1
|
Zhang Q, Liu J, Wang W, Lin W, Ahmed W, Duan W, Huang S, Zhu Z, Chen L. The role of exosomes derived from stem cells in nerve regeneration: A contribution to neurological repair. Exp Neurol 2024; 380:114882. [PMID: 39002923 DOI: 10.1016/j.expneurol.2024.114882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/27/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
Stem cell-derived exosomes have gained attention in regenerative medicine for their role in encouraging nerve regeneration and potential use in treating neurological diseases. These nanosized extracellular vesicles act as carriers of bioactive molecules, facilitating intercellular communication and enhancing the regenerative process in neural tissues. This comprehensive study explores the methods by which exosomes produced from various stem cells contribute to nerve healing, with a particular emphasis on their role in angiogenesis, inflammation, and cellular signaling pathways. By examining cutting-edge developments and exploring the potential of exosomes in delivering disease-specific miRNAs and proteins, we highlight their versatility in tailoring personalized therapeutic strategies. The findings presented here highlight the potential of stem cell-produced exosomes for use in neurological diseases therapy, establishing the door for future research into exosome-based neurotherapies.
Collapse
Affiliation(s)
- Qiankun Zhang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiale Liu
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Wang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Wentong Lin
- Department of Orthopaedics, Chaozhou Hospital of Traditional Chinese Medicine, Chaozhou, China
| | - Waqas Ahmed
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Wenjie Duan
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Songze Huang
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhihan Zhu
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Lukui Chen
- Department of Neurosurgery, Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
2
|
Song W, Guo Y, Liu W, Yao Y, Zhang X, Cai Z, Yuan C, Wang X, Wang Y, Jiang X, Wang H, Yu W, Li H, Zhu Y, Kong L, He Y. Circadian Rhythm-Regulated ADSC-Derived sEVs and a Triphasic Microneedle Delivery System to Enhance Tendon-to-Bone Healing. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408255. [PMID: 39120049 DOI: 10.1002/adma.202408255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/23/2024] [Indexed: 08/10/2024]
Abstract
Modulating the inflammatory microenvironment to reconstruct the fibrocartilaginous layer while promoting tendon repair is crucial for enhancing tendon-to-bone healing in rotator cuff repair (RCR), a persistent challenge in orthopedics. Small extracellular vesicles (sEVs) hold significant potential to modulate inflammation, yet the efficient production of highly bioactive sEVs remains a substantial barrier to their clinical application. Moreover, achieving minimally invasive local delivery of sEVs to the tendon-to-bone interface presents significant technical difficulties. Herein, the circadian rhythm of adipose-derived stem cells is modulated to increase the yield and enhance the inflammatory regulatory capacity of sEVs. Circadian rhythm-regulated sEVs (CR-sEVs) enhance the cyclic adenosine monophosphate signaling pathway in macrophage (Mφ) via platelet factor 4 delivery, thereby inhibiting Mφ M1 polarization. Subsequently, a triphasic microneedle (MN) scaffold with a tip, stem, and base is designed for the local delivery of CR-sEVs (CR-sEVs/MN) at the tendon-to-bone junction, incorporating tendon-derived decellularized extracellular matrix in the base to facilitate tendon repair. CR-sEVs/MN mitigates inflammation, promotes fibrocartilage regeneration, and enhances tendon healing, thereby improving biomechanical strength and shoulder joint function in a rat RCR model. Combining CR-sEVs with this triphasic microneedle delivery system presents a promising strategy for enhancing tendon-to-bone healing in clinical settings.
Collapse
Affiliation(s)
- Wei Song
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Ying Guo
- Department of Cardiology, Heart Center, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, P. R. China
| | - Wencai Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Yijing Yao
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, P. R. China
| | - Xuancheng Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Zhuochang Cai
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Chenrui Yuan
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Xin Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Yifei Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Xiping Jiang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Haoyuan Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Weilin Yu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Haiyan Li
- Chemical and Environmental Engineering Department, School of Engineering, STEM College, RMIT University, 124 La Trobe St., Melbourne, Victoria, 3000, Australia
| | - Yanlun Zhu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Lingzhi Kong
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
| | - Yaohua He
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, P. R. China
- Department of Orthopedic Surgery, Jinshan District Central Hospital affiliated to Shanghai University of Medicine & Health Sciences, Jinshan Branch of Shanghai Sixth People's Hospital, Shanghai, 201500, P. R. China
| |
Collapse
|
3
|
Rudnitsky E, Braiman A, Wolfson M, Muradian KK, Gorbunova V, Turgeman G, Fraifeld VE. Stem cell-derived extracellular vesicles as senotherapeutics. Ageing Res Rev 2024; 99:102391. [PMID: 38914266 DOI: 10.1016/j.arr.2024.102391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
Cellular senescence (CS) is recognized as one of the hallmarks of aging, and an important player in a variety of age-related pathologies. Accumulation of senescent cells can promote a pro-inflammatory and pro-cancerogenic microenvironment. Among potential senotherapeutics are extracellular vesicles (EVs) (40-1000 nm), including exosomes (40-150 nm), that play an important role in cell-cell communications. Here, we review the most recent studies on the impact of EVs derived from stem cells (MSCs, ESCs, iPSCs) as well as non-stem cells of various types on CS and discuss potential mechanisms responsible for the senotherapeutic effects of EVs. The analysis revealed that (i) EVs derived from stem cells, pluripotent (ESCs, iPSCs) or multipotent (MSCs of various origin), can mitigate the cellular senescence phenotype both in vitro and in vivo; (ii) this effect is presumably senomorphic; (iii) EVs display cross-species activity, without apparent immunogenic responses. In summary, stem cell-derived EVs appear to be promising senotherapeutics, with a feasible application in humans.
Collapse
Affiliation(s)
- Ekaterina Rudnitsky
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Alex Braiman
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Marina Wolfson
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Khachik K Muradian
- Department of Biology of Aging and Experimental Life Span Extension, State Institute of Gerontology of National Academy of Medical Sciences of Ukraine, Kiev 4114, Ukraine
| | - Vera Gorbunova
- Department of Biology, Rochester Aging Research Center, University of Rochester, Rochester, NY 14627, USA
| | - Gadi Turgeman
- Department of Molecular Biology, Faculty of Natural Sciences and Medical School, Ariel University, Ariel 40700, Israel.
| | - Vadim E Fraifeld
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Center for Multidisciplinary Research on Aging, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel.
| |
Collapse
|
4
|
Kim SD, Kang SA, Mun SJ, Yu HS, Roh HJ, Cho KS. SCGB1C1 Plays a Critical Role in Suppression of Allergic Airway Inflammation through the Induction of Regulatory T Cell Expansion. Int J Mol Sci 2024; 25:6282. [PMID: 38892470 PMCID: PMC11173076 DOI: 10.3390/ijms25116282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/27/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
The nanosized vesicles secreted from various cell types into the surrounding extracellular space are called extracellular vesicles (EVs). Although mesenchymal stem cell-derived EVs are known to have immunomodulatory effects in asthmatic mice, the role of identified pulmonary genes in the suppression of allergic airway inflammation remains to be elucidated. Moreover, the major genes responsible for immune regulation in allergic airway diseases have not been well documented. This study aims to evaluate the immunomodulatory effects of secretoglobin family 1C member 1 (SCGB1C1) on asthmatic mouse models. C57BL/6 mice were sensitized to ovalbumin (OVA) using intraperitoneal injection and were intranasally challenged with OVA. To evaluate the effect of SCGB1C1 on allergic airway inflammation, 5 μg/50 μL of SCGB1C1 was administrated intranasally before an OVA challenge. We evaluated airway hyperresponsiveness (AHR), total inflammatory cells, eosinophils in the bronchoalveolar lavage fluid (BALF), lung histology, serum immunoglobulin (Ig), the cytokine profiles of BALF and lung-draining lymph nodes (LLN), and the T cell populations in LLNs. The intranasal administration of SCGB1C1 significantly inhibited AHR, the presence of eosinophils in BALF, eosinophilic inflammation, goblet cell hyperplasia in the lung, and serum total and allergen-specific IgE. SCGB1C1 treatment significantly decreased the expression of interleukin (IL)-5 in the BALF and IL-4 in the LLN, but significantly increased the expression of IL-10 and transforming growth factor (TGF)-β in the BALF. Furthermore, SCGB1C1 treatment notably increased the populations of CD4+CD25+Foxp3+ regulatory T cells (Tregs) in asthmatic mice. The intranasal administration of SCGB1C1 provides a significant reduction in allergic airway inflammation and improvement of lung function through the induction of Treg expansion. Therefore, SCGB1C1 may be the major regulator responsible for suppressing allergic airway inflammation.
Collapse
Affiliation(s)
- Sung-Dong Kim
- Department of Otorhinolaryngology, Pusan National University Hospital, Busan 49241, Republic of Korea;
| | - Shin-Ae Kang
- Department of Parasitology and Tropical Medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea; (S.-A.K.); (H.-S.Y.)
| | - Sue-Jean Mun
- Department of Otorhinolaryngology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (S.-J.M.); (H.-J.R.)
| | - Hak-Sun Yu
- Department of Parasitology and Tropical Medicine, Pusan National University School of Medicine, Yangsan 50612, Republic of Korea; (S.-A.K.); (H.-S.Y.)
| | - Hwan-Jung Roh
- Department of Otorhinolaryngology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea; (S.-J.M.); (H.-J.R.)
| | - Kyu-Sup Cho
- Department of Otorhinolaryngology, Pusan National University Hospital, Busan 49241, Republic of Korea;
| |
Collapse
|
5
|
Zhou C, Wu Y, Wan S, Lou L, Gu S, Peng J, Zhao S, Hua X. Exosomes isolated from TNF-α-treated bone marrow mesenchymal stem cells ameliorate pelvic floor dysfunction in rats. J Cell Mol Med 2024; 28:e18451. [PMID: 38898783 PMCID: PMC11187403 DOI: 10.1111/jcmm.18451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024] Open
Abstract
Exosomes derived from bone marrow-derived mesenchymal stem cells (BMSCs) can alleviate the symptoms of pelvic floor dysfunction (PFD) in rats. However, the potential therapeutical effects of exosomes derived from BMSCs treated with tumour necrosis factor (TNF)-α on the symptoms of PFD in rats are unknown. Exosomes extracted from BMSCs treated with or without TNF-α were applied to treat PFD rats. Our findings revealed a significant elevation in interleukin (IL)-6 and TNF-α, and matrix metalloproteinase-2 (MMP2) levels in the vaginal wall tissues of patients with pelvic organ prolapse (POP) compared with the control group. Daily administration of exosomes derived from BMSCs, treated either with or without TNF-α (referred to as Exo and TNF-Exo), resulted in increased void volume and bladder void pressure, along with reduced peak bladder pressure and leak point pressure in PFD rats. Notably, TNF-Exo treatment demonstrated superior efficacy in restoring void volume, bladder void pressure and the mentioned parameters compared with Exo treatment. Importantly, TNF-Exo exhibited greater potency than Exo in restoring the levels of multiple proteins (Elastin, Collagen I, Collagen III, IL-6, TNF-α and MMP2) in the anterior vaginal walls of PFD rats. The application of exosomes derived from TNF-α-treated BMSCs holds promise as a novel therapeutic approach for treating PFD.
Collapse
Affiliation(s)
- Chenchen Zhou
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yuelin Wu
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Sheng Wan
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Liqun Lou
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Shengyi Gu
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Jing Peng
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Shifeng Zhao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Xiaolin Hua
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| |
Collapse
|
6
|
Karimian A, Khoshnazar SM, Kazemi T, Asadi A, Abdolmaleki A. Role of secretomes in cell-free therapeutic strategies in regenerative medicine. Cell Tissue Bank 2024; 25:411-426. [PMID: 36725732 DOI: 10.1007/s10561-023-10073-5] [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: 08/01/2022] [Accepted: 01/21/2023] [Indexed: 02/03/2023]
Abstract
After an injury, peripheral nervous system neurons have the potential to rebuild their axons by generating a complicated activation response. Signals from the damaged axon are required for this genetic transition to occur. Schwann cells (SCs) near a damaged nerve's distal stump also play a role in the local modulation of axonal programs, not only via cell-to-cell contacts but also through secreted signals (the secretome). The secretome is made up of all the proteins that the cell produces, such as cytokines, growth factors, and extracellular vesicles. The released vesicles may carry signaling proteins as well as coding and regulatory RNAs, allowing for multilayer communication. The secretome of SCs is now well understood as being critical for both orchestrating Wallerian degeneration and maintaining axonal regeneration. As a consequence, secretome has emerged as a feasible tissue regeneration alternative to cell therapy. Separate SC secretome components have been used extensively in the lab to promote peripheral nerve regeneration after injury. However, in neurological therapies, the secretome generated by mesenchymal (MSC) or other derived stem cells has been the most often used. In fact, the advantages of cell treatment have been connected to the release of bioactive chemicals and extracellular vesicles, which make up MSCs' secretome.
Collapse
Affiliation(s)
- Aida Karimian
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Seyedeh Mahdieh Khoshnazar
- Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Tahmineh Kazemi
- Department of Basic Sciences, Faculty of Veterinary Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Asadollah Asadi
- Department of Biology, Faculty of Science, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Arash Abdolmaleki
- Department of Biophysics, Faculty of Advanced Technologies, University of Mohaghegh Ardabili, Namin, Iran.
| |
Collapse
|
7
|
Mohammadi A, Shabani R, Bashiri Z, Rafiei S, Asgari H, Koruji M. Therapeutic potential of exosomes in spermatogenesis regulation and male infertility. Biol Cell 2024; 116:e2300127. [PMID: 38593304 DOI: 10.1111/boc.202300127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Spermatogenesis is a fundamental process crucial for male reproductive health and fertility. Exosomes, small membranous vesicles released by various cell types, have recently garnered attention for their role in intercellular communication. OBJECTIVE This review aims to comprehensively explore the role of exosomes in regulating spermatogenesis, focusing on their involvement in testicular development and cell-to-cell communication. METHODS A systematic examination of literature was conducted to gather relevant studies elucidating the biogenesis, composition, and functions of exosomes in the context of spermatogenesis. RESULTS Exosomes play a pivotal role in orchestrating the complex signaling networks required for proper spermatogenesis. They facilitate the transfer of key regulatory molecules between different cell populations within the testes, including Sertoli cells, Leydig cells, and germ cells. CONCLUSION The emerging understanding of exosome-mediated communication sheds light on novel mechanisms underlying spermatogenesis regulation. Further research in this area holds promise for insights into male reproductive health and potential therapeutic interventions.
Collapse
Affiliation(s)
- Amirhossein Mohammadi
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ronak Shabani
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Reproductive Sciences and Technology Research Center, Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Bashiri
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Endometrium and Endometriosis Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Omid Fertility & Infertility Clinic, Hamedan, Iran
| | - Sara Rafiei
- Department of Botany and Plant Sciences, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Hamidreza Asgari
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Koruji
- Stem Cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Anvari Y, Afrashteh A, Pourkaveh S, Salek SB, Al-Numan L, Khademnezhad S. Emerging role of mesenchymal stem cell-derived extracellular vesicles in periodontal regeneration. J Taibah Univ Med Sci 2024; 19:390-402. [PMID: 38380419 PMCID: PMC10876597 DOI: 10.1016/j.jtumed.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/17/2023] [Accepted: 01/25/2024] [Indexed: 02/22/2024] Open
Abstract
Periodontitis is a prevalent oral ailment that harms both hard and soft tissues of the periodontium, leading to loosening and eventual removal of the teeth. Current clinical treatments have limitations in achieving complete periodontal tissue regeneration. Mesenchymal stem cells (MSCs) have garnered attention due to their unique characteristics and potential as a promising new therapy for periodontitis. Research suggests that the role of MSCs in regenerative medicine primarily occurs through the paracrine pathway, involving the emission of particles encased by lipids called extracellular vesicles (EVs) abundant in bioactive compounds. These EVs play a vital function in controlling the activities of periodontal tissues and immune system cells, and by influencing the immediate surrounding, thus fostering the healing of periodontal damage and renewal of tissues. EVs obtained from MSCs (MSC-EVs), in the form of a cell-free treatment, offer advantages in terms of stability, reduced immune rejection, and ethical considerations, elevating their potential as a hopeful choice for broad clinical applications. This concise overview highlights the mechanisms of MSC-EVs and the possibilities they hold in clinical application for periodontal regeneration.
Collapse
Affiliation(s)
- Yaldasadat Anvari
- Department of Dentistry, School of Dentistry, Near East University, Nicosia, Cyprus
| | - Ahmad Afrashteh
- Department of Periodontics, School of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajjad Pourkaveh
- Department of Periodontics, School of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira B. Salek
- Department of Periodontics, School of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Lelaw Al-Numan
- Department of Dentistry, School of Dentistry, Near East University, Nicosia, Cyprus
| | - Sahar Khademnezhad
- Department of Oral and Maxillofacial Medicine, School of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
9
|
Dehghan Z, Rezaee D, Noori E, Pilehchi T, Saberi F, Taheri Z, Darya G, Mehdinejadiani S. Exosomes as modulators of embryo implantation. Mol Biol Rep 2024; 51:284. [PMID: 38324178 DOI: 10.1007/s11033-024-09282-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/23/2024] [Indexed: 02/08/2024]
Abstract
Exosomes, known as extracellular vehicles (EVs), are found in biological fluids. They have the capability to carry and transfer signaling molecules, such as nucleic acids and proteins, facilitating intercellular communication and regulating the gene expression profile in target cells. EVs have the potential to be used as biomarkers in diagnosis, prognosis and also as feasible therapeutic targets. The available evidence suggests that exosomes play critical roles in the reproductive system, particularly during implantation, which is widely recognized as a crucial step in early pregnancy. A proper molecular dialogue between a high-quality embryo and a receptive endometrium is essential for the establishment of a normal pregnancy. This review focuses on the key role of exosomes originated from various sources, including the embryo, seminal fluid, and uterus fluid, based on the available evidence. It explores their potential applications as a novel approach in assisted reproductive technologies (ART).
Collapse
Affiliation(s)
- Zeinab Dehghan
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Delsuz Rezaee
- School of Allied Medical Sciences, Ilam University of Medical Sciences, Ilam, Iran
| | - Effat Noori
- Department of Biotechnology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Tayyebeh Pilehchi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Saberi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Taheri
- Department of Biology and Biotechnology, Pavia University, Pavia, Italy
| | - Gholamhossein Darya
- Department of Comparative Biomedical Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shayesteh Mehdinejadiani
- Department of Reproductive Biology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
10
|
Tan X, Xiao H, Yan A, Li M, Wang L. Effect of Exosomes From Bone Marrow-Derived Mesenchymal Stromal Cells and Adipose-Derived Stromal Cells on Bone-Tendon Healing in a Murine Rotator Cuff Injury Model. Orthop J Sports Med 2024; 12:23259671231210304. [PMID: 38188618 PMCID: PMC10768594 DOI: 10.1177/23259671231210304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 06/06/2023] [Indexed: 01/09/2024] Open
Abstract
Background Bone-tendon injury is characterized by poor self-healing. It is established that exosomes are favorable for tissue repair and regeneration. However, their effect on bone-tendon healing has not yet been determined. Purpose To compare the effectiveness of exosomes derived from adipose-derived mesenchymal stromal cells (ADSC-Exos) and bone marrow-derived mesenchymal stromal cells (BMSC-Exos) on bone-tendon interface healing in murine rotator cuff injury model and explore the underlying mechanisms thereof. Study Design Controlled laboratory study. Methods A total of 63 male C57BL6 mice with rotator cuff injuries underwent surgery and were randomly assigned to a control group treated without exosomes (n = 21), an ADSC-Exos group (n = 21), or a BMSC-Exos group (n = 21). The mice were sacrificed 4 or 8 weeks after surgery, and tissues were collected for histologic examination and radiographic and biomechanical testing. For exosome tracing in vivo, mice were sacrificed 7 days after surgery. A series of functional assays (radiographic evaluation, proliferation assay, Alizarin Red staining, alkaline phosphatase staining and activity, Alcian blue staining, quantitative polymerase chain reaction analyses, and glycosaminoglycans quantification) were conducted to evaluate the effect of exosomes on the cellular behaviors of the BMSCs in vitro. A statistical analysis of multiple-group comparisons was performed by 1-way analysis of variance, followed by the Bonferroni post hoc test to assess the differences between the 2 groups. Results The ADSCs and BMSCs were positive for surface markers CD29 and CD90 and negative for surface markers CD34 and CD45 and could differentiate into osteoblasts, chondrocytes, and adipocytes. Exosomes showed a cup- or sphere-shaped morphology and were positive for CD63 and TGS101. Local injection of ADSC-Exos and BMSC-Exos could recruit BMSCs and promote osteogenesis, chondrogenesis, and bone-tendon healing. In vitro, ADSC-Exos and BMSC-Exos could significantly promote the proliferation, migration, osteogenic differentiation, and chondrogenic differentiation ability of BMSCs. In vivo, ADSC-Exos and BMSC-Exos significantly accelerated bone-tendon injury healing, with no significant statistical difference between them. Conclusion ADSC-Exos and BMSC-Exos exhibited similar therapeutic effects on bone-tendon healing in our murine animal model. Clinical Relevance ADSC-Exos and BMSC-Exos may be used to develop a new cell-free therapy method for promoting rotator cuff injury repair.
Collapse
Affiliation(s)
- Xiaoqian Tan
- Department of Pediatric Orthopedics, Hunan Children's Hospital, Changsha, Hunan, China
- The School of Pediatrics, University of South China, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children's Hospital, Hunan, China
| | - Han Xiao
- Department of Pediatric Orthopedics, Hunan Children's Hospital, Changsha, Hunan, China
- The School of Pediatrics, University of South China, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children's Hospital, Hunan, China
| | - An Yan
- Department of Pediatric Orthopedics, Hunan Children's Hospital, Changsha, Hunan, China
- The School of Pediatrics, University of South China, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children's Hospital, Hunan, China
| | - Miao Li
- Department of Pediatric Orthopedics, Hunan Children's Hospital, Changsha, Hunan, China
- The School of Pediatrics, University of South China, Changsha, Hunan, China
- Hunan Provincial Key Laboratory of Pediatric Orthopedics, Hunan Children's Hospital, Hunan, China
| | - Linfeng Wang
- Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
11
|
Yin Z, Gong G, Liu X, Yin J. Mechanism of regulating macrophages/osteoclasts in attenuating wear particle-induced aseptic osteolysis. Front Immunol 2023; 14:1274679. [PMID: 37860014 PMCID: PMC10582964 DOI: 10.3389/fimmu.2023.1274679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/22/2023] [Indexed: 10/21/2023] Open
Abstract
Joint replacement surgery is the most effective treatment for end-stage arthritis. Aseptic loosening caused by periprosthetic osteolysis is a common complication after joint replacement. Inflammation induced by wear particles derived from prosthetic biomaterials is a major cause of osteolysis. We emphasize that bone marrow-derived macrophages and their fusion-derived osteoclasts play a key role in this pathological process. Researchers have developed multiple intervention approaches to regulate macrophage/osteoclast activation. Aiming at wear particle-induced periprosthetic aseptic osteolysis, this review separately discusses the molecular mechanism of regulation of ROS formation and inflammatory response through intervention of macrophage/osteoclast RANKL-MAPKs-NF-κB pathway. These molecular mechanisms regulate osteoclast activation in different ways, but they are not isolated from each other. There is also a lot of crosstalk among the different mechanisms. In addition, other bone and joint diseases related to osteoclast activation are also briefly introduced. Therefore, we discuss these new findings in the context of existing work with a view to developing new strategies for wear particle-associated osteolysis based on the regulation of macrophages/osteoclasts.
Collapse
Affiliation(s)
- Zhaoyang Yin
- Department of Orthopedics, The Affiliated Lianyungang Hospital of Xuzhou Medical University (The First People’s Hospital of Lianyungang), Lianyungang, China
| | - Ge Gong
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinhui Liu
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Jian Yin
- Department of Orthopedics, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
Manley AL, Chen J, Fitzgerald W, Feng X, Young NS. Immunosuppressive Activity of Exosomes from Granulocytic Myeloid-Derived Suppressor Cells in a Murine Model of Immune Bone Marrow Failure. Int J Mol Sci 2023; 24:14661. [PMID: 37834110 PMCID: PMC10572857 DOI: 10.3390/ijms241914661] [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: 08/17/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
We previously reported that granulocytic myeloid-derived suppressor cells (G-MDSCs) suppressed T-cell activation and attenuated bone marrow failure (BMF) in a minor histocompatibility (minor-H) antigen mismatched murine aplastic anemia (AA) model. In the current study, we tested the hypothesis that exosomes, a subset of extracellular vesicles, are responsible at least partially for G-MDSCs' therapeutic efficacy. Indeed, exosomes isolated from GMDSCs (G-MDSC-exos) suppressed CD4+ and CD8+ T-cell proliferation in vitro and mildly attenuated immune BMF in the minor-H mismatched AA model. G-MDSC-exos treatment significantly increased red blood cells, hemoglobin, and total bone marrow (BM) cells, and moderately reduced BM CD8+ T cells. G-MDSC-exos' effects were associated with upregulations in an array of lymphocyte-suppression-related miRNAs such as hsa-miR-142-5p, miR-19a-3p, and miR-19b-3p in both BM CD4+ and CD8+ T cells. We concluded that G-MDSC-exos attenuate immune BMF via modulating the delivery of immunosuppressive miRNAs into activated T lymphocytes.
Collapse
Affiliation(s)
- Ash Lee Manley
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (A.L.M.); (J.C.); (N.S.Y.)
| | - Jichun Chen
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (A.L.M.); (J.C.); (N.S.Y.)
| | - Wendy Fitzgerald
- Intracellular Interactions, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (A.L.M.); (J.C.); (N.S.Y.)
| | - Neal S. Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (A.L.M.); (J.C.); (N.S.Y.)
| |
Collapse
|
13
|
Chen Q, Yao L, Liu Q, Hou J, Qiu X, Chen M, Wu Z, Hu D, Cui F, Yan T. Exosome-coated polydatin nanoparticles in the treatment of radiation-induced intestinal damage. Aging (Albany NY) 2023; 15:6905-6920. [PMID: 37466428 PMCID: PMC10415572 DOI: 10.18632/aging.204882] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 06/22/2023] [Indexed: 07/20/2023]
Abstract
This study aimed to develop an exosome-coated polydatin (PD) nanoparticles (exo-PD) for improving the water solubility and bioavailability of polydatin and explore its salutary effects on intestinal radiation injury. Exosomes (exo) were extracted from the medium of human amniotic fluid stem cells (hAFSc). Mice were divided into control group, irradiation (IR) group, irradiation+PD (IR+PD) group, irradiation+exo (IR+exo) group and irradiation+exo-PD (IR+exo-PD) group. The results of characterization of protein markers, particle size, morphology and cellular uptake ability confirmed that exosomes were effectively isolated using ultracentrifugation. Compared with the IR group, exo-PD improved cell viability, prolonged survival of mice, improved leukocyte count and reduced diarrhea rate. Histological results showed that the exo-PD group had significant improvements in small intestinal villus length and crypt number and less crypt cell damage. exo-PD could reduce IL-1α and IL-6 levels, reduced γ-H2AX expression, increased mitochondrial membrane potential, enhanced oxidative phosphorylation, and delayed cellular senescence. exo-PD could alleviate intestinal injury by improving mitochondrial function through PI3K-AKT pathway. The exo-PD was able to reduce radiation damage to intestinal cells and could be a potential candidate for salvage of intestinal radiation damage.
Collapse
Affiliation(s)
- Qiu Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Lei Yao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Quanbin Liu
- Rocket Force Specialty Medical Center PLA, Beijing 100088, China
| | - Jun Hou
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xinyu Qiu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Mengyuan Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Zhuojun Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Duanmin Hu
- Department of Gastroenterology, The Second Affiliated Hospital of Soochow University, Suzhou 215123, China
| | - Fengmei Cui
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China
- Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou 215123, China
| | - Tao Yan
- Rocket Force Specialty Medical Center PLA, Beijing 100088, China
| |
Collapse
|
14
|
Sun ZL, You T, Zhang BH, Liu Y, Liu J. Bone marrow mesenchymal stem cell-derived exosomes miR-202-5p inhibited pyroptosis to alleviate lung ischemic-reperfusion injury by targeting CMPK2. Kaohsiung J Med Sci 2023; 39:688-698. [PMID: 37092308 DOI: 10.1002/kjm2.12688] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 03/03/2023] [Accepted: 03/30/2023] [Indexed: 04/25/2023] Open
Abstract
Bone mesenchymal stem cell-derived exosome (BMSC-exosome) is a potential candidate for lung ischemia-reperfusion injury (LIRI) treatment. This study aims to investigate the anti-pyroptosis effect of BMSC-exosomes in LIRI. The LIRI cell model was established by hypoxia/reoxygenation (H/R) treatment. Interleukin (IL)-1β and IL-18 levels were examined by enzyme-linked immunosorbent assay. Cell viability was assessed by 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide assay. Lactate dehydrogenase (LDH) release was examined using a LDH assay kit. The interaction between microRNA (miR)-202-5p and cytidine monophosphate kinase 2 (CMPK2) was analyzed using dual-luciferase reporter assay and RNA immunoprecipitation. BMSC-exosomes promoted cell viability and suppressed pyroptosis in H/R-treated mouse lung epithelial. miR-202-5p was enriched in BMSC-exosomes, and exosomal miR-202-5p inhibition upregulated pyroptosis-associated proteins, including cleaved N-terminal Gasdermin D, nucleotide-binding domain-like receptor family member pyrin domain-containing protein 3, and Caspase1. Meanwhile, miR-202-5p suppressed CMPK2 expression by directly targeting CMPK2. Expectedly, CMPK2 knockdown reversed the promoting effect of exosomal miR-202-5p inhibition on pyroptosis in LIRI. Therefore, BMSC-derived exosome miR-202-5p repressed pyroptosis to inhibit LIRI progression by targeting CMPK2.
Collapse
Affiliation(s)
- Zhi-Lu Sun
- The First Affiliated Hospital, Emergency Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Ting You
- The First Affiliated Hospital, Emergency Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Bi-Hong Zhang
- The First Affiliated Hospital, Emergency Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Yu Liu
- The First Affiliated Hospital, Emergency Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Jing Liu
- The First Affiliated Hospital, Department of Rehabilitation, Hengyang Medical School, University of South China, Hengyang, Hunan Province, People's Republic of China
| |
Collapse
|
15
|
Ding J, Pan Y, Raj S, Schaffrick L, Wong J, Nguyen A, Manchikanti S, Unsworth L, Kwan P, Tredget E. Characteristics of Serum Exosomes after Burn Injury and Dermal Fibroblast Regulation by Exosomes In Vitro. Cells 2023; 12:1738. [PMID: 37443772 PMCID: PMC10341298 DOI: 10.3390/cells12131738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
(1) Background: Exosomes (EXOs) have been considered a new target thought to be involved in and treat wound healing. More research is needed to fully understand EXO characteristics and the mechanisms of EXO-mediated wound healing, especially wound healing after burn injury. (2) Methods: All EXOs were isolated from 85 serum samples of 29 burn patients and 13 healthy individuals. We characterized the EXOs for morphology and density, serum concentration, protein level, marker expression, size distribution, and cytokine content. After a confirmation of EXO uptake by dermal fibroblasts, we also explored the functional regulation of primary human normal skin and hypertrophic scar fibroblast cell lines by the EXOs in vitro, including cell proliferation and apoptosis. (3) Results: EXOs dynamically changed their morphology, density, size, and cytokine level during wound healing in burn patients, which were correlated with burn severity and the stages of wound healing. EXOs both from burn patients and healthy individuals stimulated dermal fibroblast proliferation and apoptosis. (4) Conclusions: EXO features may be important signals that influence wound healing after burn injury; however, to understand the mechanisms by which EXOs regulates the fibroblasts in healing wounds, further studies will be required.
Collapse
Affiliation(s)
- Jie Ding
- Wound Healing Research Group, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada; (Y.P.); (L.S.); (J.W.); (A.N.); (S.M.); (P.K.); (E.T.)
| | - Yingying Pan
- Wound Healing Research Group, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada; (Y.P.); (L.S.); (J.W.); (A.N.); (S.M.); (P.K.); (E.T.)
| | - Shammy Raj
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 2S2, Canada; (S.R.); (L.U.)
| | - Lindy Schaffrick
- Wound Healing Research Group, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada; (Y.P.); (L.S.); (J.W.); (A.N.); (S.M.); (P.K.); (E.T.)
| | - Jolene Wong
- Wound Healing Research Group, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada; (Y.P.); (L.S.); (J.W.); (A.N.); (S.M.); (P.K.); (E.T.)
| | - Antoinette Nguyen
- Wound Healing Research Group, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada; (Y.P.); (L.S.); (J.W.); (A.N.); (S.M.); (P.K.); (E.T.)
| | - Sharada Manchikanti
- Wound Healing Research Group, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada; (Y.P.); (L.S.); (J.W.); (A.N.); (S.M.); (P.K.); (E.T.)
| | - Larry Unsworth
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 2S2, Canada; (S.R.); (L.U.)
| | - Peter Kwan
- Wound Healing Research Group, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada; (Y.P.); (L.S.); (J.W.); (A.N.); (S.M.); (P.K.); (E.T.)
| | - Edward Tredget
- Wound Healing Research Group, Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2S2, Canada; (Y.P.); (L.S.); (J.W.); (A.N.); (S.M.); (P.K.); (E.T.)
| |
Collapse
|
16
|
Csobonyeiova M, Smolinska V, Harsanyi S, Ivantysyn M, Klein M. The Immunomodulatory Role of Cell-Free Approaches in SARS-CoV-2-Induced Cytokine Storm-A Powerful Therapeutic Tool for COVID-19 Patients. Biomedicines 2023; 11:1736. [PMID: 37371831 DOI: 10.3390/biomedicines11061736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Currently, there is still no effective and definitive cure for the coronavirus disease 2019 (COVID-19) caused by the infection of the novel highly contagious severe acute respiratory syndrome virus (SARS-CoV-2), whose sudden outbreak was recorded for the first time in China in late December 2019. Soon after, COVID-19 affected not only the vast majority of China's population but the whole world and caused a global health public crisis as a new pandemic. It is well known that viral infection can cause acute respiratory distress syndrome (ARDS) and, in severe cases, can even be lethal. Behind the inflammatory process lies the so-called cytokine storm (CS), which activates various inflammatory cytokines that damage numerous organ tissues. Since the first outbreak of SARS-CoV-2, various research groups have been intensively trying to investigate the best treatment options; however, only limited outcomes have been achieved. One of the most promising strategies represents using either stem cells, such as mesenchymal stem cells (MSCs)/induced pluripotent stem cells (iPSCs), or, more recently, using cell-free approaches involving conditioned media (CMs) and their content, such as extracellular vesicles (EVs) (e.g., exosomes or miRNAs) derived from stem cells. As key mediators of intracellular communication, exosomes carry a cocktail of different molecules with anti-inflammatory effects and immunomodulatory capacity. Our comprehensive review outlines the complex inflammatory process responsible for the CS, summarizes the present results of cell-free-based pre-clinical and clinical studies for COVID-19 treatment, and discusses their future perspectives for therapeutic applications.
Collapse
Affiliation(s)
- Maria Csobonyeiova
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
- Apel, Dunajská 52, 811 08 Bratislava, Slovakia
- Regenmed Ltd., Medená 29, 811 08 Bratislava, Slovakia
| | - Veronika Smolinska
- Regenmed Ltd., Medená 29, 811 08 Bratislava, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | - Stefan Harsanyi
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| | | | - Martin Klein
- Institute of Histology and Embryology, Faculty of Medicine, Comenius University, Sasinkova 4, 811 08 Bratislava, Slovakia
| |
Collapse
|
17
|
Şimşek FB, Şencan A, Vatansever HS. Exosomes obtained from adipose mesenchymal stem cells prevent ischemia-reperfusion injury after torsion-detorsion in rat testes. Pediatr Surg Int 2023; 39:204. [PMID: 37222876 DOI: 10.1007/s00383-023-05487-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/10/2023] [Indexed: 05/25/2023]
Abstract
PURPOSE To investigate the effect of exosomes obtained from adipose-derived mesenchymal stem cells (AD-MSCs) on testicular ischemia-reperfusion (I/R) injury. METHODS AD-MSCs from rat adipose tissue were cultured. Characterization of cells was evaluated with CD44, CD90, CD34 and CD45 antibodies. Exosomes from AD-MSCs were obtained with the miRCURY exosome isolation kit. 21 rats were divided into 3 groups. The I/R model was created as 720° torsion for 4 h and reperfusion for 4 h. In the Sham group (SG), only scrotal incision was made. 100 µl of medium in the torsion-control group (T-CG) and 100 µl of exosome in the treatment group (TG) were injected into the testicular parenchyma after detorsion. Johnsen scores of testicles were determined. Apoptosis was evaluated by the TUNEL method. RESULTS It was observed that the seminiferous tubule structures were partially disrupted in T-CG, but normal in SG and TG. Johnsen scores in SG, T-CG, and TG were 8.64 ± 0.39, 7.71 ± 0.37, and 8.57 ± 0.39, respectively. Apoptotic cell distribution was 11.28 ± 5.25%, 60.58% ± 1.68% and 17.71 ± 8.34% in SG, T-CG and TG, respectively. In both parameters, the difference between SG and TG was insignificant (p > 0.05), the difference between T-CG/TG and SG/T-CG was significant (p < 0.05). CONCLUSION Exosomes obtained from AD-MSCs are effective in preventing testicular I/R injury. This effect appears to occur because of suppression of apoptotic activity.
Collapse
Affiliation(s)
- Fatma Bilgecan Şimşek
- Department of Pediatric Surgery, Faculty of Medicine, Manisa Celal Bayar University, Manisa, Türkiye
| | - Aydın Şencan
- Department of Pediatric Surgery, Faculty of Medicine, Manisa Celal Bayar University, Manisa, Türkiye.
| | - H Seda Vatansever
- Department of Histology-Embryology, Faculty of Medicine, Manisa Celal Bayar University, Manisa, Türkiye
| |
Collapse
|
18
|
Xie J, Hu Y, Li H, Wang Y, Fan X, Lu W, Liao R, Wang H, Cheng Y, Yang Y, Wang J, Liang S, Ma T, Su W. Targeted therapy for peri-prosthetic osteolysis using macrophage membrane-encapsulated human urine-derived stem cell extracellular vesicles. Acta Biomater 2023; 160:297-310. [PMID: 36773884 DOI: 10.1016/j.actbio.2023.02.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
Aseptic loosening of the prosthesis is a severe complication after joint replacement. It is of great practical significance and social value to discover the prevention and treatment strategies for this condition. Exosomes from urine-derived stem cells (Exos) have great potential in promoting bone repair, reconstruction, and regulating bone metabolism. However, they are easily eliminated by macrophages and incapable of targeting the osteolysis zone. In this study, based on macrophage "homing" into periprosthetic osteolysis region and cell membrane encapsulating nanotechnology, exosomes from urine-derived stem cells were encapsulated with macrophage membrane (MM) to prevent periprosthetic osteolysis. We found that macrophage membrane encapsulated urine-derived stem cell-derived exosomes (MM-Exos) can be targeted delivery to the osteolysis zone and enhance the therapeutic effectiveness of Exos, which alleviated wear particles-induced calvarial osteolysis. Furthermore, MM-Exos could provide immunological camouflage and allow the Exos to avoid phagocytosis by macrophages and stimulate cellular uptake by bone marrow-derived stem cells (BMSCs). Therefore, we demonstrated the unique ability of the macrophage membrane as a targeted transport of exosomes from urine-derived stem cells for the prevention and treatment of periprosthetic osteolysis. These biomimetic nanoparticles provided a new therapeutic exosome delivery system for preventing wear particles-induced osteolysis. STATEMENT OF SIGNIFICANCE: Macrophage membrane encapsulated urine-derived stem cell-derived exosomes (MM-Exos) can be targeted delivery to the osteolysis zone and enhance the therapeutic effect of Exos on peri‑prosthetic osteolysis prevention. MM-Exos could allow the Exos to avoid phagocytosis by macrophages and promote the uptake of Exos by BMSCs.
Collapse
Affiliation(s)
- Jie Xie
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Yihe Hu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, China; Department of Orthopedics, Xiangya Hospital, Central South University, China
| | - Hui Li
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Yinan Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, China
| | - Xiaolei Fan
- Department of Orthopedics, Xiangya Hospital, Central South University, China
| | - Wei Lu
- Department of Orthopedics, Xiangya Hospital, Central South University, China
| | - Runzhi Liao
- Department of Orthopedics, Xiangya Hospital, Central South University, China
| | - Haoyi Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, China
| | - Yurui Cheng
- Department of Orthopedics, Xiangya Hospital, Central South University, China
| | - Yute Yang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, China
| | - Jiahao Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, China
| | - Shuailong Liang
- Department of Orthopedics, Xiangya Hospital, Central South University, China
| | - Tianliang Ma
- Department of Orthopedics, Xiangya Hospital, Central South University, China.
| | - Weiping Su
- Department of Orthopedics, The 3rd Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
19
|
Guillot AJ, Martínez-Navarrete M, Garrigues TM, Melero A. Skin drug delivery using lipid vesicles: A starting guideline for their development. J Control Release 2023; 355:624-654. [PMID: 36775245 DOI: 10.1016/j.jconrel.2023.02.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/02/2023] [Accepted: 02/02/2023] [Indexed: 02/14/2023]
Abstract
Lipid vesicles can provide a cost-effective enhancement of skin drug absorption when vesicle production process is optimised. It is an important challenge to design the ideal vesicle, since their properties and features are related, as changes in one affect the others. Here, we review the main components, preparation and characterization methods commonly used, and the key properties that lead to highly efficient vesicles for transdermal drug delivery purposes. We stand by size, deformability degree and drug loading, as the most important vesicle features that determine the further transdermal drug absorption. The interest in this technology is increasing, as demonstrated by the exponential growth of publications on the topic. Although long-term preservation and scalability issues have limited the commercialization of lipid vesicle products, freeze-drying and modern escalation methods overcome these difficulties, thus predicting a higher use of these technologies in the market and clinical practice.
Collapse
Affiliation(s)
- Antonio José Guillot
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicente A. Estelles SN, Burjassot (Valencia), Spain
| | - Miquel Martínez-Navarrete
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicente A. Estelles SN, Burjassot (Valencia), Spain
| | - Teresa M Garrigues
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicente A. Estelles SN, Burjassot (Valencia), Spain
| | - Ana Melero
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Avda. Vicente A. Estelles SN, Burjassot (Valencia), Spain.
| |
Collapse
|
20
|
Lu W, Zhang J, Wu Y, Sun W, Jiang Z, Luo X. Engineered NF-κB siRNA-encapsulating exosomes as a modality for therapy of skin lesions. Front Immunol 2023; 14:1109381. [PMID: 36845116 PMCID: PMC9945116 DOI: 10.3389/fimmu.2023.1109381] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/10/2023] [Indexed: 02/10/2023] Open
Abstract
Introduction Despite the protection and management of skin has been paid more and more attention, effective countermeasures are still lacking for patients suffering from UV or chemotherapy with damaged skin. Recently, gene therapy by small interfering RNA (siRNA) has emerged as a new therapeutic strategy for skin lesions. However, siRNA therapy has not been applied to skin therapy due to lack of effective delivery vector. Methods Here, we develop a synthetic biology strategy that integrates the exosomes with artificial genetic circuits to reprogram the adipose mesenchymal stem cell to express and assemble siRNAs into exosomes and facilitate in vivo delivery siRNAs for therapy of mouse models of skin lesions. Results Particularly, siRNA enriched exosomes (si-ADMSC-EXOs) could be directly taken up by the skin cells to inhibit the expression of skin injury related genes. When mice with skin lesions were smeared with si-ADMSC-EXOs, the repair of lesioned skin became faster and the expression of inflammatory cytokines were decreased. Discussion Overall, this study establishes a feasible therapeutic strategy for skin injury, which may offer an alternative to conventional biological therapies requiring two or more independent compounds.
Collapse
Affiliation(s)
- Wei Lu
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, Zhejiang, China
| | - Jinzhong Zhang
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, Zhejiang, China
| | - Yungang Wu
- Department of the Orthopedics of Traditional Chinese Medicine (TCM), the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenxue Sun
- Hemodialysis Room, Department of Nephrology, the First Hospital Affiliated of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zipei Jiang
- Department of Ophthalmology, the First Hospital Affiliated of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xu Luo
- Wenzhou Medical University, Wenzhou, Zhejiang, China,Department of Wounds and Burns, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Zhejiang Engineering Research Center for Hospital Emergency and Process Digitization, Wenzhou, Zhejiang, China,*Correspondence: Xu Luo,
| |
Collapse
|
21
|
Rezayat F, Esmaeil N, Rezaei A. Potential Therapeutic Effects of Human Amniotic Epithelial Cells on Gynecological Disorders Leading to Infertility or Abortion. Stem Cell Rev Rep 2023; 19:368-381. [PMID: 36331801 DOI: 10.1007/s12015-022-10464-3] [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: 10/09/2022] [Indexed: 11/06/2022]
Abstract
The induction of feto-maternal tolerance, fetal non-immunogenicity, and the regulation of mother's immune system are essential variables in a successful pregnancy. Fetal membranes have been used as a source of stem cells and biological components in recent decades. Human amniotic epithelial cells (hAEC) have stem/progenitor characteristics like those found in the amniotic membrane. Based on their immunomodulatory capabilities, recent studies have focused on the experimental and therapeutic applications of hAECs in allograft transplantation, autoimmune disorders, and gynecological problems such as recurrent spontaneous abortion (RSA), recurrent implantation failure (RIF), and premature ovarian failure (POF). This review discusses some of the immunomodulatory features and therapeutic potential of hAECs in preventing infertility, miscarriage, and implantation failure by controlling the maternal immune system.
Collapse
Affiliation(s)
- Fatemeh Rezayat
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran. .,Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran. .,Department of Immunology, School of Medicine, Environment Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, 81744-176, Isfahan, Iran.
| | - Abbas Rezaei
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
22
|
Adipose-derived stem cells exosome and its potential applications in autologous fat grafting. J Plast Reconstr Aesthet Surg 2023; 76:219-229. [PMID: 36527904 DOI: 10.1016/j.bjps.2022.10.050] [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: 06/23/2022] [Accepted: 10/25/2022] [Indexed: 12/23/2022]
Abstract
Recently, there has been renewed interest in autologous fat grafting both for its filler and regenerative traits. The universal application, however, has been impeded by the unstable survival rates and complications. There has been substantial research undertaken on the role of adipose-derived stem cells (ADSCs) involved in fat graft fates including angiogenesis, adipogenesis, and inflammatory regulation. As the effectors of their parental cells, ADSC-derived exosomes (ADSC-exos) encapsulating multiple bioactive cargoes mediate cell-to-cell communication in a paracrine manner. ADSC-exos have received much attention for their biocompatible and efficient therapeutic potentials as "cell-free therapy" in plastic surgery, including increasing fat grafting survival rates. In this review, we summarize the current knowledge about the biological basis of ADSC-exos, ADSC-related mechanisms of fat survival, research updates of ADSC-exos in autologous fat grafting, and discuss some challenges along with research prospects.
Collapse
|
23
|
Jalan-Sakrikar N, Brevini T, Huebert RC, Sampaziotis F. Organoids and regenerative hepatology. Hepatology 2023; 77:305-322. [PMID: 35596930 PMCID: PMC9676408 DOI: 10.1002/hep.32583] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 02/03/2023]
Abstract
The burden of liver diseases is increasing worldwide, with liver transplantation remaining the only treatment option for end-stage liver disease. Regenerative medicine holds great potential as a therapeutic alternative, aiming to repair or replace damaged liver tissue with healthy functional cells. The properties of the cells used are critical for the efficacy of this approach. The advent of liver organoids has not only offered new insights into human physiology and pathophysiology, but also provided an optimal source of cells for regenerative medicine and translational applications. Here, we discuss various historical aspects of 3D organoid culture, how it has been applied to the hepatobiliary system, and how organoid technology intersects with the emerging global field of liver regenerative medicine. We outline the hepatocyte, cholangiocyte, and nonparenchymal organoids systems available and discuss their advantages and limitations for regenerative medicine as well as future directions.
Collapse
Affiliation(s)
- Nidhi Jalan-Sakrikar
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota, USA
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota, USA
| | - Teresa Brevini
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
| | - Robert C. Huebert
- Division of Gastroenterology and Hepatology, Mayo Clinic and Foundation, Rochester, Minnesota, USA
- Gastroenterology Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota, USA
| | - Fotios Sampaziotis
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- Cambridge Liver Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
24
|
Kim SD, Cho KS. Immunomodulatory Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles in Allergic Airway Disease. LIFE (BASEL, SWITZERLAND) 2022; 12:life12121994. [PMID: 36556359 PMCID: PMC9786036 DOI: 10.3390/life12121994] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022]
Abstract
Mesenchymal stem cells (MSCs) have been reported as promising candidates for the treatment of various diseases, especially allergic diseases, as they have the capacity to differentiate into various cells. However, MSCs itself have several limitations such as creating a risk of aneuploidy, difficulty in handling them, immune rejection, and tumorigenicity, so interest in the extracellular vesicles (EVs) released from MSCs are increasing, and many studies have been reported. Previous studies have shown that extracellular vesicles (EVs) produced by MSCs are as effective as the MSCs themselves in suppression of allergic airway inflammation through the suppression of Th2 cytokine production and the induction of regulatory T cells (Treg) expansion. EVs are one of the substances secreted by paracrine induction from MSCs, and because it exerts its effect by delivering contents such as mRNA, microRNA, and proteins to the receptor cell, it can reduce the problems or risks related to stem cell therapy. This article reviews the immunomodulatory properties of MSCs-derived EVs and their therapeutic implications for allergic airway disease.
Collapse
Affiliation(s)
- Sung-Dong Kim
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, 179 Gudeok-Ro, Seo-gu, Busan 602-739, Republic of Korea
| | - Kyu-Sup Cho
- Department of Otorhinolaryngology and Biomedical Research Institute, Pusan National University School of Medicine, Pusan National University Hospital, 179 Gudeok-Ro, Seo-gu, Busan 602-739, Republic of Korea
| |
Collapse
|
25
|
Roshandel E, Mehravar M, Nikoonezhad M, Alizadeh AM, Majidi M, Salimi M, Hajifathali A. Cell-Based Therapy Approaches in Treatment of Non-obstructive Azoospermia. Reprod Sci 2022; 30:1482-1494. [PMID: 36380137 PMCID: PMC9666961 DOI: 10.1007/s43032-022-01115-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022]
Abstract
The rate of infertility has globally increased in recent years for a variety of reasons. One of the main causes of infertility in men is azoospermia that is defined by the absence of sperm in the ejaculate and classified into two categories: obstructive azoospermia and non-obstructive azoospermia. In non-obstructive azoospermia, genital ducts are not obstructed, but the testicles do not produce sperm at all, due to various reasons. Non-obstructive azoospermia in most cases has no therapeutic options other than assisted reproductive techniques, which in most cases require sperm donors. Here we discuss cell-based therapy approaches to restore fertility in men with non-obstructive azoospermia including cell-based therapies of non-obstructive azoospermia using regenerative medicine and cell-based therapies of non-obstructive azoospermia by paracrine and anti-inflammatory pathway, technical and ethical challenges for using different cell sources and alternative options will be described, and then the more effectual approaches will be mentioned as future trends.
Collapse
Affiliation(s)
- Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985711151, Tehran, Iran
| | - Maryam Mehravar
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985711151, Tehran, Iran
| | - Maryam Nikoonezhad
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985711151, Tehran, Iran
| | - Afshin Mohammad Alizadeh
- Department of Internal Medicine, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Majidi
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Salimi
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985711151, Tehran, Iran
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, P.O. Box: 1985711151, Tehran, Iran
| |
Collapse
|
26
|
Miller CM, L Enninga EA, Rizzo SA, Phillipps J, Guerrero-Cazares H, Destephano CC, Peterson TE, Stalboerger PG, Behfar A, Khan Z. Platelet-derived exosomes induce cell proliferation and wound healing in human endometrial cells. Regen Med 2022; 17:805-817. [PMID: 36193669 DOI: 10.2217/rme-2022-0095] [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] [Indexed: 11/21/2022] Open
Abstract
Aim: To investigate the regenerative effects of a platelet-derived purified exosome product (PEP) on human endometrial cells. Materials & methods: Endometrial adenocarcinoma cells (HEC-1A), endometrial stromal cells (T HESC) and menstrual blood-derived stem cells (MenSC) were assessed for exosome absorption and subsequent changes in cell proliferation and wound healing properties over 48 h. Results: Cell proliferation increased in PEP treated T HESC (p < 0.0001) and MenSC (p < 0.001) after 6 h and in HEC-1A (p < 0.01) after 12 h. PEP improved wound healing after 6 h in HEC-1A (p < 0.01) and MenSC (p < 0.0001) and in T HESC between 24 and 36 h (p < 0.05). Conclusion: PEP was absorbed by three different endometrial cell types. PEP treatment increased cell proliferation and wound healing capacity.
Collapse
Affiliation(s)
- Colleen M Miller
- Division of Reproductive Endocrinology & Infertility, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Skylar A Rizzo
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.,Van Cleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN 55905, USA.,Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN 55905, USA
| | - Jordan Phillipps
- Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | - Timothy E Peterson
- Van Cleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Paul G Stalboerger
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Atta Behfar
- Department of Molecular Pharmacology & Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.,Van Cleve Cardiac Regenerative Medicine Program, Center for Regenerative Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Zaraq Khan
- Division of Reproductive Endocrinology & Infertility, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
27
|
Lee CH, Hunt D, Roth JG, Chiu CC, Suhar RA, LeSavage BL, Seymour AJ, Lindsay C, Krajina B, Chen YT, Chang KH, Hsieh IC, Chu PH, Wen MS, Heilshorn SC. Tuning pro-survival effects of human induced pluripotent stem cell-derived exosomes using elastin-like polypeptides. Biomaterials 2022; 291:121864. [DOI: 10.1016/j.biomaterials.2022.121864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/03/2022] [Accepted: 10/17/2022] [Indexed: 11/28/2022]
|
28
|
Zhang X, Wang D, Wang Z, Ling SKK, Yung PSH, Tuan RS, Ker DFE. Clinical perspectives for repairing rotator cuff injuries with multi-tissue regenerative approaches. J Orthop Translat 2022; 36:91-108. [PMID: 36090820 PMCID: PMC9428729 DOI: 10.1016/j.jot.2022.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/15/2022] [Accepted: 06/21/2022] [Indexed: 11/25/2022] Open
Abstract
Background In the musculoskeletal system, bone, tendon, and muscle form highly integrated multi-tissue units such as the rotator cuff complex, which facilitates functional and dynamic movement of the shoulder joint. Understanding the intricate interplay among these tissues within clinical, biological, and engineering contexts is vital for addressing challenging issues in treatment of musculoskeletal disorders and injuries. Methods A wide-ranging literature search was performed, and findings related to the socioeconomic impact of rotator cuff tears, the structure-function relationship of rotator cuff bone-tendon-muscle units, pathophysiology of injury, current clinical treatments, recent state-of-the-art advances (stem cells, growth factors, and exosomes) as well as their regulatory approval, and future strategies aimed at engineering bone-tendon-muscle musculoskeletal units are outlined. Results Rotator cuff injuries are a significant socioeconomic burden on numerous healthcare systems that may be addressed by treating the rotator cuff as a single complex, given its highly integrated structure-function relationship as well as degenerative pathophysiology and limited healing in bone-tendon-muscle musculoskeletal tissues. Current clinical practices for treating rotator cuff injuries, including the use of commercially available devices and evolving trends in surgical management have benefited patients while advances in application of stem/progenitor cells, growth factors, and exosomes hold clinical potential. However, such efforts do not emphasize targeted regeneration of bone-tendon-muscle units. Strategies aimed at regenerating bone-tendon-muscle units are thus expected to address challenging issues in rotator cuff repair. Conclusions The rotator cuff is a highly integrated complex of bone-tendon-muscle units that when injured, has severe consequences for patients and healthcare systems. State-of-the-art clinical treatment as well as recent advances have resulted in improved patient outcome and may be further enhanced by engineering bone-tendon-muscle multi-tissue grafts as a potential strategy for rotator cuff injuries. Translational Potential of this Article This review aims to bridge clinical, tissue engineering, and biological aspects of rotator cuff repair and propose a novel therapeutic strategy by targeted regeneration of multi-tissue units. The presentation of these wide-ranging and multi-disciplinary concepts are broadly applicable to regenerative medicine applications for musculoskeletal and non-musculoskeletal tissues.
Collapse
Affiliation(s)
- Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, Hong Kong
- School of Biomedical Sciences, Hong Kong
| | - Dan Wang
- Institute for Tissue Engineering and Regenerative Medicine, Hong Kong
- School of Biomedical Sciences, Hong Kong
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Hong Kong
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong
| | - Zuyong Wang
- College of Materials Science and Engineering, Hunan University, Changsha, China
| | - Samuel Ka-kin Ling
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong
| | - Patrick Shu-hang Yung
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong
| | - Rocky S. Tuan
- Institute for Tissue Engineering and Regenerative Medicine, Hong Kong
- School of Biomedical Sciences, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, Hong Kong
- School of Biomedical Sciences, Hong Kong
- Ministry of Education Key Laboratory for Regenerative Medicine, School of Biomedical Sciences, Hong Kong
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Hong Kong
| |
Collapse
|
29
|
Wang W, Chen D, Wang J, Wen L. Cellular Homeostasis and Repair in the Biliary Tree. Semin Liver Dis 2022; 42:271-282. [PMID: 35672015 DOI: 10.1055/a-1869-7714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
During biliary tree homeostasis, BECs are largely in a quiescent state and their turnover is slow for maintaining normal tissue homeostasis. BTSCs continually replenish new BECs in the luminal surface of EHBDs. In response to various types of biliary injuries, distinct cellular sources, including HPCs, BTSCs, hepatocytes, and BECs, repair or regenerate the injured bile duct. BEC, biliary epithelial cell; BTSC, biliary tree stem/progenitor cell; EHBD, extrahepatic bile ducts; HPC, hepatic progenitor cell.The biliary tree comprises intrahepatic bile ducts and extrahepatic bile ducts lined with epithelial cells known as biliary epithelial cells (BECs). BECs are a common target of various cholangiopathies for which there is an unmet therapeutic need in clinical hepatology. The repair and regeneration of biliary tissue may potentially restore the normal architecture and function of the biliary tree. Hence, the repair and regeneration process in detail, including the replication of existing BECs, expansion and differentiation of the hepatic progenitor cells and biliary tree stem/progenitor cells, and transdifferentiation of the hepatocytes, should be understood. In this paper, we review biliary tree homeostasis, repair, and regeneration and discuss the feasibility of regenerative therapy strategies for cholangiopathy treatment.
Collapse
Affiliation(s)
- Wei Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Dongfeng Chen
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Jun Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Liangzhi Wen
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
30
|
Zelli V, Compagnoni C, Capelli R, Corrente A, Di Vito Nolfi M, Zazzeroni F, Alesse E, Tessitore A. Role of exosomal microRNAs in cancer therapy and drug resistance mechanisms: focus on hepatocellular carcinoma. Front Oncol 2022; 12:940056. [PMID: 35912267 PMCID: PMC9334682 DOI: 10.3389/fonc.2022.940056] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), defined as intercellular messengers that carry their cargos between cells, are involved in several physiological and pathological processes. These small membranous vesicles are released by most cells and contain biological molecules, including nucleic acids, proteins and lipids, which can modulate signaling pathways of nearby or distant recipient cells. Exosomes, one the most characterized classes of EVs, include, among others, microRNAs (miRNAs), small non-coding RNAs able to regulate the expression of several genes at post-transcriptional level. In cancer, exosomal miRNAs have been shown to influence tumor behavior and reshape tumor microenvironment. Furthermore, their possible involvement in drug resistance mechanisms has become evident in recent years. Hepatocellular carcinoma (HCC) is the major type of liver cancer, accounting for 75-85% of all liver tumors. Although the improvement in HCC treatment approaches, low therapeutic efficacy in patients with intermediate-advanced HCC is mainly related to the development of tumor metastases, high risk of recurrence and drug resistance. Exosomes have been shown to be involved in pathogenesis and progression of HCC, as well as in drug resistance, by regulating processes such as cell proliferation, epithelial-mesenchymal transition and immune response. Herein, we summarize the current knowledge about the involvement of exosomal miRNAs in HCC therapy, highlighting their role as modulators of therapeutic response, particularly chemotherapy and immunotherapy, as well as possible therapeutic tools.
Collapse
Affiliation(s)
- Veronica Zelli
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
- Center for Molecular Diagnostics and Advanced Therapies, University of L’Aquila, L’Aquila, Italy
| | - Chiara Compagnoni
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Roberta Capelli
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Alessandra Corrente
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Mauro Di Vito Nolfi
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Francesca Zazzeroni
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Edoardo Alesse
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
| | - Alessandra Tessitore
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, L’Aquila, Italy
- Center for Molecular Diagnostics and Advanced Therapies, University of L’Aquila, L’Aquila, Italy
- *Correspondence: Alessandra Tessitore,
| |
Collapse
|
31
|
Wang Y, Cheng L, Zhao H, Li Z, Chen J, Cen Y, Zhang Z. The Therapeutic Role of ADSC-EVs in Skin Regeneration. Front Med (Lausanne) 2022; 9:858824. [PMID: 35755023 PMCID: PMC9218955 DOI: 10.3389/fmed.2022.858824] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/20/2022] [Indexed: 02/05/2023] Open
Abstract
Large skin defects caused by burns, unhealing chronic wounds, and trauma, are still an intractable problem for clinicians and researchers. Ideal skin regeneration includes several intricate and dynamic stages of wound repair and regeneration of skin physiological function. Adipose-derived stem cells (ADSCs), a type of mesenchymal stem cells (MSCs) with abundant resources and micro-invasive extraction protocols, have been reported to participate in each stage of promoting skin regeneration via paracrine effects. As essential products secreted by ADSCs, extracellular vesicles (EVs) derived from ADSCs (ADSC-EVs) inherit such therapeutic potential. However, ADSC-EVs showed much more clinical superiorities than parental cells. ADSC-EVs carry various mRNAs, non-coding RNAs, proteins, and lipids to regulate the activities of recipient cells and eventually accelerate skin regeneration. The beneficial role of ADSCs in wound repair has been widely accepted, while a deep comprehension of the mechanisms of ADSC-EVs in skin regeneration remains unclear. In this review, we provided a basic profile of ADSC-EVs. Moreover, we summarized the latest mechanisms of ADSC-EVs on skin regeneration from the aspects of inflammation, angiogenesis, cell proliferation, extracellular matrix (ECM) remodeling, autophagy, and oxidative stress. Hair follicle regeneration and skin barrier repair stimulated by ADSC-EVs were also reviewed. The challenges and prospects of ADSC-EVs-based therapies were discussed at the end of this review.
Collapse
Affiliation(s)
- Yixi Wang
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lihui Cheng
- Department of Central Sterile Supply, West China Hospital, Sichuan University, Chengdu, China
| | - Hanxing Zhao
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhengyong Li
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Junjie Chen
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ying Cen
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenyu Zhang
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
32
|
Joo SY, Cho YS, Lee SY, Seo CH. Regenerative effect of combined laser and human stem cell-conditioned medium therapy on hypertrophic burn scar. Burns 2022; 49:870-876. [PMID: 35842273 DOI: 10.1016/j.burns.2022.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 05/23/2022] [Accepted: 06/17/2022] [Indexed: 01/06/2023]
Abstract
PURPOSE This study aimed to determine the effect of combined treatment with non-ablative laser and human stem cell-conditioned medium (HSCM) on tissue regeneration after burn-induced hypertrophic scar (HTS) formation. METHODS Fourteen patients with HTSs on both sides of the same body part were subjected to three sessions of non-ablative laser treatment, with an interval of four weeks between each treatment. Following laser treatment, HSCM and normal saline were applied to the HTSs of the right (experimental) and left side (control), respectively. Over the next 6 days, HSCM and moisturizer were applied to experimental scars, while only moisturizer was applied to control scars. Skin characteristics were evaluated before laser treatment and on the seventh day after the third laser treatment. RESULTS No significant intergroup differences were noted in the initial evaluation (P > 0.05). We found significant differences between the pre- and post-treatment measurements of erythema (P < 0.001), trans-epidermal water loss (TEWL; P < 0.001), and Cutometer® parameters (all parameters; P <0.05) of experimental scars. Control scars also showed significant differences between pre- and post-treatment measurements of thickness (P = 0.01), erythema (P < 0.001), TEWL (P < 0.001), and Cutometer® parameters (all parameters; P < 0.05). Changes (pre- to post-treatment) in scar thickness between the experimental (-0.003 ± 0.09) and control scars (0.04 ± 0.12) were significant (P = 0.01). CONCLUSION These results suggest that HSCM has a positive effect on short-term results when applied after laser treatment of hypertrophic scars.
Collapse
Affiliation(s)
- So Young Joo
- Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, Seoul, the Republic of Korea
| | - Yoon Soo Cho
- Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, Seoul, the Republic of Korea
| | - Seung Yeol Lee
- Department of Physical Medicine and Rehabilitation, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, the Republic of Korea
| | - Cheong Hoon Seo
- Department of Rehabilitation Medicine, Hangang Sacred Heart Hospital, College of Medicine, Hallym University, Seoul, the Republic of Korea.
| |
Collapse
|
33
|
Bone marrow mesenchymal stem cells-derived exosomes containing miR-539-5p inhibit pyroptosis through NLRP3/caspase-1 signalling to alleviate inflammatory bowel disease. Inflamm Res 2022; 71:833-846. [PMID: 35637388 DOI: 10.1007/s00011-022-01577-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/22/2022] [Accepted: 04/14/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Exosomes derived from bone mesenchymal stem cells (BMSCs) are potential candidates for inflammatory bowel disease (IBD) treatment. The present study investigated the therapeutic effect and potential mechanism of BMSCs-derived exosomes on pyroptosis in IBD. METHODS We induced IBD in mice and cell models through dextran sulfate sodium (DSS) and LPS, respectively. The mRNA and protein expression levels were assessed by qRT-PCR, Western blotting, IF and IHC. The concentrations of IL-1β, IL-18 and TNFα were assessed using ELISA. ROS levels were determined using DCFH-DA staining. Cell proliferation of mIECs was analysed using an MTT assay. In addition, a flow cytometry assay was performed to detect pyroptosis. Finally, the binding relationship between miR-539-5p and NLRP3 was verified by a dual luciferase reporter gene assay. RESULTS Our results revealed that intraperitoneal injection of BMSCs-derived exosomes inhibited DSS-induced pyroptosis as well as IBD symptoms in mice. In addition, BMSCs-derived exosome treatment suppressed pyroptosis, ROS levels and the concentrations of proinflammatory cytokines (IL-1β, IL-18 and TNFα) in LPS-treated mIECs in a miR-539-5p-dependent manner. Further research found that miR-539-5p suppressed NLRP3 expression in mIECs by directly targeting NLRP3. As expected, pyroptosis in LPS-treated mIECs was significantly reduced by NLRP3 knockdown. In addition, NLRP3 silencing restored the inhibitory effect of exosomes derived from BMSCs transfected with miR-539-5p inhibitor on pyroptosis in LPS-treated mIECs. CONCLUSION The present study demonstrated that BMSCs-derived exosomal miR-539-5p suppresses pyroptosis through NLRP3/caspase-1 signalling to inhibit IBD progression.
Collapse
|
34
|
Rad LM, Yumashev AV, Hussen BM, Jamad HH, Ghafouri-Fard S, Taheri M, Rostami S, Niazi V, Hajiesmaeili M. Therapeutic Potential of Microvesicles in Cell Therapy and Regenerative Medicine of Ocular Diseases With an Especial Focus on Mesenchymal Stem Cells-Derived Microvesicles. Front Genet 2022; 13:847679. [PMID: 35422841 PMCID: PMC9001951 DOI: 10.3389/fgene.2022.847679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 02/28/2022] [Indexed: 12/13/2022] Open
Abstract
These days, mesenchymal stem cells (MSCs), because of immunomodulatory and pro-angiogenic abilities, are known as inevitable factors in regenerative medicine and cell therapy in different diseases such as ocular disorder. Moreover, researchers have indicated that exosome possess an essential potential in the therapeutic application of ocular disease. MSC-derived exosome (MSC-DE) have been identified as efficient as MSCs for treatment of eye injuries due to their small size and rapid diffusion all over the eye. MSC-DEs easily transfer their ingredients such as miRNAs, proteins, and cytokines to the inner layer in the eye and increase the reconstruction of the injured area. Furthermore, MSC-DEs deliver their immunomodulatory cargos in inflamed sites and inhibit immune cell migration, resulting in improvement of autoimmune uveitis. Interestingly, therapeutic effects were shown only in animal models that received MSC-DE. In this review, we summarized the therapeutic potential of MSCs and MSC-DE in cell therapy and regenerative medicine of ocular diseases.
Collapse
Affiliation(s)
- Lina Moallemi Rad
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Alexey V Yumashev
- Department of Prosthetic Dentistry, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq.,Center of Research and Strategic Studies, Lebanese French University, Kurdistan Region, Erbil, Iraq
| | - Hazha Hadayat Jamad
- Department of Biology, College of Education, Salahaddin University-Erbil, Kurdistan Region, Erbil, Iraq
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Samaneh Rostami
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciecnes, Zanjan, Iran
| | - Vahid Niazi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Hajiesmaeili
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Critical Care Quality Improvement Research Center, Loghman Hakin Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Zhu Q, Tang S, Zhu Y, Chen D, Huang J, Lin J. Exosomes Derived From CTF1-Modified Bone Marrow Stem Cells Promote Endometrial Regeneration and Restore Fertility. Front Bioeng Biotechnol 2022; 10:868734. [PMID: 35497344 PMCID: PMC9043110 DOI: 10.3389/fbioe.2022.868734] [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: 02/03/2022] [Accepted: 03/28/2022] [Indexed: 11/27/2022] Open
Abstract
Background: Thin endometrial tissue is a leading cause of embryo transfer failure, potentially contributing to sustained infertility and associated adverse outcomes. The application of exosomes derived from autologous or allogeneic bone marrow-derived stem cells (BMSCs) has been used to promote uterine repair following injury, and there is also prior evidence that stem cell transplantation can bolster fertility. Genetic modifications represent a primary approach to enhancing exosomal therapy strategies. The present study thus explored the effects of Cardiotrophin-1 (CTF1)-modified BMSCs-exo on fertility-related outcomes. Methods: An adenoviral vector was used to generate CTF1-overexpressing BMSCs (C-BMSCs), after which exosomes were isolated from control BMSCs (BMSC-exos) and C-BMSCs (C-BMSC-exos). The angiogenic effects of C-BMSC-exo treatment were assessed through analyses of endothelial cell proliferation and tube formation. Model rats exhibiting endometrial thinning were administered C-BMSCs-exo, after which the effects of such treatment were assessed through H&E staining, Masson’s trichrome staining, and immunofluorescence analyses. The mechanistic basis for the proangiogenic effects of CTF1 as a driver of endometrial regeneration was additionally explored. Results: C-BMSC-exo treatment of HUVECs was associated with enhanced neovascularization, as evidenced by improved in vitro proliferation, migration, and tube formation. Importantly, such treatment was also linked to tissue regeneration, neovascularization, and the suppression of localized tissue fibrosis in vivo. Regenerated endometrial tissue exhibited higher embryo receptivity and was associated with higher birth rates in treated rats. The upregulation of the JAK/PI3K/mTOR/STAT3 signaling pathways in C-BMSC-exo-treated rats may underscore the mechanistic basis whereby CTF1 can positively impact endometrial angiogenesis and regeneration. Conclusion: Our data suggest that exosomes produced by CTF1-modified BMSCs can more effectively promote the regeneration of endometrial and myometrial tissues, driving neovascularization in a manner that improves endometrial receptivity in a rat model system, highlighting the therapeutic promise of this approach for patients diagnosed with endometrial thinning.
Collapse
Affiliation(s)
- Qianqian Zhu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengluan Tang
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanwen Zhu
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Chen
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jialyu Huang
- Center for Reproductive Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang University School of Medicine, Nanchang, China
- *Correspondence: Jialyu Huang, ; Jiaying Lin,
| | - Jiaying Lin
- Department of Assisted Reproduction, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Jialyu Huang, ; Jiaying Lin,
| |
Collapse
|
36
|
Gholami L, Khorsandi K, Taghdiri Nooshabadi V, Shahabi S, Jazaeri M, Esfahani H, Rabiei Faradonbeh D, Veisi Malekshahi Z, Afsartala Z, Mostafa N. Effect of Photobiomodulation on Structure and Function of Extracellular Vesicle Secreted from Mesenchymal Stem Cells. Photochem Photobiol 2022; 98:1447-1458. [DOI: 10.1111/php.13633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 04/02/2022] [Accepted: 04/03/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Leila Gholami
- Department of periodontics, Dental Research Center Hamadan University of Medical Sciences Hamadan Iran
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry University of British Columbia Canada
| | - Khatereh Khorsandi
- Department of Photodynamic, Medical Laser Research Center Yara Institute ACECR Tehran Iran
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences The George Washington University Washington DC 20037 USA
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine Semnan University of Medical Science Iran
| | - Shiva Shahabi
- Student Research Committee, School of Dentistry Hamadan University of Medical Sciences Iran
| | - Marzieh Jazaeri
- Student Research Committee, School of Dentistry Hamadan University of Medical Sciences Iran
| | - HomaSadat Esfahani
- Department of Photodynamic, Medical Laser Research Center Yara Institute ACECR Tehran Iran
| | - Davood Rabiei Faradonbeh
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine Tehran University of Medical Sciences Tehran Iran
| | - Ziba Veisi Malekshahi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine Tehran University of Medical Sciences Tehran Iran
| | - Zohreh Afsartala
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute Tehran University of Medical Science Tehran Iran
| | - Nesrine Mostafa
- Department of Oral Health Sciences, Faculty of Dentistry University of British Columbia Canada
| |
Collapse
|
37
|
Application of exosomes in the diagnosis and treatment of pancreatic diseases. Stem Cell Res Ther 2022; 13:153. [PMID: 35395948 PMCID: PMC8994331 DOI: 10.1186/s13287-022-02826-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/22/2022] [Indexed: 12/20/2022] Open
Abstract
Pancreatic diseases, a serious threat to human health, have garnered considerable research interest, as they are associated with a high mortality rate. However, owing to the uncertain etiology and complex pathophysiology, the treatment of pancreatic diseases is a challenge for clinicians and researchers. Exosomes, carriers of intercellular communication signals, play an important role in the diagnosis and treatment of pancreatic diseases. Exosomes are involved in multiple stages of pancreatic disease development, including apoptosis, immune regulation, angiogenesis, cell migration, and cell proliferation. Thus, extensive alterations in the quantity and variety of exosomes may be indicative of abnormal biological behaviors of pancreatic cells. This phenomenon could be exploited for the development of exosomes as a new biomarker or target of new treatment strategies. Several studies have demonstrated the diagnostic and therapeutic effects of exosomes in cancer and inflammatory pancreatic diseases. Herein, we introduce the roles of exosomes in the diagnosis and treatment of pancreatic diseases and discuss directions for future research and perspectives of their applications.
Collapse
|
38
|
Kim D, Kim S, Sung A, Patel N, Wong N, Conboy MJ, Conboy IM. Autologous treatment for ALS with implication for broad neuroprotection. Transl Neurodegener 2022; 11:16. [PMID: 35272709 PMCID: PMC8915496 DOI: 10.1186/s40035-022-00290-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 02/18/2022] [Indexed: 01/20/2023] Open
Abstract
Background Amyotrophic lateral sclerosis (ALS) is characterized by a progressive loss of motor neurons (MNs), leading to paralysis, respiratory failure and death within 2–5 years of diagnosis. The exact mechanisms of sporadic ALS, which comprises 90% of all cases, remain unknown. In familial ALS, mutations in superoxide dismutase (SOD1) cause 10% of cases. Methods ALS patient-derived human-induced pluripotent stem cells (ALS hiPSCs, harboring the SOD1AV4 mutation), were differentiated to MNs (ALS-MNs). The neuroprotective effects of conditioned medium (CM) of hESCs (H9), wt hiPSCs (WTC-11) and the ALS iPSCs, on MN apoptosis and viability, formation and maintenance of neurites, mitochondrial activity and expression of inflammatory genes, were examined. For in vivo studies, 200 μl of CM from the ALS iPSCs (CS07 and CS053) was injected subcutaneously into the ALS model mice (transgenic for the human SOD1G93A mutation). Animal agility and strength, muscle innervation and mass, neurological score, onset of paralysis and lifespan of the ALS mice were assayed. After observing significant disease-modifying effects, the CM was characterized biochemically by fractionation, comparative proteomics, and epigenetic screens for the dependence on pluripotency. CM of fibroblasts that were differentiated from the wt hiPSCs lacked any neuroprotective activity and was used as a negative control throughout the studies. Results The secretome of PSCs including the ALS patient iPSCs was neuroprotective in the H2O2 model. In the model with pathogenic SOD1 mutation, ALS iPSC-CM attenuated all examined hallmarks of ALS pathology, rescued human ALS-MNs from denervation and death, restored mitochondrial health, and reduced the expression of inflammatory genes. The ALS iPSC-CM also improved neuro-muscular health and function, and delayed paralysis and morbidity in ALS mice. Compared side by side, cyclosporine (CsA), a mitochondrial membrane blocker that prevents the leakage of mitochondrial DNA, failed to avert the death of ALS-MNs, although CsA and ALS iPSC-CM equally stabilized MN mitochondria and attenuated inflammatory genes. Biochemical characterization, comparative proteomics, and epigenetic screen all suggested that it was the interactome of several key proteins from different fractions of PSC-CM that delivered the multifaceted neuroprotection. Conclusions This work introduces and mechanistically characterizes a new biologic for treating ALS and other complex neurodegenerative diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s40035-022-00290-5.
Collapse
Affiliation(s)
- Daehwan Kim
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Subin Kim
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Ashley Sung
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Neetika Patel
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Nathan Wong
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Michael J Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA
| | - Irina M Conboy
- Department of Bioengineering and QB3 Institute, University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
39
|
Zhao AG, Shah K, Cromer B, Sumer H. Comparative analysis of extracellular vesicles isolated from human mesenchymal stem cells by different isolation methods and visualisation of their uptake. Exp Cell Res 2022; 414:113097. [PMID: 35276207 DOI: 10.1016/j.yexcr.2022.113097] [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: 11/08/2021] [Revised: 02/09/2022] [Accepted: 03/07/2022] [Indexed: 12/15/2022]
Abstract
Various types of cells secrete extracellular vesicle (EVs) which contain proteins, lipids and nucleic acids and play important roles in inter-cellular signalling and pathological processes to impact the recipient cells. EVs have demonstrated their potential as biomarkers for disease and as therapeutic agents in regenerative medicine. In recent times, EVs derived from mesenchymal stem cells (MSCs), which are widely used as a promising medicinal product in many clinical applications, are being tested in many preclinical trials. However, the lack of standardization of MSC-derived EV isolation and analysis methods, restricts the utility of MSC-derived EVs in the clinical setting. Here, we focused on optimising the isolation method for EVs derived from MSCs. Four samples of EVs were isolated from human adipose derived MSC culture medium by differential ultracentrifugation with three different ultracentrifuge durations to investigate the influence of ultracentrifuge time on quality and quantity of MSC-derived EVs. Additionally, we used a commercial kit to extract EVs from MSC cultured medium and compared it with the ultracentrifugation method. The EV samples were then characterised for particle concentration, protein concentration, size distribution and the presence of known EV protein markers, by western blot and flow cytometry. A comparison of these results for the five samples demonstrated that 1 h of differential ultracentrifugation was optimal to isolate high quality and quantity of MSC-derived EVs from MSC cultured medium. Additionally, fluorescence imaging of the freshly isolated vs frozen EVs showed that freshly isolated EVs are taken up by cells more efficiently than frozen EVs. These finding establish a simple and reliable method of EV isolation from MSCs.
Collapse
Affiliation(s)
- Ashley G Zhao
- Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology, John St, Hawthorn, VIC, 3122, Australia
| | - Kiran Shah
- Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology, John St, Hawthorn, VIC, 3122, Australia; Australian Veterinary Stem Cells and Magellan Stem Cells P/L, 116-118 Thames St, Box Hill, VIC, 3129, Australia
| | - Brett Cromer
- Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology, John St, Hawthorn, VIC, 3122, Australia
| | - Huseyin Sumer
- Department of Chemistry and Biotechnology, Faculty of Science, Engineering and Technology, Swinburne University of Technology, John St, Hawthorn, VIC, 3122, Australia.
| |
Collapse
|
40
|
Exosomes in the Pathogenesis, Progression, and Treatment of Osteoarthritis. Bioengineering (Basel) 2022; 9:bioengineering9030099. [PMID: 35324788 PMCID: PMC8945849 DOI: 10.3390/bioengineering9030099] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 12/18/2022] Open
Abstract
Osteoarthritis (OA) is a prevalent and debilitating age-related joint disease characterized by articular cartilage degeneration, synovial membrane inflammation, osteophyte formation, as well as subchondral bone sclerosis. OA drugs at present are mainly palliative and do not halt or reverse disease progression. Currently, no disease-modifying OA drugs (DMOADs) are available and total joint arthroplasty remains a last resort. Therefore, there is an urgent need for the development of efficacious treatments for OA management. Among all novel pharmaco-therapeutical options, exosome-based therapeutic strategies are highly promising. Exosome cargoes, which include proteins, lipids, cytokines, and various RNA subtypes, are potentially capable of regulating intercellular communications and gene expression in target cells and tissues involved in OA development. With extensive research in recent years, exosomes in OA studies are no longer limited to classic, mesenchymal stem cell (MSC)-derived vesicles. New origins, structures, and functions of exosomes are constantly being discovered and investigated. This review systematically summarizes the non-classic origins, biosynthesis, and extraction of exosomes, describes modification and delivery techniques, explores their role in OA pathogenesis and progression, and discusses their therapeutic potential and hurdles to overcome in OA treatment.
Collapse
|
41
|
Keshtkar S, Kaviani M, Soleimanian S, Azarpira N, Asvar Z, Pakbaz S. Stem Cell-Derived Exosome as Potential Therapeutics for Microbial Diseases. Front Microbiol 2022; 12:786111. [PMID: 35237239 PMCID: PMC8882917 DOI: 10.3389/fmicb.2021.786111] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes, as the smallest extracellular vesicles that carry a cargo of nucleic acids, lipids, and proteins and mediate intercellular communication, have attracted much attention in diagnosis and treatment in the field of medicine. The contents of exosomes vary depending on the cell type and physiological conditions. Among exosomes derived from several cell types, stem cell-derived exosomes (stem cell-Exo) are increasingly being explored due to their immunomodulatory properties, regenerative capacity, anti-inflammatory and anti-microbial functions. Administration of stem cell-Exo, as a cell-free therapy for various diseases, has gained great promise. Indeed, the advantages of exosomes secreted from stem cells outweigh those of their parent cells owing to their small size, high stability, less immunogenicity, no risk of tumorigenesis, and easier condition for storage. Recently, the use of stem cell-Exo has been proposed in the field of microbial diseases. Pathogens including bacteria, viruses, fungi, and parasites can cause various diseases in humans with acute and chronic complications, sometimes resulting in mortality. On the other hand, treatments based on antibiotics and other chemical compounds have many side effects and the strains become resistant to drugs in some cases. Hence, this review aimed to highlight the effect of stem cell-derived extracellular vesicles including stem cell-Exo on microbial diseases. Although most published studies are preclinical, the avenue of clinical application of stem cell-Exo is under way to reach clinical applications. The challenges ahead of this cell-free treatment that might be applied as a therapeutic alternative to stem cells for translation from bench to bed were emphasized, as well.
Collapse
Affiliation(s)
- Somayeh Keshtkar
- Molecular Dermatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maryam Kaviani
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeede Soleimanian
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Asvar
- Nanotechnology School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sara Pakbaz
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Pathology, University Health Network, Toronto, ON, Canada
| |
Collapse
|
42
|
Zhou Q, Cheng Y, Sun F, Shen J, Nasser MI, Zhu P, Zhang X, Li Y, Yin G, Wang Y, Wu X, Zhao M. A Comprehensive Review of the Therapeutic Value of Urine-Derived Stem Cells. Front Genet 2022; 12:781597. [PMID: 35047009 PMCID: PMC8762167 DOI: 10.3389/fgene.2021.781597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/30/2021] [Indexed: 12/17/2022] Open
Abstract
Stem cells possess regenerative powers and multidirectional differentiation potential and play an important role in disease treatment and basic medical research. Urine-derived stem cells (USCs) represent a newly discovered type of stem cell with biological characteristics similar to those of mesenchymal stromal cells (MSCs), including their doubling time and immunophenotype. USCs are noninvasive and can be readily obtained from voided urine and steadily cultured. Based on advances in this field, USCs and their secretions have increasingly emerged as ideal sources. USCs may play regulatory roles in the cellular immune system, oxidative stress, revascularization, apoptosis and autophagy. This review summarizes the applications of USCs in tissue regeneration and various disease treatments. Furthermore, by analysing their limitations, we anticipate the development of more feasible therapeutic strategies to promote USC-based individualized treatment.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yiyu Cheng
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Fang Sun
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jie Shen
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - M I Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xueyan Zhang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuxiang Li
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Guangming Yin
- Department of Urology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yuequn Wang
- The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiushan Wu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,The Center for Heart Development, State Key Laboratory of Development Biology of Freshwater Fish, Key Laboratory of MOE for Development Biology and Protein Chemistry, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Mingyi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
43
|
Medenica S, Abazovic D, Ljubić A, Vukovic J, Begovic A, Cucinella G, Zaami S, Gullo G. The Role of Cell and Gene Therapies in the Treatment of Infertility in Patients with Thyroid Autoimmunity. Int J Endocrinol 2022; 2022:4842316. [PMID: 36081621 PMCID: PMC9448571 DOI: 10.1155/2022/4842316] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/03/2022] [Indexed: 12/02/2022] Open
Abstract
There is a rising incidence of infertility worldwide, and many couples experience difficulties conceiving nowadays. Thyroid autoimmunity (TAI) is recognized as one of the major female infertility causes related to a diminished ovarian reserve and potentially impaired oocyte maturation and embryo development, causing adverse pregnancy outcomes. Growing evidence has highlighted its impact on spontaneously achieved pregnancy and pregnancy achieved by in vitro fertilization. Despite the influence of thyroid hormones on the male reproductive system, there is insufficient data on the association between TAI and male infertility. In past years, significant progress has been achieved in cell and gene therapies as emerging treatment options for infertility. Cell therapies utilize living cells to restore healthy tissue microenvironment and homeostasis and usually involve platelet-rich plasma and various stem cells. Using stem cells as therapeutic agents has many advantages, including simple sampling, abundant sources, poor immunogenicity, and elimination of ethical concerns. Mesenchymal Stem Cells (MSCs) represent a heterogeneous fraction of self-renewal, multipotent non-hematopoietic stem cells that display profound immunomodulatory and immunosuppressive features and promising therapeutic effects. Infertility has a genetic component in about half of all cases, although most of its genetic causes are still unknown. Hence, it is essential to identify genes involved in meiosis, DNA repair, ovarian development, steroidogenesis, and folliculogenesis, as well as those involved in spermatogenesis in order to develop potential gene therapies for infertility. Despite advances in therapy approaches such as biological agents, autoimmune disorders remain impossible to cure. Recent research demonstrates the remarkable therapeutic effectiveness of MSCs in a wide array of autoimmune diseases. TAI is one of many autoimmune disorders that can benefit from the use of MSCs, which can be derived from bone marrow and adipose tissue. Cell and gene therapies hold great potential for treating autoimmune conditions, although further research is still needed.
Collapse
Affiliation(s)
- Sanja Medenica
- Department of Endocrinology, Internal Medicine Clinic, Clinical Center of Montenegro, School of Medicine, University of Montenegro, Podgorica, Montenegro
| | | | - Aleksandar Ljubić
- Biocell Hospital, Belgrade, Serbia
- Special Gynecology Hospital with Maternity Ward Jevremova, Belgrade, Serbia
- Libertas International University, Dubrovnik, Croatia
| | | | | | - Gaspare Cucinella
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF UNIT, University of Palermo, Palermo, Italy
| | - Simona Zaami
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, “Sapienza” University of Rome, Rome, Italy
| | - Giuseppe Gullo
- Department of Obstetrics and Gynecology, Villa Sofia Cervello Hospital, IVF UNIT, University of Palermo, Palermo, Italy
| |
Collapse
|
44
|
Intranasally Administered Extracellular Vesicles from Adipose Stem Cells Have Immunomodulatory Effects in a Mouse Model of Asthma. Stem Cells Int 2021; 2021:6686625. [PMID: 34899920 PMCID: PMC8664544 DOI: 10.1155/2021/6686625] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 06/02/2021] [Accepted: 07/14/2021] [Indexed: 12/27/2022] Open
Abstract
Asthma is a chronic eosinophilic airway disease characterized by type 2 helper T cell-driven inflammation. Adipose stem cells (ASCs) and the ASC culture supernatant are known to improve allergic airway inflammation; however, the immunomodulatory effects of ASC-derived extracellular vesicles (EVs) on allergic airway diseases remain unclear. Thus, we assessed the effects of ASC-derived EVs on allergic airway inflammation in a mouse model of asthma. EVs were isolated from the culture supernatant of murine ASCs and characterized. Six-week-old female C57BL/6 mice were sensitized to ovalbumin (OVA) by intraperitoneal injection and challenged intranasally with OVA. Before the OVA challenge, 10 μg/50 μl of ASC-derived EVs was administered intranasally to the experimental group. ASC-derived EVs significantly attenuated airway hyperresponsiveness (AHR) in asthmatic mice (p = 0.023). ASC-derived EVs resulted in a remarkable reduction of the total number of inflammatory cells (p = 0.005) and eosinophils (p = 0.023) in the bronchoalveolar lavage fluid (BALF), the degree of eosinophilic lung inflammation (p < 0.001), and the serum total and OVA-specific immunoglobulin (Ig)E (p = 0.048 and p = 0.001) and total IgG1 (p < 0.001). Interleukin- (IL-) 4 was significantly inhibited with ASC-derived EV pretreatment in the BALF and lung draining lymph nodes (LLNs) (p = 0.040 and p = 0.011). Furthermore, ASC-derived EV administration resulted in a significant increase of the regulatory T cell (Treg) populations in LLNs. ASC-derived EVs alleviated AHR and allergic airway inflammation caused by the induction of Treg expansion in a mouse model of asthma. There seems to be a role for ASC-derived EVs as a modifier in allergic airway disease.
Collapse
|
45
|
Kos IA, Thurner L, Bittenbring JT, Christofyllakis K, Kaddu-Mulindwa D. Advances in Lymphoma Molecular Diagnostics. Diagnostics (Basel) 2021; 11:diagnostics11122174. [PMID: 34943410 PMCID: PMC8699850 DOI: 10.3390/diagnostics11122174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022] Open
Abstract
Lymphomas encompass a diverse group of malignant lymphoid neoplasms. Over recent years much scientific effort has been undertaken to identify and understand molecular changes in lymphomas, resulting in a wide range of genetic alterations that have been reported across all types of lymphomas. As many of these changes are now incorporated into the World Health Organization’s defined criteria for the diagnostic evaluation of patients with lymphoid neoplasms, their accurate identification is crucial. Even if many alterations are not routinely evaluated in daily clinical practice, they may still have implications in risk stratification, treatment, prognosis or disease monitoring. Moreover, some alterations can be used for targeted treatment. Therefore, these advances in lymphoma molecular diagnostics in some cases have led to changes in treatment algorithms. Here, we give an overview of and discuss advances in molecular techniques in current clinical practice, as well as highlight some of them in a clinical context.
Collapse
|
46
|
Exosome-Induced Vaginal Tissue Regeneration in a Porcine Mesh Exposure Model. Female Pelvic Med Reconstr Surg 2021; 27:609-615. [PMID: 34554143 DOI: 10.1097/spv.0000000000001005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
OBJECTIVES The purpose of this study was to explore the utility of an injectable purified exosome product derived from human apheresis blood to (1) augment surgical closure of vaginal mesh exposures, and (2) serve as a stand-alone therapy for vaginal mesh exposure. METHODS Sixteen polypropylene meshes (1×1-3×3 cm) were implanted in the vaginas of 7 Yorkshire-crossed pigs by urogynecologic surgeons (day 0). On day 7, group 1 underwent surgical intervention via vaginal tissue suture reclosure with (n=2 pigs, n=4 meshes) or without (n=2 pigs, n=4 meshes) exosome injection; group 2 underwent medical intervention with an exosome injection (n=3, n=8 meshes). One animal in group 2 was given oral 2'-deoxy-5-ethynyluridine to track cellular regeneration. Euthansia occurred at 5 weeks. RESULTS Mesh exposures treated with surgical closure alone experienced reexposure of the mesh. Exosome treatment with or without surgical closure resulted in partial to full mesh exposure resolution up to 3×3 cm. Exosome-treated tissues had significantly thicker regenerated epithelial tissue (208 μm exosomes-only and 217 μm surgery+exosomes, versus 80 μm for surgery-only; P < 0.05); evaluation of 2'-deoxy-5-ethynyluridine confirmed de novo regeneration throughout the epithelium and underlying tissues. Capillary density was significantly higher in the surgery+exosomes group (P = 0.03). Surgery-only tissues had a higher inflammatory and fibrosis response as compared with exosome-treated tissues. CONCLUSIONS In this pilot study, exosome treatment augmented healing in the setting of vaginal mesh exposure, reducing the incidence of mesh reexposure after suture closure and decreasing the area of mesh exposure through de novo tissue regeneration after exosome injection only. Further study of varied local tissue conditions and mesh configurations is warranted.
Collapse
|
47
|
Mesenchymal Stem Cells in the Treatment of COVID-19, a Promising Future. Cells 2021; 10:cells10102588. [PMID: 34685567 PMCID: PMC8533906 DOI: 10.3390/cells10102588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/11/2021] [Accepted: 09/17/2021] [Indexed: 12/20/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stem cells present in virtually all tissues; they have a potent self-renewal capacity and can differentiate into multiple cell types. They also affect the ambient tissue by the paracrine secretion of numerous factors in vivo, including the induction of other stem cells’ differentiation. In vitro, the culture media supernatant is named secretome and contains soluble molecules and extracellular vesicles that retain potent biological function in tissue regeneration. MSCs are considered safe for human treatment; their use does not involve ethical issues, as embryonic stem cells do not require genetic manipulation as induced pluripotent stem cells, and after intravenous injection, they are mainly found in the lugs. Therefore, these cells are currently being tested in various preclinical and clinical trials for several diseases, including COVID-19. Several affected COVID-19 patients develop induced acute respiratory distress syndrome (ARDS) associated with an uncontrolled inflammatory response. This condition causes extensive damage to the lungs and may leave serious post-COVID-19 sequelae. As the disease may cause systemic alterations, such as thromboembolism and compromised renal and cardiac function, the intravenous injection of MSCs may be a therapeutic alternative against multiple pathological manifestations. In this work, we reviewed the literature about MSCs biology, focusing on their function in pulmonary regeneration and their use in COVID-19 treatment.
Collapse
|
48
|
Tiwari A, Singh A, Verma S, Stephenson S, Bhowmick T, Sangwan VS. Mini Review: Current Trends and Understanding of Exosome Therapeutic Potential in Corneal Diseases. Front Pharmacol 2021; 12:684712. [PMID: 34489693 PMCID: PMC8417240 DOI: 10.3389/fphar.2021.684712] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are a subset of extracellular vesicles (EVs) that are secreted by most cell types. They are nanosized EVs ranging from 30 to 150 nm. The membrane-enclosed bodies originate by the process of endocytosis and mainly comprise DNA, RNA, protein, and lipids. Exosomes not only act as cell-to-cell communication signaling mediators but also have the potential to act as biomarkers for clinical application and as a promising carrier for drug delivery. Unfortunately, the purification methods for exosomes remain an obstacle. While most of the exosome researches are mainly focused on cancer, there are limited studies highlighting the importance of exosomes in ocular biology, specifically cornea-associated pathologies. Here, we summarize a brief description of exosome biogenesis, roles of exosomes and exosome-based therapies in corneal pathologies, and exosome bioengineering for tissue-specific therapy.
Collapse
Affiliation(s)
- Anil Tiwari
- Department of Cornea and Uveitis, Dr. Shroff's Charity Eye Hospital, New Delhi, India
| | - Aastha Singh
- Department of Cornea and Uveitis, Dr. Shroff's Charity Eye Hospital, New Delhi, India
| | - Sudhir Verma
- Department of Zoology, Deen Dayal Upadhyaya College (University of Delhi), New Delhi, India
| | - Sarah Stephenson
- Pandorum Technologies Ltd., Bangalore Bioinnovation Centre, Bangalore, India.,Department of Surgery/Division of Transplant Surgery, The Medical University of South Carolina, Charleston, SC, United States
| | - Tuhin Bhowmick
- Pandorum Technologies Ltd., Bangalore Bioinnovation Centre, Bangalore, India
| | | |
Collapse
|
49
|
Yuan R, Dai X, Li Y, Li C, Liu L. Exosomes from miR-29a-modified adipose-derived mesenchymal stem cells reduce excessive scar formation by inhibiting TGF-β2/Smad3 signaling. Mol Med Rep 2021; 24:758. [PMID: 34476508 PMCID: PMC8436211 DOI: 10.3892/mmr.2021.12398] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 06/02/2021] [Indexed: 12/31/2022] Open
Abstract
Pathological scars mainly refer to hypertrophic scars and keloids, and have a high incidence. Moreover, these scars seriously affect the patient's appearance and are associated with significant pain. The present study aimed to investigate the inhibitory effect of microRNA (miR)-29a from human adipose-derived mesenchymal stem cells (hADSCs) exosomes on scar formation. Firstly, the expression of miR-29a in thermal skin tissues of mice and human hypertrophic scar fibroblasts (HSFBs) was detected via reverse transcription-quantitative PCR. Exosomes derived from miR-29a-modified hADSCs were extracted and the influence of miR-29a-modified hADSCs-exo on the proliferation and function of HSFBs was determined. Lastly, the effect of miR-29a-modified hADSCs-exo on scar formation was determined using a thermal mouse model. The results demonstrated that miR-29a was downregulated in scar tissues after scalding and in HSFBs. After treating HSFBs with miR-29a-modified hADSC exosomes, miR-29a-overexpressing hADSC exosomes inhibited the proliferation and migration of HSFBs. Moreover, it was found that TGF-β2 was the target of miR-29a, and that hADSC exosome-derived miR-29a inhibited the fibrosis of HSFBs and scar hyperplasia after scalding in mice by targeting the TGF-β2/Smad3 signaling pathway. In summary, the current data indicated that miR-29a-modified hADSC exosome therapy can decrease scar formation by inhibiting the TGF-β2/Smad3 signaling pathway via its derived exogenous miR-29a, and this may be useful for the future treatment of pathological scars by providing a potential molecular basis.
Collapse
Affiliation(s)
- Ruihong Yuan
- Department of Plastic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Xiaoming Dai
- Department of Plastic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yisong Li
- Department of Plastic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Chunshan Li
- Department of Plastic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Liu Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
50
|
Sahu R, Sahu SK, Nishank SS. de novo transcriptome profile of two earthworms Lampito mauritii and Drawida calebi during regeneration. Biochem Biophys Rep 2021; 27:101092. [PMID: 34409173 PMCID: PMC8361223 DOI: 10.1016/j.bbrep.2021.101092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 11/26/2022] Open
Abstract
Earthworms have remarkable ability to regenerate its tail and head region. However the list of genes expressed in this regeneration process has been less explored baring a few species. The current study involves the de novo transcriptome sequencing of intact tail and regenerating tail (15 day post amputation) of earthworms belonging to two different genera Lampito mauritii (Kinberg, 1867) and Drawida calebi (Gates, 1945). This study contains one de-novo and one reference based transcriptome analysis each from one genus of two earthworm genera. From a total of 119.92 million (150 × 2) reads, 112.95 million high quality adapter free reads were utilized in analysis. Assembly of high-quality reads was performed separately for Lampito mauritii (LM sample) and Drawida calebi (DC sample) that resulted in 66368 and 1,61,289 transcripts respectively. About 25.21% of transcripts were functionally annotated for DC sample and 38.27% for LM samples against Annelida sequences. A total of 239 genes were expressed exclusively in regenerated tissue compared to intact sample in DC whereas about 241 genes were exclusively expressed in regenerated tissue of LM compared to its intact sample. Majority of genes in Drawida and Lampito were dedicated to immune response, maintenance of cytoskeleton, resisting oxidative stress and promoting neuronal regeneration for cell-cell communication during tail regeneration. Upregulation of genes such as beta catenin, Sox, notch, FGF, frizzled. Similarity with annelid worm Capitella telesta.
Collapse
Affiliation(s)
- Ranjan Sahu
- Post Graduate Department of Zoology, Utkal University, Vani Vihar, Bhubaneswar, 751004, Odisha, India
| | - Sanjat Kumar Sahu
- Dept. of Environment Science, Sambalpur University, Jyoti Vihar, Burla, 768019, Odisha, India
| | - Sudhansu Sekhar Nishank
- Post Graduate Department of Zoology, Utkal University, Vani Vihar, Bhubaneswar, 751004, Odisha, India
| |
Collapse
|