1
|
Kheradmand F, Yasaman Rahimzadeh SF, Esmaeili SA, Negah SS, Farkhad NK, Nazari SE, Hajinejad M, Khodadoust MA, Fadaee A, Afshari JT, Khazaei M. Efficacy of umbilical cord-derived mesenchymal stem cells and exosomes in conjunction with standard IBD drug on immune responses in an IBD mouse model. Stem Cell Res Ther 2025; 16:5. [PMID: 39773498 PMCID: PMC11707839 DOI: 10.1186/s13287-024-04062-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 11/11/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a persistent inflammation of the digestive system, and Mesenchymal Stem Cells (MSCs) and their exosomes have demonstrated potential as treatments for this condition. The objective of this research was to examine the possible effectiveness of intraperitoneal injection of umbilical cord-MSCs (UC-MSCs) and their exosomes through a two-time injection regimen in a mouse model. METHOD In this study, an animal model of a specific type of IBD in C57BL/6 mice, induced by dextran sulfate sodium (DSS), was utilized. The mice were treated with MSCs, exosomes, Mesalazine, and a combination of them. Upon sacrificing the mice, colon and spleen tissues were isolated to assess the changes in the mice's weight, colon length, spleen weight, and colitis' pathological symptoms. IL-10 and IL-17 levels were measured, and Treg and Th17 cell percentages were determined as well. Furthermore, colon tissue was stained to investigate histopathological changes. RESULTS In the groups that received MSCs, there was a significant reduction in the disease activity index and their combinations with exosomes and Mesalazine compared to the colitis group. Colon length increased in all groups except the exosome group. Histological measures were notably reduced in the MSC groups and their combinations. Significant increases in the IL-10 level of colon tissue and the proportion of Treg present in the spleen were observed in the groups receiving MSC and combination treatment. Furthermore, these groups showed a notable reduction in the percentage of spleen Th17 cells. However, IL17A decreased non-significantly in all groups. CONCLUSION The results showed that intraperitoneal injection of UC-MSCs and their combination with exosome and Mesalazine in a murine colitis model improved the disease's symptoms. Therefore, MSCs and their combination with exosomes can be a promising therapeutic approach along with other common drugs for IBD, but exosomes alone could not significantly reduce the symptoms of colitis.
Collapse
Affiliation(s)
- Fatemeh Kheradmand
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Fatemeh Yasaman Rahimzadeh
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Islamic Azad University, Garmsar Branch, Faculty of Veterinary Medicine, Tehran, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Najmeh Kaffash Farkhad
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Elnaz Nazari
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Hajinejad
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Qaen Faculty of Medical Science, Birjand University of Medical Science, Birjand, Iran
| | - Mohammad Ali Khodadoust
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsane Fadaee
- Immunology Research Center, Inflammation and Inflammatory Diseases Division, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Tavakol Afshari
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Majid Khazaei
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Chen J, Chen J, Li Q, Hu M, Zhong X, Yu L, Zhang X, Huang H, Liu J, Huang Z, Liu X, Xiong W. Astragaloside promotes the secretion of MSC-derived exosomal miR-146a-5p by regulating TRAF6/NF-κB pathway to attenuate inflammation in high glucose-impaired endothelial cells. In Vitro Cell Dev Biol Anim 2025; 61:93-106. [PMID: 39441504 DOI: 10.1007/s11626-024-00984-2] [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: 05/29/2024] [Accepted: 09/04/2024] [Indexed: 10/25/2024]
Abstract
This study aimed to explore the potential of using mesenchymal stem cell (MSC)-derived exosomes (MSC-Exos) pre-treated with Astragaloside IV (ASIV) to alleviate inflammation in high glucose (HG)-damaged endothelial cells. MSC-Exos were isolated from untreated MSCs and ASIV-pre-treated MSCs, and their characteristics were assessed. The expression of miR-146a-5p in MSC-Exos was determined, and it was found that ASIV treatment enhanced its expression. In order to assess the impact of highly miR-146a-5p-expressing MSC-Exos on HG-injured endothelial cells, we established a model of HG-induced inflammation using human umbilical vein endothelial cells (HUVECs). The study measured cell viability, apoptosis, tube formation, and levels of inflammatory cytokines among the different treatment groups. It was found that transferring MSC-Exos with high miR-146a-5p expression to HG-damaged HUVECs increased cell viability and tube formation ability while reducing the number of apoptotic cells. Additionally, changes in inflammatory factors indicated a reduction in the inflammatory response. Further investigation demonstrated that miR-146a-5p inhibited the expression of TNF receptor associated factor 6 (TRAF6) and phosphorylated NF-κB, which are involved in the inflammatory response. This resulted in the alleviation of inflammation in HG-damaged endothelial cells. In summary, our findings indicate that ASIV treatment stimulated the secretion of MSC-Exos that exhibited increased levels of miR-146a-5p. These exosomes, in turn, regulated the TRAF6/NF-κB pathway. As a result of this modulation, the inflammatory response in HG-damaged endothelial cells was alleviated. These findings offer a fresh approach to addressing vascular complications associated with diabetes, which could lead to novel treatment strategies in the field.
Collapse
Affiliation(s)
- Jiye Chen
- Burn and Plastic Surgery Department of Yiyang Central Hospital in Hunan Province, Yiyang, 413000, China
| | - Jiayao Chen
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410003, China
| | - Qinxia Li
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410003, China
| | - Minxia Hu
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410003, China
| | - Xingxing Zhong
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410003, China
| | - Liang Yu
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410003, China
| | - Xi Zhang
- Clinical Medical School of Hunan University of Chinese Medicine, Hunan Brain Hospital, Yiyang, Changsha, 410007, China
| | - Hongyu Huang
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, 410003, China
| | - Jing Liu
- Burn and Plastic Surgery Department of Yiyang Central Hospital in Hunan Province, Yiyang, 413000, China
| | - Ziyi Huang
- College of Acupuncture, Massage and Rehabilitation, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xinyi Liu
- College of Traditional Chinese Medicie, Hunan University of Chinese Medicine, Changsha, 410003, China
| | - Wu Xiong
- Department of Breast Surgery, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410003, China.
| |
Collapse
|
3
|
Wu Y, Wang X, Zeng Y, Liu X. Exosomes are the mediators between the tumor microenvironment and prostate cancer (Review). Exp Ther Med 2024; 28:439. [PMID: 39355518 PMCID: PMC11443591 DOI: 10.3892/etm.2024.12728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/23/2024] [Indexed: 10/03/2024] Open
Abstract
Prostate cancer poses a serious threat to the well-being of men worldwide, with the leading cause of mortality being primarily through metastasis. Prostate cancer metastasis is dependent on cell communication, which is an essential component of this process; yet its exact mechanism remains obscure. Nonetheless, cell-to-cell communication plays a critical part in prostate cancer metastasis. Exosomes play an indispensable role in the development of metastatic growth by promoting intercellular communication. They are pivotal regulatory agents for both prostate cancer cells as well as their microenvironment. The present study investigated the makeup and function of exosomes in the tumor microenvironment, highlighting their significance to prostate cancer metastasis.
Collapse
Affiliation(s)
- Yiqi Wu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiao Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yan Zeng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
4
|
Wu Y, Liang Z, Li K, Feng J. Knockdown of HOTAIR Alleviates High Glucose-Induced Apoptosis and Inflammation in Retinal Pigment Epithelial Cells. Appl Biochem Biotechnol 2024:10.1007/s12010-024-05083-2. [PMID: 39607470 DOI: 10.1007/s12010-024-05083-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
Diabetic retinopathy (DR) is one of the most common microvascular complications in diabetes. Accumulating evidence demonstrated that long non-coding RNAs (lncRNAs) played critical regulatory roles in DR. However, the role of lncRNA HOX Transcript Antisense Intergenic RNA (HOTAIR) in the high glucose (HG)-induced human retinal pigment epithelial (RPE) cell injury remains unclear. Herein, we found the expression of HOTAIR was increased in the retina of DR rats and HG-induced ARPE-19 cells. Knockdown of HOTAIR improved viability, inhibited apoptosis, increased Bcl-2 protein levels, and decreased Bax and cleaved caspase 3 protein levels in HG-treated ARPE-19 cells. Moreover, enzyme-linked immunosorbent assay showed that HOTAIR silencing reduced interleukin 6 and tumor necrosis factor-α release of ARPE-19 cells under HG conditions. Mechanistically, luciferase reporter assay and RNA immunoprecipitation assay validated that HOTAIR could directly sponge miR-326 to upregulate transcription factor 4 (TCF4) expression. Furthermore, rescue experiments confirmed that HOTAIR promoted apoptosis and inflammation of HG-treated ARPE-19 cells by the miR-326/TCF4 axis. In summary, HOTAIR enhanced HG-induced retinal pigment epithelial cell injury by promoting apoptosis and inflammation, shedding light on the importance of HOTAIR as a novel potential target for DR treatment.
Collapse
Affiliation(s)
- Yanping Wu
- Department of Pediatric Ophthalmology, Cangzhou Central Hospital, No. 16, Xinhua West Street, Cangzhou, 061000, Hebei Province, China.
| | - Zenghui Liang
- Department of Interventional Medicine, Cangzhou People's Hospital, Cangzhou, China
| | - Kun Li
- Department of Pediatric Ophthalmology, Cangzhou Central Hospital, No. 16, Xinhua West Street, Cangzhou, 061000, Hebei Province, China
| | - Junli Feng
- Department of Pediatric Ophthalmology, Cangzhou Central Hospital, No. 16, Xinhua West Street, Cangzhou, 061000, Hebei Province, China
| |
Collapse
|
5
|
Tang S, Feng W, Li Z, Liu X, Yang T, Wei F, Ding G. Extracellular Vesicles Derived from Lipopolysaccharide-Pretreated Periodontal Ligament Stem Cells Ameliorate Inflammatory Responses in Experimental Colitis via the PI3K/AKT Signaling Pathway. Int J Nanomedicine 2024; 19:11997-12013. [PMID: 39583323 PMCID: PMC11583767 DOI: 10.2147/ijn.s494321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024] Open
Abstract
Introduction Inflammatory bowel disease is a complex chronic inflammatory condition characterized by dysbiosis of the gut microbiota and dysregulation of immune system. In recent years, extracellular vesicles (EVs) derived from mesenchymal stem cells have garnered significant attention for their beneficial potentials in immune modulation and tissue repair. This study aims to evaluate the therapeutic effects and underlying mechanisms of EVs derived from lipopolysaccharide (LPS)-pretreated periodontal ligament stem cells (PDLSCs) in mice with colitis. Methods A mouse model of colitis was established using 3.0% dextran sulfate sodium (DSS). Following the induction of colitis, mice were treated via tail vein injection with either conventional PDLSC-derived EVs (P-EVs) or LPS-pretreated PDLSC-derived EVs (LPS pre-EVs). The EVs were characterized using transmission electron microscopy, nanoparticle tracking analysis, and Western blot analysis. The therapeutic effects and mechanisms were evaluated through a combination of small animal live imaging, disease activity index (DAI) scoring, histopathological staining, qRT-PCR, 16S rRNA gene sequencing, and mass spectrometry analysis. Results The LPS pre-EVs exhibited typical EVs characteristics in terms of morphology, particle size distribution, and marker protein expression. Compared to P-EVs, LPS pre-EVs significantly ameliorated weight loss, DAI scores, colon length, and perianal symptoms in DSS-induced murine colitis. Additionally, LPS pre-EVs up-regulated the expression of Arginase-1, a typical M2 macrophage marker, and tight junction proteins, including ZO-1, Occludin, and Claudin-1, enhanced gut microbial diversity, and significantly regulated intestinal protein expression and activation of the PI3K/AKT signaling pathway. Conclusion LPS pre-EVs exhibit significant anti-inflammatory and tissue repair effects in a mouse model of colitis. The underlying mechanisms may involve the regulation of macrophage polarization, maintenance of intestinal barrier function, modulation of the gut microbiota, and activation of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Shuai Tang
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong, 261053, People’s Republic of China
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Wenyu Feng
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Zekun Li
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong, 261053, People’s Republic of China
| | - Xinjuan Liu
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong, 261053, People’s Republic of China
| | - Tong Yang
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong, 261053, People’s Republic of China
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Fulan Wei
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Shandong Key Laboratory of Oral Tissue Regeneration, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
- Shandong Provincial Clinical Research Center for Oral Diseases, Shandong University, Jinan, Shandong, 250012, People’s Republic of China
| | - Gang Ding
- School of Stomatology, Shandong Second Medical University, Weifang, Shandong, 261053, People’s Republic of China
| |
Collapse
|
6
|
Clua‐Ferré L, Suau R, Vañó‐Segarra I, Ginés I, Serena C, Manyé J. Therapeutic potential of mesenchymal stem cell-derived extracellular vesicles: A focus on inflammatory bowel disease. Clin Transl Med 2024; 14:e70075. [PMID: 39488745 PMCID: PMC11531661 DOI: 10.1002/ctm2.70075] [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: 07/09/2024] [Revised: 09/13/2024] [Accepted: 10/16/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have emerged as key regulators of intercellular communication, orchestrating essential biological processes by delivering bioactive cargoes to target cells. Available evidence suggests that MSC-EVs can mimic the functions of their parental cells, exhibiting immunomodulatory, pro-regenerative, anti-apoptotic, and antifibrotic properties. Consequently, MSC-EVs represent a cell-free therapeutic option for patients with inflammatory bowel disease (IBD), overcoming the limitations associated with cell replacement therapy, including their non-immunogenic nature, lower risk of tumourigenicity, cargo specificity and ease of manipulation and storage. MAIN TOPICS COVERED This review aims to provide a comprehensive examination of the therapeutic efficacy of MSC-EVs in IBD, with a focus on their mechanisms of action and potential impact on treatment outcomes. We examine the advantages of MSC-EVs over traditional therapies, discuss methods for their isolation and characterisation, and present mechanistic insights into their therapeutic effects through transcriptomic, proteomic and lipidomic analyses of MSC-EV cargoes. We also discuss available preclinical studies demonstrating that MSC-EVs reduce inflammation, promote tissue repair and restore intestinal homeostasis in IBD models, and compare these findings with those of clinical trials. CONCLUSIONS Finally, we highlight the potential of MSC-EVs as a novel therapy for IBD and identify challenges and opportunities associated with their translation into clinical practice. HIGHLIGHTS The source of mesenchymal stem cells (MSCs) strongly influences the composition and function of MSC-derived extracellular vesicles (EVs), affecting their therapeutic potential. Adipose-derived MSC-EVs, known for their immunoregulatory properties and ease of isolation, show promise as a treatment for inflammatory bowel disease (IBD). MicroRNAs are consistently present in MSC-EVs across cell types and are involved in pathways that are dysregulated in IBD, making them potential therapeutic agents. For example, miR-let-7a is associated with inhibition of apoptosis, miR-100 supports cell survival, miR-125b helps suppress pro-inflammatory cytokines and miR-20 promotes anti-inflammatory M2 macrophage polarisation. Preclinical studies in IBD models have shown that MSC-EVs reduce intestinal inflammation by suppressing pro-inflammatory mediators (e.g., TNF-α, IL-1β, IL-6) and increasing anti-inflammatory factors (e.g., IL-4, IL-10). They also promote mucosal healing and strengthen the integrity of the gut barrier, suggesting their potential to address IBD pathology.
Collapse
Affiliation(s)
- Laura Clua‐Ferré
- Germans Trias i Pujol Research Institute IGTPInflammatory Bowel DiseasesBadalonaSpain
| | - Roger Suau
- Germans Trias i Pujol Research Institute IGTPInflammatory Bowel DiseasesBadalonaSpain
| | - Irene Vañó‐Segarra
- Hospital Universitari Joan XXIIIInstitut d'investigació sanitària Pere VirgiliTarragonaSpain
| | - Iris Ginés
- Hospital Universitari Joan XXIIIInstitut d'investigació sanitària Pere VirgiliTarragonaSpain
| | - Carolina Serena
- Hospital Universitari Joan XXIIIInstitut d'investigació sanitària Pere VirgiliTarragonaSpain
| | - Josep Manyé
- Germans Trias i Pujol Research Institute IGTPInflammatory Bowel DiseasesBadalonaSpain
- Centro de Investigación Biomédica en RedMadridSpain
| |
Collapse
|
7
|
Klyucherev TO, Peshkova MA, Revokatova DP, Serejnikova NB, Fayzullina NM, Fayzullin AL, Ershov BP, Khristidis YI, Vlasova II, Kosheleva NV, Svistunov AA, Timashev PS. The Therapeutic Potential of Exosomes vs. Matrix-Bound Nanovesicles from Human Umbilical Cord Mesenchymal Stromal Cells in Osteoarthritis Treatment. Int J Mol Sci 2024; 25:11564. [PMID: 39519121 PMCID: PMC11545893 DOI: 10.3390/ijms252111564] [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: 09/14/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease with limited therapeutic options, where inflammation plays a critical role in disease progression. Extracellular vesicles (EV) derived from mesenchymal stromal cells (MSC) have shown potential as a therapeutic approach for OA by modulating inflammation and alleviating degenerative processes in the joint. This study evaluated the therapeutic effects for the treatment of OA of two types of EV-exosomes and matrix-bound nanovesicles (MBV)-both derived from the human umbilical cord MSC (UC-MSC) via differential ultracentrifugation. Different phenotypes of human monocyte-derived macrophages (MDM) were used to study the anti-inflammatory properties of EV in vitro, and the medial meniscectomy-induced rat model of knee osteoarthritis (MMx) was used in vivo. The study found that both EV reduced pro-inflammatory cytokines IL-6 and TNF-α in MDM. However, exosomes showed superior results, preserving the extracellular matrix (ECM) of hyaline cartilage, and reducing synovitis more effectively than MBVs. Additionally, exosomes downregulated inflammatory markers (TNF-α, iNOS) and increased Arg-1 expression in macrophages and synovial fibroblasts, indicating a stronger anti-inflammatory effect. These results suggest UC-MSC exosomes as a promising therapeutic option for OA, with the potential for modulating inflammation and promoting joint tissue regeneration.
Collapse
Affiliation(s)
- Timofey O. Klyucherev
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Maria A. Peshkova
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Daria P. Revokatova
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Natalia B. Serejnikova
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Laboratory of Digital Microscopic Analysis, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Nafisa M. Fayzullina
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Alexey L. Fayzullin
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Laboratory of Digital Microscopic Analysis, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Boris P. Ershov
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Yana I. Khristidis
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Irina I. Vlasova
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Nastasia V. Kosheleva
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | | | - Peter S. Timashev
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| |
Collapse
|
8
|
Kumar P, Kedia S, Ahuja V. Target potential of miRNAs in ulcerative colitis: what do we know? Expert Opin Ther Targets 2024; 28:829-841. [PMID: 39307951 DOI: 10.1080/14728222.2024.2408423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION The global rise in ulcerative colitis (UC) incidence highlights the urgent need for enhanced diagnostic and therapeutic strategies. Recent advances in genome-wide association studies (GWAS) have identified genetic loci associated with UC, providing insights into the disease's molecular mechanisms, including immune modulation, mucosal defense, and epithelial barrier function. Despite these findings, many GWAS signals are located in non-coding regions and are linked to low risk, suggesting that protein-coding genes alone do not fully explain UC's pathophysiology. Emerging research emphasizes the potential of microRNAs (miRNAs) as biomarkers and therapeutic targets due to their crucial role in UC. This review explores the current understanding of miRNAs in UC, including their mechanisms of action and their potential as both biomarkers and therapeutic targets. The present review provides the latest update on their potential as a biomarker and therapeutic target. AREAS COVERED This review synthesizes an extensive literature search on miRNAs in UC, focusing on their roles in the mucosal barrier, innate and adaptive immunity, and their potential applications as biomarkers and therapeutic modalities. EXPERT OPINION While miRNAs present promising opportunities as biomarkers and novel therapeutic agents in UC, challenges in validation, specificity, delivery, and clinical application need to be addressed through rigorous, large-scale studies.
Collapse
Affiliation(s)
- Peeyush Kumar
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical sciences, New Delhi, India
| | - Saurabh Kedia
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical sciences, New Delhi, India
| | - Vineet Ahuja
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical sciences, New Delhi, India
| |
Collapse
|
9
|
Li YJ, Yu ZY, Zhang D, Zhang FR, Zhang DM, Chen M. Extracellular vesicles for the treatment of ulcerative colitis: A systematic review and meta-analysis of animal studies. Heliyon 2024; 10:e36890. [PMID: 39281542 PMCID: PMC11400994 DOI: 10.1016/j.heliyon.2024.e36890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/18/2024] Open
Abstract
Background Extracellular vesicles (EVs) are being considered as a potential therapeutic option for ulcerative colitis (UC), and numerous preclinical studies have been conducted on the use of EVs for UC. Methods A systematic review was conducted to compare the therapeutic effects of mammalian EVs and placebo on UC in animal models, along with a meta-analysis comparing naïve (unmodified) EVs and placebo. The search was performed in four databases (PubMed, Web of Science, Scopus, and EMBASE) up to September 13th, 2023. The primary outcomes included disease activity index (DAI), colonic mucosal damage index (CMDI), and adverse effects (PROSPERO ID: CRD42023458039). Results A total of 69 studies were included based on pre-determined criteria, involving 1271 animals. Of these studies, 51 measured DAI scores, with 98 % reporting that EVs could reduce DAI scores. Additionally, 5 studies reported CMDI and all showed that EVs could significantly reduce CMDI. However, only 3 studies assessed adverse effects and none reported any significant adverse effects. The meta-analysis of these studies (40 studies involving 1065 animals) revealed that naïve EVs could significantly decrease the DAI score (SMD = -3.00; 95 % CI: -3.52 to -2.48) and CMDI (SMD = -2.10; 95 % CI: -2.85 to -1.35). Conclusion The results indicate that mammalian EVs have demonstrated therapeutic benefits in animal models of UC; however, the safety profile of EVs remains inadequate which highlights the need for further research on safety outcomes.
Collapse
Affiliation(s)
- Yu-Jing Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, NO.11 North Third Ring Road East, Chaoyang District, Beijing, 100029, China
| | - Ze-Yu Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, NO.11 North Third Ring Road East, Chaoyang District, Beijing, 100029, China
- Centre for Evidence-Based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Di Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, NO.11 North Third Ring Road East, Chaoyang District, Beijing, 100029, China
| | - Fu-Rong Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, NO.11 North Third Ring Road East, Chaoyang District, Beijing, 100029, China
| | - Dong-Mei Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, No.5 Haiyuncang Road, Dongcheng District, Beijing, 101121, China
| | - Meng Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, NO.11 North Third Ring Road East, Chaoyang District, Beijing, 100029, China
| |
Collapse
|
10
|
Xu X, Peng J, Wang N, Ocansey DKW, Zhang X, Mao F. hucMSC-Ex alleviates inflammatory bowel disease in mice by enhancing M2-type macrophage polarization via the METTL3-Slc37a2-YTHDF1 axis. Cell Biol Toxicol 2024; 40:74. [PMID: 39259386 PMCID: PMC11390928 DOI: 10.1007/s10565-024-09921-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
Human umbilical cord mesenchymal stem cell-derived exosomes (hucMSC-Ex) have emerged as a new treatment strategy for inflammatory bowel disease (IBD) due to their immunoregulatory function. N6-methyladenosine (m6A) plays a crucial role in regulating intestinal immunity, especially in IBD where macrophages play an important role, although its mechanism is not yet fully understood. From this perspective, this research aimed to evaluate the effect of hucMSC-Ex on m6A modification of macrophages in IBD. In the process of alleviating inflammation, hucMSC-Ex promotes macrophage polarization toward the M2 type and regulates intracellular m6A levels by upregulating the expression of m6A "Writer" METTL3 and "Reader" YTHDF1. Solute Carrier Family 37 Member 2 (Slc37a2) was identified by Methylation RNA immunoprecipitation sequencing as the target molecule of the hucMSC-Ex. Mechanically, hucMSC-Ex promoted the binding of METTL3 to the Slc37a2 mRNA complex, and enhanced the binding of Slc37a2 to YTHDF1 to upregulate the intracellular expression of Slc37a2, thereby attenuating the pro-inflammatory function of macrophage. This study confirms the modulatory role of hucMSC-Ex on the m6A modification of macrophages in IBD, providing a new scientific basis for the treatment of IBD with hucMSC-Ex.
Collapse
Affiliation(s)
- Xinwei Xu
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, 222006, Jiangsu, P.R. China
- Clinical Laboratory, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, 221000, Jiangsu, China
| | - Jianhua Peng
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, 212300, Jiangsu, P.R. China
| | - Naijian Wang
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, 222006, Jiangsu, P.R. China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, CC0959347, Cape Coast, Ghana
| | - Xu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China.
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, 222006, Jiangsu, P.R. China.
| |
Collapse
|
11
|
Chen LP, Gui XD, Tian WD, Kan HM, Huang JZ, Ji FH. Botulinum toxin type A-targeted SPP1 contributes to neuropathic pain by the activation of microglia pyroptosis. World J Psychiatry 2024; 14:1254-1266. [PMID: 39165552 PMCID: PMC11331382 DOI: 10.5498/wjp.v14.i8.1254] [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: 04/03/2024] [Revised: 05/29/2024] [Accepted: 07/02/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Neuropathic pain (NP) is the primary symptom of various neurological conditions. Patients with NP often experience mood disorders, particularly depression and anxiety, that can severely affect their normal lives. Microglial cells are associated with NP. Excessive inflammatory responses, especially the secretion of large amounts of pro-inflammatory cytokines, ultimately lead to neuroinflammation. Microglial pyroptosis is a newly discovered form of inflammatory cell death associated with immune responses and inflammation-related diseases of the central nervous system. AIM To investigate the effects of botulinum toxin type A (BTX-A) on microglial pyroptosis in terms of NP and associated mechanisms. METHODS Two models, an in vitro lipopolysaccharide (LPS)-stimulated microglial cell model and a selective nerve injury model using BTX-A and SPP1 knockdown treatments, were used. Key proteins in the pyroptosis signaling pathway, NLRP3-GSDMD, were assessed using western blotting, real-time quantitative polymerase chain reaction, and immunofluorescence. Inflammatory factors [interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α] were assessed using enzyme-linked immunosorbent assay. We also evaluated microglial cell proliferation and apoptosis. Furthermore, we measured pain sensation by assessing the delayed hind paw withdrawal latency using thermal stimulation. RESULTS The expression levels of ACS and GSDMD-N and the mRNA expression of TNF-α, IL-6, and IL-1β were enhanced in LPS-treated microglia. Furthermore, SPP1 expression was also induced in LPS-treated microglia. Notably, BTX-A inhibited SPP1 mRNA and protein expression in the LPS-treated microglia. Additionally, depletion of SPP1 or BTX-A inhibited cell viability and induced apoptosis in LPS-treated microglia, whereas co-treatment with BTX-A enhanced the effect of SPP1 short hairpin (sh)RNA in LPS-treated microglia. Finally, SPP1 depletion or BTX-A treatment reduced the levels of GSDMD-N, NLPRP3, and ASC and suppressed the production of inflammatory factors. CONCLUSION Notably, BTX-A therapy and SPP1 shRNA enhance microglial proliferation and apoptosis and inhibit microglial death. It improves pain perception and inhibits microglial activation in rats with selective nerve pain.
Collapse
Affiliation(s)
- Li-Ping Chen
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Xiao-Die Gui
- Department of Pain, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Wen-Di Tian
- Department of Pain, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Hou-Ming Kan
- Faculty of Medicine, Macao University of Science and Technology, Macau 999078, China
| | - Jin-Zhao Huang
- Department of Pain, Xuzhou Medical University, Xuzhou 221004, Jiangsu Province, China
| | - Fu-Hai Ji
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
12
|
Liao HX, Mao X, Wang L, Wang N, Ocansey DKW, Wang B, Mao F. The role of mesenchymal stem cells in attenuating inflammatory bowel disease through ubiquitination. Front Immunol 2024; 15:1423069. [PMID: 39185411 PMCID: PMC11341407 DOI: 10.3389/fimmu.2024.1423069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Inflammatory bowel disease (IBD), a condition of the digestive tract and one of the autoimmune diseases, is becoming a disease of significant global public health concern and substantial clinical burden. Various signaling pathways have been documented to modulate IBD, but the exact activation and regulatory mechanisms have not been fully clarified; thus, a need for constant exploration of the molecules and pathways that play key roles in the development of IBD. In recent years, several protein post-translational modification pathways, such as ubiquitination, phosphorylation, methylation, acetylation, and glycolysis, have been implicated in IBD. An aberrant ubiquitination in IBD is often associated with dysregulated immune responses and inflammation. Mesenchymal stem cells (MSCs) play a crucial role in regulating ubiquitination modifications through the ubiquitin-proteasome system, a cellular machinery responsible for protein degradation. Specifically, MSCs have been shown to influence the ubiquitination of key signaling molecules involved in inflammatory pathways. This paper reviews the recent research progress in MSC-regulated ubiquitination in IBD, highlighting their therapeutic potential in treating IBD and offering a promising avenue for developing targeted interventions to modulate the immune system and alleviate inflammatory conditions.
Collapse
Affiliation(s)
- Hong Xi Liao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, China
| | - Xiaojun Mao
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu, China
| | - Lan Wang
- Department of Laboratory Medicine, Danyang Blood Station, Zhenjiang, Jiangsu, China
| | - Naijian Wang
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Bo Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Jiangsu University, Lianyungang, Jiangsu, China
| |
Collapse
|
13
|
Arbade G, Jose JV, Gulbake A, Kadam S, Kashte SB. From stem cells to extracellular vesicles: a new horizon in tissue engineering and regenerative medicine. Cytotechnology 2024; 76:363-401. [PMID: 38933869 PMCID: PMC11196501 DOI: 10.1007/s10616-024-00631-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 04/07/2024] [Indexed: 06/28/2024] Open
Abstract
In the fields of tissue engineering and regenerative medicine, extracellular vesicles (EVs) have become viable therapeutic tools. EVs produced from stem cells promote tissue healing by regulating the immune system, enhancing cell proliferation and aiding remodeling processes. Recently, EV has gained significant attention from researchers due to its ability to treat various diseases. Unlike stem cells, stem cell-derived EVs show lower immunogenicity, are less able to overcome biological barriers, and have a higher safety profile. This makes the use of EVs derived from cell-free stem cells a promising alternative to whole-cell therapy. This review focuses on the biogenesis, isolation, and characterization of EVs and highlights their therapeutic potential for bone fracture healing, wound healing, and neuronal tissue repair and treatment of kidney and intestinal diseases. Additionally, this review discusses the potential of EVs for the treatment of cancer, COVID-19, and HIV. In summary, the use of EVs derived from stem cells offers a new horizon for applications in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
| | | | - Arvind Gulbake
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, (NIPER G), Guwahati, Assam 781101 India
| | - Sachin Kadam
- Sophisticated Analytical and Technical Help Institute, Indian Institute of Technology, Delhi, New Delhi 110016 India
| | - Shivaji B. Kashte
- Department of Stem Cell and Regenerative Medicine, Centre for Interdisciplinary Research, D. Y. Patil Education Society (Institution Deemed to be University), Kolhapur, MS 416006 India
| |
Collapse
|
14
|
Akanyibah FA, Zhu Y, Jin T, Ocansey DKW, Mao F, Qiu W. The Function of Necroptosis and Its Treatment Target in IBD. Mediators Inflamm 2024; 2024:7275309. [PMID: 39118979 PMCID: PMC11306684 DOI: 10.1155/2024/7275309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 06/22/2024] [Accepted: 07/13/2024] [Indexed: 08/10/2024] Open
Abstract
Inflammatory bowel disease (IBD), which encompasses Crohn's disease (CD) and ulcerative colitis (UC), is a complicated illness whose exact cause is yet unknown. Necroptosis is associated with IBD pathogenesis, leading to intestinal barrier abnormalities and uncontrolled inflammation. Molecules involved in necroptosis, however, exhibit different expression levels in IBD and its associated colorectal cancer. Multiple studies have shown that inhibiting these molecules alleviates necroptosis-induced IBD. Moreover, due to the severe scarcity of clinical medications for treating IBD caused by necroptosis, we review the various functions of crucial necroptosis molecules in IBD, the stimuli regulating necroptosis, and the current emerging therapeutic strategies for treating IBD-associated necroptosis. Eventually, understanding the pathogenesis of necroptosis in IBD will enable the development of additional therapeutic approaches for the illness.
Collapse
Affiliation(s)
- Francis Atim Akanyibah
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu ProvinceDepartment of Laboratory MedicineSchool of MedicineJiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Yi Zhu
- The People's Hospital of DanyangAffiliated Danyang Hospital of Nantong University, Zhenjiang 212300, Jiangsu, China
| | - Tao Jin
- Department of Gastrointestinal and EndoscopyThe Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu ProvinceDepartment of Laboratory MedicineSchool of MedicineJiangsu University, Zhenjiang 212013, Jiangsu, China
- Directorate of University Health ServicesUniversity of Cape Coast, Cape Coast CC0959347, Ghana
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu ProvinceDepartment of Laboratory MedicineSchool of MedicineJiangsu University, Zhenjiang 212013, Jiangsu, China
| | - Wei Qiu
- Nanjing Jiangning Hospital, Nanjing 211100, Jiangsu, China
| |
Collapse
|
15
|
Li N, Zhao L, Geng X, Liu J, Zhang X, Hu Y, Qi J, Chen H, Qiu J, Zhang X, Jin S. Stimulation by exosomes from hypoxia-preconditioned hair follicle mesenchymal stem cells facilitates mitophagy by inhibiting the PI3K/AKT/mTOR signaling pathway to alleviate ulcerative colitis. Theranostics 2024; 14:4278-4296. [PMID: 39113800 PMCID: PMC11303078 DOI: 10.7150/thno.96038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/24/2024] [Indexed: 08/10/2024] Open
Abstract
Background: Ulcerative colitis (UC) is an intestinal inflammatory disease that is strongly associated with mitochondrial damage and dysfunction as well as mitophagy and lacks of satisfactory treatments. Hair follicle mesenchymal stem cell (HF-MSC)-derived exosomes owe benefit effectiveness on inflammatory therapies. Hypoxia-preconditioned HF-MSCs exhibit enhanced proliferation and migration abilities, and their exosomes exert stronger effects than normal exosomes. However, the therapeutic function of Hy-Exos in UC is unknown. Methods: The inflammation model was established with LPS-treated MODE-K cells, and the mouse UC model was established by dextran sulfate sodium (DSS) administration. The therapeutic effects of HF-MSC-derived exosomes (Exos) and hypoxia-preconditioned HF-MSC-derived exosomes (Hy-Exos) were compared in vitro and in vivo. Immunofluorescence staining and western blotting were used to explore the effects of Hy-Exos on mitochondrial function, mitochondrial fission and fusion and mitophagy. MiRNA sequencing analysis was applied to investigate the differences in components between Exos and Hy-Exos. Results: Hy-Exos had a better therapeutic effect on LPS-treated MODE-K cells and DSS-induced UC mice. Hy-Exos promoted colonic tight junction proteins expression, suppressed the oxidative stress response, and reduced UC-related inflammatory injury. Hy-Exos may exert these effects via miR-214-3p-mediated inhibition of the PI3K/AKT/mTOR signaling pathway, maintenance of mitochondrial dynamic stability, alleviation of mitochondrial dysfunction and enhancement of mitophagy. Conclusion: This study revealed a vital role for Hy-Exos in suppressing inflammatory progression in UC and suggested that miR-214-3p is a potential critical target for Hy-Exos in alleviating UC.
Collapse
Affiliation(s)
- Ning Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Lei Zhao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xinyu Geng
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Jingyang Liu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xu Zhang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Ying Hu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Jihan Qi
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Hongliang Chen
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Jiawei Qiu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Xiaoyu Zhang
- Department of Infectious Diseases, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| | - Shizhu Jin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital of Harbin Medical University, Harbin 150086, China
| |
Collapse
|
16
|
Wei Z, Tang X, Yi C, Ocansey DKW, Mao F, Mao Z. HucMSC-Ex alleviates DSS-induced colitis in mice by decreasing mast cell activation via the IL-33/ST2 axis. Am J Transl Res 2024; 16:2727-2744. [PMID: 39006299 PMCID: PMC11236658 DOI: 10.62347/exze5413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic inflammatory disease that poses challenges in terms of treatment. The precise mechanism underlying the role of human umbilical cord mesenchymal stem cell-derived exosome (HucMSC-Ex) in the inflammatory repair process of IBD remains elusive. Mucosal mast cells accumulate within the intestinal tract and exert regulatory functions in IBD, thus presenting a novel target for addressing this intestinal disease. METHODS A mouse model of Dextran Sulfate Sodium (DSS)-induced colitis was established and hucMSC-Ex were administered to investigate their impact on the regulation of intestinal mast cells. An in vitro co-culture model using the human clonal colorectal adenocarcinoma cell line (Caco-2) and human mast cell line (LAD2) was also established for further exploration of the effect of hucMSC-Ex. RESULTS We observed the accumulation of mast cells in the intestines of patients with IBD as well as mice. In colitis mice, there was an upregulation of mast cell-related tryptase, interleukin-33 (IL-33), and suppression of tumorigenicity 2 receptor (ST2 or IL1RL1), and the function of the intestinal mucosal barrier related to intestinal tight junction protein was weakened. HucMSC-Ex treatment significantly reduced mast cell infiltration and intestinal damage. In the co-culture model, a substantial number of mast cells interact with the epithelial barrier, triggering activation of the IL-33/IL1RL1 (ST2) pathway and subsequent release of inflammatory factors and trypsin. This disruption leads to aberrant expression of tight junction proteins, which can be alleviated by supplementation with hucMSC-Ex. CONCLUSION Our results suggest that hucMSC-Ex may reduce the release of mast cell mediators via the IL-33/IL1RL1 (ST2) axis, thereby mitigating its detrimental effects on intestinal barrier function.
Collapse
Affiliation(s)
- Zhiping Wei
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu UniversityZhenjiang 212002, Jiangsu, P. R. China
- Department of Clinical Laboratory, The Third People’s Hospital of Xindu DistrictChengdu 610500, Sichuan, P. R. China
| | - Xiaohua Tang
- Department of Orthopaedics, The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong UniversityZhenjiang 212300, Jiangsu, P. R. China
| | - Chengxue Yi
- School of Medical Technology, Zhenjiang CollegeZhenjiang 212028, Jiangsu, P. R. China
| | - Dickson Kofi Wiredu Ocansey
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu UniversityZhenjiang 212002, Jiangsu, P. R. China
- Department of Medical Laboratory Science, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape CoastCape Coast CC0959347, Ghana
| | - Fei Mao
- Department of Laboratory Medicine, The Affiliated People’s Hospital, Jiangsu UniversityZhenjiang 212002, Jiangsu, P. R. China
| | - Zhenwei Mao
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People’s Hospital of Jiangsu UniversityZhenjiang 212002, Jiangsu, P. R. China
| |
Collapse
|
17
|
Li X, Hu Y, Wu Y, Yang Z, Liu Y, Liu H. Exosomal let-7a-5p derived from human umbilical cord mesenchymal stem cells alleviates coxsackievirus B3-induced cardiomyocyte ferroptosis via the SMAD2/ZFP36 signal axis. J Zhejiang Univ Sci B 2024; 25:422-437. [PMID: 38725341 PMCID: PMC11087186 DOI: 10.1631/jzus.b2300077] [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: 02/07/2023] [Accepted: 06/29/2023] [Indexed: 05/13/2024]
Abstract
Viral myocarditis (VMC) is one of the most common acquired heart diseases in children and teenagers. However, its pathogenesis is still unclear, and effective treatments are lacking. This study aimed to investigate the regulatory pathway by which exosomes alleviate ferroptosis in cardiomyocytes (CMCs) induced by coxsackievirus B3 (CVB3). CVB3 was utilized for inducing the VMC mouse model and cellular model. Cardiac echocardiography, left ventricular ejection fraction (LVEF), and left ventricular fractional shortening (LVFS) were implemented to assess the cardiac function. In CVB3-induced VMC mice, cardiac insufficiency was observed, as well as the altered levels of ferroptosis-related indicators (glutathione peroxidase 4 (GPX4), glutathione (GSH), and malondialdehyde (MDA)). However, exosomes derived from human umbilical cord mesenchymal stem cells (hucMSCs-exo) could restore the changes caused by CVB3 stimulation. Let-7a-5p was enriched in hucMSCs-exo, and the inhibitory effect of hucMSCs-exolet-7a-5p mimic on CVB3-induced ferroptosis was higher than that of hucMSCs-exomimic NC (NC: negative control). Mothers against decapentaplegic homolog 2 (SMAD2) increased in the VMC group, while the expression of zinc-finger protein 36 (ZFP36) decreased. Let-7a-5p was confirmed to interact with SMAD2 messenger RNA (mRNA), and the SMAD2 protein interacted directly with the ZFP36 protein. Silencing SMAD2 and overexpressing ZFP36 inhibited the expression of ferroptosis-related indicators. Meanwhile, the levels of GPX4, solute carrier family 7, member 11 (SLC7A11), and GSH were lower in the SMAD2 overexpression plasmid (oe-SMAD2)+let-7a-5p mimic group than in the oe-NC+let-7a-5p mimic group, while those of MDA, reactive oxygen species (ROS), and Fe2+ increased. In conclusion, these data showed that ferroptosis could be regulated by mediating SMAD2 expression. Exo-let-7a-5p derived from hucMSCs could mediate SMAD2 to promote the expression of ZFP36, which further inhibited the ferroptosis of CMCs to alleviate CVB3-induced VMC.
Collapse
Affiliation(s)
- Xin Li
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610041, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Yanan Hu
- Department of Pediatrics, the Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yueting Wu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610041, China
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu 610041, China
| | - Zuocheng Yang
- Department of Pediatrics, the Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yang Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China.
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610041, China.
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu 610041, China.
| | - Hanmin Liu
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu 610041, China. ,
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu 610041, China. ,
- NHC Key Laboratory of Chronobiology (Sichuan University), Chengdu 610041, China. ,
- The Joint Laboratory for Lung Development and Related Diseases of West China Second University Hospital, Sichuan University and School of Life Sciences of Fudan University, West China Institute of Women and Children's Health, West China Second University Hospital, Sichuan University, Chengdu 610041, China. ,
- Sichuan Birth Defects Clinical Research Center, West China Second University Hospital, Sichuan University, Chengdu 610041, China. ,
| |
Collapse
|
18
|
Zhang X, Che X, Zhang S, Wang R, Li M, Jin Y, Wang T, Song Y. Mesenchymal stem cell-derived extracellular vesicles for human diseases. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:64-82. [PMID: 39698413 PMCID: PMC11648454 DOI: 10.20517/evcna.2023.47] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/26/2024] [Accepted: 02/01/2024] [Indexed: 12/20/2024]
Abstract
Stem cell therapy is a novel approach for treating various severe and intractable diseases, including autoimmune disorders, organ transplants, tumors, and neurodegenerative diseases. Nevertheless, the extensive utilization of stem cells is constrained by potential tumorigenicity, challenges in precise differentiation, rejection concerns, and ethical considerations. Extracellular vesicles possess the ability to carry diverse bioactive factors from stem cells and deliver them to specific target cells or tissues. Moreover, they offer the advantage of low immunogenicity. Consequently, they have the potential to facilitate the therapeutic potential of stem cells, mitigating the risks associated with direct stem cell application. Therefore, the use of stem cell extracellular vesicles in clinical diseases has received increasing attention. This review summarizes advances in the use of extracellular vesicles from mesenchymal stem cells (MSC). MSC extracellular vesicles are used in the treatment of inflammatory diseases such as rheumatoid arthritis, liver injury, COVID-19, and allergies; in the repair of tissue damage in heart disease, kidney injury, and osteoarthritic diseases; as a carrier in the treatment of tumors; and as a regenerative agent in neurodegenerative disorders such as Alzheimer's and Parkinson's.
Collapse
Affiliation(s)
- Xiaofang Zhang
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Cancer Hospital of Dalian University of Technology, Faculty of Medicine, Dalian University of Technology, Shenyang 110042, Liaoning, China
- Authors contributed equally
| | - Xiaofang Che
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang 110001, Liaoning, China
- Authors contributed equally
| | - Sibo Zhang
- The Fourth Hospital of China Medical University, Shenyang 110032, Liaoning, China
- Authors contributed equally
| | - Runze Wang
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Cancer Hospital of Dalian University of Technology, Faculty of Medicine, Dalian University of Technology, Shenyang 110042, Liaoning, China
| | - Mo Li
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Cancer Hospital of Dalian University of Technology, Faculty of Medicine, Dalian University of Technology, Shenyang 110042, Liaoning, China
| | - Yi Jin
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Cancer Hospital of Dalian University of Technology, Faculty of Medicine, Dalian University of Technology, Shenyang 110042, Liaoning, China
| | - Tianlu Wang
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Cancer Hospital of Dalian University of Technology, Faculty of Medicine, Dalian University of Technology, Shenyang 110042, Liaoning, China
| | - Yingqiu Song
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Cancer Hospital of Dalian University of Technology, Faculty of Medicine, Dalian University of Technology, Shenyang 110042, Liaoning, China
| |
Collapse
|
19
|
Miron RJ, Estrin NE, Sculean A, Zhang Y. Understanding exosomes: Part 2-Emerging leaders in regenerative medicine. Periodontol 2000 2024; 94:257-414. [PMID: 38591622 DOI: 10.1111/prd.12561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
Exosomes are the smallest subset of extracellular signaling vesicles secreted by most cells with the ability to communicate with other tissues and cell types over long distances. Their use in regenerative medicine has gained tremendous momentum recently due to their ability to be utilized as therapeutic options for a wide array of diseases/conditions. Over 5000 publications are currently being published yearly on this topic, and this number is only expected to dramatically increase as novel therapeutic strategies continue to be developed. Today exosomes have been applied in numerous contexts including neurodegenerative disorders (Alzheimer's disease, central nervous system, depression, multiple sclerosis, Parkinson's disease, post-traumatic stress disorders, traumatic brain injury, peripheral nerve injury), damaged organs (heart, kidney, liver, stroke, myocardial infarctions, myocardial infarctions, ovaries), degenerative processes (atherosclerosis, diabetes, hematology disorders, musculoskeletal degeneration, osteoradionecrosis, respiratory disease), infectious diseases (COVID-19, hepatitis), regenerative procedures (antiaging, bone regeneration, cartilage/joint regeneration, osteoarthritis, cutaneous wounds, dental regeneration, dermatology/skin regeneration, erectile dysfunction, hair regrowth, intervertebral disc repair, spinal cord injury, vascular regeneration), and cancer therapy (breast, colorectal, gastric cancer and osteosarcomas), immune function (allergy, autoimmune disorders, immune regulation, inflammatory diseases, lupus, rheumatoid arthritis). This scoping review is a first of its kind aimed at summarizing the extensive regenerative potential of exosomes over a broad range of diseases and disorders.
Collapse
Affiliation(s)
- Richard J Miron
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Nathan E Estrin
- Advanced PRF Education, Venice, Florida, USA
- School of Dental Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Anton Sculean
- Department of Periodontology, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- Department of Oral Implantology, University of Wuhan, Wuhan, China
| |
Collapse
|
20
|
Kim SH, Keum B, Kwak S, Byun J, Shin JM, Kim TH. Therapeutic Applications of Extracellular Vesicles in Inflammatory Bowel Disease. Int J Mol Sci 2024; 25:745. [PMID: 38255819 PMCID: PMC10815267 DOI: 10.3390/ijms25020745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/28/2023] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
The treatment landscape for inflammatory bowel disease (IBD) has undergone substantial advancements with the introduction of biologics. However, a considerable number of patients either show an immediate lack of response or lose responsiveness over time, necessitating the development of innovative and effective treatment approaches. Extracellular vesicles (EVs) are small lipid bilayer-enclosed structures that facilitate cell-to-cell molecular transfer and are integral to the pathogenesis of IBD. They play pivotal roles in maintaining the integrity of the intestinal epithelial barrier and the expulsion of cellular metabolites. The potential use of EVs as drug carriers or therapeutic agents has opened up a plethora of clinical applications. This review investigates the creation and content of EVs, their role in IBD development, and advances in their isolation and analytical techniques. Furthermore, the therapeutic promise they hold for IBD is explored, along with the latest research on their roles as IBD drug delivery systems.
Collapse
Affiliation(s)
- Sang Hyun Kim
- Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (S.H.K.)
| | - Bora Keum
- Department of Internal Medicine, Korea University College of Medicine, Seoul 02841, Republic of Korea; (S.H.K.)
| | - Sooun Kwak
- Department of Otorhinolaryngology—Head & Neck Surgery, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Junhyoung Byun
- Department of Otorhinolaryngology—Head & Neck Surgery, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Mucosal Immunology Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jae Min Shin
- Department of Otorhinolaryngology—Head & Neck Surgery, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Mucosal Immunology Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Tae Hoon Kim
- Department of Otorhinolaryngology—Head & Neck Surgery, Korea University College of Medicine, Seoul 02841, Republic of Korea
- Mucosal Immunology Institute, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
21
|
Ma F, Zhang S, Akanyibah FA, Zhang W, Chen K, Ocansey DKW, Lyu C, Mao F. Exosome-mediated macrophage regulation for inflammatory bowel disease repair: a potential target of gut inflammation. Am J Transl Res 2023; 15:6970-6987. [PMID: 38186999 PMCID: PMC10767518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/14/2023] [Indexed: 01/09/2024]
Abstract
Inflammatory bowel disease (IBD), which includes Crohn's disease (CD) and ulcerative colitis (UC), is a complex condition without a definite cause. During IBD, immune cells such as macrophages release proinflammatory cytokines and chemokines, contributing to intestinal barrier integrity dysfunction. IBD is largely influenced by macrophages, which are classified into subtypes M1 and M2. M1 macrophages have been found to contribute to the development of IBD, whereas M2 macrophages alleviate IBD. Hence, agents that cause increased polarization of the M2 phenotype could help repair IBD. Exosomes, as ubiquitous conveyors of intercellular messages, are involved in immune responses and immune-mediated disease processes. Exosomes and their microRNA (miRNA) from healthy cells have been found to polarize macrophages to M2 to repair IBD due to their anti-inflammatory properties; however, those from inflammatory-driven cells and disease cells promote M1 macrophages to perpetuate IBD. Here, we review the biogenesis, biochemical composition, and sources of exosomes, as well as the roles of exosomes as extracellular vesicles in regulation of macrophages to repair IBD.
Collapse
Affiliation(s)
- Feifei Ma
- School of Medical Technology, Shangqiu Medical CollegeShangqiu 476100, Henan, P. R. China
| | - Shiheng Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, P. R. China
| | - Francis Atim Akanyibah
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, P. R. China
| | - Weibin Zhang
- School of Medical Technology, Shangqiu Medical CollegeShangqiu 476100, Henan, P. R. China
| | - Kangjing Chen
- School of Medical Technology, Shangqiu Medical CollegeShangqiu 476100, Henan, P. R. China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, P. R. China
- Directorate of University Health Services, University of Cape CoastCape Coast CC0959347, Ghana
| | - Changkun Lyu
- School of Medical Technology, Shangqiu Medical CollegeShangqiu 476100, Henan, P. R. China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, P. R. China
| |
Collapse
|
22
|
Liu W, Liu A, Li X, Sun Z, Sun Z, Liu Y, Wang G, Huang D, Xiong H, Yu S, Zhang X, Fan C. Dual-engineered cartilage-targeting extracellular vesicles derived from mesenchymal stem cells enhance osteoarthritis treatment via miR-223/NLRP3/pyroptosis axis: Toward a precision therapy. Bioact Mater 2023; 30:169-183. [PMID: 37593145 PMCID: PMC10429745 DOI: 10.1016/j.bioactmat.2023.06.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 05/24/2023] [Accepted: 06/19/2023] [Indexed: 08/19/2023] Open
Abstract
Osteoarthritis (OA) is the most common disabling joint disease with no effective disease modifying drugs. Extracellular vesicles released by several types of mesenchymal stem cells could promote cartilage repair and ameliorate OA pathology in animal models, representing a novel therapeutic strategy. In this study, we demonstrated that extracellular vesicles derived from human umbilical cord mesenchymal stem cells (hUC-EVs) could maintain chondrocyte homeostasis and alleviate OA, and further revealed a novel molecular mechanism of this therapeutic effect. miR-223, which could directly bind with the 3'UTR of NLRP3 mRNA, was found to be a key miRNA for hUC-EVs to exert beneficial effects on inflammation inhibiting and cartilage protecting. For enhancing the effect on mitigating osteoarthritis, exogenous miR-223 was loaded into hUC-EVs by electroporation, and a collagen II-targeting peptide (WYRGRL) was modified onto the surface of hUC-EVs by genetic engineering to achieve a more targeted and efficient RNA delivery to the cartilage. The dual-engineered EVs showed a maximal effect on inhibiting the NLRP3 inflammasome activation and chondrocyte pyroptosis, and offered excellent results for the treatment of OA. This study provides a novel theoretical basis and a promising therapeutic strategy for the application of engineered extracellular vesicles in OA treatment.
Collapse
Affiliation(s)
- Weixuan Liu
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
- Shanghai Engineering Research Center for Orthopedic Material Innovation and Tissue Regeneration, Shanghai, 201306, China
| | - Anqi Liu
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, 200001, China
| | - Xujun Li
- Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Ziyang Sun
- Shanghai Engineering Research Center for Orthopedic Material Innovation and Tissue Regeneration, Shanghai, 201306, China
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zhenghua Sun
- Shanghai Engineering Research Center for Orthopedic Material Innovation and Tissue Regeneration, Shanghai, 201306, China
| | - Yaru Liu
- Shanghai Engineering Research Center for Orthopedic Material Innovation and Tissue Regeneration, Shanghai, 201306, China
| | - Gang Wang
- Shanghai Engineering Research Center for Orthopedic Material Innovation and Tissue Regeneration, Shanghai, 201306, China
| | - Dan Huang
- Shanghai Engineering Research Center for Orthopedic Material Innovation and Tissue Regeneration, Shanghai, 201306, China
| | - Hao Xiong
- Shanghai Engineering Research Center for Orthopedic Material Innovation and Tissue Regeneration, Shanghai, 201306, China
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Shiyang Yu
- Shanghai Engineering Research Center for Orthopedic Material Innovation and Tissue Regeneration, Shanghai, 201306, China
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Xintao Zhang
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Cunyi Fan
- Shanghai Engineering Research Center for Orthopedic Material Innovation and Tissue Regeneration, Shanghai, 201306, China
- Department of Orthopedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
23
|
Didamoony MA, Soubh AA, Atwa AM, Ahmed LA. Innovative preconditioning strategies for improving the therapeutic efficacy of extracellular vesicles derived from mesenchymal stem cells in gastrointestinal diseases. Inflammopharmacology 2023; 31:2973-2993. [PMID: 37874430 PMCID: PMC10692273 DOI: 10.1007/s10787-023-01350-6] [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: 08/28/2023] [Accepted: 09/20/2023] [Indexed: 10/25/2023]
Abstract
Gastrointestinal (GI) diseases have become a global health issue and an economic burden due to their wide distribution, late prognosis, and the inefficacy of recent available medications. Therefore, it is crucial to search for new strategies for their management. In the recent decades, mesenchymal stem cells (MSCs) therapy has attracted attention as a viable option for treating a myriad of GI disorders such as hepatic fibrosis (HF), ulcerative colitis (UC), acute liver injury (ALI), and non-alcoholic fatty liver disease (NAFLD) due to their regenerative and paracrine properties. Importantly, recent studies have shown that MSC-derived extracellular vesicles (MSC-EVs) are responsible for most of the therapeutic effects of MSCs. In addition, EVs have revealed several benefits over their parent MSCs, such as being less immunogenic, having a lower risk of tumour formation, being able to cross biological barriers, and being easier to store. MSC-EVs exhibited regenerative, anti-oxidant, anti-inflammatory, anti-apoptotic, and anti-fibrotic effects in different experimental models of GI diseases. However, a key issue with their clinical application is the maintenance of their stability and efficacy following in vivo transplantation. Preconditioning of MSC-EVs or their parent cells is one of the novel methods used to improve their effectiveness and stability. Herein, we discuss the application of MSC-EVs in several GI disorders taking into account their mechanism of action. We also summarise the challenges and restrictions that need to be overcome to promote their clinical application in the treatment of various GI diseases as well as the recent developments to improve their effectiveness. A representation of the innovative preconditioning techniques that have been suggested for improving the therapeutic efficacy of MSC-EVs in GI diseases. The pathological conditions in various GI disorders (ALI, UC, HF and NAFLD) create a harsh environment for EVs and their parents, increasing the risk of apoptosis and senescence of MSCs and thereby diminishing MSC-EVs yield and restricting their large-scale applications. Preconditioning with pharmacological agents or biological mediators can improve the therapeutic efficacy of MSC-EVs through their adaption to the lethal environment to which they are subjected. This can result in establishment of a more conducive environment and activation of numerous vital trajectories that act to improve the immunomodulatory, reparative and regenerative activities of the derived EVs, as a part of MSCs paracrine system. ALI, acute liver injury; GI diseases, gastrointestinal diseases; HF, hepatic fibrosis; HSP, heat shock protein; miRNA, microRNA; mRNA, messenger RNA; MSC-EVs, mesenchymal stem cell-derived extracellular vesicles; NAFLD, non-alcoholic fatty liver disease; UC, ulcerative colitis.
Collapse
Affiliation(s)
- Manar A Didamoony
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Egyptian Russian University, Cairo, 11829, Egypt.
| | - Ayman A Soubh
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Ahram Canadian University, 4th Industrial Zone, Banks Complex, 6th of October City, Giza, 12451, Egypt
| | - Ahmed M Atwa
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Egyptian Russian University, Cairo, 11829, Egypt
| | - Lamiaa A Ahmed
- Faculty of Pharmacy, Pharmacology and Toxicology Department, Cairo University, Cairo, 11562, Egypt.
| |
Collapse
|
24
|
Hou JJ, Li WW, Wang XL, Ma AH, Qin YH. Efficacy of extracellular vesicles as a cell-free therapy in colitis: a systematic review and meta-analysis of animal studies. Front Pharmacol 2023; 14:1260134. [PMID: 37954844 PMCID: PMC10637393 DOI: 10.3389/fphar.2023.1260134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/09/2023] [Indexed: 11/14/2023] Open
Abstract
Background: Extracellular vesicles (EVs) mediate inflammation, immune responses, gut barrier integrity, and intestinal homeostasis. Recently, the application of EVs in the treatment of inflammatory bowel disease (IBD) has been under intensive focus. Some studies have been conducted in animal models of colitis, while systematic reviews and meta-analyses are lacking. The current study aimed to conduct a systematic review and meta-analysis of studies investigating the efficacy of EVs on IBD. Methods: A systematic retrieval of all studies in PubMed, EMBASE, MEDLINE, Web of Science, and Cochrane Library reported the effects of EVs in the colitis model up to 22 June 2023. The methodological quality was assessed based on SYRCLE's risk of bias (RoB) tool. Disease activity index (DAI), myeloperoxidase activity (MPO), histopathological score (HS), and inflammatory cytokines (TNF-α, NF-κB, IL-1β, IL-6, and IL-10) were extracted as analysis indicators by Web Plot Digitizer 4.5. A meta-analysis was performed to calculate the standardized mean difference and 95% confidence interval using random-effect models by Review Manager 5.3 and STATA 14.0 software. Results: A total of 21 studies were included in this meta-analysis. Although the heterogeneity between studies and the potential publication bias limits confidence in the extent of the benefit, EV treatment was superior to the control in the colitis evaluation with reduced DAI, HS, MPO activity, and pro-inflammatory cytokines, including TNF-α, NF-κB, IL-1β, and IL-6, while increasing the content of anti-inflammatory cytokine IL-10 (all p < 0.05). Conclusion: Our meta-analysis results supported the protective effect of EVs on colitis rodent models based on their potential role in IBD therapy and propelling the field toward clinical studies.
Collapse
Affiliation(s)
| | | | | | | | - Yue-Hua Qin
- Department of Gastroenterology, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
25
|
Zheng X, Zhu Y, Zhao Z, Chu Y, Yang W. The role of amino acid metabolism in inflammatory bowel disease and other inflammatory diseases. Front Immunol 2023; 14:1284133. [PMID: 37936710 PMCID: PMC10626463 DOI: 10.3389/fimmu.2023.1284133] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/10/2023] [Indexed: 11/10/2023] Open
Abstract
Inflammation is a characteristic symptom of the occurrence and development of many diseases, which is mainly characterized by the infiltration of inflammatory cells such as macrophages and granulocytes, and the increased release of proinflammatory factors. Subsequently, macrophage differentiates and T cells and other regulated factors exhibit anti-inflammatory function, releasing pro- and anti-inflammatory factors to maintain homeostasis. Although reports define various degrees of metabolic disorders in both the inflamed and non-inflamed parts of inflammatory diseases, little is known about the changes in amino acid metabolism in such conditions. This review aims to summarize amino acid changes and mechanisms involved in the progression of inflammatory bowel disease (IBD) and other inflammatory diseases. Since mesenchymal stem cells (MSCs) and their derived exosomes (MSC-EXO) have been found to show promising effects in the treatment of IBD and other inflammatory diseases,their potential in the modulation of amino acid metabolism in the treatment of inflammation is also discussed.
Collapse
Affiliation(s)
- Xiaowen Zheng
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yi Zhu
- The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu, China
| | - Zihan Zhao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ying Chu
- Changzhou Key Laboratory of Molecular Diagnostics and Precision Cancer Medicine, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
| | - Wenjing Yang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
26
|
Ntampakis G, Pramateftakis MG, Ioannidis O, Bitsianis S, Christidis P, Symeonidis S, Koliakos G, Karakota M, Bekiari C, Tsakona A, Cheva A, Aggelopoulos S. The Role of Adipose Tissue Mesenchymal Stem Cells in Colonic Anastomosis Healing in Inflammatory Bowel Disease: Experimental Study in Rats. J Clin Med 2023; 12:6336. [PMID: 37834980 PMCID: PMC10573964 DOI: 10.3390/jcm12196336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 10/15/2023] Open
Abstract
(1) Background: A surgical operation on an inflamed bowel is, diachronically, a challenge for the surgeon, especially for patients with inflammatory bowel disease. Adipose tissue-derived mesenchymal stromal cells are already in use in clinical settings for their anti-inflammatory properties. The rationale of the current study was to use AdMSCs in high-risk anastomoses to monitor if they attenuate inflammation and prevent anastomotic leak. (2) Methods: a total of 4 groups of rats were subjected to a surgical transection of the large intestine and primary anastomosis. In two groups, DSS 5% was administered for 7 days prior to the procedure, to induce acute intestinal inflammation. After the anastomosis, 5 × 106 autologous AdMSCs or an acellular solution was injected locally. Macroscopic evaluation, bursting pressure, hydroxyproline, and inflammatory cytokine expression were the parameters measured on the 8th post-operative day. (3) Results: Significantly less intra-abdominal complications, higher bursting pressures, and a decrease in pro-inflammatory markers were found in the groups that received AdMSCs. No difference in VEGF expression was observed on the 8th post-operative day. (4) Conclusions: AdMSCs attenuate inflammation in cases of acutely inflamed anastomosis.
Collapse
Affiliation(s)
- Georgios Ntampakis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.N.)
| | | | - Orestis Ioannidis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.N.)
| | - Stefanos Bitsianis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.N.)
| | - Panagiotis Christidis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.N.)
| | - Savvas Symeonidis
- 4th Department of General Surgery, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.N.)
| | - Georgios Koliakos
- Laboratory of Biochemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Maria Karakota
- Laboratory of Biochemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Chrysanthi Bekiari
- Experimental and Research Center, Papageorgiou General Hospital of Thessaloniki, 56403 Thessaloniki, Greece
- Laboratory of Anatomy and Histology, Veterinary School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Anastasia Tsakona
- Pathology Department, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Angeliki Cheva
- Pathology Department, Faculty of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Stamatios Aggelopoulos
- 4th Department of General Surgery, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (G.N.)
| |
Collapse
|
27
|
Liu R, Wu S, Liu W, Wang L, Dong M, Niu W. microRNAs delivered by small extracellular vesicles in MSCs as an emerging tool for bone regeneration. Front Bioeng Biotechnol 2023; 11:1249860. [PMID: 37720323 PMCID: PMC10501734 DOI: 10.3389/fbioe.2023.1249860] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Bone regeneration is a dynamic process that involves angiogenesis and the balance of osteogenesis and osteoclastogenesis. In bone tissue engineering, the transplantation of mesenchymal stem cells (MSCs) is a promising approach to restore bone homeostasis. MSCs, particularly their small extracellular vesicles (sEVs), exert therapeutic effects due to their paracrine capability. Increasing evidence indicates that microRNAs (miRNAs) delivered by sEVs from MSCs (MSCs-sEVs) can alter gene expression in recipient cells and enhance bone regeneration. As an ideal delivery vehicle of miRNAs, MSCs-sEVs combine the high bioavailability and stability of sEVs with osteogenic ability of miRNAs, which can effectively overcome the challenge of low delivery efficiency in miRNA therapy. In this review, we focus on the recent advancements in the use of miRNAs delivered by MSCs-sEVs for bone regeneration and disorders. Additionally, we summarize the changes in miRNA expression in osteogenic-related MSCs-sEVs under different microenvironments.
Collapse
Affiliation(s)
| | | | | | | | - Ming Dong
- School of Stomatology, Dalian Medical University, Dalian, China
| | - Weidong Niu
- School of Stomatology, Dalian Medical University, Dalian, China
| |
Collapse
|
28
|
Zhang J, Li A, Gu R, Tong Y, Cheng J. Role and regulatory mechanism of microRNA mediated neuroinflammation in neuronal system diseases. Front Immunol 2023; 14:1238930. [PMID: 37637999 PMCID: PMC10457161 DOI: 10.3389/fimmu.2023.1238930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs with the unique ability to degrade or block specific RNAs and regulate many cellular processes. Neuroinflammation plays the pivotal role in the occurrence and development of multiple central nervous system (CNS) diseases. The ability of miRNAs to enhance or restrict neuroinflammatory signaling pathways in CNS diseases is an emerging and important research area, including neurodegenerative diseases, stroke, and traumatic brain injury (TBI). In this review, we summarize the roles and regulatory mechanisms of recently identified miRNAs involved in neuroinflammation-mediated CNS diseases, aiming to explore and provide a better understanding and direction for the treatment of CNS diseases.
Collapse
Affiliation(s)
| | | | | | | | - Jinbo Cheng
- Center on Translational Neuroscience, College of Life and Environmental Science, Minzu University of China, Beijing, China
| |
Collapse
|
29
|
Greenberg ZF, Graim KS, He M. Towards artificial intelligence-enabled extracellular vesicle precision drug delivery. Adv Drug Deliv Rev 2023:114974. [PMID: 37356623 DOI: 10.1016/j.addr.2023.114974] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 06/27/2023]
Abstract
Extracellular Vesicles (EVs), particularly exosomes, recently exploded into nanomedicine as an emerging drug delivery approach due to their superior biocompatibility, circulating stability, and bioavailability in vivo. However, EV heterogeneity makes molecular targeting precision a critical challenge. Deciphering key molecular drivers for controlling EV tissue targeting specificity is in great need. Artificial intelligence (AI) brings powerful prediction ability for guiding the rational design of engineered EVs in precision control for drug delivery. This review focuses on cutting-edge nano-delivery via integrating large-scale EV data with AI to develop AI-directed EV therapies and illuminate the clinical translation potential. We briefly review the current status of EVs in drug delivery, including the current frontier, limitations, and considerations to advance the field. Subsequently, we detail the future of AI in drug delivery and its impact on precision EV delivery. Our review discusses the current universal challenge of standardization and critical considerations when using AI combined with EVs for precision drug delivery. Finally, we will conclude this review with a perspective on future clinical translation led by a combined effort of AI and EV research.
Collapse
Affiliation(s)
- Zachary F Greenberg
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, 32610, USA
| | - Kiley S Graim
- Department of Computer & Information Science & Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, Florida, 32610, USA
| | - Mei He
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, 32610, USA.
| |
Collapse
|
30
|
Wei Z, Hang S, Wiredu Ocansey DK, Zhang Z, Wang B, Zhang X, Mao F. Human umbilical cord mesenchymal stem cells derived exosome shuttling mir-129-5p attenuates inflammatory bowel disease by inhibiting ferroptosis. J Nanobiotechnology 2023; 21:188. [PMID: 37303049 DOI: 10.1186/s12951-023-01951-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 06/02/2023] [Indexed: 06/13/2023] Open
Abstract
BACKGROUND Ferroptosis, a unique form of non-apoptotic cell death, is dependent on iron and lipoperoxidation, and has been shown to be associated with the pathogenesis of inflammatory bowel disease (IBD). Human umbilical cord mesenchymal stem cell-derived exosomes (hucMSC-Ex) are involved in cell survival, immune conditioning, and damage repair. However, the relationship between hucMSC-Ex, IBD, and ferroptosis is unknown. This paper explores the role of hucMSC-Ex in the repair of IBD through the regulation of the ferroptosis signaling pathway. RESULTS In this study, we used small RNA sequencing to find that miR-129-5p was highly expressed in hucMSC-Ex, and by predicting its targeting to ACSL4, we verified the effect of miR-129-5p on mice IBD in vitro and human colonic epithelial cells (HCoEpiC) in vivo. We found that miR-129-5p reduces ferroptosis in intestinal epithelial cells by targeting ACSL4 to repair IBD, which provides new strategies for the prevention and treatment of IBD. CONCLUSION In conclusion, our results demonstrate that hucMSC-Ex relieves IBD by targeting ACSL4 with miR-129-5p to inhibit lipid peroxidation (LPO) and ferroptosis, reducing intestinal inflammation and repairing damages. Mechanism of hucMSC-Ex inhibiting ferroptosis in intestinal epithelial cells. System Xc- mediates the transport of extracellular cystine into the cell, which gets reduced to cysteine to participate in GSH-mediated metabolism. GPX4 strongly inhibits ferroptosis by helping scavenge reactive oxygen species. The depletion of GSH correlates with decreased GPX4, and the imbalance of the antioxidant system leads to the formation of toxic phospholipid hydroperoxide, which promotes the occurrence of ferroptosis with the participation of irons. HucMSC-Ex has the ability to relieve GSH and GPX4 depletion and repair the intracellular antioxidant system. Ferric ions enter the cytosol through DMT1 and participate in lipid peroxidation. HucMSC-Ex can reduce the expression of DMT1 and alleviate this process. HucMSC-Ex-derived miR-129-5p targets ACSL4 and reduces the expression of ACSL4, an enzyme that mediates the conversion of PUFAs into phospholipids in intestinal epithelial cells, and is a positive regulator of lipid peroxidation. ABBREVIATIONS GSH, glutathione; GPX4, glutathione peroxidase 4; GSSG, oxidized glutathione; DMT1, divalent metal transporter 1; ACSL4, acyl-CoA synthetase long-chain family member 4; PUFAs, polyunsaturated fatty acids; ALOXs, lipoxygenases; CoA, coenzyme A; PL, phospholipid; PLOOH, hydroperoxides, LOH, phospholipid alcohols; LPO, lipid peroxidation.
Collapse
Affiliation(s)
- Zhiping Wei
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
| | - Sanhua Hang
- The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Zhenjiang, Jiangsu, 212300, P.R. China
| | - Dickson Kofi Wiredu Ocansey
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
| | - Zhaoyang Zhang
- Clinical Lab, Taicang Hospital of Traditional Chinese Medicine, Suzhou, Jiangsu, 215400, P.R. China
| | - Bo Wang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
| | - Xu Zhang
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, P.R. China.
| |
Collapse
|
31
|
Chen L, Ou Q, Kou X. Extracellular vesicles and their indispensable roles in pathogenesis and treatment of inflammatory bowel disease: A comprehensive review. Life Sci 2023; 327:121830. [PMID: 37286163 DOI: 10.1016/j.lfs.2023.121830] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
Inflammatory bowel disease (IBD) is a global disease with rising incidence worldwide, and its debilitating symptoms and dissatisfactory therapies have brought heavy burdens for patients. Extracellular vesicles (EVs), a heterogeneous population of lipid bilayer membranes containing abundant bioactive molecules, have been indicated to play important roles in the pathogenesis and treatment of many diseases. However, to our knowledge, comprehensive reviews summarizing the various roles of diverse source-derived EVs in the pathogenesis and treatment of IBD are still lacking. This review, not only summarizes the EV characteristics, but also focuses on the multiple roles of diverse EVs in IBD pathogenesis and their treatment potential. In addition, hoping to push forward the research frontiers, we point out several challenges that the researchers are faced, about EVs in current IBD research and future therapeutic applications. We also put forward our prospects on future exploration regarding EVs in IBD treatment, including developing IBD vaccines and paying more attention on apoptotic vesicles. This review is aimed to enrich the knowledge on the indispensable roles of EVs in IBD pathogenesis and treatment, providing ideas and reference for future therapeutic strategy for IBD treatment.
Collapse
Affiliation(s)
- Linling Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, 510055 Guangzhou, China
| | - Qianmin Ou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, 510055 Guangzhou, China
| | - Xiaoxing Kou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, 510055 Guangzhou, China; Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou, Guangdong 510055, China.
| |
Collapse
|
32
|
Thomaidou AC, Goulielmaki M, Tsintarakis A, Zoumpourlis P, Toya M, Christodoulou I, Zoumpourlis V. miRNA-Guided Regulation of Mesenchymal Stem Cells Derived from the Umbilical Cord: Paving the Way for Stem-Cell Based Regeneration and Therapy. Int J Mol Sci 2023; 24:ijms24119189. [PMID: 37298143 DOI: 10.3390/ijms24119189] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/19/2023] [Accepted: 05/21/2023] [Indexed: 06/12/2023] Open
Abstract
The human body is an abundant source of multipotent cells primed with unique properties that can be exploited in a multitude of applications and interventions. Mesenchymal stem cells (MSCs) represent a heterogenous population of undifferentiated cells programmed to self-renew and, depending on their origin, differentiate into distinct lineages. Alongside their proven ability to transmigrate toward inflammation sites, the secretion of various factors that participate in tissue regeneration and their immunoregulatory function render MSCs attractive candidates for use in the cytotherapy of a wide spectrum of diseases and conditions, as well as in different aspects of regenerative medicine. In particular, MSCs that can be found in fetal, perinatal, or neonatal tissues possess additional capabilities, including predominant proliferation potential, increased responsiveness to environmental stimuli, and hypoimmunogenicity. Since microRNA (miRNA)-guided gene regulation governs multiple cellular functions, miRNAs are increasingly being studied in the context of driving the differentiation process of MSCs. In the present review, we explore the mechanisms of miRNA-directed differentiation of MSCs, with a special focus on umbilical cord-derived mesenchymal stem cells (UCMSCs), and we identify the most relevant miRNAs and miRNA sets and signatures. Overall, we discuss the potent exploitations of miRNA-driven multi-lineage differentiation and regulation of UCMSCs in regenerative and therapeutic protocols against a range of diseases and/or injuries that will achieve a meaningful clinical impact through maximizing treatment success rates, while lacking severe adverse events.
Collapse
Affiliation(s)
- Arsinoe C Thomaidou
- Laboratory of Clinical Virology, Medical School, University of Crete, 71500 Heraklion, Greece
| | - Maria Goulielmaki
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Antonis Tsintarakis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
| | - Panagiotis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
| | - Marialena Toya
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
| | - Ioannis Christodoulou
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
| |
Collapse
|
33
|
Ulpiano C, da Silva CL, Monteiro GA. Bioengineered Mesenchymal-Stromal-Cell-Derived Extracellular Vesicles as an Improved Drug Delivery System: Methods and Applications. Biomedicines 2023; 11:biomedicines11041231. [PMID: 37189850 DOI: 10.3390/biomedicines11041231] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/30/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nano-sized lipid membranous structures that modulate cell-cell communication by transporting a variety of biologically active cellular components. The potential of EVs in delivering functional cargos to targeted cells, their capacity to cross biological barriers, as well as their high modification flexibility, make them promising drug delivery vehicles for cell-free therapies. Mesenchymal stromal cells (MSCs) are known for their great paracrine trophic activity, which is largely sustained by the secretion of EVs. MSC-derived EVs (MSC-EVs) retain important features of the parental cells and can be bioengineered to improve their therapeutic payload and target specificity, demonstrating increased therapeutic potential in numerous pre-clinical animal models, including in the treatment of cancer and several degenerative diseases. Here, we review the fundamentals of EV biology and the bioengineering strategies currently available to maximize the therapeutic value of EVs, focusing on their cargo and surface manipulation. Then, a comprehensive overview of the methods and applications of bioengineered MSC-EVs is presented, while discussing the technical hurdles yet to be addressed before their clinical translation as therapeutic agents.
Collapse
Affiliation(s)
- Cristiana Ulpiano
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gabriel A Monteiro
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
34
|
Liu A, Hefley B, Escandon P, Nicholas SE, Karamichos D. Salivary Exosomes in Health and Disease: Future Prospects in the Eye. Int J Mol Sci 2023; 24:ijms24076363. [PMID: 37047335 PMCID: PMC10094317 DOI: 10.3390/ijms24076363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/30/2023] Open
Abstract
Exosomes are a group of vesicles that package and transport DNA, RNA, proteins, and lipids to recipient cells. They can be derived from blood, saliva, urine, and/or other biological tissues. Their impact on several diseases, such as neurodegenerative, autoimmune, and ocular diseases, have been reported, but not fully unraveled. The exosomes that are derived from saliva are less studied, but offer significant advantages over exosomes from other sources, due to their accessibility and ease of collection. Thus, their role in the pathophysiology of diseases is largely unknown. In the context of ocular diseases, salivary exosomes have been under-utilized, thus creating an enormous gap in the literature. The current review discusses the state of exosomes research on systemic and ocular diseases and highlights the role and potential of salivary exosomes as future ocular therapeutic vehicles.
Collapse
Affiliation(s)
- Angela Liu
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Brenna Hefley
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Paulina Escandon
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Sarah E. Nicholas
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | - Dimitrios Karamichos
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3430 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- Department of Pharmaceutical Sciences, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
- Correspondence: ; Tel.: +1-817-735-2101
| |
Collapse
|
35
|
Zhang X, Liu Y, Zhang T, Tan Y, Dai X, Yang YG, Zhang X. Advances in the potential roles of Cullin-RING ligases in regulating autoimmune diseases. Front Immunol 2023; 14:1125224. [PMID: 37006236 PMCID: PMC10064048 DOI: 10.3389/fimmu.2023.1125224] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/28/2023] [Indexed: 03/19/2023] Open
Abstract
Cullin-RING ligases (CRLs) are the largest class of E3 ubiquitin ligases regulating the stability and subsequent activity of a large number of important proteins responsible for the development and progression of various diseases, including autoimmune diseases (AIDs). However, the detailed mechanisms of the pathogenesis of AIDs are complicated and involve multiple signaling pathways. An in-depth understanding of the underlying regulatory mechanisms of the initiation and progression of AIDs will aid in the development of effective therapeutic strategies. CRLs play critical roles in regulating AIDs, partially by affecting the key inflammation-associated pathways such as NF-κB, JAK/STAT, and TGF-β. In this review, we summarize and discuss the potential roles of CRLs in the inflammatory signaling pathways and pathogenesis of AIDs. Furthermore, advances in the development of novel therapeutic strategies for AIDs through targeting CRLs are also highlighted.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Yu’e Liu
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Tong Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Yuying Tan
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
| | - Xiangpeng Dai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
- *Correspondence: Xiangpeng Dai, ; Yong-Guang Yang, ; Xiaoling Zhang,
| | - Yong-Guang Yang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
- International Center of Future Science, Jilin University, Changchun, China
- *Correspondence: Xiangpeng Dai, ; Yong-Guang Yang, ; Xiaoling Zhang,
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital, Jilin University, Changchun, China
- *Correspondence: Xiangpeng Dai, ; Yong-Guang Yang, ; Xiaoling Zhang,
| |
Collapse
|
36
|
Innocenti T, Bigagli E, Lynch EN, Galli A, Dragoni G. MiRNA-Based Therapies for the Treatment of Inflammatory Bowel Disease: What Are We Still Missing? Inflamm Bowel Dis 2023; 29:308-323. [PMID: 35749310 DOI: 10.1093/ibd/izac122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Indexed: 02/05/2023]
Abstract
Micro-RNAs (miRNAs) are noncoding RNAs usually 24-30 nucleotides long that play a central role in epigenetic mechanisms of inflammatory diseases and cancers. Recently, several studies have assessed the involvement of miRNAs in the pathogenesis of inflammatory bowel disease (IBD) and colitis-associated neoplasia. Particularly, it has been shown that many members of miRNAs family are involved in the pathways of inflammation and fibrogenesis of IBD; therefore, their use as inflammatory and fibrosis biomarkers has been postulated. In light of these results, the role of miRNAs in IBD therapy has been proposed and is currently under investigation with many in vitro and in vivo studies, murine models, and a phase 2a trial. The accumulating data have pushed miRNA-based therapy closer to clinical practice, although many open questions remain. With this systematic review, we discuss the current knowledge about the therapeutic effects of miRNAs mimicking and inhibition, and we explore the new potential targets of miRNA family for the treatment of inflammation and fibrosis in IBD.
Collapse
Affiliation(s)
- Tommaso Innocenti
- IBD Referral Center, Gastroenterology Department, Careggi University Hospital, Florence, Italy.,Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Elisabetta Bigagli
- Section of Pharmacology and Toxicology, Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), University of Florence, Florence, Italy
| | - Erica Nicola Lynch
- IBD Referral Center, Gastroenterology Department, Careggi University Hospital, Florence, Italy.,Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Andrea Galli
- Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Gabriele Dragoni
- IBD Referral Center, Gastroenterology Department, Careggi University Hospital, Florence, Italy.,Gastroenterology Research Unit, Department of Experimental and Clinical Biochemical Sciences "Mario Serio", University of Florence, Florence, Italy
| |
Collapse
|
37
|
miRNA Molecules-Late Breaking Treatment for Inflammatory Bowel Diseases? Int J Mol Sci 2023; 24:ijms24032233. [PMID: 36768556 PMCID: PMC9916785 DOI: 10.3390/ijms24032233] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023] Open
Abstract
MicroRNAs (miRNAs) are a group of non-coding RNAs that play a critical role in regulating epigenetic mechanisms in inflammation-related diseases. Inflammatory bowel diseases (IBDs), which primarily include ulcerative colitis (UC) and Crohn's disease (CD), are characterized by chronic recurrent inflammation of intestinal tissues. Due to the multifactorial etiology of these diseases, the development of innovative treatment strategies that can effectively maintain remission and alleviate disease symptoms is a major challenge. In recent years, evidence for the regulatory role of miRNAs in the pathogenetic mechanisms of various diseases, including IBD, has been accumulating. In light of these findings, miRNAs represent potential innovative candidates for therapeutic application in IBD. In this review, we discuss recent findings on the role of miRNAs in regulating inflammatory responses, maintaining intestinal barrier integrity, and developing fibrosis in clinical and experimental IBD. The focus is on the existing literature, indicating potential therapeutic application of miRNAs in both preclinical experimental IBD models and translational data in the context of clinical IBD. To date, a large and diverse data set, which is growing rapidly, supports the potential use of miRNA-based therapies in clinical practice, although many questions remain unanswered.
Collapse
|
38
|
Guo J, Wang F, Hu Y, Luo Y, Wei Y, Xu K, Zhang H, Liu H, Bo L, Lv S, Sheng S, Zhuang X, Zhang T, Xu C, Chen X, Su J. Exosome-based bone-targeting drug delivery alleviates impaired osteoblastic bone formation and bone loss in inflammatory bowel diseases. Cell Rep Med 2023; 4:100881. [PMID: 36603578 PMCID: PMC9873828 DOI: 10.1016/j.xcrm.2022.100881] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/12/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023]
Abstract
Systematic bone loss is commonly complicated with inflammatory bowel diseases (IBDs) with unclear pathogenesis and uncertain treatment. In experimental colitis mouse models established by dextran sulfate sodium and IL-10 knockout induced with piroxicam, bone mass and quality are significantly decreased. Colitis mice demonstrate a lower bone formation rate and fewer osteoblasts in femur. Bone marrow mesenchymal stem/stromal cells (BMSCs) from colitis mice tend to differentiate into adipocytes rather than osteoblasts. Serum from patients with IBD promotes adipogenesis of human BMSCs. RNA sequencing reveals that colitis downregulates Wnt signaling in BMSCs. For treatment, exosomes with Golgi glycoprotein 1 inserted could carry Wnt agonist 1 and accumulate in bone via intravenous administration. They could alleviate bone loss, promote bone formation, and accelerate fracture healing in colitis mice. Collectively, BMSC commitment in inflammatory microenvironment contributes to lower bone quantity and quality and could be rescued by redirecting differentiation toward osteoblasts through bone-targeted drug delivery.
Collapse
Affiliation(s)
- Jiawei Guo
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Fuxiao Wang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Yan Hu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Ying Luo
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Hao Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Han Liu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
| | - Lumin Bo
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shunli Lv
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shihao Sheng
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xinchen Zhuang
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Tao Zhang
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Can Xu
- Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Xiao Chen
- Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Department of Orthopedics Trauma, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Organoid Research Center, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
39
|
Cano A, Muñoz-Morales Á, Sánchez-López E, Ettcheto M, Souto EB, Camins A, Boada M, Ruíz A. Exosomes-Based Nanomedicine for Neurodegenerative Diseases: Current Insights and Future Challenges. Pharmaceutics 2023; 15:298. [PMID: 36678926 PMCID: PMC9863585 DOI: 10.3390/pharmaceutics15010298] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/09/2023] [Accepted: 01/12/2023] [Indexed: 01/17/2023] Open
Abstract
Neurodegenerative diseases constitute a group of pathologies whose etiology remains unknown in many cases, and there are no treatments that stop the progression of such diseases. Moreover, the existence of the blood-brain barrier is an impediment to the penetration of exogenous molecules, including those found in many drugs. Exosomes are extracellular vesicles secreted by a wide variety of cells, and their primary functions include intercellular communication, immune responses, human reproduction, and synaptic plasticity. Due to their natural origin and molecular similarities with most cell types, exosomes have emerged as promising therapeutic tools for numerous diseases. Specifically, neurodegenerative diseases have shown to be a potential target for this nanomedicine strategy due to the difficult access to the brain and the strategy's pathophysiological complexity. In this regard, this review explores the most important biological-origin drug delivery systems, innovative isolation methods of exosomes, their physicochemical characterization, drug loading, cutting-edge functionalization strategies to target them within the brain, the latest research studies in neurodegenerative diseases, and the future challenges of exosomes as nanomedicine-based therapeutic tools.
Collapse
Affiliation(s)
- Amanda Cano
- Ace Alzheimer Center Barcelona—International University of Catalunya (UIC), 08028 Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), 08028 Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Álvaro Muñoz-Morales
- Ace Alzheimer Center Barcelona—International University of Catalunya (UIC), 08028 Barcelona, Spain
| | - Elena Sánchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), 08028 Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- Unit of Synthesis and Biomedical Applications of Peptides, IQAC-CSIC, 08034 Barcelona, Spain
| | - Miren Ettcheto
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain
| | - Eliana B. Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- REQUIMTE/UCIBIO, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Antonio Camins
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, 08028 Barcelona, Spain
| | - Mercè Boada
- Ace Alzheimer Center Barcelona—International University of Catalunya (UIC), 08028 Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| | - Agustín Ruíz
- Ace Alzheimer Center Barcelona—International University of Catalunya (UIC), 08028 Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), 28029 Madrid, Spain
| |
Collapse
|
40
|
TNF- α Enhances the Therapeutic Effects of MenSC-Derived Small Extracellular Vesicles on Inflammatory Bowel Disease through Macrophage Polarization by miR-24-3p. Stem Cells Int 2023; 2023:2988907. [PMID: 36895784 PMCID: PMC9991477 DOI: 10.1155/2023/2988907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/18/2022] [Accepted: 11/24/2022] [Indexed: 03/06/2023] Open
Abstract
Human menstrual blood-derived mesenchymal stem cells (MenSCs) and their secreted small extracellular vesicles (EVs) had been proven to relieve inflammation, tissue damage, and fibrosis in various organs. The microenvironment induced by inflammatory cytokines can promote mesenchymal stem cells (MSCs) to secrete more substances (including EVs) that could regulate inflammation. Inflammatory bowel disease (IBD) is a chronic idiopathic intestinal inflammation, the etiology and mechanism of which are unclear. At present, the existing therapeutic methods are ineffective for many patients and have obvious side effects. Hence, we explored the role of tumor necrosis factor α- (TNF-α-) pretreated MenSC-derived small EV (MenSCs-sEVTNF-α ) in a mouse model of dextran sulfate sodium- (DSS-) induced colitis, expecting to find better therapeutic alterations. In this research, the small EVs of MenSCs were obtained by ultracentrifugation. MicroRNAs of small EVs derived from MenSCs before and after TNF-α treatment were sequenced, and the differential microRNAs were analyzed by bioinformatics. The small EVs secreted by TNF-α-stimulating MenSCs were more effective in colonic mice than those secreted directly by MenSCs, as evidenced by the results of histopathology analysis of colonic tissue, immunohistochemistry for tight junction proteins, and enzyme-linked immunosorbent assay (ELISA) for cytokine expression profiles in vivo. The process of MenSCs-sEVTNF-α relieving colonic inflammation was accompanied by the polarization of M2 macrophages in the colon and miR-24-3p upregulation in small EVs. In vitro, both MenSC-derived sEV (MenSCs-sEV) and MenSCs-sEVTNF-α reduced the expression of proinflammatory cytokines, and MenSCs-sEVTNF-α can increase the portion of M2 macrophages. In conclusion, after TNF-α stimulation, the expression of miR-24-3p in small EVs derived from MenSCs was upregulated. MiR-24-3p was proved to target and downregulate interferon regulatory factor 1 (IRF1) expression in the murine colon and then promoted the polarization of M2 macrophages. The polarization of M2 macrophages in colonic tissues then reduced the damage caused by hyperinflammation.
Collapse
|
41
|
Ma X, Liu B, Fan L, Liu Y, Zhao Y, Ren T, Li Y, Li Y. Native and engineered exosomes for inflammatory disease. NANO RESEARCH 2022; 16:6991-7006. [PMID: 36591564 PMCID: PMC9793369 DOI: 10.1007/s12274-022-5275-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 05/24/2023]
Abstract
Exosomes are extracellular vesicles which carry specific molecular information from donor cells and act as an intercellular communication vehicle, which have emerged as a novel cell-free strategy for the treatment of many diseases including inflammatory disease. Recently, rising studies have developed exosome-based strategies for novel inflammation therapy due to their biocompatibility and bioactivity. Researchers not only use native exosomes as therapeutic agents for inflammation, but also strive to make up for the natural defects of exosomes through engineering methods to improve and update the property of exosomes for enhanced therapeutic effects. The engineered exosomes can improve cargo-loading efficiency, targeting ability, stability, etc., to achieve combined and diverse treatment strategies in inflammation diseases. Herein, a comprehensive overview of the recent advances in application studies of native and engineered exosomes as well as the engineered methods is provided. Meanwhile, potential application prospects, possible challenges, and the development of clinical researches of exosome treatment strategy are concluded from plentiful examples, which may be able to provide guidance and suggestions for the future research and application of exosomes.
Collapse
Affiliation(s)
- Xiaoyi Ma
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200092 China
| | - Bingbing Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200092 China
| | - Limin Fan
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200092 China
| | - Yiqiong Liu
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200092 China
| | - Yuge Zhao
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200092 China
| | - Tianbin Ren
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200092 China
| | - Yan Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200092 China
| | - Yongyong Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200092 China
| |
Collapse
|
42
|
Liu J, Xing F, Fu Q, He B, Jia Z, Du J, Li Y, Zhang X, Chen X. hUC-MSCs exosomal miR-451 alleviated acute lung injury by modulating macrophage M2 polarization via regulating MIF-PI3K-AKT signaling pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:2819-2831. [PMID: 35997581 DOI: 10.1002/tox.23639] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 06/15/2023]
Abstract
In the previous study, we have proved that exosomal miR-451 from human umbilical cord mesenchymal stem cells (hUC-MSCs) attenuated burn-induced acute lung injury (ALI). However, the mechanism of exosomal miR-451 in ALI remains unclear. Therefore, this study aimed to study the molecular mechanism of hUC-MSCs-derived exosomal miR-451 on ALI by regulating macrophage polarization. Exosomes were isolated and identified by transmission electron microscope (TEM) and nanoparticle tracking analysis (NTA). The expression of miR-451, macrophage migration inhibitory factor (MIF) and PI3K/AKT signaling pathway proteins were detected by qRT-PCR and western blot. Flow cytometry was used to detect the CD80 and CD206 positive cells. Severe burn rat model was established and HE was used to detect the inflammatory cell infiltration and inflammatory injury. Dual luciferase reporter system was used to detect the regulation of miR-451 to MIF. The contents of cytokines were detected by ELISA. The results showed that hUC-MSCs exosomes promoted macrophage M1 to M2 polarization. Furthermore, hUC-MSCs-derived exosomal miR-451 alleviated ALI development and promoted macrophage M1 to M2 polarization. Moreover, MIF was a direct target of miR-451. Downregulation of MIF regulated by miR-451 alleviated ALI development promoted macrophage M1 to M2 polarization. In addition, we found that MIF and hUC-MSCs-derived exosomal miR-451 participated in ALI by regulating PI3K/AKT signaling pathway. In conclusion, we indicated that hUC-MSCs-derived exosomal miR-451 alleviated ALI by modulating macrophage M2 polarization via regulating MIF-PI3K-AKT signaling pathway, which provided great scientific significance and clinical application value for the treatment of burn-induced ALI.
Collapse
Affiliation(s)
- Jisong Liu
- Department of Burn and Plastic Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Fuxi Xing
- Department of Burn and Plastic Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Quanyou Fu
- Department of Burn and Plastic Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Bo He
- Department of Burn and Plastic Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Zhigang Jia
- Department of Burn and Plastic Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Juan Du
- Department of Minimally Invasive Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Yong Li
- Department of Burn and Plastic Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Xiangzhou Zhang
- Department of Burn and Plastic Surgery, Third Hospital of Bengbu, Bengbu, Anhui, China
| | - Xulin Chen
- Department of Burns, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
43
|
Liu C, Helsper S, Marzano M, Chen X, Muok L, Esmonde C, Zeng C, Sun L, Grant SC, Li Y. Human Forebrain Organoid-Derived Extracellular Vesicle Labeling with Iron Oxides for In Vitro Magnetic Resonance Imaging. Biomedicines 2022; 10:3060. [PMID: 36551816 PMCID: PMC9775717 DOI: 10.3390/biomedicines10123060] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/20/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
The significant roles of extracellular vesicles (EVs) as intracellular mediators, disease biomarkers, and therapeutic agents, make them a scientific hotspot. In particular, EVs secreted by human stem cells show significance in treating neurological disorders, such as Alzheimer’s disease and ischemic stroke. However, the clinical applications of EVs are limited due to their poor targeting capabilities and low therapeutic efficacies after intravenous administration. Superparamagnetic iron oxide (SPIO) nanoparticles are biocompatible and have been shown to improve the targeting ability of EVs. In particular, ultrasmall SPIO (USPIO, <50 nm) are more suitable for labeling nanoscale EVs due to their small size. In this study, induced forebrain neural progenitor cortical organoids (iNPCo) were differentiated from human induced pluripotent stem cells (iPSCs), and the iNPCo expressed FOXG1, Nkx2.1, α-catenin, as well as β-tubulin III. EVs were isolated from iNPCo media, then loaded with USPIOs by sonication. Size and concentration of EV particles were measured by nanoparticle tracking analysis, and no significant changes were observed in size distribution before and after sonication, but the concentration decreased after labeling. miR-21 and miR-133b decreased after sonication. Magnetic resonance imaging (MRI) demonstrated contrast visualized for the USPIO labeled EVs embedded in agarose gel phantoms. Upon calculation, USPIO labeled EVs exhibited considerably shorter relaxation times, quantified as T2 and T2* values, reducing the signal intensity and generating higher MRI contrast compared to unlabeled EVs and gel only. Our study demonstrated that USPIO labeling was a feasible approach for in vitro tracking of brain organoid-derived EVs, which paves the way for further in vivo examination.
Collapse
Affiliation(s)
- Chang Liu
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
| | - Shannon Helsper
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL 32310, USA
| | - Mark Marzano
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
| | - Xingchi Chen
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
- High Performance Materials Institute, Florida State University, Tallahassee, FL 32310, USA
| | - Laureana Muok
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
| | - Colin Esmonde
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
| | - Changchun Zeng
- High Performance Materials Institute, Florida State University, Tallahassee, FL 32310, USA
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32310, USA
| | - Samuel C. Grant
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL 32310, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32310, USA
| |
Collapse
|
44
|
From Hair to Colon: Hair Follicle-Derived MSCs Alleviate Pyroptosis in DSS-Induced Ulcerative Colitis by Releasing Exosomes in a Paracrine Manner. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9097530. [PMID: 36160717 PMCID: PMC9507792 DOI: 10.1155/2022/9097530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/09/2022] [Accepted: 04/22/2022] [Indexed: 11/18/2022]
Abstract
Ulcerative colitis (UC) has attracted intense attention due to its high recurrence rate and the difficulty of treatment. Pyroptosis has been suggested to be crucial in the development of UC. Although mesenchymal stem cells (MSCs) are broadly used for UC therapy, they have rarely been studied in the context of UC pyroptosis. Hair follicle-derived MSCs (HFMSCs) are especially understudied with regard to UC and pyroptosis. In this study, we aimed to discover the effects and potential mechanisms of HFMSCs in UC. We administered HFMSCs to dextran sulfate sodium- (DSS-) treated mice and found that the HFMSCs significantly inhibited pyroptosis to alleviate DSS-induced UC. A transwell system and GW4869, an exosome inhibitor, were used to prove the paracrine mechanism of HFMSCs. HFMSC supernatant reduced pyroptosis-related protein expression and promoted cell viability, but these effects were attenuated by GW4869, suggesting a role for HFMSC-released exosomes (Exos) in pyroptosis. Next, Exos were extracted and administered in vitro and in vivo to explore their roles in pyroptosis and UC. In addition, the biodistribution of Exos in mice was tracked using an imaging system and immunofluorescence. The results suggested that Exos not only improved DSS-induced pyroptosis and UC but also were internalized into the injured colon. Furthermore, the therapeutic efficacy of Exos was dose dependent. Among the Exo treatments, administration of 400 μg of Exos per mouse twice a week exhibited the highest efficacy. The differentially expressed miRNAs (DEmiRNAs) between MSCs and MSC-released Exos suggested that Exos might inhibit pyroptosis through tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) signalling and interferon- (IFN-) gamma pathways. Our study reveals that HFMSCs can alleviate pyroptosis in UC by releasing DEmiRNA-containing Exos in a paracrine manner. This finding may lead to new treatments for UC.
Collapse
|
45
|
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
|
46
|
Liang Y, Miao Y, Xiang J. Circular RNA circESPL1 knockdown alleviates lipopolysaccharide (LPS)-induced lung cell injury via sponging miR-326 to regulate MAPK14. Int Immunopharmacol 2022; 112:109146. [PMID: 36030691 DOI: 10.1016/j.intimp.2022.109146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/25/2022] [Accepted: 08/05/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND Infantile pneumonia (IP) is a common inflammatory disease, which brings a heavy burden to young children's health. Previous studies suggested that circular RNA (circRNA) hsa_circ_0026579 (also called circESPL1) was significantly upregulated in pneumonia patients, which was associated with the disease severity. This subject aimed to explore the functional effects and potential regulatory mechanism of circESPL1 on lipopolysaccharide (LPS)-induced lung cell injury. METHODS WI-38 and MRC-5 cells were stimulated by LPS to mimic the inflammatory injury model. CircESPL1, microRNA-326 (miR-326), and Mitogen-Activated Protein Kinase 14 (MAPK14)levels were measured using real-time quantitative polymerase chain reaction (RT-qPCR). Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), and flow cytometry assays were performed to assess cell proliferation and apoptosis. Western blot analysis of B-cell lymphoma-2 (Bcl-2), Bcl-2 related X protein (Bax), C-caspase 3, and MAPK14 protein levels. Tumor necrosis factor-α (TNF-α), Interleukin-6 (IL-6), and IL-1β levels were examined using an Enzyme-linked immunosorbent assay (ELISA). Using Starbase analysis, the binding between miR-326 and circESPL1 or MAPK14 was predicted, followed by confirmation using a dual-luciferase reporter and RNA Immunoprecipitation (RIP) assays. RESULTS Increased circESPL1 and MAPK14, and reduced miR-326 were observed in serum samples from preeclampsia sufferers and LPS-treated lung cells (P < 0.05). Furthermore, circESPL1 deficiency overturned LPS-mediated cell proliferation, apoptosis, and inflammatory response in vitro (P < 0.05). In terms of molecular mechanisms, circESPL1 worked as a sponge of miR-326, and miR-326 absence reversed the protective role of circESPL1 silencing on LPS-triggered lung cell injury (P < 0.05). Also, miR-326 directly targeted MAPK14, and MAPK14 overexpression abolished miR-326-mediated impacts under LPS treatment (P < 0.05). CONCLUSION CircESPL1 knockdown might attenuate LPS-caused lung cell injury by regulating the miR-326/ MAPK14 axis, providing useful insight for exploring a novel therapeutic approach for IP.
Collapse
Affiliation(s)
- Yamei Liang
- Department of Pediatrics, Taizhou First People's Hospital, China
| | - Yingying Miao
- Department of Pediatrics, Taizhou First People's Hospital, China
| | - Jingjing Xiang
- Department of Pediatrics, Taizhou First People's Hospital, China.
| |
Collapse
|
47
|
Tailored Extracellular Vesicles: Novel Tool for Tissue Regeneration. Stem Cells Int 2022; 2022:7695078. [PMID: 35915850 PMCID: PMC9338735 DOI: 10.1155/2022/7695078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 05/10/2022] [Accepted: 07/05/2022] [Indexed: 11/18/2022] Open
Abstract
Extracellular vesicles (EVs) play an essential part in multiple pathophysiological processes including tissue injury and regeneration because of their inherent characteristics of small size, low immunogenicity and toxicity, and capability of carrying a variety of bioactive molecules and mediating intercellular communication. Nevertheless, accumulating studies have shown that the application of EVs faces many challenges such as insufficient therapeutic efficacy, a lack of targeting capability, low yield, and rapid clearance from the body. It is known that EVs can be engineered, modified, and designed to encapsulate therapeutic cargos like proteins, peptides, nucleic acids, and drugs to improve their therapeutic efficacy. Targeted peptides, antibodies, aptamers, magnetic nanoparticles, and proteins are introduced to modify various cell-derived EVs for increasing targeting ability. In addition, extracellular vesicle mimetics (EMs) and self-assembly EV-mimicking nanocomplex are applied to improve production and simplify EV purification process. The combination of EVs with biomaterials like hydrogel, and scaffolds dressing endows EVs with long-term therapeutic efficacy and synergistically enhanced regenerative outcome. Thus, we will summarize recent developments of EV modification strategies for more extraordinary regenerative effect in various tissue injury repair. Subsequently, opportunities and challenges of promoting the clinical application of engineered EVs will be discussed.
Collapse
|
48
|
Apocynin loaded silver nanoparticles displays potent in vitro biological activities and mitigates pyrogallol-induced hepatotoxicity. Chem Biol Interact 2022; 365:110069. [DOI: 10.1016/j.cbi.2022.110069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 01/24/2023]
|
49
|
Qiu Z, Zhong Z, Zhang Y, Tan H, Deng B, Meng G. Human umbilical cord mesenchymal stem cell-derived exosomal miR-335-5p attenuates the inflammation and tubular epithelial-myofibroblast transdifferentiation of renal tubular epithelial cells by reducing ADAM19 protein levels. Stem Cell Res Ther 2022; 13:373. [PMID: 35902972 PMCID: PMC9330665 DOI: 10.1186/s13287-022-03071-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/20/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Renal tubular epithelial-myofibroblast transdifferentiation (EMT) plays a key role in the regulation of renal fibrosis. Exosomes derived from human umbilical cord mesenchymal stem cells (hucMSCs) play a crucial role in alleviating renal fibrosis and injury. Additionally, hucMSC-derived exosomes contain numerous microRNAs (miRNAs). However, it is unclear whether mesenchymal stem cells can regulate the transforming growth factor (TGF)-β1-induced EMT of human renal tubular epithelial cells (RTECs) through exosomal miRNAs. METHOD HK-2, a human RTEC line, was co-treated with TGF-β1 and hucMSC-derived exosomes. Additionally, TGF-β1-treated HK-2 cells were transfected with a miR-335-5p mimic and disintegrin and metalloproteinase domain-containing protein 19 (ADAM19)-overexpression plasmid. miR-335-5p expression and ADAM19 protein and inflammation levels were measured via quantitative reverse transcription polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assays, respectively. RESULTS TGF-β1 treatment changed the shape of HK-2 cells from a cobblestone morphology to a long spindle shape, accompanied by an increase in interleukin (IL)-6, tumor necrosis factor-α, IL-1β, collagen I, collagen III, α-smooth muscle actin, vimentin, and N-cadherin protein levels, whereas E-cadherin protein levels were reduced in these HK-2 cells, suggesting that TGF-β1 treatment induced the inflammation and EMT of HK-2 cells. HucMSC-exosomes improved the inflammation and EMT phenotype of TGF-β1-induced HK-2 cells by transferring miR-335-5p. miR-335-5p was found to bind the ADAM19 3'-untranslated region to reduce ADAM19 protein levels. Additionally, miR-335-5p improved the inflammation and EMT phenotype of HK-2 cells by reducing ADAM19 protein levels with TGF-β1 induction. CONCLUSIONS HucMSC-derived exosomal miR-335-5p attenuates the inflammation and EMT of HK-2 cells by reducing ADAM19 protein levels upon TGF-β1 induction. This study provides a potential therapeutic strategy and identifies targets for clinically treating renal fibrosis.
Collapse
Affiliation(s)
- Zhenhua Qiu
- Department of Laboratory Medicine, The People's Hospital of Gaozhou, Maoming, 525200, China.
| | - Zhihui Zhong
- Department of Laboratory Medicine, The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Yuehan Zhang
- Department of Laboratory Medicine, The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Haoling Tan
- Department of Laboratory Medicine, The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Bo Deng
- Department of Laboratory Medicine, The People's Hospital of Gaozhou, Maoming, 525200, China
| | - Guohuang Meng
- Department of Laboratory Medicine, The People's Hospital of Gaozhou, Maoming, 525200, China
| |
Collapse
|
50
|
Wu F, Gao J, Kang J, Wang X, Niu Q, Liu J, Zhang L. Knowledge Mapping of Exosomes in Autoimmune Diseases: A Bibliometric Analysis (2002–2021). Front Immunol 2022; 13:939433. [PMID: 35935932 PMCID: PMC9353180 DOI: 10.3389/fimmu.2022.939433] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/24/2022] [Indexed: 12/14/2022] Open
Abstract
Background Autoimmune diseases (AIDs) are a class of chronic disabling diseases characterized by inflammation and damage to muscles, joints, bones, and internal organs. Recent studies have shown that much progress has been made in the research of exosomes in AIDs. However, there is no bibliometric analysis in this research field. This study aims to provide a comprehensive overview of the knowledge structure and research hotspots of exosomes in AIDs through bibliometrics. Method Publications related to exosomes in AIDs from 2002 to 2021 were searched on the web of science core collection (WoSCC) database. VOSviewers, CiteSpace and R package “bibliometrix” were used to conduct this bibliometric analysis. Results 312 articles from 48 countries led by China and the United States were included. The number of publications related to exosomes in AIDs is increasing year by year. Central South University, Sun Yat Sen University, Tianjin Medical University and University of Pennsylvania are the main research institutions. Frontiers in immunology is the most popular journal in this field, and Journal of Immunology is the most co-cited journal. These publications come from 473 authors among which Ilias Alevizos, Qianjin Lu, Wei Wei, Jim Xiang and Ming Zhao had published the most papers and Clotilde Théry was co-cited most often. Studying the mechanism of endogenous exosomes in the occurrence and development of AIDs and the therapeutic strategy of exogenous exosomes in AIDs are the main topics in this research field. “Mesenchymal stem cells”, “microRNA”, “biomarkers”, “immunomodulation”, and “therapy” are the primary keywords of emerging research hotspots. Conclusion This is the first bibliometric study that comprehensively summarizes the research trends and developments of exosomes in AIDs. This information identifies recent research frontiers and hot directions, which will provide a reference for scholars studying exosomes.
Collapse
Affiliation(s)
- Fengping Wu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Jinfang Gao
- Department of Rheumatology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Jie Kang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Xuexue Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Qing Niu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Jiaxi Liu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Liyun Zhang
- Department of Rheumatology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- *Correspondence: Liyun Zhang,
| |
Collapse
|