1
|
Zhang Y, Xia Y, Zhang R, Zhou X, Jiang J. Urine-Derived Stem Cells Reverse Bleomycin‑Induced Experimental Pulmonary Fibrosis by Inhibition of the TGF-β1-Smad2/3 Pathway. Cytotherapy 2024; 26:1236-1244. [PMID: 38852093 DOI: 10.1016/j.jcyt.2024.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 05/07/2024] [Accepted: 05/14/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is characterized by progressive lung interstitial lesions with the disease pathophysiology incompletely understood, which is a serious and fatal disorder with limited treatment options. Mesenchymal stem cells (MSCs) have exhibited promising therapeutic capability for IPF. While most types of MSCs are obtained invasively, urine-derived stem cells (USCs) can be gained in a safe, noninvasive, and inexpensive procedure, which are readily available and reported to exhibit no risk of teratoma formation or oncogenic potential in vivo, sounding alternative to other MSCs. This study aims to investigate the therapeutic effect and mechanism of USCs on IPF, using a bleomycin (BLM)-induced IPF model in mice. METHODS Cell surface marker examination by flow cytometry analysis and cell differentiation culture were used to characterize USCs obtained from healthy individuals. BLM was instilled endotracheally in adult C57BL/6 mice, followed by USCs or human bone marrow-derived mesenchymal stem cells (BMSCs) treatment by tail vein injection on day 14. Mice were euthanized on day 14 before administration or day 21 for the evaluation of pulmonary histopathology and hydroxyproline (HYP) content. Inflammatory factors of the lung, including transforming growth factor (TGF)-β1, TNF-α, IL-6, MMP2 were analyzed by quantitative real-time PCR (qRT-PCR). Additionally, immunohistochemistry (IHC) and western blotting (WB) were applied to evaluate the expression of α-SMA and activation of TGF-β1-Smad2/3 in lung. RESULTS USCs highly expressed CD29 and CD90, showing negative expression of hematopoietic stem cell markers (CD45, CD34) and could differentiate into, at least, bone and fat in vitro. In mice challenged with BLM, septal thickening and prominent fibrosis were observed on day 14, with higher HYP content and mRNA levels of TGF-β1, TNF-α and IL-6 exhibited, compared to untreated mice. USCs could migrate to lung and accumulate there in mouse model after intravenous injection. Transplantation of USCs into BLM-induced mice improved their pulmonary histopathology, decreasing Ashcroft score, Szapiel score, HYP content and mRNA levels of TGF-β1 and MMP2 of lung, similar to the effects of BMSCs. IHC and WB further revealed that USCs could inhibit activation of the TGF‑β1-Smad2/3 pathway of lung in vivo. CONCLUSIONS Transplantation of USCs effectively reverses pulmonary fibrotic phenotype in an experimental IPF model, inhibiting the TGF-β1-Smad2/3 pathway, a key driver of fibrosis. These results suggest the therapeutic application of USCs for IPF, instead of other types of MSCs obtained invasively.
Collapse
Affiliation(s)
- Yanju Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China; Infection Management Office, Affiliated Hospital of Nantong University, Nantong, China; Department of Pulmonary and Critical Care Medicine, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China
| | - Yunfei Xia
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
| | - Rui Zhang
- Department of Pulmonary and Critical Care Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaodi Zhou
- Infection Management Office, Affiliated Hospital of Nantong University, Nantong, China
| | - Junhong Jiang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China; Department of Pulmonary and Critical Care Medicine, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, China.
| |
Collapse
|
2
|
Gao Y, Liu MF, Li Y, Liu X, Cao YJ, Long QF, Yu J, Li JY. Mesenchymal stem cells-extracellular vesicles alleviate pulmonary fibrosis by regulating immunomodulators. World J Stem Cells 2024; 16:670-689. [PMID: 38948098 PMCID: PMC11212550 DOI: 10.4252/wjsc.v16.i6.670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/22/2024] [Accepted: 05/11/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a chronic interstitial lung disease characterized by fibroblast proliferation and extracellular matrix formation, causing structural damage and lung failure. Stem cell therapy and mesenchymal stem cells-extracellular vesicles (MSC-EVs) offer new hope for PF treatment. AIM To investigate the therapeutic potential of MSC-EVs in alleviating fibrosis, oxidative stress, and immune inflammation in A549 cells and bleomycin (BLM)-induced mouse model. METHODS The effect of MSC-EVs on A549 cells was assessed by fibrosis markers [collagen I and α-smooth muscle actin (α-SMA), oxidative stress regulators [nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), and inflammatory regulators [nuclear factor-kappaB (NF-κB) p65, interleukin (IL)-1β, and IL-2]. Similarly, they were assessed in the lungs of mice where PF was induced by BLM after MSC-EV transfection. MSC-EVs ion PF mice were detected by pathological staining and western blot. Single-cell RNA sequencing was performed to investigate the effects of the MSC-EVs on gene expression profiles of macrophages after modeling in mice. RESULTS Transforming growth factor (TGF)-β1 enhanced fibrosis in A549 cells, significantly increasing collagen I and α-SMA levels. Notably, treatment with MSC-EVs demonstrated a remarkable alleviation of these effects. Similarly, the expression of oxidative stress regulators, such as Nrf2 and HO-1, along with inflammatory regulators, including NF-κB p65 and IL-1β, were mitigated by MSC-EV treatment. Furthermore, in a parallel manner, MSC-EVs exhibited a downregulatory impact on collagen deposition, oxidative stress injuries, and inflammatory-related cytokines in the lungs of mice with PF. Additionally, the mRNA sequencing results suggested that BLM may induce PF in mice by upregulating pulmonary collagen fiber deposition and triggering an immune inflammatory response. The findings collectively highlight the potential therapeutic efficacy of MSC-EVs in ameliorating fibrotic processes, oxidative stress, and inflammatory responses associated with PF. CONCLUSION MSC-EVs could ameliorate fibrosis in vitro and in vivo by downregulating collagen deposition, oxidative stress, and immune-inflammatory responses.
Collapse
Affiliation(s)
- Ying Gao
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial Rehabilitation Hospital, Xi'an 710000, Shaanxi Province, China
| | - Mei-Fang Liu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Ningxia Medical University (The First People's Hospital of Yinchuan), Yinchuan 750001, Ningxia Hui Autonomous Region, China
| | - Yang Li
- School of Clinical Medicine, Xi'an Medical University, Xi'an 710021, Shaanxi Province, China
| | - Xi Liu
- Department of Respiratory and Critical Care Medicine, Xi'an Central Hospital, Xi'an 710000, Shaanxi Province, China
| | - Yu-Jie Cao
- Department of Respiratory and Critical Care Medicine, Xi'an Central Hospital, Xi'an 710000, Shaanxi Province, China
| | - Qian-Fa Long
- Department of Neurosurgery, Xi'an Central Hospital, Xi'an 710000, Shaanxi Province, China
| | - Jun Yu
- Department of Emergency, Xi'an Central Hospital, Xi'an 710000, Shaanxi Province, China
| | - Jian-Ying Li
- Department of Respiratory and Critical Care Medicine, Xi'an Central Hospital, Xi'an 710000, Shaanxi Province, China.
| |
Collapse
|
3
|
Gao Y, Liu MF, Li Y, Liu X, Cao YJ, Long QF, Yu J, Li JY. Mesenchymal stem cells-extracellular vesicles alleviate pulmonary fibrosis by regulating immunomodulators. World J Stem Cells 2024; 16:669-688. [DOI: 10.4252/wjsc.v16.i6.669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/22/2024] [Accepted: 05/11/2024] [Indexed: 06/25/2024] Open
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a chronic interstitial lung disease characterized by fibroblast proliferation and extracellular matrix formation, causing structural damage and lung failure. Stem cell therapy and mesenchymal stem cells-extracellular vesicles (MSC-EVs) offer new hope for PF treatment.
AIM To investigate the therapeutic potential of MSC-EVs in alleviating fibrosis, oxidative stress, and immune inflammation in A549 cells and bleomycin (BLM)-induced mouse model.
METHODS The effect of MSC-EVs on A549 cells was assessed by fibrosis markers [collagen I and α-smooth muscle actin (α-SMA), oxidative stress regulators [nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), and inflammatory regulators [nuclear factor-kappaB (NF-κB) p65, interleukin (IL)-1β, and IL-2]. Similarly, they were assessed in the lungs of mice where PF was induced by BLM after MSC-EV transfection. MSC-EVs ion PF mice were detected by pathological staining and western blot. Single-cell RNA sequencing was performed to investigate the effects of the MSC-EVs on gene expression profiles of macrophages after modeling in mice.
RESULTS Transforming growth factor (TGF)-β1 enhanced fibrosis in A549 cells, significantly increasing collagen I and α-SMA levels. Notably, treatment with MSC-EVs demonstrated a remarkable alleviation of these effects. Similarly, the expression of oxidative stress regulators, such as Nrf2 and HO-1, along with inflammatory regulators, including NF-κB p65 and IL-1β, were mitigated by MSC-EV treatment. Furthermore, in a parallel manner, MSC-EVs exhibited a downregulatory impact on collagen deposition, oxidative stress injuries, and inflammatory-related cytokines in the lungs of mice with PF. Additionally, the mRNA sequencing results suggested that BLM may induce PF in mice by upregulating pulmonary collagen fiber deposition and triggering an immune inflammatory response. The findings collectively highlight the potential therapeutic efficacy of MSC-EVs in ameliorating fibrotic processes, oxidative stress, and inflammatory responses associated with PF.
CONCLUSION MSC-EVs could ameliorate fibrosis in vitro and in vivo by downregulating collagen deposition, oxidative stress, and immune-inflammatory responses.
Collapse
Affiliation(s)
- Ying Gao
- Department of Respiratory and Critical Care Medicine, Shaanxi Provincial Rehabilitation Hospital, Xi’an 710000, Shaanxi Province, China
| | - Mei-Fang Liu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Ningxia Medical University (The First People’s Hospital of Yinchuan), Yinchuan 750001, Ningxia Hui Autonomous Region, China
| | - Yang Li
- School of Clinical Medicine, Xi’an Medical University, Xi’an 710021, Shaanxi Province, China
| | - Xi Liu
- Department of Respiratory and Critical Care Medicine, Xi’an Central Hospital, Xi’an 710000, Shaanxi Province, China
| | - Yu-Jie Cao
- Department of Respiratory and Critical Care Medicine, Xi’an Central Hospital, Xi’an 710000, Shaanxi Province, China
| | - Qian-Fa Long
- Department of Neurosurgery, Xi’an Central Hospital, Xi’an 710000, Shaanxi Province, China
| | - Jun Yu
- Department of Emergency, Xi’an Central Hospital, Xi’an 710000, Shaanxi Province, China
| | - Jian-Ying Li
- Department of Respiratory and Critical Care Medicine, Xi’an Central Hospital, Xi’an 710000, Shaanxi Province, China
| |
Collapse
|
4
|
Feng X, Qi F, Wang H, Li W, Gan Y, Qi C, Lin Z, Chen L, Wang P, Hu Z, Miao Y. Sorting Technology for Mesenchymal Stem Cells from a Single Tissue Source. Stem Cell Rev Rep 2024; 20:524-537. [PMID: 38112926 DOI: 10.1007/s12015-023-10635-w] [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] [Accepted: 09/23/2023] [Indexed: 12/21/2023]
Abstract
Mesenchymal stem cells (MSCs) are adult stem cells that can be obtained, enriched and proliferated in vitro. They owned enormous potential in fields like regenerative medicine, tissue engineering and immunomodulation. However, though isolated from the same origin, MSCs are still essentially heterogeneous cell populations with different phenotypes and functions. This heterogeneity of MSCs significantly affects their therapeutic efficacy and brings obstacles to scientific research. Thus, reliable sorting technology which can isolate or purify MSC subpopulations with various potential and differentiation pathways is urgently needed. This review summarized principles, application status and clinical implications for these sorting methods, aiming at improving the understanding of MSC heterogeneity as well as providing fresh perspectives for subsequent clinical applications.
Collapse
Affiliation(s)
- Xinyi Feng
- The First Clinical School of Southern Medical University, Guangzhou, China
| | - Fangfang Qi
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Hailin Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Wenzhen Li
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yuyang Gan
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Caiyu Qi
- The First Clinical School of Southern Medical University, Guangzhou, China
| | - Zhen Lin
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Lu Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Piao Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China.
| | - Yong Miao
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
5
|
He YC, Yuan GD, Li N, Ren MF, Qian-Zhang, Deng KN, Wang LC, Xiao WL, Ma N, Stamm C, Felthaus O, Prantl L, Nie J, Wang G. Recent advances in mesenchymal stem cell therapy for myocardial infarction. Clin Hemorheol Microcirc 2024; 87:383-398. [PMID: 38578884 DOI: 10.3233/ch-249101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2024]
Abstract
Myocardial infarction refers to the ischemic necrosis of myocardium, characterized by a sharp reduction or interruption of blood flow in the coronary arteries due to the coronary artery occlusion, resulting in severe and prolonged ischemia in the corresponding myocardium and ultimately leading to ischemic necrosis of the myocardium. Given its high risk, it is considered as one of the most serious health threats today. In current clinical practice, multiple approaches have been explored to diminish myocardial oxygen consumption and alleviate symptoms, but notable success remains elusive. Accumulated clinical evidence has showed that the implantation of mesenchymal stem cell for treating myocardial infarction is both effective and safe. Nevertheless, there persists controversy and variability regarding the standardizing MSC transplantation protocols, optimizing dosage, and determining the most effective routes of administration. Addressing these remaining issues will pave the way of integration of MSCs as a feasible mainstream cardiac treatment.
Collapse
Affiliation(s)
- Yu-Chuan He
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Guo-Dong Yuan
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Nan Li
- Shijiazhuang Obstetrics and Gynecology Hospital, Shijiazhuang, Hebei, China
| | - Mei-Fang Ren
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Qian-Zhang
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Kai-Ning Deng
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Le-Chuan Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Wei-Ling Xiao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Nan Ma
- Helmholtz-Zentrum Hereon, Institute of Active Polymers, Teltow, Germany
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | | | - Oliver Felthaus
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Lukas Prantl
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Jia Nie
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| | - Gang Wang
- Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang, Hebei, China
| |
Collapse
|
6
|
Pham DV, Nguyen TK, Nguyen BL, Kim JO, Jeong JH, Choi I, Park PH. Adiponectin restores the obesity-induced impaired immunomodulatory function of mesenchymal stromal cells via glycolytic reprogramming. Acta Pharm Sin B 2024; 14:273-291. [PMID: 38261813 PMCID: PMC10793097 DOI: 10.1016/j.apsb.2023.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 08/07/2023] [Accepted: 10/18/2023] [Indexed: 01/25/2024] Open
Abstract
Obesity has been known to negatively modulate the life-span and immunosuppressive potential of mesenchymal stromal cells (MSC). However, it remains unclear what drives the compromised potency of obese MSC. In this study, we examined the involvement of adiponectin, an adipose tissue-derived hormone, in obesity-induced impaired therapeutic function of MSC. Diet-induced obesity leads to a decrease in serum adiponectin, accompanied by impairment of survival and immunomodulatory effects of adipose-derived MSC (ADSC). Interestingly, priming with globular adiponectin (gAcrp) improved the immunomodulatory potential of obese ADSC. Similar effects were also observed in lean ADSC. In addition, gAcrp potentiated the therapeutic effectiveness of ADSC in a mouse model of DSS-induced colitis. Mechanistically, while obesity inhibited the glycolytic capacity of MSC, gAcrp treatment induced a metabolic shift toward glycolysis through activation of adiponectin receptor type 1/p38 MAPK/hypoxia inducible factor-1α axis. These findings suggest that activation of adiponectin signaling is a promising strategy for enhancing the therapeutic efficacy of MSC against immune-mediated disorders.
Collapse
Affiliation(s)
- Duc-Vinh Pham
- Department of Pharmacology, Hanoi University of Pharmacy, Hanoi 100000, Viet Nam
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Thi-Kem Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Bao-Loc Nguyen
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jong-Oh Kim
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
7
|
Ali Mohammed S, Elbaramawy A, Hassan Abd-Allah S, Elkholy A, Ibrahim Elsayed N, Hussein S. Therapeutic potentials of mesenchymal stem cells in the treatment of inflammatory bowel disease in rats. J Biochem Mol Toxicol 2024; 38:e23532. [PMID: 37676835 DOI: 10.1002/jbt.23532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/19/2023] [Accepted: 08/18/2023] [Indexed: 09/09/2023]
Abstract
Interleukin-1beta (IL-1β) and interleukin-17A (IL-17A) have strong pro-inflammatory activities that are involved in inflammatory bowel diseases (IBDs). Mesenchymal stem cell (MSC) therapy is considered a promising treatment for IBD. This study was performed to understand the role of rat Nlrp3 inflammasome, Hmgb1, and pro-inflammatory cytokines (IL-1β and IL-17a) in the pathogenesis of IBD. Also, to evaluate the role of human umbilical cord blood-MSCs (hUCB-MSCs) in the management of IBD. The rats were in four groups: normal controls, indomethacin-induced IBD group, indomethacin-induced IBD rats that received phosphate-buffered saline (PBS), and the IBD group that received hUCB-MSCs as a treatment. The messenger RNA (mRNA) expression levels of rat Nlrp3, Hmgb1, IL-1β, and IL-17a were evaluated by quantitative real-time polymerase chain reaction. Histopathological examination of the small intestinal tissues of the studied rats was performed. There was a significant upregulation of the rat Nlrp3, IL-1β, IL-17a mRNA expression (p < 0.001 for the three parameters), and Hmgb1 (p < 0.05) in the untreated IBD group compared to the normal control group. In the MSC-treated group, IL-1β, IL-17a, and rat Nlrp3 mRNA expression significantly decreased compared to both the untreated IBD group and PBS group (p < 0.05 for all). hUCB-MSCs ameliorated IBD in rats by downregulating the pro-inflammatory cytokines (IL-1β and IL-17a) and other inflammatory mediators such as Hmgb1 and rat Nlrp3.
Collapse
Affiliation(s)
- Shuzan Ali Mohammed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Azza Elbaramawy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Somia Hassan Abd-Allah
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Adel Elkholy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Nashwa Ibrahim Elsayed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Samia Hussein
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
8
|
Yu S, Lu J. The potential of mesenchymal stem cells to induce immune tolerance to allogeneic transplants. Transpl Immunol 2023; 81:101939. [PMID: 37866668 DOI: 10.1016/j.trim.2023.101939] [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: 05/03/2023] [Revised: 10/04/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Organ allograft transplantation is an effective treatment plan for patients with organ failure. Although the application of continuous immunosuppressants makes successful allograft survival possible, the patients' long-term survival rate and quality of life are not ideal. Therefore, it is necessary to find a new strategy to alleviate transplant rejection by developing therapies for permanent allograft acceptance. One promising approach is the application of tolerogenic mesenchymal stem cells (MSCs). Extensive research on MSCs has revealed that MSCs have potent differentiation potential and immunomodulatory properties. This review describes the molecular markers and functional properties of MSCs as well as the immunomodulatory mechanisms of MSCs in transplantation, focuses on the research progress in clinical trials of MSCs, and expounds on the future development prospects and possible limitations.
Collapse
Affiliation(s)
- Shaochen Yu
- Department of Emergency and Critical Care Medicine, Guangdong Second Provincial General Hospital, No. 466, Xingang Middle Road, Haizhu District, Guangzhou, Guangdong 510317, China.
| | - Jian Lu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, Anhui 230022, China.
| |
Collapse
|
9
|
Sababathy M, Ramanathan G, Abd Rahaman NY, Ramasamy R, Biau FJ, Qi Hao DL, Hamid NFS. A 'one stone, two birds' approach with mesenchymal stem cells for acute respiratory distress syndrome and Type II diabetes mellitus. Regen Med 2023; 18:913-934. [PMID: 38111999 DOI: 10.2217/rme-2023-0193] [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] [Indexed: 12/20/2023] Open
Abstract
This review explores the intricate relationship between acute respiratory distress syndrome (ARDS) and Type II diabetes mellitus (T2DM). It covers ARDS epidemiology, etiology and pathophysiology, along with current treatment trends and challenges. The lipopolysaccharides (LPS) role in ARDS and its association between non-communicable diseases and COVID-19 are discussed. The review highlights the therapeutic potential of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) for ARDS and T2DM, emphasizing their immunomodulatory effects. This review also underlines how T2DM exacerbates ARDS pathophysiology and discusses the potential of hUC-MSCs in modulating immune responses. In conclusion, the review highlights the multidisciplinary approach to managing ARDS and T2DM, focusing on inflammation, oxidative stress and potential therapy of hUC-MSCs in the future.
Collapse
Affiliation(s)
- Mogesh Sababathy
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Ghayathri Ramanathan
- Faculty of Computer Science & Information Technology, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Nor Yasmin Abd Rahaman
- Department of Veterinary Laboratory Diagnostics, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Laboratory of Vaccines & Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Rajesh Ramasamy
- Department of Pathology, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Foo Jhi Biau
- Centre for Drug Discovery & Molecular Pharmacology (CDDMP), Faculty of Health & Medical Sciences, Taylor's University, Selangor, Subang Jaya, 47500, Malaysia
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, Selangor, Subang Jaya, 47500, Malaysia
| | - Daniel Looi Qi Hao
- My Cytohealth Sdn. Bhd., 18-2, Jalan Radin Bagus 1, Bandar Seri Petaling, Kuala Lumpur, 57000, Malaysia
| | - Nur-Fazila Saulol Hamid
- Department of Veterinary Pathology & Microbiology, Faculty of Veterinary Medicine, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
- Laboratory of Vaccines & Biomolecules, Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| |
Collapse
|
10
|
Zheng X, Zhao D, Liu Y, Jin Y, Liu T, Li H, Liu D. Regeneration and anti-inflammatory effects of stem cells and their extracellular vesicles in gynecological diseases. Biomed Pharmacother 2023; 168:115739. [PMID: 37862976 DOI: 10.1016/j.biopha.2023.115739] [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: 08/25/2023] [Revised: 10/10/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023] Open
Abstract
There are many gynecological diseases, among which breast cancer (BC), cervical cancer (CC), endometriosis (EMs), and polycystic ovary syndrome (PCOS) are common and difficult to cure. Stem cells (SCs) are a focus of regenerative medicine. They are commonly used to treat organ damage and difficult diseases because of their potential for self-renewal and multidirectional differentiation. SCs are also commonly used for difficult-to-treat gynecological diseases because of their strong directional differentiation ability with unlimited possibilities, their tendency to adhere to the diseased tissue site, and their use as carriers for drug delivery. SCs can produce exosomes in a paracrine manner. Exosomes can be produced in large quantities and have the advantage of easy storage. Their safety and efficacy are superior to those of SCs, which have considerable potential in gynecological treatment, such as inhibiting endometrial senescence, promoting vascular reconstruction, and improving anti-inflammatory and immune functions. In this paper, we review the mechanisms of the regenerative and anti-inflammatory capacity of SCs and exosomes in incurable gynecological diseases and the current progress in their application in genetic engineering to provide a foundation for further research.
Collapse
Affiliation(s)
- Xu Zheng
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Dan Zhao
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun 130000, China
| | - Yang Liu
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun 130000, China
| | - Ye Jin
- Changchun University of Chinese Medicine, Changchun 130117, China
| | - Tianjia Liu
- Changchun University of Chinese Medicine, Changchun 130117, China; Baicheng Medical College, Baicheng 137000, China.
| | - Huijing Li
- Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Da Liu
- Changchun University of Chinese Medicine, Changchun 130117, China.
| |
Collapse
|
11
|
Kim Y, An SB, Lee SH, Lee JJ, Kim SB, Ahn JC, Hwang DY, Han I. Enhanced Intervertebral Disc Repair via Genetically Engineered Mesenchymal Stem Cells with Tetracycline Regulatory System. Int J Mol Sci 2023; 24:16024. [PMID: 38003216 PMCID: PMC10671788 DOI: 10.3390/ijms242216024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/28/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
The therapeutic potential of Mesenchymal stem cells (MSCs) for the treatment of Intervertebral disc (IVD) degeneration can be enhanced by amplifying specific cytokines and proteins. This study aimed to investigate the therapeutic potential of tetracycline-off system-engineered tonsil-derived mesenchymal stem cells (ToMSC-Tetoff-TGFβ1-IGF1-BMP7) for treating intervertebral disc (IVD) degeneration. ToMSCs were isolated from a tonsillectomy patient and genetically modified with four distinct plasmids via CRISPR/Cas9-mediated knock-in gene editing. Transgene expression was confirmed through immunofluorescence, western blots, and an enzyme-linked immunosorbent assay for transforming growth factor beta 1 (TGFβ1) protein secretion, and the effect of MSC-TetOff-TGFβ1-IGF1-BMP7 on disc injury was assessed in a rat model. The ToMSC-Tetoff-TGFβ1-IGF1-BMP7 treatment exhibited superior therapeutic effects compared to ToMSC-TGFβ1, and ToMSC-SDF1α implantation groups, stimulating the regeneration of nucleus pulposus (NP) cells crucial for IVD. The treatment showed potential to restore the structural integrity of the extracellular matrix (ECM) by upregulating key molecules such as aggrecan and type II collagen. It also exhibited anti-inflammatory properties and reduced pain-inducing neuropeptides. ToMSC-Tetoff-TGFβ1-IGF1-BMP7 holds promise as a novel treatment for IVD degeneration. It appears to promote NP cell regeneration, restore ECM structure, suppress inflammation, and reduce pain. However, more research and clinical trials are required to confirm its therapeutic potential.
Collapse
Affiliation(s)
- Yeji Kim
- Research Competency Milestones Program of School of Medicine, CHA University School of Medicine, Seongnam-si 13496, Republic of Korea;
| | - Seong Bae An
- Department of Biomedical Science, Graduate School of CHA University, Seongnam-si 13496, Republic of Korea;
| | - Sang-Hyuk Lee
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si 13496, Republic of Korea;
| | - Jong Joo Lee
- Department of Medicine, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea;
- Department of Neurosurgery, Kangbuk Samsung Hospital, Sungkyunkwan University College of Medicine, Seoul 03181, Republic of Korea
| | - Sung Bum Kim
- Department of Neurosurgery, Kyung Hee University, Seoul 02453, Republic of Korea;
| | - Jae-Cheul Ahn
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si 13496, Republic of Korea
| | - Dong-Youn Hwang
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si 13496, Republic of Korea;
- Department of Microbiology, School of Medicine, CHA University, Seongnam-si 13496, Republic of Korea
| | - Inbo Han
- Department of Biomedical Science, Graduate School of CHA University, Seongnam-si 13496, Republic of Korea;
| |
Collapse
|
12
|
Huang H, Liu X, Wang J, Suo M, Zhang J, Sun T, Zhang W, Li Z. Umbilical cord mesenchymal stem cells for regenerative treatment of intervertebral disc degeneration. Front Cell Dev Biol 2023; 11:1215698. [PMID: 37601097 PMCID: PMC10439242 DOI: 10.3389/fcell.2023.1215698] [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: 05/02/2023] [Accepted: 07/27/2023] [Indexed: 08/22/2023] Open
Abstract
Intervertebral disc degeneration is thought to be a major contributor to low back pain, the etiology of which is complex and not yet fully understood. To compensate for the lack of drug and surgical treatment, mesenchymal stem cells have been proposed for regenerative treatment of intervertebral discs in recent years, and encouraging results have been achieved in related trials. Mesenchymal stem cells can be derived from different parts of the body, among which mesenchymal stem cells isolated from the fetal umbilical cord have excellent performance in terms of difficulty of acquisition, differentiation potential, immunogenicity and ethical risk. This makes it possible for umbilical cord derived mesenchymal stem cells to replace the most widely used bone marrow-derived and adipose tissue derived mesenchymal stem cells as the first choice for regenerating intervertebral discs. However, the survival of umbilical cord mesenchymal stem cells within the intervertebral disc is a major factor affecting their regenerative capacity. In recent years biomaterial scaffolds in tissue engineering have aided the survival of umbilical cord mesenchymal stem cells by mimicking the natural extracellular matrix. This seems to provide a new idea for the application of umbilical cord mesenchymal stem cells. This article reviews the structure of the intervertebral disc, disc degeneration, and the strengths and weaknesses of common treatment methods. We focus on the cell source, cell characteristics, mechanism of action and related experiments to summarize the umbilical cord mesenchymal stem cells and explore the feasibility of tissue engineering technology of umbilical cord mesenchymal stem cells. Hoping to provide new ideas for the treatment of disc degeneration.
Collapse
Affiliation(s)
- Huagui Huang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xin Liu
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jinzuo Wang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Moran Suo
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jing Zhang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tianze Sun
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Wentao Zhang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhonghai Li
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Dalian, Liaoning, China
| |
Collapse
|
13
|
Chen W, Lv L, Chen N, Cui E. Immunogenicity of mesenchymal stromal/stem cells. Scand J Immunol 2023; 97:e13267. [PMID: 39007962 DOI: 10.1111/sji.13267] [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: 08/29/2022] [Revised: 03/06/2023] [Accepted: 03/17/2023] [Indexed: 04/07/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) possess the ability to self-renew and differentiate into other cell types. Because of their anti-inflammatory and immunomodulatory abilities, as well as their more ready availability compared to other stem cell sources, MSCs hold great promise for the treatment of many diseases, such as haematological defects, acute respiratory distress syndrome, autoimmunity, cardiovascular diseases, etc. However, immune rejection remains an important problem. MSCs are considered to have low immunogenicity, but they do not have full immunological privilege. This review analyzes and discusses the safety of MSCs from the perspective of their immunogenicity, with the aim of providing a reference for future research and clinical application.
Collapse
Affiliation(s)
- Wenyan Chen
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, 313000, Zhejiang, No. 1558, Third Ring North Road, Huzhou, China
| | - Lu Lv
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, 313000, Zhejiang, No. 1558, Third Ring North Road, Huzhou, China
| | - Na Chen
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, 313000, Zhejiang, No. 1558, Third Ring North Road, Huzhou, China
| | - Enhai Cui
- Department of Respiratory and Critical Care Medicine, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, 313000, Zhejiang, No. 1558, Third Ring North Road, Huzhou, China
| |
Collapse
|
14
|
Cimino M, Parreira P, Leiro V, Sousa A, Gonçalves RM, Barrias CC, Martins MCL. Enhancement of hMSC In Vitro Proliferation by Surface Immobilization of a Heparin-Binding Peptide. Molecules 2023; 28:molecules28083422. [PMID: 37110656 PMCID: PMC10146743 DOI: 10.3390/molecules28083422] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/29/2023] Open
Abstract
The use of human Mesenchymal Stem Cells (hMSC) as therapeutic agents for advanced clinical therapies relies on their in vitro expansion. Over the last years, several efforts have been made to optimize hMSC culture protocols, namely by mimicking the cell physiological microenvironment, which strongly relies on signals provided by the extracellular matrix (ECM). ECM glycosaminoglycans, such as heparan-sulfate, sequester adhesive proteins and soluble growth factors at the cell membrane, orchestrating signaling pathways that control cell proliferation. Surfaces exposing the synthetic polypeptide poly(L-lysine, L-leucine) (pKL) have previously been shown to bind heparin from human plasma in a selective and concentration-dependent manner. To evaluate its effect on hMSC expansion, pKL was immobilized onto self-assembled monolayers (SAMs). The pKL-SAMs were able to bind heparin, fibronectin and other serum proteins, as demonstrated by quartz crystal microbalance with dissipation (QCM-D) studies. hMSC adhesion and proliferation were significantly increased in pKL-SAMs compared to controls, most probably related to increased heparin and fibronectin binding to pKL surfaces. This proof-of-concept study highlights the potential of pKL surfaces to improve hMSC in vitro expansion possible through selective heparin/serum protein binding at the cell-material interface.
Collapse
Affiliation(s)
- Maura Cimino
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Faculdade de Engenharia, Departamento de Engenharia Metalúrgica e de Materiais, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Paula Parreira
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Victoria Leiro
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Aureliana Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Raquel M Gonçalves
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Cristina C Barrias
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - M Cristina L Martins
- i3S-Instituto de Investigação e Inovação em Saúde, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| |
Collapse
|
15
|
Li Z, Wu Y, Tan G, Xu Z, Xue H. Exosomes and exosomal miRNAs: A new therapy for intervertebral disc degeneration. Front Pharmacol 2022; 13:992476. [PMID: 36160436 PMCID: PMC9492865 DOI: 10.3389/fphar.2022.992476] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/22/2022] [Indexed: 11/15/2022] Open
Abstract
Low back pain has been found as a major cause of global disease burden and disability. Intervertebral disc degeneration is recognized as the vital factor causing low back pain. Intervertebral disc degeneration has a complex mechanism and cannot be avoided. Traditional strategies for the treatment of intervertebral disc degeneration cannot meet the needs of intervertebral disc regeneration, so novel treatment methods are urgently required. Exosomes refer to extracellular vesicles that can be released by most cells, and play major roles in intercellular material transport and information transmission. MicroRNAs have been identified as essential components in exosomes, which can be selectively ingested by exosomes and delivered to receptor cells for the regulation of the physiological activities and functions of receptor cells. Existing studies have progressively focused on the role of exosomes and exosomal microRNAs in the treatment of intervertebral disc degeneration. The focus on this paper is placed on the changes of microenvironment during intervertebral disc degeneration and the biogenesis and mechanism of action of exosomes and exosomal microRNAs. The research results and deficiencies of exosomes and exosomal microRNAs in the regulation of apoptosis, extracellular matrix homeostasis, inflammatory response, oxidative stress, and angiogenesis in intervertebral disc degeneration are primarily investigated. The aim of this paper is to identify the latest research results, potential applications and challenges of this emerging treatment strategy.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Wu
- Department of Orthopedics, The First Affiliated Hospital of Shandong First Medcial Unversity, Jinan, China
| | - Guoqing Tan
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhanwang Xu
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haipeng Xue
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Haipeng Xue,
| |
Collapse
|
16
|
Bhujel B, Shin HE, Choi DJ, Han I. Mesenchymal Stem Cell-Derived Exosomes and Intervertebral Disc Regeneration: Review. Int J Mol Sci 2022; 23:7306. [PMID: 35806304 PMCID: PMC9267028 DOI: 10.3390/ijms23137306] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 02/07/2023] Open
Abstract
Intervertebral disc degeneration (IVDD) is a common cause of lower back pain (LBP), which burdens individuals and society as a whole. IVDD occurs as a result of aging, mechanical trauma, lifestyle factors, and certain genetic abnormalities, leads to loss of nucleus pulposus, alteration in the composition of the extracellular matrix, excessive oxidative stress, and inflammation in the intervertebral disc. Pharmacological and surgical interventions are considered a boon for the treatment of IVDD, but the effectiveness of those strategies is limited. Mesenchymal stem cells (MSCs) have recently emerged as a possible promising regenerative therapy for IVDD due to their paracrine effect, restoration of the degenerated cells, and capacity for differentiation into disc cells. Recent investigations have shown that the pleiotropic effect of MSCs is not related to differentiation capacity but is mediated by the secretion of soluble paracrine factors. Early studies have demonstrated that MSC-derived exosomes have therapeutic potential for treating IVDD by promoting cell proliferation, tissue regeneration, modulation of the inflammatory response, and reduced apoptosis. This paper highlights the current state of MSC-derived exosomes in the field of treatment of IVDD with further possible future developments, applications, and challenges.
Collapse
Affiliation(s)
- Basanta Bhujel
- Department of Biomedical Science, College of Life Sciences, CHA University, Seongnam-si 13496, Korea; (B.B.); (H.-E.S.)
| | - Hae-Eun Shin
- Department of Biomedical Science, College of Life Sciences, CHA University, Seongnam-si 13496, Korea; (B.B.); (H.-E.S.)
| | - Dong-Jun Choi
- Department of Medicine, CHA Univerity School of Medicine, Seongnam-si 13496, Korea;
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si 13496, Korea
| |
Collapse
|
17
|
He JY, Cheng M, Ye JL, Peng CH, Chen J, Luo B, Zhang XY, Fu Q. YY1-induced lncRNA XIST inhibits cartilage differentiation of BMSCs by binding with TAF15 to stabilizing FUT1 expression. Regen Ther 2022; 20:41-50. [PMID: 35402663 PMCID: PMC8968204 DOI: 10.1016/j.reth.2022.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/22/2022] [Accepted: 02/15/2022] [Indexed: 12/17/2022] Open
Abstract
Introduction The functional roles and mechanism of the XIST in osteoarthritis and the chondrogenic differentiation of BMSCs were clarified. Methods The expression levels of XIST, TAF15, FUT1 and YY1 were detected through quantitative RT-PCR. The protein expression of Sox9, ACAN, COL2A1 and FUT1 were detected by western blot and immunohistochemistry. The damage of cartilage tissue was detected by HE staining, and Safranin O-fast green. Alcian-Blue and Alizarin red S staining were performed to evaluate BMSCs chondrogenic differentiation. The relationship between XIST and TAF15, XIST and TAF15 were analyzed by RNA immunoprecipitation assay. Luciferase reporter assays and chromatin immunoprecipitation were performed to detect the interaction relationship between XIST and YY1. In addition, osteoarthritis mice were built to assess the function of XIST in vivo. Results The levels of XIST, TAF15 and FUT1 were upregulated in cartilage tissues from osteoarthritis patient. The level of XIST was decreased in BMSCs during chondrogenic differentiation. XIST overexpression inhibited the chondrogenic differentiation of BMSCs. Moreover, silencing of FUT1 reversed the effects of XIST overexpression on BMSCs chondrogenic differentiation. Mechanistically, in BMSCs, YY1 induced the expression of XIST in BMSCs, and XIST regulated FUT1 mRNA stability through targeting TAF15. Furthermore, silencing of XIST alleviated the symptoms of cartilage injury in OA mice. Conclusion Taken together, these results suggested that YY1 induced XIST was closely related to the chondrogenic differentiation of BMSCs and the progression of osteoarthritis by TAF15/FUT1 axis, and may be a new OA therapeutic target. XIST and TAF15 expression were upregulated in osteoarthritis. Overexpression of XIST suppressed the chondrogenic differentiation of BMSCs. XIST regulated BMSCs chondrogenic differentiation through enhancing FUT1 mRNA stability via TAF15. XIST silencing relieved cartilage damage in OA mice.
Collapse
Affiliation(s)
- Jian-Ying He
- Orthopedics Department, JiangXi Provinvcial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi Province, PR China
| | - Min Cheng
- Orthopedics Department, People's Hospital of Poyang County, Shangrao, 333100, Jiangxi Province, PR China
| | - Jia-Lian Ye
- Orthopedics Department, People's Hospital of Poyang County, Shangrao, 333100, Jiangxi Province, PR China
| | - Chuan-Hua Peng
- Orthopedics Department, People's Hospital of Poyang County, Shangrao, 333100, Jiangxi Province, PR China
| | - Jian Chen
- Orthopedics Department, People's Hospital of Poyang County, Shangrao, 333100, Jiangxi Province, PR China
| | - Bin Luo
- Orthopedics Department, People's Hospital of Poyang County, Shangrao, 333100, Jiangxi Province, PR China
| | - Xian-Yu Zhang
- Orthopedics Department, Shangrao People's Hospital, Shangrao, 333400, Jiangxi Province, PR China
| | - Qiang Fu
- Department of Rheumatology, JiangXi Provinvcial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, 330006, Jiangxi Province, PR China
- Corresponding author. Department of Rheumatology, JiangXi Provinvcial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, No. 92, Aiguo Road, Donghu District, Nanchang, 330006, Jiangxi Province, PR China.
| |
Collapse
|
18
|
Trophic and immunomodulatory effects of adipose tissue derived stem cells in a preclinical murine model of endometriosis. Sci Rep 2022; 12:8031. [PMID: 35577867 PMCID: PMC9110373 DOI: 10.1038/s41598-022-11891-5] [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: 08/29/2021] [Accepted: 04/25/2022] [Indexed: 11/24/2022] Open
Abstract
Endometriosis, which exhibits enigmatic pathological features such as stromal fibrosis and proliferation of ectopic epithelial cells, is known as a refractory disease. Mesenchymal stem cells modulate the fibrosis in stromal tissues through their trophic and immunomodulatory properties. To investigate the potential of stem cells in treating endometriosis, we examined the secondary morphology and molecular alterations in endometriosis-like lesions after the administration of adipose tissue-derived stem cells (ASCs) to an experimental murine model of endometriosis. The infused ASCs were found integrated in the endometriosis-like lesions. Accompanied by the suppression of stromal fibrosis and proliferation of endometriotic epithelial cells, the infusion of ASCs with stemness potential (early passage of ASCs) suppressed the growth of endometriosis-like lesions and inhibited the expression of pro-inflammatory and pro-fibrotic cytokines, whereas no significant attenuation of endometriosis-like lesions occurred after the infusion of ASCs without stemness potential (late passage of ASCs). Accordingly, the trophic and immunomodulatory properties of ASCs may regulate fibrosis in endometriosis-like lesions, suggesting that regenerative medicine could be recognized as an innovative treatment for patients with endometriosis through the accumulation of evidence of preclinical efficacy.
Collapse
|
19
|
Lu L, Xu A, Gao F, Tian C, Wang H, Zhang J, Xie Y, Liu P, Liu S, Yang C, Ye Z, Wu X. Mesenchymal Stem Cell-Derived Exosomes as a Novel Strategy for the Treatment of Intervertebral Disc Degeneration. Front Cell Dev Biol 2022; 9:770510. [PMID: 35141231 PMCID: PMC8818990 DOI: 10.3389/fcell.2021.770510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/31/2021] [Indexed: 12/15/2022] Open
Abstract
Intervertebral disc degeneration (IVDD) has been reported to be the most prevalent contributor to low back pain, posing a significant strain on the healthcare systems on a global scale. Currently, there are no approved therapies available for the prevention of the progressive degeneration of intervertebral disc (IVD); however, emerging regenerative strategies that aim to restore the normal structure of the disc have been fundamentally promising. In the last decade, mesenchymal stem cells (MSCs) have received a significant deal of interest for the treatment of IVDD due to their differentiation potential, immunoregulatory capabilities, and capability to be cultured and regulated in a favorable environment. Recent investigations show that the pleiotropic impacts of MSCs are regulated by the production of soluble paracrine factors. Exosomes play an important role in regulating such effects. In this review, we have summarized the current treatments for disc degenerative diseases and their limitations and highlighted the therapeutic role and its underlying mechanism of MSC-derived exosomes in IVDD, as well as the possible future developments for exosomes.
Collapse
Affiliation(s)
- Lin Lu
- Department of Orthopaedics Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aoshuang Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Gao
- Department of Orthopaedics Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenjun Tian
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- The First Hospital of Lanzhou University, Lanzhou, China
| | - Honglin Wang
- Department of Orthopaedics Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiayao Zhang
- Department of Orthopaedics Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Xie
- Department of Orthopaedics Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengran Liu
- Department of Orthopaedics Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songxiang Liu
- Department of Orthopaedics Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cao Yang
- Department of Orthopaedics Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhewei Ye
- Department of Orthopaedics Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zhewei Ye, ; Xinghuo Wu,
| | - Xinghuo Wu
- Department of Orthopaedics Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zhewei Ye, ; Xinghuo Wu,
| |
Collapse
|
20
|
Al Mushafi A, Ooi JD, Odobasic D. Crescentic Glomerulonephritis: Pathogenesis and Therapeutic Potential of Human Amniotic Stem Cells. Front Physiol 2021; 12:724186. [PMID: 34721059 PMCID: PMC8554237 DOI: 10.3389/fphys.2021.724186] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/24/2021] [Indexed: 12/15/2022] Open
Abstract
Chronic kidney disease (CKD) leads to significant morbidity and mortality worldwide. Glomerulonephritis (GN) is the second leading cause of CKD resulting in end stage renal failure. The most severe and rapidly progressive type of GN is characterized by glomerular crescent formation. The current therapies for crescentic GN, which consist of broad immunosuppressive drugs, are partially effective, non-specific, toxic and cause many serious side effects including infections, cancer, and cardiovascular problems. Therefore, new and safer therapies are needed. Human amniotic epithelial cells (hAECs) are a type of stem cell which are isolated from the placenta after birth. They represent an attractive and novel therapeutic option for the treatment of various inflammatory conditions owing to their unique and selective immunosuppressive ability, as well as their excellent safety profile and clinical applicability. In this review, we will discuss the immunopathogenesis of crescentic GN, issues with currently available treatments and how hAECs offer potential to become a new and harmless treatment option for this condition.
Collapse
Affiliation(s)
- Ahmed Al Mushafi
- Department of Medicine, Monash Medical Centre, Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia
| | - Joshua D Ooi
- Department of Medicine, Monash Medical Centre, Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia
| | - Dragana Odobasic
- Department of Medicine, Monash Medical Centre, Centre for Inflammatory Diseases, Monash University, Clayton, VIC, Australia
| |
Collapse
|
21
|
Wiśniewska J, Sadowska A, Wójtowicz A, Słyszewska M, Szóstek-Mioduchowska A. Perspective on Stem Cell Therapy in Organ Fibrosis: Animal Models and Human Studies. Life (Basel) 2021; 11:life11101068. [PMID: 34685439 PMCID: PMC8538998 DOI: 10.3390/life11101068] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/17/2022] Open
Abstract
Tissue fibrosis is characterized by excessive deposition of extracellular matrix (ECM) components that result from the disruption of regulatory processes responsible for ECM synthesis, deposition, and remodeling. Fibrosis develops in response to a trigger or injury and can occur in nearly all organs of the body. Thus, fibrosis leads to severe pathological conditions that disrupt organ architecture and cause loss of function. It has been estimated that severe fibrotic disorders are responsible for up to one-third of deaths worldwide. Although intensive research on the development of new strategies for fibrosis treatment has been carried out, therapeutic approaches remain limited. Since stem cells, especially mesenchymal stem cells (MSCs), show remarkable self-renewal, differentiation, and immunomodulatory capacity, they have been intensively tested in preclinical studies and clinical trials as a potential tool to slow down the progression of fibrosis and improve the quality of life of patients with fibrotic disorders. In this review, we summarize in vitro studies, preclinical studies performed on animal models of human fibrotic diseases, and recent clinical trials on the efficacy of allogeneic and autologous stem cell applications in severe types of fibrosis that develop in lungs, liver, heart, kidney, uterus, and skin. Although the results of the studies seem to be encouraging, there are many aspects of cell-based therapy, including the cell source, dose, administration route and frequency, timing of delivery, and long-term safety, that remain open areas for future investigation. We also discuss the contemporary status, challenges, and future perspectives of stem cell transplantation for therapeutic options in fibrotic diseases as well as we present recent patents for stem cell-based therapies in organ fibrosis.
Collapse
|
22
|
Zhang X, van Rijt S. 2D biointerfaces to study stem cell-ligand interactions. Acta Biomater 2021; 131:80-96. [PMID: 34237424 DOI: 10.1016/j.actbio.2021.06.044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/18/2021] [Accepted: 06/28/2021] [Indexed: 02/07/2023]
Abstract
Stem cells have great potential in the field of tissue engineering and regenerative medicine due to their inherent regenerative capabilities. However, an ongoing challenge within their clinical translation is to elicit or predict the desired stem cell behavior once transplanted. Stem cell behavior and function are regulated by their interaction with biophysical and biochemical signals present in their natural environment (i.e., stem cell niches). To increase our understanding about the interplay between stem cells and their resident microenvironments, biointerfaces have been developed as tools to study how these substrates can affect stem cell behaviors. This article aims to review recent developments on fabricating cell-instructive interfaces to control cell adhesion processes towards directing stem cell behavior. After an introduction on stem cells and their natural environment, static surfaces exhibiting predefined biochemical signals to probe the effect of chemical features on stem cell behaviors are discussed. In the third section, we discuss more complex dynamic platforms able to display biochemical cues with spatiotemporal control using on-off ligand display, reversible ligand display, and ligand mobility. In the last part of the review, we provide the reader with an outlook on future designs of biointerfaces. STATEMENT OF SIGNIFICANCE: Stem cells have great potential as treatments for many degenerative disorders prevalent in our aging societies. However, an ongoing challenge within their clinical translation is to promote stem cell mediated regeneration once they are transplanted in the body. Stem cells reside within our bodies where their behavior and function are regulated by interactions with their natural environment called the stem cell niche. To increase our understanding about the interplay between stem cells and their niche, 2D materials have been developed as tools to study how specific signals can affect stem cell behaviors. This article aims to review recent developments on fabricating cell-instructive interfaces to control cell adhesion processes towards directing stem cell behavior.
Collapse
|
23
|
Lee S, Kim HS, Min BH, Kim BG, Kim SA, Nam H, Lee M, Kim M, Hwang HY, Leesong AI, Leesong MM, Kim JH, Shin JS. Enhancement of anti-inflammatory and immunomodulatory effects of adipose-derived human mesenchymal stem cells by making uniform spheroid on the new nano-patterned plates. Biochem Biophys Res Commun 2021; 552:164-169. [PMID: 33751933 DOI: 10.1016/j.bbrc.2021.03.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/05/2021] [Indexed: 02/08/2023]
Abstract
Human mesenchymal stem cells (MSCs) are known to have anti-inflammatory and immunomodulatory functions; thus, several MSC products have been applied as cell therapy in clinical trials worldwide. Recent studies have demonstrated that MSC spheroids have superior anti-inflammatory and immunomodulatory functions to a single cell suspension. Current methods to prepare MSC spheroids include hanging drop, concave microwell aggregation, spinner flask, and gravity circulation. However, all these methods have limitations such as low scalability, easy cell clumping, low viability, and irregular size distribution. Here, we present a nano-patterned culture plasticware named PAMcell™ 3D plate to overcome these limitations. Nano-sized silica particles (700 nm) coated with RGD peptide were arrayed into fusiform onto the PLGA film. This uniform array enabled the seeded MSCs to grow only on the silica particles, forming uniform-sized semi-spheroids within 48 h. These MSC spheroids have been shown to have enhanced stemness, anti-inflammatory, and immunomodulatory functions, as revealed by the increased expression of stem cell markers (Oct4, Sox2, and Nanog), anti-inflammatory (IL-10, TSG6, and IDO), and immunomodulatory molecules (HGF, VEGF, CXCR4) both at mRNA and protein expression levels. Furthermore, these MSC spheroids demonstrated an increased palliative effect on glycemic control in a multiple low-dose streptozotocin-induced diabetes model compared with the same number of MSC single cell suspensions. Taken together, this study presents a new method to produce uniform-sized MSC spheroids with enhanced anti-inflammatory and immunomodulatory functions in vitro and in vivo.
Collapse
Affiliation(s)
- Sangho Lee
- R&D Center, YidoBio, Inc., 123 Beolmal-ro, Anyang-si, Gyeonggi-do, 14056, South Korea
| | - Hyo-Sop Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Byoung-Hoon Min
- R&D Center, YidoBio, Inc., 123 Beolmal-ro, Anyang-si, Gyeonggi-do, 14056, South Korea
| | - Byoung Geun Kim
- R&D Center, YidoBio, Inc., 123 Beolmal-ro, Anyang-si, Gyeonggi-do, 14056, South Korea
| | - Shin Ae Kim
- R&D Center, YidoBio, Inc., 123 Beolmal-ro, Anyang-si, Gyeonggi-do, 14056, South Korea
| | - Hyeyoung Nam
- R&D Center, YidoBio, Inc., 123 Beolmal-ro, Anyang-si, Gyeonggi-do, 14056, South Korea
| | - Minsuk Lee
- R&D Center, YidoBio, Inc., 123 Beolmal-ro, Anyang-si, Gyeonggi-do, 14056, South Korea
| | - Minsun Kim
- R&D Center, YidoBio, Inc., 123 Beolmal-ro, Anyang-si, Gyeonggi-do, 14056, South Korea
| | - Hye Yeon Hwang
- R&D Center, YidoBio, Inc., 123 Beolmal-ro, Anyang-si, Gyeonggi-do, 14056, South Korea
| | - Alex Inkeun Leesong
- R&D Center, YidoBio, Inc., 123 Beolmal-ro, Anyang-si, Gyeonggi-do, 14056, South Korea
| | | | - Jae-Ho Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea.
| | - Jun-Seop Shin
- R&D Center, YidoBio, Inc., 123 Beolmal-ro, Anyang-si, Gyeonggi-do, 14056, South Korea.
| |
Collapse
|
24
|
Harrell CR, Volarevic V. Mesenchymal Stem Cell-Derived Secretome: A New Remedy for the Treatment of Autoimmune and Inflammatory Diseases. Stem Cells 2021. [DOI: 10.1007/978-3-030-77052-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|