, SDF-1 increased the autophagy of chondrocytes in rabbits and promoted the development of OA. Compared with the negative control, miR-146a-5p significantly reduced the morphological abnormalities of the rabbit cartilage that were induced by SDF-1, as well as the number of LC3-II-positive cells, protein expression of LC3-II and Beclin 1, and mRNA expression of CXCR4 in osteochondral tissue. These effects were reversed by the autophagy agonist rapamycin. CONCLUSIONS SDF-1/CXCR4 promotes OA development by enhancing chondrocyte autophagy. MicroRNA-146a-5p may alleviate OA by suppressing CXCR4 mRNA expression and SDF-1/CXCR4-induced chondrocyte autophagy.
Collapse
Affiliation(s)
- Tengyun Yang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Canzhang Li
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Yanlin Li
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China.
| | - Guofeng Cai
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Guoliang Wang
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Lu He
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| | - Chuan He
- Department of Sports Medicine, The First Affiliated Hospital, Kunming Medical University, Kunming 650032, Yunnan, China
| |
Collapse
|
37
|
Secretome of hESC-Derived MSC-like Immune and Matrix Regulatory Cells Mitigate Pulmonary Fibrosis through Antioxidant and Anti-Inflammatory Effects. Biomedicines 2023; 11:biomedicines11020463. [PMID: 36830999 PMCID: PMC9953085 DOI: 10.3390/biomedicines11020463] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Oxidative stress and inflammation are major drivers in the pathogenesis and progression of pulmonary fibrosis (PF). The mesenchymal stem cell (MSC) secretome has regenerative potential and immunomodulatory functions. Human embryonic stem cell (hESC)-derived MSC-like immune and matrix regulatory cells (IMRCs) are manufacturable with large-scale good manufacturing practice (GMP) preparation. In the present study, the antioxidative and anti-inflammatory properties and the therapeutic effect of the secretome of hESC-MSC-IMRC-derived conditioned culture medium (CM) (hESC-MSC-IMRC-CM) were investigated. Results revealed the capacities of hESC-MSC-IMRC-CM to reduce bleomycin (BLM)-induced reactive oxygen species (ROS), extracellular matrix (ECM) deposition, and epithelial-mesenchymal transition (EMT) in A549 cells. The administration of concentrated hESC-MSC-IMRC-CM significantly alleviated the pathogenesis of PF in lungs of BLM-injured mice, as accessed by pathohistological changes and the expression of ECM and EMT. A mechanistic study further demonstrated that the hESC-MSC-IMRC-CM was able to inhibit BLM-induced ROS and pro-inflammatory cytokines, accompanied by a reduced expression of Nox4, Nrf2, Ho-1, and components of the Tlr4/MyD88 signaling cascade. These results provide a proof of concept for the hESC-MSC-IMRC-derived secretome treatment of PF, in part mediated by their antioxidative and anti-inflammatory effects. This study thus reinforces the development of ready-to-use, cell-free hESC-MSC-IMRC secretome biomedicine for the treatment of PF in clinical settings.
Collapse
|
38
|
Oveili E, Vafaei S, Bazavar H, Eslami Y, Mamaghanizadeh E, Yasamineh S, Gholizadeh O. The potential use of mesenchymal stem cells-derived exosomes as microRNAs delivery systems in different diseases. Cell Commun Signal 2023; 21:20. [PMID: 36690996 PMCID: PMC9869323 DOI: 10.1186/s12964-022-01017-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/14/2022] [Indexed: 01/24/2023] Open
Abstract
MicroRNAs (miRNAs) are a group of small non-coding RNAs that regulate gene expression by targeting mRNA. Moreover, it has been shown that miRNAs expression are changed in various diseases, such as cancers, autoimmune disease, infectious diseases, and neurodegenerative Diseases. The suppression of miRNA function can be easily attained by utilizing of anti-miRNAs. In contrast, an enhancement in miRNA function can be achieved through the utilization of modified miRNA mimetics. The discovery of appropriate miRNA carriers in the body has become an interesting subject for investigators. Exosomes (EXOs) therapeutic efficiency and safety for transferring different cellular biological components to the recipient cell have attracted significant attention for their capability as miRNA carriers. Mesenchymal stem cells (MSCs) are recognized to generate a wide range of EXOs (MSC-EXOs), showing that MSCs may be effective for EXO generation in a clinically appropriate measure as compared to other cell origins. MSC-EXOs have been widely investigated because of their immune attributes, tumor-homing attributes, and flexible characteristics. In this article, we summarized the features of miRNAs and MSC-EXOs, including production, purification, and miRNA loading methods of MSC-EXOs, and the modification of MSC-EXOs for targeted miRNA delivery in various diseases. Video abstract.
Collapse
Affiliation(s)
- Elham Oveili
- Department of Pharmaceutical Science, Azad Islamic University of Medical Sciences, Tehran, Iran
| | - Somayeh Vafaei
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Haniyeh Bazavar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yeganeh Eslami
- Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ehsan Mamaghanizadeh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saman Yasamineh
- Department of Biotechnology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Omid Gholizadeh
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Bacteriology and Virology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
39
|
Padilha CS, Antunes BM, Jiménez-Maldonado A, St-Pierre DH, Lira FS. Impact of Breaking up of Sitting Time on Anti-inflammatory Response Induced by Extracellular Vesicles. Curr Pharm Des 2023; 29:2524-2533. [PMID: 37921133 DOI: 10.2174/0113816128244442231018070639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 06/21/2023] [Accepted: 08/31/2023] [Indexed: 11/04/2023]
Abstract
Physical inactivity and sedentary behaviors (SB) have promoted a dramatic increase in the incidence of a host of chronic disorders over the last century. The breaking up of sitting time (i.e., sitting to standing up transition) has been proposed as a promising solution in several epidemiological and clinical studies. In parallel to the large interest it initially created, there is a growing body of evidence indicating that breaking up prolonged sedentary time (i.e., > 7 h in sitting time) could reduce overall mortality risks by normalizing the inflammatory profile and cardiometabolic functions. Recent advances suggest that the latter health benefits, may be mediated through the immunomodulatory properties of extracellular vesicles. Primarily composed of miRNA, lipids, mRNA and proteins, these vesicles would influence metabolism and immune system functions by promoting M1 to M2 macrophage polarization (i.e., from a pro-inflammatory to anti-inflammatory phenotype) and improving endothelial function. The outcomes of interrupting prolonged sitting time may be attributed to molecular mechanisms induced by circulating angiogenic cells. Functionally, circulating angiogenic cells contribute to repair and remodel the vasculature. This effect is proposed to be mediated through the secretion of paracrine factors. The present review article intends to clarify the beneficial contributions of breaking up sitting time on extracellular vesicles formation and macrophage polarization (M1 and M2 phenotypes). Hence, it will highlight key mechanistic information regarding how breaking up sitting time protocols improves endothelial health by promoting antioxidant and anti-inflammatory responses in human organs and tissues.
Collapse
Affiliation(s)
- Camila S Padilha
- Exercise and Immunometabolism Research Group, Post-graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Bárbara M Antunes
- Facultad de Deportes Campus Ensenada, Universidad Autónoma de Baja California, Ensenada, Mexico
| | | | - David H St-Pierre
- Department of Kinesiology, Université du Québec à Montréal (UQAM), Montreal QC, Canada
| | - Fabio S Lira
- Exercise and Immunometabolism Research Group, Post-graduation Program in Movement Sciences, Department of Physical Education, Universidade Estadual Paulista (UNESP), Presidente Prudente, São Paulo, Brazil
| |
Collapse
|
40
|
Proteomics profile of mesenchymal stromal cells and extracellular vesicles in normoxic and hypoxic conditions. Cytotherapy 2022; 24:1211-1224. [PMID: 36192337 DOI: 10.1016/j.jcyt.2022.08.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 08/11/2022] [Accepted: 08/27/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AIMS Although bone marrow-derived mesenchymal stromal cells (MSCs) have demonstrated success in pre-clinical studies, they have shown only mild therapeutic effects in clinical trials. Hypoxia pre-conditioning may optimize the performance of bone marrow-derived MSCs because it better reflects the physiological conditions of their origin. It is not known whether changes in the protein profile caused by hypoxia in MSCs can be extended to the extracellular vesicles (EVs) released from them. The aim of this study was to evaluate the proteomics profile of MSCs and their EVs under normoxic and hypoxic conditions. METHODS Bone marrow-derived MSCs were isolated from six healthy male Wistar rats. After achieving 80% confluence, MSCs were subjected to normoxia (MSC-Norm) (21% oxygen, 5% carbon dioxide, 74% nitrogen) or hypoxia (MSC-Hyp) (1% oxygen, 5% carbon dioxide, 94% nitrogen) for 48 h. Cell viability and oxygen consumption rate were assessed. EVs were extracted from MSCs for each condition (EV-Norm and EV-Hyp) by ultracentrifugation. Total proteins were isolated from MSCs and EVs and prepared for mass spectrometry. EVs were characterized by nanoparticle tracking analysis. Proteomics data were analyzed by PatternLab 4.0, Search Tool for the Retrieval of Interacting Genes/Proteins, Gene Ontology, MetaboAnalyst and Reactome software. RESULTS Cell viability was higher in MSC-Hyp than MSC-Norm (P = 0.007). Basal respiration (P = 0.001), proton leak (P = 0.004) and maximal respiration (P = 0.014) were lower in MSC-Hyp than MSC-Norm, and no changes in adenosine triphosphate-linked and residual respiration were observed. The authors detected 2177 proteins in MSC-Hyp and MSC-Norm, of which 147 were identified in only MSC-Hyp and 512 were identified in only MSC-Norm. Furthermore, 718 proteins were identified in EV-Hyp and EV-Norm, of which 293 were detected in only EV-Hyp and 30 were detected in only EV-Norm. Both MSC-Hyp and EV-Hyp showed enrichment of pathways and biological processes related to glycolysis, the immune system and extracellular matrix organization. CONCLUSIONS MSCs subjected to hypoxia showed changes in their survival and metabolic activity. In addition, MSCs under hypoxia released more EVs, and their content was related to expression of regulatory proteins of the immune system and extracellular matrix organization. Because of the upregulation of proteins involved in glycolysis, gluconeogenesis and glucose uptake during hypoxia, production of reactive oxygen species and expression of immunosuppressive properties may be affected.
Collapse
|
41
|
Wang Z, Zhang X, Qi L, Feng W, Gu Y, Ding Y. Olfactory mucosa tissue-derived mesenchymal stem cells lysate ameliorates LPS-induced acute liver injury in mice. BMC Pulm Med 2022; 22:414. [DOI: 10.1186/s12890-022-02204-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 10/25/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Acute liver injury (ALI) induced by sepsis seriously endangers the health of human beings every year. Mesenchymal stem cells (MSCs) lysate containing various regulators had a positive effect on anti-inflammation, hoping to provide a promising strategy in ALI.
Methods
Olfactory mucosa-derived mesenchymal stem cells (OM-MSCs) were extracted and identified. The collected OM-MSCs were prepared after repeated freeze–thaw in phosphate buffer solution (PBS). Then, OM-MSCs lysate was filtered for future experiments. To understand the composes of OM-MSCs clearly, we detected the components of OM-MSCs lysate by western blotting. In vitro, OM-MSCs lysate was applied to evaluate the effects on normal human liver cells (LO-2) under stimulation of LPS. Lipopolysaccharide (LPS) was also injected intraperitoneally to build ALI model in mice. We further assessed the anti-inflammatory capacity of OM-MSCs lysate on ALI in vivo by aminotransferase determination, pathology observation, and immunohistochemical staining. Moreover, the immunoblot technique was performed to recognize the changes in inflammatory factors and related proteins.
Results
In this study, we found that OM-MSCs lysate could protect structure effectively, improve the plasma aminotransferases, diminish inflammation by releasing interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β). A significant decrease in tumor necrosis factor-α (TNF-α) also occurred under the treatment of OM-MSCs lysate. In addition, trophic factors originating from OM-MSCs lysate provided a supportive micro-environment for liver recovery. Especially, up-expression of vascular endothelial growth factor (VEGF) in vivo revealed that OM-MSCs might have a great potential for healing.
Conclusions
Our results demonstrated that OM-MSCs lysate could alleviate LPS-induced ALI via decreasing inflammatory cytokines and promoting recovery.
Collapse
|
42
|
Luo Y, Li Z, Wang X, Wang J, Duan X, Li R, Peng Y, Ye Q, He Y. Characteristics of culture-condition stimulated exosomes or their loaded hydrogels in comparison with other extracellular vesicles or MSC lysates. Front Bioeng Biotechnol 2022; 10:1016833. [PMID: 36185445 PMCID: PMC9523448 DOI: 10.3389/fbioe.2022.1016833] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/05/2022] [Indexed: 12/12/2022] Open
Abstract
Recently, it has become popular to study the use of extracellular vesicles (EVs) secreted by stem cells to repair damaged tissues or lost cells. Various cell types and physiological fluids release EVs, and they play an important role in cell-to-cell communication. Moreover, EVs have been implicated in important processes, such as immune responses, homeostasis maintenance, coagulation, inflammation, cancer progression, angiogenesis, and antigen presentation. Thus, EVs participate in both physiological and pathological progression. The main classes of EVs include exosomes, microvesicles (MVs), and apoptotic bodies (ApoBDs). Exosomes, which carry a mass of signal molecules such as RNA, DNA, proteins, and lipids, are the most important of these EVs subsets. Currently, exosomes are generating substantial interest in the scientific community. Exosomes loaded hydrogels or under different cultural environments exhibit different properties and functions. Therefore, the exosomes obtained from different sources and conditions are worth reviewing. More importantly, no review article has compared the different EVs, such as exosomes, MVs, ApoBDs, and mesenchymal stem cell (MSC) lysates, which are special soluble substances. The differentiation between EVs and MSC lysates is a logical approach. Accordingly, this review provides an update on the latest progress in studying the roles of culture-condition stimulated exosomes or their loaded hydrogels and the differentiation between exosomes, MVs, ApoBDs, and MSC lysates. Published studies were retrieved from the PubMed® database for review.
Collapse
Affiliation(s)
- Yu Luo
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihua Li
- Department of Orthodontics, School and Hospital of Stomatology, Nanchang University, Nanchang, China
| | - Xinxin Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Juan Wang
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xingxiang Duan
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ruohan Li
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Youjian Peng
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingsong Ye
- Center of Regenerative Medicine and Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Orthodontics, School and Hospital of Stomatology, Nanchang University, Nanchang, China
- *Correspondence: Qingsong Ye, ; Yan He,
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- *Correspondence: Qingsong Ye, ; Yan He,
| |
Collapse
|
43
|
Zhang Y, Huo M, Wang Y, Xiao L, Wu J, Ma Y, Zhang D, Lang X, Wang X. A tailored bioactive 3D porous poly(lactic-acid)-exosome scaffold with osteo-immunomodulatory and osteogenic differentiation properties. J Biol Eng 2022; 16:22. [PMID: 35996115 PMCID: PMC9394013 DOI: 10.1186/s13036-022-00301-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/08/2022] [Indexed: 11/10/2022] Open
Abstract
Polylactic acid (PLA) is a versatile and biodegradable scaffold widely used in biomedical fields to repair tissue defects. Exosomes derived from mesenchymal stem cells (MSCs) are nano-sized extracellular vesicles, which play an important role in tissue engineering in recent years. The primary focus of this study was to develop a bioactive 3D PLA scaffold using exosome-based strategy to improve its osteogenic and immunoregulatory potential. We firstly successfully isolated MSC-derived exosomes (MSC-Exo). Morphological analysis revealed that MSC-Exo exhibits a typical cup-shaped morphology with high expression of exosomal marker CD63. MSC-Exo internalization into recipient cells were also investigated using flow cytometry and confocal laser scanning microscopy. Porous 3D PLA scaffold coated MSC-Exo were used for immunoregulatory and osteogenic testing. Exosomes released from 3D PLA scaffold were validated in RAW264.7 and hBMSCs. The cell proliferation and live/dead assay indicated high biocompatibility for PLA-Exo scaffold. Additionally, PLA-Exo scaffold could reduce the pro-inflammatory marker expression and reactive oxygen species (ROS) production, indicating potential immunoregulatory potential. It is also confirmed that PLA-Exo scaffold could potentiate osteogenic differentiation in the osteogenesis assay. In conclusion, our results demonstrate this bioactive 3D-printed PLA scaffolds with MSC-Exo modification holds immunoregulatory potential and favor osteogenic differentiation, thus having potential applications in bone tissue regeneration.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Hygiene Toxicology, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Mengjie Huo
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Yi Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Lan Xiao
- School of Mechanical, Medical & Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia.,Australia China Centre for Tissue Engineering and Regenerative Medicine, Kelvin Grove, Brisbane, Queensland, 4059, Australia
| | - Jianmei Wu
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Dingmei Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China
| | - Xuemei Lang
- Department of Pre-hospital Emergency, Central Hospital of Chongqing University / Chongqing Emergency Medical Center, Chongqing, Chongqing, 400014, China.
| | - Xin Wang
- Department of Hygiene Toxicology, Zunyi Medical University, Zunyi, 563000, Guizhou, China. .,Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, 563003, Guizhou, China. .,School of Mechanical, Medical & Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology (QUT), Brisbane, QLD, 4000, Australia. .,Australia China Centre for Tissue Engineering and Regenerative Medicine, Kelvin Grove, Brisbane, Queensland, 4059, Australia.
| |
Collapse
|
44
|
Tan MI, Alfarafisa NM, Septiani P, Barlian A, Firmansyah M, Faizal A, Melani L, Nugrahapraja H. Potential Cell-Based and Cell-Free Therapy for Patients with COVID-19. Cells 2022; 11:2319. [PMID: 35954162 PMCID: PMC9367488 DOI: 10.3390/cells11152319] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023] Open
Abstract
Since it was first reported, the novel coronavirus disease 2019 (COVID-19) remains an unresolved puzzle for biomedical researchers in different fields. Various treatments, drugs, and interventions were explored as treatments for COVID. Nevertheless, there are no standard and effective therapeutic measures. Meanwhile, mesenchymal stem cell (MSC) therapy offers a new approach with minimal side effects. MSCs and MSC-based products possess several biological properties that potentially alleviate COVID-19 symptoms. Generally, there are three classifications of stem cell therapy: cell-based therapy, tissue engineering, and cell-free therapy. This review discusses the MSC-based and cell-free therapies for patients with COVID-19, their potential mechanisms of action, and clinical trials related to these therapies. Cell-based therapies involve the direct use and injection of MSCs into the target tissue or organ. On the other hand, cell-free therapy uses secreted products from cells as the primary material. Cell-free therapy materials can comprise cell secretomes and extracellular vesicles. Each therapeutic approach possesses different benefits and various risks. A better understanding of MSC-based and cell-free therapies is essential for supporting the development of safe and effective COVID-19 therapy.
Collapse
Affiliation(s)
- Marselina Irasonia Tan
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| | - Nayla Majeda Alfarafisa
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Sumedang 45363, Indonesia;
| | - Popi Septiani
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| | - Anggraini Barlian
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| | - Mochamad Firmansyah
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| | - Ahmad Faizal
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| | - Lili Melani
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| | - Husna Nugrahapraja
- School of Life Sciences and Technology, Institut Teknologi Bandung, Bandung 40132, Indonesia; (P.S.); (A.B.); (M.F.); (A.F.); (L.M.); (H.N.)
| |
Collapse
|
45
|
Műzes G, Sipos F. Mesenchymal Stem Cell-Derived Secretome: A Potential Therapeutic Option for Autoimmune and Immune-Mediated Inflammatory Diseases. Cells 2022; 11:2300. [PMID: 35892597 PMCID: PMC9367576 DOI: 10.3390/cells11152300] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/16/2022] [Accepted: 07/26/2022] [Indexed: 02/05/2023] Open
Abstract
Immune-mediated inflammatory diseases (IMIDs) encompass several entities such as "classic" autoimmune disorders or immune-mediated diseases with autoinflammatory characteristics. Adult stem cells including mesenchymal stem cells (MSCs) are by far the most commonly used type in clinical practice. However, due to the possible side effects of MSC-based treatments, there is an increase in interest in the MSC-secretome (containing large extracellular vesicles, microvesicles, and exosomes) as an alternative therapeutic option in IMIDs. A wide spectrum of MSC-secretome-related biological activities has been proven thus far including anti-inflammatory, anti-apoptotic, and immunomodulatory properties. In comparison with MSCs, the secretome is less immunogenic but exerts similar biological actions, so it can be considered as an ideal cell-free therapeutic alternative. Additionally, since the composition of the MSC-secretome can be engineered, for a future perspective, it could also be viewed as part of a potential delivery system within nanomedicine, allowing us to specifically target dysfunctional cells or tissues. Although many encouraging results from pre-clinical studies have recently been obtained that strongly support the application of the MSC-secretome in IMIDs, human studies with MSC-secretome administration are still in their infancy. This article reviews the immunomodulatory effects of the MSC-secretome in IMIDs and provides insight into the interpretation of its beneficial biological actions.
Collapse
Affiliation(s)
- Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, Szentkirályi Street 46, 1088 Budapest, Hungary;
| | | |
Collapse
|
46
|
Ren YZ, Ding SS, Jiang YP, Wen H, Li T. Application of exosome-derived noncoding RNAs in bone regeneration: Opportunities and challenges. World J Stem Cells 2022; 14:473-489. [PMID: 36157529 PMCID: PMC9350624 DOI: 10.4252/wjsc.v14.i7.473] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 05/15/2022] [Accepted: 07/11/2022] [Indexed: 02/06/2023] Open
Abstract
With advances in the fields of regenerative medicine, cell-free therapy has received increased attention. Exosomes have a variety of endogenous properties that provide stability for molecular transport across biological barriers to cells, as a form of cell-to-cell communication that regulates function and phenotype. In addition, exosomes are an important component of paracrine signaling in stem-cell-based therapy and can be used as a stand-alone therapy or as a drug delivery system. The remarkable potential of exosomes has paved the pathway for cell-free treatment in bone regeneration. Exosomes are enriched in distinct noncoding RNAs (ncRNAs), including microRNAs, long ncRNAs and circular RNAs. Different ncRNAs have multiple functions. Altered expression of ncRNA in exosomes is associated with the regenerative potential and development of various diseases, such as femoral head osteonecrosis, myocardial infarction, and cancer. Although there is increasing evidence that exosome-derived ncRNAs (exo-ncRNAs) have the potential for bone regeneration, the detailed mechanisms are not fully understood. Here, we review the biogenesis of exo-ncRNA and the effects of ncRNAs on angiogenesis and osteoblast- and osteoclast-related pathways in different diseases. However, there are still many unsolved problems and challenges in the clinical application of ncRNA; for instance, production, storage, targeted delivery and therapeutic potency assessment. Advancements in exo-ncRNA methods and design will promote the development of therapeutics, revolutionizing the present landscape.
Collapse
Affiliation(s)
- Yuan-Zhong Ren
- Department of Emergency Trauma Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471000, Henan Province, China
| | - Shan-Shan Ding
- Department of Geriatrics, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471000, Henan Province, China
| | - Ya-Ping Jiang
- Department of Oral Implantology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong Province, China
| | - Hui Wen
- Department of Emergency Trauma Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang 471000, Henan Province, China
| | - Tao Li
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| |
Collapse
|
47
|
Salehipour Bavarsad S, Jalali MT, Bijan Nejad D, Alypoor B, Babaahmadi Rezaei H, Mohammadtaghvaei N. TGFβ1-Pretreated Exosomes of Wharton Jelly Mesenchymal Stem Cell as a Therapeutic Strategy for Improving Liver Fibrosis. HEPATITIS MONTHLY 2022; 22. [DOI: 10.5812/hepatmon-123416] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/14/2022] [Accepted: 05/26/2022] [Indexed: 01/03/2025]
Abstract
Background: Mesenchymal stem cells (MSCs) are the most promising tools for cell treatment and human tissue regeneration, e.g., in liver fibrosis. Mesenchymal stem cells repair tissue damage through paracrine mediators such as exosomes. Types and concentrations of inflammatory mediators, including transforming growth factor-beta (TGFβ1), in MSCs microenvironment can affect MSCs’ function and therapeutic potency. Objectives: This experimental study aimed to explore the effects of Wharton jelly MSCs (WJ-MSCs) exosomes on fibrotic gene expression and Smad2/3 phosphorylation (phospho-Smad2/3 (p-Smad2/3)). Moreover, we further investigated whether WJ-MSCs pretreatment with different concentrations of TGFβ1 changes the anti-fibrotic properties of their exosomes. Methods: After isolation from the umbilical cord, WJ-MSCs were characterized by observing differentiation and measuring surface biomarkers using flowcytometry. The WJ-MSC-derived exosomes were extracted and identified using transmission electron microscopy (TEM), dynamic light scattering (DLS), and western blotting. Real-time PCR and western blot for extracellular matrix (ECM) and p-Smad2/3 expression detection were used to investigate the effect of exosomes from untreated and TGFβ1-pretreated WJ-MSCs on activated hepatic stellate cells (HSCs). Results: Phospho-Smad2/3, α-smooth muscle actin (α-SMA), and collagen1α1 levels were enhanced following treatment with TGFβ1, whereas E-cadherin was decreased. However, the outcomes were reversed after treatment with WJ-MSC-derived exosomes. Exosomes from TGFβ1-pretreated WJ-MSCs induced a significant decrease in p-Smad2/3 levels in activated HSCs, accompanied by the upregulation of E-cadherin gene expression and downregulation of α-SMA and collagen1α1 when compared to untreated WJ-MSC-derived exosomes. The p-Smad2/3 proteins were significantly decreased (fold change: 0.23, P-value < 0.0001) after exposure to low-dose TGFβ1-pretreated WJ-MSC-derived exosomes (0.1 ng/mL), showing the best effect on activated HSCs. Conclusions: Exosomes derived from untreated WJ-MSCs could regress TGFβ-Smad2/3 signaling and the expression of fibrotic markers in activated LX-2 cells. However, these effects were significantly profound with applying exosomes derived from 0.1 ng/mL TGFβ-pretreated WJ-MSCs. We also observed the dose-response effects of TGFβ on WJ-MSCs-derived exosomes. Therefore, exosomes derived from TGFβ-pretreated WJ-MSCs may be critical in improving fibrosis and benefit liver fibrosis patients.
Collapse
|
48
|
Sipos F, Műzes G. Disagreements in the therapeutic use of mesenchymal stem cell-derived secretome. World J Stem Cells 2022; 14:365-371. [PMID: 35949398 PMCID: PMC9244954 DOI: 10.4252/wjsc.v14.i6.365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/15/2022] [Accepted: 05/27/2022] [Indexed: 02/06/2023] Open
Abstract
In a recent article, the authors provide a detailed summary of the characteristics and biological functions of mesenchymal stem cells (MSCs), as well as a discussion on the potential mechanisms of action of MSC-based therapies. They describe the morphology, biogenesis, and current isolation techniques of exosomes, one of the most important fractions of the MSC-derived secretome. They also summarize the characteristics of MSC-derived exosomes and highlight their functions and therapeutic potential for tissue/organ regeneration and for kidney, liver, cardiovascular, neurological, and musculoskeletal diseases, as well as cutaneous wound healing. Despite the fact that MSCs are regarded as an important pillar of regenerative medicine, their regenerative potential has been demonstrated to be limited in a number of pathological conditions. The negative effects of MSC-based cell therapy have heightened interest in the therapeutic use of MSC-derived secretome. On the other hand, MSC-derived exosomes and microvesicles possess the potential to have a significant impact on disease development, including cancer. MSCs can interact with tumor cells and promote mutual exchange and induction of cellular markers by exchanging secretome. Furthermore, enzymes secreted into and activated within exosomes can result in tumor cells acquiring new properties. As a result, therapeutic applications of MSC-derived secretomes must be approached with extreme caution.
Collapse
Affiliation(s)
- Ferenc Sipos
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest 1088, Hungary.
| | - Györgyi Műzes
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest 1088, Hungary
| |
Collapse
|
49
|
S S, Dahal S, Bastola S, Dayal S, Yau J, Ramamurthi A. Stem Cell Based Approaches to Modulate the Matrix Milieu in Vascular Disorders. Front Cardiovasc Med 2022; 9:879977. [PMID: 35783852 PMCID: PMC9242410 DOI: 10.3389/fcvm.2022.879977] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/20/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) represents a complex and dynamic framework for cells, characterized by tissue-specific biophysical, mechanical, and biochemical properties. ECM components in vascular tissues provide structural support to vascular cells and modulate their function through interaction with specific cell-surface receptors. ECM–cell interactions, together with neurotransmitters, cytokines, hormones and mechanical forces imposed by blood flow, modulate the structural organization of the vascular wall. Changes in the ECM microenvironment, as in post-injury degradation or remodeling, lead to both altered tissue function and exacerbation of vascular pathologies. Regeneration and repair of the ECM are thus critical toward reinstating vascular homeostasis. The self-renewal and transdifferentiating potential of stem cells (SCs) into other cell lineages represents a potentially useful approach in regenerative medicine, and SC-based approaches hold great promise in the development of novel therapeutics toward ECM repair. Certain adult SCs, including mesenchymal stem cells (MSCs), possess a broader plasticity and differentiation potential, and thus represent a viable option for SC-based therapeutics. However, there are significant challenges to SC therapies including, but not limited to cell processing and scaleup, quality control, phenotypic integrity in a disease milieu in vivo, and inefficient delivery to the site of tissue injury. SC-derived or -inspired strategies as a putative surrogate for conventional cell therapy are thus gaining momentum. In this article, we review current knowledge on the patho-mechanistic roles of ECM components in common vascular disorders and the prospects of developing adult SC based/inspired therapies to modulate the vascular tissue environment and reinstate vessel homeostasis in these disorders.
Collapse
|
50
|
Bladder Cancer Cells Exert Pleiotropic Effects on Human Adipose-Derived Stem Cells. Life (Basel) 2022; 12:life12040549. [PMID: 35455040 PMCID: PMC9025060 DOI: 10.3390/life12040549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/29/2022] [Accepted: 04/05/2022] [Indexed: 12/12/2022] Open
Abstract
Stem cell-based therapies are considered one of the most promising disciplines in biomedicine. Bladder cancer patients could benefit from therapies directed to promote healing after invasive surgeries or to lessen urinary incontinence, a common side effect of both cancer itself and the treatment. However, the local delivery of cells producing large amounts of paracrine factors may alter interactions within the microenvironment. For this reason, reconstructive cellular therapies for patients with a history of cancer carry a potential risk of tumor reactivation. We used an indirect co-culture model to characterize the interplay between adipose-derived stem cells and bladder cancer cells. Incubation with ASCs increased MCP-1 secretion by bladder cancer cells (from 2.1-fold to 8.1-fold, depending on the cell line). Cancer cell-derived factors altered ASC morphology. Cells with atypical shapes and significantly enlarged volumes appeared within the monolayer. Incubation in a conditioned medium (CM) containing soluble mediators secreted by 5637 and HB-CLS-1 bladder cancer cell lines decreased ASC numbers by 47.5% and 45.7%. A significant increase in adhesion to ECM components, accompanied by reduced motility and sheet migration, was also observed after incubation in CM from 5637 and HB-CLS-1 cells. No differences were observed when ASCs were co-cultured with HT-1376 cells. Our previous and present results indicate that soluble mediators secreted by ASCs and bladder cancer cells induce opposite effects influencing cells that represent the non-muscle-invasive urinary bladder.
Collapse
|