1
|
Wang H, Zhang Q, Wu S, Pan D, Ning Y, Wang C, Guo J, Gu Y. Mesenchymal stem cell therapy in eosinophilic granulomatosis with polyangiitis-related lower limb gangrene: a case report. Stem Cell Res Ther 2024; 15:307. [PMID: 39285456 PMCID: PMC11406883 DOI: 10.1186/s13287-024-03924-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Eosinophilic granulomatosis with polyangiitis (EGPA), a rare but life-threatening systemic vasculitis, is distinguished by marked eosinophilia and presents with diverse symptoms, including asthma, cutaneous purpura, ecchymosis, skin necrosis, cardiac lesions, peripheral neuropathy, and necrotizing vasculitis. The etiology of EGPA involves a complex interaction among humoral, adaptive, innate, and allergic immune responses. Standard treatment employs prolonged high-dose glucocorticoid therapy, which is critical for survival; however, some patients' symptoms cannot be relieved. CASE REPORT This case report details the medical management of an 11-year-old patient with EGPA, who was at risk of bilateral lower limb amputation due to differential arterial occlusion and severe, necrotizing vasculitis-induced gangrene in both feet. Treatment modalities administered included systemic infusion of Umbilical Cord Mesenchymal Stem Cells (UC-MSCs), targeted gastrocnemius muscle injections, and application of a Placenta-Derived Mesenchymal Stem Cells (PD-MSCs) hydrogel. RESULTS After receiving a four-month regimen of allogeneic mesenchymal stem cell therapy via intravenous and local administration, the patient showed normalized eosinophil counts, reestablished blood flow in the dorsal arteries, and marked improvement in foot ulcerations. CONCLUSION Mesenchymal stem cell therapy is a promising option for severe EGPA cases refractory to glucocorticoids.
Collapse
Affiliation(s)
- Hui Wang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China
- ShangRao Jingkai Health-Biotech United Hospital, ShangRao, 334000, Jiangxi, China
| | - Qian Zhang
- ShangRao Jingkai Health-Biotech United Hospital, ShangRao, 334000, Jiangxi, China
- Shangrao Normal University, ShangRao, 334000, Jiangxi, China
| | - Sensen Wu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China
| | - Dikang Pan
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China
| | - Yachan Ning
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China
| | - Cong Wang
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China
| | - Jianming Guo
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China.
| | - Yongquan Gu
- Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, No. 45, Changchun Street, Beijing, 100053, China.
| |
Collapse
|
2
|
Xiong Y, Wang F, Mu H, Zhang A, Zhao Y, Han K, Zhang J, Zhang H, Wang Z, Ma J, Wei R, Luan X. hPMSCs prevent erythrocytes dysfunction caused by graft versus host disease via promoting GSH synthesis. Int Immunopharmacol 2024; 139:112689. [PMID: 39029234 DOI: 10.1016/j.intimp.2024.112689] [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: 12/21/2023] [Revised: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Oxidative stress is increased in allogeneic hematopoietic stem cell transplant (allo-HSCT) recipients and leads to the development of graft versus host disease (GVHD). Mesenchymal stromal cells (MSCs) can ameliorate GVHD by regulating the function of T cells. However, whether MSCs can modulate erythrocyte antioxidant metabolism and thus reduce GVHD is not known. METHODS Forty female BALB/c mice were randomly assigned to four groups: the control, GVHDhigh, hPMSC, and PBS groups. A hypoxanthine/xanthine oxidase system was used to steadily and gradually produce superoxide in an in vitro experiment. A scanning microscope was used to examine the ultrastructure of erythrocytes. Laser diffraction analyses were used to analyze erythrocyte deformability. Western blotting was used to measure the expression of the erythrocyte membrane skeleton proteins Band 3 and β-Spectrin. Corresponding kits were used to assess the levels of oxidative damage and the activity of antioxidant enzymes. RESULTS Morphological and deformability defects were significantly increased in erythrocytes from GVHD patients. Band 3 and β-Spectrin expression was also reduced in GVHD patients and model mice. Furthermore, we observed significantly increased oxidative stress-induce injury and decreased antioxidant capability in erythrocytes from both GVHD patients and model mice. Subsequent research showed that human placenta-derived MSC (hPMSC) therapy decreased the GVHD-induced redox imbalance in erythrocytes. Furthermore, our findings suggested that upregulating glucose metabolism promoted both the de novo synthesis and recycling of GSH, which is the primary mechanism by which hPMSCs mediate the increase in antioxidant capacity in erythrocytes. CONCLUSION Together, our findings suggest that hPMSCs can increase antioxidant capacity by increasing erythrocyte GSH production and thus ameliorate GVHD.
Collapse
Affiliation(s)
- Yanlian Xiong
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Feifei Wang
- Department of Anesthesiology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, PR China
| | - Huanmei Mu
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Aiping Zhang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Yaxuan Zhao
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Kaiyue Han
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Jiashen Zhang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Hengchao Zhang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Zhuoya Wang
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Junjie Ma
- Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong Province 264000, PR China
| | - Rongxia Wei
- Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong Province 264000, PR China.
| | - Xiying Luan
- Department of Immunology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China.
| |
Collapse
|
3
|
Zhang ZS, Ren HC, Gu X, Liang QR, Fei H, Yang YH, Yang S, He LY, Liu LL. Interleukin-1 beta (IL-1β) as adjuvant enhances the immune effects of Aeromonas veronii inactivated vaccine in largemouth bass (Micropterus salmoides). Int J Biol Macromol 2024; 273:133135. [PMID: 38876231 DOI: 10.1016/j.ijbiomac.2024.133135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Largemouth bass (Micropterus salmoides) has emerged as a significant economic fish species, with a rise in Aeromonas veronii infections in farming. However, research on adjuvants for vaccines against A. veronii in largemouth bass remains scarce. In present study, recombinant largemouth bass IL-1β (LbIL-1β) was expressed to explore its adjuvant effect on the A. veronii inactivated vaccine. Following vaccination with recombinant LbIL-1β (rLbIL-1β) and the inactivated A. veronii, higher serum SOD levels and lysozyme activities were observed in largemouth bass from inactivated A. veronii + rLbIL-1β vaccinated group. Furthermore, it was discovered that rLbIL-1β was able to boost the serum-specific antibody levels induced by the inactivated A. veronii. The qRT-PCR analysis revealed that rLbIL-1β also enhanced the expression of IgM, CD4, and MHC II in largemouth bass triggered by the inactivated A. veronii. After challenged with live A. veronii, the outcomes demonstrated that the relative percentage survival (RPS) for largemouth bass resulting from the inactivated A. veronii in combination with rLbIL-1β was 76.67 %, surpassing the RPS of 60 % in the inactivated A. veronii group. Collectively, these findings indicate that rLbIL-1β enhances the protective effect of the A. veronii inactivated vaccine on largemouth bass, showcasing potential as an adjuvant for further development.
Collapse
Affiliation(s)
- Ze-Sheng Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Heng-Chu Ren
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Xie Gu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Qian-Rong Liang
- Zhejiang Fisheries Test and Aquatic Disease Prevention Center, Zhejiang Fisheries Technical Extension Center, Hangzhou 310023, China
| | - Hui Fei
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Yu-Hong Yang
- Zhejiang Fisheries Test and Aquatic Disease Prevention Center, Zhejiang Fisheries Technical Extension Center, Hangzhou 310023, China
| | - Shun Yang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China; College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China; Zhejiang Huadi Pharmaceutical Group Co., Ltd, Hangzhou 313300, China.
| | - Liang-Yin He
- College of Marine Sciences, Ningde Normal University, Ningde 352100, China.
| | - Li-Li Liu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China.
| |
Collapse
|
4
|
Li W, Si Y, Wang Y, Chen J, Huo X, Xu P, Jiang B, Li Z, Shang K, Luo Q, Xiong Y. hUCMSC-derived exosomes protect against GVHD-induced endoplasmic reticulum stress in CD4 + T cells by targeting the miR-16-5p/ATF6/CHOP axis. Int Immunopharmacol 2024; 135:112315. [PMID: 38805908 DOI: 10.1016/j.intimp.2024.112315] [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: 01/19/2024] [Revised: 05/18/2024] [Accepted: 05/19/2024] [Indexed: 05/30/2024]
Abstract
Exosomes generated from mesenchymal stem cells (MSCs) are thought to be a unique therapeutic strategy for several autoimmune deficiency illnesses. The purpose of this study was to elucidate the protective effects of human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-Exo) on CD4+ T cells dysfunction during graft-versus-host disease (GVHD) and to identify the underlying processes involved. Here, we showed that hUCMSC-Exo treatment can effectively attenuate GVHD injury by alleviating redox metabolism disorders and inflammatory cytokine bursts in CD4+ T cells. Furthermore, hUCMSC-Exo ameliorate ER stress and ATF6/CHOP signaling-mediated apoptosis in CD4+ T cells and promote the development of CD4+IL-10+ T cells during GVHD. Moreover, downregulating miR-16-5p in hUCMSC-Exo impaired their ability to prevent CD4+ T cells apoptosis and weakened their ability to promote the differentiation of CD4+IL-10+ T cells. Collectively, the obtained data suggested that hUCMSC-Exo suppress ATF6/CHOP signaling-mediated ER stress and apoptosis in CD4+ T cells, enhance the differentiation of CD4+IL-10+ T cells, and reverse the imbalance of immune homeostasis in the GVHD process by transferring miR-16-5p. Our study provided further evidence that GVHD patients can benefit from hUCMSC-Exo-mediated therapy.
Collapse
Affiliation(s)
- Weihan Li
- Xu Rongxiang Regenerative Medicine Research Center, Binzhou Medical University, Yantai, PR China; Shanghai Mebo Life Science & Technology Co., Shanghai, PR China
| | - Yaru Si
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Yueming Wang
- Xu Rongxiang Regenerative Medicine Research Center, Binzhou Medical University, Yantai, PR China
| | - Juntong Chen
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Xingyu Huo
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Pengzhan Xu
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Bingzhen Jiang
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Zile Li
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Kangdi Shang
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China
| | - Qianqian Luo
- Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China.
| | - Yanlian Xiong
- Xu Rongxiang Regenerative Medicine Research Center, Binzhou Medical University, Yantai, PR China; Department of Histology and Embryology, School of Basic Medicine, Binzhou Medical University, Yantai, PR China.
| |
Collapse
|
5
|
Zhang H, Han K, Li H, Zhang J, Zhao Y, Wu Y, Wang B, Ma J, Luan X. hPMSCs Regulate the Level of TNF-α and IL-10 in Th1 Cells and Improve Hepatic Injury in a GVHD Mouse Model via CD73/ADO/Fyn/Nrf2 Axis. Inflammation 2024; 47:244-263. [PMID: 37833615 DOI: 10.1007/s10753-023-01907-1] [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: 06/19/2023] [Revised: 08/18/2023] [Accepted: 09/13/2023] [Indexed: 10/15/2023]
Abstract
Mesenchymal stem cells (MSCs) ameliorate graft-versus-host disease (GVHD)-induced tissue damage by exerting immunosuppressive effects. However, the related mechanism remains unclear. Here, we explored the therapeutic effect and mechanism of action of human placental-derived MSCs (hPMSCs) on GVHD-induced mouse liver tissue damage, which shows association with inflammatory responses, fibrosis accompanied by hepatocyte tight junction protein loss, the upregulation of Bax, and the downregulation of Bcl-2. It was observed in GVHD mice and Th1 cell differentiation system that hPMSCs treatment increased IL-10 levels and decreased TNF-α levels in the Th1 subsets via CD73. Moreover, hPMSCs treatment reduced tight junction proteins loss and inhibited hepatocyte apoptosis in the livers of GVHD mice via CD73. ADO level analysis in GVHD mice and the Th1 cell differentiation system showed that hPMSCs could also upregulate ADO levels via CD73. Moreover, hPMSCs enhanced Nrf2 expression and diminished Fyn expression via the CD73/ADO pathway in Th1, TNF-α+, and IL-10+ cells. These results indicated that hPMSCs promoted and inhibited the secretion of IL-10 and TNF-α, respectively, during Th1 cell differentiation through the CD73/ADO/Fyn/Nrf2 axis signaling pathway, thereby alleviating liver tissue injury in GVHD mice.
Collapse
Affiliation(s)
- Hengchao Zhang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong Province, China
| | - Kaiyue Han
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong Province, China
| | - Heng Li
- Traditional Chinese Medicine Hospital of Muping District of Yantai City, Yantai, 264100, Shandong Province, China
| | - Jiashen Zhang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong Province, China
| | - Yaxuan Zhao
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong Province, China
| | - Yunhua Wu
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong Province, China
| | - Bin Wang
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong Province, China
| | - Junjie Ma
- Yuhuangding Hospital Affiliated to Qingdao University, Yantai, 264000, Shandong Province, China.
| | - Xiying Luan
- Department of Immunology, Binzhou Medical University, Yantai, 264003, Shandong Province, China.
| |
Collapse
|
6
|
Chen Z, Yao MW, Ao X, Gong QJ, Yang Y, Liu JX, Lian QZ, Xu X, Zuo LJ. The expression mechanism of programmed cell death 1 ligand 1 and its role in immunomodulatory ability of mesenchymal stem cells. Chin J Traumatol 2024; 27:1-10. [PMID: 38065706 PMCID: PMC10859298 DOI: 10.1016/j.cjtee.2023.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 10/30/2023] [Accepted: 11/13/2023] [Indexed: 02/05/2024] Open
Abstract
Programmed cell death 1 ligand 1 (PD-L1) is an important immunosuppressive molecule, which inhibits the function of T cells and other immune cells by binding to the receptor programmed cell death-1. The PD-L1 expression disorder plays an important role in the occurrence, development, and treatment of sepsis or other inflammatory diseases, and has become an important target for the treatment of these diseases. Mesenchymal stem cells (MSCs) are a kind of pluripotent stem cells with multiple differentiation potential. In recent years, MSCs have been found to have a strong immunosuppressive ability and are used to treat various inflammatory insults caused by hyperimmune diseases. Moreover, PD-L1 is deeply involved in the immunosuppressive events of MSCs and plays an important role in the treatment of various diseases. In this review, we will summarize the main regulatory mechanism of PD-L1 expression, and discuss various biological functions of PD-L1 in the immune regulation of MSCs.
Collapse
Affiliation(s)
- Zhuo Chen
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China; College of Basic Medical Sciences, Army Medical University, Chongqing, 400038, China
| | - Meng-Wei Yao
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiang Ao
- Department of Orthopedics, 953 Hospital of PLA, Shigatse Branch of Xinqiao Hospital, Army Medical University, Shigatse, 857000, Tibet Autonomous Region, China
| | - Qing-Jia Gong
- College of Basic Medical Sciences, Army Medical University, Chongqing, 400038, China
| | - Yi Yang
- Department of Rheumatology and Immunology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jin-Xia Liu
- Department of Obstetrics and Gynecology, Chongqing People's Hospital, Chongqing, 401121, China
| | - Qi-Zhou Lian
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xiang Xu
- Department of Stem Cell & Regenerative Medicine, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Ling-Jing Zuo
- Department of Nuclear Medicine, The First People's Hospital of Yunnan province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650034, China.
| |
Collapse
|
7
|
Hoseinzadeh A, Rezaieyazdi Z, Afshari JT, Mahmoudi A, Heydari S, Moradi R, Esmaeili SA, Mahmoudi M. Modulation of Mesenchymal Stem Cells-Mediated Adaptive Immune Effectors' Repertoire in the Recovery of Systemic Lupus Erythematosus. Stem Cell Rev Rep 2023; 19:322-344. [PMID: 36272020 DOI: 10.1007/s12015-022-10452-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2022] [Indexed: 02/07/2023]
Abstract
The breakdown of self-tolerance of the immune response can lead to autoimmune conditions in which chronic inflammation induces tissue damage. Systemic lupus erythematosus (SLE) is a debilitating multisystemic autoimmune disorder with a high prevalence in women of childbearing age; however, SLE incidence, prevalence, and severity are strongly influenced by ethnicity. Although the mystery of autoimmune diseases remains unsolved, disturbance in the proportion and function of B cell subsets has a major role in SLE's pathogenesis. Additionally, colocalizing hyperactive T helper cell subgroups within inflammatory niches are indispensable. Despite significant advances in standard treatments, nonspecific immunosuppression, the risk of serious infections, and resistance to conventional therapies in some cases have raised the urgent need for new treatment strategies. Without the need to suppress the immune system, mesenchymal stem cells (MSCs), as ''smart" immune modulators, are able to control cellular and humoral auto-aggression responses by participating in precursor cell development. In lupus, due to autologous MSCs disorder, the ability of allogenic engrafted MSCs in tissue regeneration and resetting immune homeostasis with the provision of a new immunocyte repertoire has been considered simultaneously. In Brief The bone marrow mesenchymal stem cells (BM-MSCs) lineage plays a critical role in maintaining the hematopoietic stem-cell microstructure and modulating immunocytes. The impairment of BM-MSCs and their niche partially contribute to the pathogenesis of SLE-like diseases. Allogenic MSC transplantation can reconstruct BM microstructure, possibly contributing to the recovery of immunocyte phenotype restoration of immune homeostasis. In terms of future prospects of MSCs, artificially gained by ex vivo isolation and culture adaptation, the wide variety of potential mediators and mechanisms might be linked to the promotion of the immunomodulatory function of MSCs.
Collapse
Affiliation(s)
- Akram Hoseinzadeh
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Rezaieyazdi
- Department of Rheumatology, Ghaem Hospital, Mashhad University of Medical Science, Mashhad, Iran.,Rheumatic Diseases Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Tavakol Afshari
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Mahmoudi
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Heydari
- Department of Physiology and Pharmacology, Faculty of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Reza Moradi
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Immunology, Mashhad University of Medical Sciences, Azadi Square, Kalantari Blvd, Pardi's campusMashhad, Iran.
| |
Collapse
|
8
|
Najafi-Ghalehlou N, Feizkhah A, Mobayen M, Pourmohammadi-Bejarpasi Z, Shekarchi S, Roushandeh AM, Roudkenar MH. Plumping up a Cushion of Human Biowaste in Regenerative Medicine: Novel Insights into a State-of-the-Art Reserve Arsenal. Stem Cell Rev Rep 2022; 18:2709-2739. [PMID: 35505177 PMCID: PMC9064122 DOI: 10.1007/s12015-022-10383-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 12/03/2022]
Abstract
Major breakthroughs and disruptive methods in disease treatment today owe their thanks to our inch by inch developing conception of the infinitive aspects of medicine since the very beginning, among which, the role of the regenerative medicine can on no account be denied, a branch of medicine dedicated to either repairing or replacing the injured or diseased cells, organs, and tissues. A novel means to accomplish such a quest is what is being called "medical biowaste", a large assortment of biological samples produced during a surgery session or as a result of physiological conditions and biological activities. The current paper accentuating several of a number of promising sources of biowaste together with their plausible applications in routine clinical practices and the confronting challenges aims at inspiring research on the existing gap between clinical and basic science to further extend our knowledge and understanding concerning the potential applications of medical biowaste.
Collapse
Affiliation(s)
- Nima Najafi-Ghalehlou
- Department of Medical Laboratory Sciences, Faculty of Paramedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Feizkhah
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammadreza Mobayen
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Pourmohammadi-Bejarpasi
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Shima Shekarchi
- Anatomical Sciences Department, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Amaneh Mohammadi Roushandeh
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| | - Mehryar Habibi Roudkenar
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran.
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
9
|
The Influence of Intervertebral Disc Microenvironment on the Biological Behavior of Engrafted Mesenchymal Stem Cells. Stem Cells Int 2022; 2022:8671482. [DOI: 10.1155/2022/8671482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
Intervertebral disc degeneration is the main cause of low back pain. Traditional treatment methods cannot repair degenerated intervertebral disc tissue. The emergence of stem cell therapy makes it possible to regenerate and repair degenerated intervertebral disc tissue. At present, mesenchymal stem cells are the most studied, and different types of mesenchymal stem cells have their own characteristics. However, due to the harsh and complex internal microenvironment of the intervertebral disc, it will affect the biological behaviors of the implanted mesenchymal stem cells, such as viability, proliferation, migration, and chondrogenic differentiation, thereby affecting the therapeutic effect. This review is aimed at summarizing the influence of each intervertebral disc microenvironmental factor on the biological behavior of mesenchymal stem cells, so as to provide new ideas for using tissue engineering technology to assist stem cells to overcome the influence of the microenvironment in the future.
Collapse
|
10
|
Zhang L, Wang L, Xiang S, Hu Y, Zhao S, Liao Y, Zhu Z, Wu X. CRISPR/Cas9-mediated gene knockout of Sj16 in Schistosoma japonicum eggs upregulates the host-to-egg immune response. FASEB J 2022; 36:e22615. [PMID: 36273308 DOI: 10.1096/fj.202200600rr] [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: 05/07/2022] [Revised: 09/15/2022] [Accepted: 10/04/2022] [Indexed: 11/11/2022]
Abstract
Schistosomiasis is an important, neglected tropical disease. Schistosoma japonicum can evade host attacks by regulating the host's immunity, causing continuous infection. However, interactions between the host's immune system and S. japonicum are unclear. Our previous research found that the Sj16 protein isolated from S. japonicum has an anti-inflammatory effect in the host. However, the role of Sj16 in the regulation of host immunity in S. japonicum infection is not clear. Here, we applied the CRISPR/Cas9 technique to knockout Sj16 in S. japonicum eggs and investigated the effect of Sj16 in regulating host immunity. We found egg viability decreased after Sj16 knockout. In addition, we found granulomatous inflammation increased, the T-cell immune response enhanced and the immune microenvironment changed in mice model injected with Sj16-knockout eggs by tail vein. These findings suggested that S. japonicum could regulate host immunity through Sj16 to evade the host immune attack and cause continuous infection. In addition, we confirmed the application of CRISPR/Cas9-mediated gene reprogramming for functional genomics in S. japonicum.
Collapse
Affiliation(s)
- Lichao Zhang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Lifu Wang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Suoyu Xiang
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Yunyi Hu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Siyu Zhao
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Yao Liao
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Zifeng Zhu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-sen University, Guangzhou, China.,Provincial Engineering Technology Research Center for Biological Vector Control, Guangzhou, China
| | - Xiaoying Wu
- Department of Parasitology of Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Department of Gastroenterology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
11
|
Mirfakhraie R, Ardakani MT, Hajifathali A, Karami S, Moshari MR, Hassani M, Firouz SM, Roshandel E. Highlighting the interaction between immunomodulatory properties of mesenchymal stem cells and signaling pathways contribute to Graft Versus Host Disease management. Transpl Immunol 2022; 71:101524. [PMID: 34990789 DOI: 10.1016/j.trim.2021.101524] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/29/2021] [Accepted: 12/29/2021] [Indexed: 12/11/2022]
Abstract
Background Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) has been increasingly used as a therapeutic approach for hematological malignancies. Several potential strategies have been developed for treating or preventing allo-HSCT complications, specifically graft-versus-host disease (GVHD). GVHD could significantly affect the morbidity and mortality of patients after allo-HSCT. Curative treatment and prophylaxis regimens for GVHD could reduce GVHD incidence and improve survival rate. Among these therapeutic strategies, mesenchymal stem cell (MSCs) mediated immunomodulation has been explored widely in clinical trials. MSCs immunomodulation ability in GVHD correlates with the interactions of MSCs with innate and adaptive immune cells. However, signaling pathways responsible for MSCs' impact on GVHD regulation, like JAK/STAT, NOTCH, MAPK/ERK, and NFκβ signaling pathways, have not been clearly described yet. This review aims to illuminate the effect of MSCs-mediated immunomodulation in GVHD management after allo-HSCT representing the role of MSCs therapy on signaling pathways in GVHD. Conclusion MSCs could potentially modulate immune responses, prevent GVHD, and improve survival after allo-HSCT. Previous studies have investigated different signaling pathways' contributions to MSCs immunoregulatory ability. Accordingly, targeting signaling pathways components involved in MSCs related GVHD regulation is proven to be beneficial.
Collapse
Affiliation(s)
- Reza Mirfakhraie
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maria Tavakoli Ardakani
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Karami
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Reza Moshari
- Department of Anesthesiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassani
- Department of General Surgery, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Mashayekhi Firouz
- Department of Immunology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Gao R, Kong L, Qing P, Cheng K, Chen H, Zhang S, Hu X, Hu Z, Yu F, Qin L. Interleukin-1β as clinically predictive risk marker for recurrent pregnancy loss in women positive for antinuclear antibody. Int J Clin Pract 2021; 75:e14887. [PMID: 34538020 DOI: 10.1111/ijcp.14887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/16/2021] [Indexed: 02/05/2023] Open
Abstract
AIMS Antinuclear antibody (ANA) was found to be associated with recurrent pregnancy loss (RPL). This study was designed to explore the immunological predictive indicators of RPL in women with positive ANA. METHODS A retrospective case-control study in a university hospital from March 2020 to January 2021 was performed, including 56 cases of women with RPL and 56 controls matched for age, all of which were positive for ANA. Levels of cytokines, lymphocyte subsets, immune globulin and complement in peripheral blood among two groups were compared. Statistical analyses in this study were performed using SPSS 25.0. RESULTS The level of IL-1β was higher in RPL women compared with controls according to univariable analysis and multivariable regression logistic analysis (7.67 [5.86-11.35] vs 5.00 [5.00-5.00], P = .000). After the cut-off value of IL-1β was set as 5.66 pg/mL, the prevalence of RPL was significantly elevated in the high IL-1β group as compared with the low IL-1β group (P < .05). The sensitivity and specificity of IL-1β predicting RPL in ANA-positive women were 76.8% and 91.1%, respectively. CONCLUSION Increased IL-1β may play roles in the occurrence of RPL among women with positive ANA. The level of the IL-1β has the potential to be a predictive indicator of RPL in women with positive ANA.
Collapse
Affiliation(s)
- Rui Gao
- The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Linglingli Kong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Pingying Qing
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Kemin Cheng
- Department of Outpatients, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Hanxiao Chen
- The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Sirui Zhang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Xiao Hu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Zhengyan Hu
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Fan Yu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lang Qin
- The Reproductive Medical Center, Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Yu YL, Tseng WK, Fan CW, Chang PH, Kuo HC, Pan YP, Yeh KY. Pretreatment Nutrition-Inflammation Biomarkers Correlated with Differential Cytokine Profiles in Taiwanese Patients with Colorectal Cancer. Nutr Cancer 2021; 74:1614-1624. [PMID: 34323132 DOI: 10.1080/01635581.2021.1957130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Systemic inflammation plays a pivotal role in colorectal cancer (CRC) development. Two hallmarks reflect the severity of inflammation-circulating cytokines and nutrition-inflammation biomarkers (NIBs); however, their interplay has not been fully investigated. In total, 128 CRC patients were included. Ten circulating cytokines (TNF-α, TGF-β, IFN-γ, IL-1β, IL-4, IL-6, IL-10, IL-12, IL-13, and IL-23) and NIBs were analyzed. The relationship between cytokines, NIBs, clinicopathological variables, and overall survival (OS) was assessed using univariate and multivariate analyses. Three NIBs (CRP-to-albumin ratio [CAR]), neutrophil-to-lymphocyte ratio [NLR]), and prognostic nutritional index [PNI]) were associated with OS in univariate analysis; however, CAR was better for OS prediction in multivariate analysis (P = 0.015). None of the serum cytokines analyzed showed a significant association with OS. High CAR (≥0.25) and high IL-10 (≥76.6 pg/mL), high NLR (≥8.2) and high IL-23 (≥51.2 pg/mL), and high PNI (≥42.4) and high IL-1β (≥14.3 pg/mL) values were correlated. CAR, NLR, and PNI were not correlated with each other, whereas circulating cytokines were closely interrelated. High CAR was an independent predictor of poor OS in patients with CRC. Different NIBs have unique cytokine profiles, but show no correlation with each other. There is a close association among the circulating cytokines.
Collapse
Affiliation(s)
- Yen-Lin Yu
- Division of Colorectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Wen-Ko Tseng
- Division of Colorectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chung-Wei Fan
- Division of Colorectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Pei-Hung Chang
- Division of Hemato-oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Hsuan-Chih Kuo
- Division of Hemato-oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Yi-Ping Pan
- Department of Nutrition, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Kun-Yun Yeh
- Division of Hemato-oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| |
Collapse
|
14
|
Zhang A, Zhang J, Li X, Zhang H, Xiong Y, Wang Z, Zhao N, Wang F, Luan X. hPMSCs inhibit the expression of PD-1 in CD4 +IL-10 + T cells and mitigate liver damage in a GVHD mouse model by regulating the crosstalk between Nrf2 and NF-κB signaling pathway. Stem Cell Res Ther 2021; 12:368. [PMID: 34187557 PMCID: PMC8240402 DOI: 10.1186/s13287-021-02407-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 05/21/2021] [Indexed: 02/07/2023] Open
Abstract
Background The activation of T cells and imbalanced redox metabolism enhances the development of graft-versus-host disease (GVHD). Human placenta-derived mesenchymal stromal cells (hPMSCs) can improve GVHD through regulating T cell responses. However, whether hPMSCs balance the redox metabolism of CD4+IL-10+ T cells and liver tissue and alleviate GVHD remains unclear. This study aimed to investigate the effect of hPMSC-mediated treatment of GVHD associated with CD4+IL-10+ T cell generation via control of redox metabolism and PD-1 expression and whether the Nrf2 and NF-κB signaling pathways were both involved in the process. Methods A GVHD mouse model was induced using 6–8-week-old C57BL/6 and Balb/c mice, which were treated with hPMSCs. In order to observe whether hPMSCs affect the generation of CD4+IL-10+ T cells via control of redox metabolism and PD-1 expression, a CD4+IL-10+ T cell culture system was induced using human naive CD4+ T cells. The percentage of CD4+IL-10+ T cells and their PD-1 expression levels were determined in vivo and in vitro using flow cytometry, and Nrf2, HO-1, NQO1, GCLC, GCLM, and NF-κB levels were determined by western blotting, qRT-PCR, and immunofluorescence, respectively. Hematoxylin-eosin, Masson’s trichrome, and periodic acid-Schiff staining methods were employed to analyze the changes in hepatic tissue. Results A decreased activity of superoxide dismutase (SOD) and a proportion of CD4+IL-10+ T cells with increased PD-1 expression were observed in GVHD patients and the mouse model. Treatment with hPMSCs increased SOD activity and GCL and GSH levels in the GVHD mouse model. The percentage of CD4+IL-10+ T cells with decreased PD-1 expression, as well as Nrf2, HO-1, NQO1, GCLC, and GCLM levels, both in the GVHD mouse model and in the process of CD4+IL-10+ T cell generation, were also increased, but NF-κB phosphorylation and nuclear translocation were inhibited after treatment with hPMSCs, which was accompanied by improvement of hepatic histopathological changes. Conclusions The findings suggested that hPMSC-mediated redox metabolism balance and decreased PD-1 expression in CD4+IL-10+ T cells were achieved by controlling the crosstalk between Nrf2 and NF-κB, which further provided evidence for the application of hPMSC-mediated treatment of GVHD.
Collapse
Affiliation(s)
- Aiping Zhang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China
| | - Jiashen Zhang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China
| | - Xiaohua Li
- Department of Component, Yantai Central Blood Station, Yantai, Shandong Province, 264003, People's Republic of China
| | - Hengchao Zhang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China
| | - Yanlian Xiong
- Department of Anatomy, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China
| | - Zhuoya Wang
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China
| | - Nannan Zhao
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China
| | - Feifei Wang
- Department of Anesthesiology, Yantai Affiliated Hospital of Binzhou Medical University, Shandong Province, 264003, Yantai, People's Republic of China.
| | - Xiying Luan
- Department of Immunology, Binzhou Medical University, Yantai, Shandong Province, 264003, People's Republic of China.
| |
Collapse
|
15
|
Pethe P, Kale V. Placenta: A gold mine for translational research and regenerative medicine. Reprod Biol 2021; 21:100508. [PMID: 33930790 DOI: 10.1016/j.repbio.2021.100508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 02/06/2023]
Abstract
Stem cell therapy has gained much impetus in regenerative medicine due to some of the encouraging results obtained in the laboratory as well as in translational/clinical studies. Although stem cells are of various types and their therapeutic potential has been documented in several studies, mesenchymal stromal/stem cells (MSCs) have an edge, as in addition to being multipotent, these cells are easy to obtain and expand, pose fewer ethical issues, and possess immense regenerative potential when used in a scientifically correct manner. Currently, MSCs are being sourced from various tissues such as bone marrow, cord, cord blood, adipose tissue, dental tissue, etc., and, quite often, the choice depends on the availability of the source. One such rich source of tissue suitable for obtaining good quality MSCs in large numbers is the placenta obtained in a full-term delivery leading to a healthy child's birth. Several studies have demonstrated the regenerative potential of human placenta-derived MSCs (hPMSC), and most show that these MSCs possess comparable, in some instances, even better, therapeutic potential as that shown by human bone marrow-derived (hBMSC) or human umbilical cord-derived (hUC-MSC) MSCs. The placenta can be easily sourced from the OB/GYN department of any hospital, and if its derivatives such as hPMSC or their EVs are produced under GMP conditions, it could serve as a gold mine for translational/clinical research. Here, we have reviewed recent studies revealing the therapeutic potential of hPMSC and their extracellular vesicles (EVs) published over the past three years.
Collapse
Affiliation(s)
- Prasad Pethe
- Symbiosis Centre for Stem Cell Research, Symbiosis International University, Pune, 412115, India
| | - Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis International University, Pune, 412115, India.
| |
Collapse
|
16
|
Wang LT, Liu KJ, Sytwu HK, Yen ML, Yen BL. Advances in mesenchymal stem cell therapy for immune and inflammatory diseases: Use of cell-free products and human pluripotent stem cell-derived mesenchymal stem cells. Stem Cells Transl Med 2021; 10:1288-1303. [PMID: 34008922 PMCID: PMC8380447 DOI: 10.1002/sctm.21-0021] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/31/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cell therapy (MSCT) for immune and inflammatory diseases continues to be popular based on progressive accumulation of preclinical mechanistic evidence. This has led to further expansion in clinical indications from graft rejection, autoimmune diseases, and osteoarthritis, to inflammatory liver and pulmonary diseases including COVID‐19. A clear trend is the shift from using autologous to allogeneic MSCs, which can be immediately available as off‐the‐shelf products. In addition, new products such as cell‐free exosomes and human pluripotent stem cell (hPSC)‐derived MSCs are exciting developments to further prevalent use. Increasing numbers of trials have now published results in which safety of MSCT has been largely demonstrated. While reports of therapeutic endpoints are still emerging, efficacy can be seen for specific indications—including graft‐vs‐host‐disease, strongly Th17‐mediated autoimmune diseases, and osteoarthritis—which are more robustly supported by mechanistic preclinical evidence. In this review, we update and discuss outcomes in current MSCT clinical trials for immune and inflammatory disease, as well as new innovation and emerging trends in the field.
Collapse
Affiliation(s)
- Li-Tzu Wang
- Department of Obstetrics & Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, NTU, Taipei, Taiwan, Republic of China
| | - Ko-Jiunn Liu
- National Institute of Cancer Research, National Health Research Institutes (NHRI), Tainan, Taiwan, Republic of China
| | - Huey-Kang Sytwu
- National Institute of Infectious Diseases & Vaccinology, NHRI, Zhunan, Taiwan, Republic of China.,Department & Graduate Institute of Microbiology & Immunology, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Men-Luh Yen
- Department of Obstetrics & Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, NTU, Taipei, Taiwan, Republic of China
| | - B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, NHRI, Zhunan, Taiwan, Republic of China
| |
Collapse
|
17
|
Li M, Soder R, Abhyankar S, Abdelhakim H, Braun MW, Trinidad CV, Pathak HB, Pessetto Z, Deighan C, Ganguly S, Dawn B, McGuirk J, Dunavin N, Godwin AK. WJMSC-derived small extracellular vesicle enhance T cell suppression through PD-L1. J Extracell Vesicles 2021; 10:e12067. [PMID: 33598108 PMCID: PMC7869022 DOI: 10.1002/jev2.12067] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 12/04/2020] [Accepted: 01/13/2021] [Indexed: 12/30/2022] Open
Abstract
Both mesenchymal stem cells (MSCs) and their corresponding small extracellular vesicles (sEVs, commonly referred to as exosomes) share similar immunomodulatory properties that are potentially beneficial for the treatment of acute graft versus host disease (aGvHD). We report that clinical grade Wharton's Jelly‐derived MSCs (WJMSCs) secrete sEVs enriched in programmed death‐ligand 1 (PD‐L1), an essential ligand for an inhibitory immune checkpoint. A rapid increase in circulating sEV‐associated PD‐L1 was observed in patients with aGvHD and was directly associated with the infusion time of clinical grade WJMSCs. In addition, in vitro inhibitory antibody mediated blocking of sEV‐associated PD‐L1 restored T cell activation (TCA), suggesting a functional inhibitory role of sEVs‐PD‐L1. PD‐L1‐deficient sEVs isolated from WJMSCs following CRISPR‐Cas9 gene editing fail to inhibit TCA. Furthermore, we found that PD‐L1 is essential for WJMSC‐derived sEVs to modulate T cell receptors (TCRs). Our study reveals an important mechanism by which therapeutic WJMSCs modulate TCR‐mediated TCA through sEVs or sEV‐carried immune checkpoints. In addition, our clinical data suggest that sEV‐associated PD‐L1 may be not only useful in predicting the outcomes from WJMSC clinical administration, but also in developing cell‐independent therapy for aGvHD patients.
Collapse
Affiliation(s)
- Meizhang Li
- Department of Pathology and Laboratory Medicine University of Kansas Medical Center Kansas City Kansas USA
| | - Rupal Soder
- Midwest Stem Cell Therapy Center University of Kansas Medical Center Kansas City Kansas USA
| | - Sunil Abhyankar
- Midwest Stem Cell Therapy Center University of Kansas Medical Center Kansas City Kansas USA.,Division of Hematologic Malignancies and Cellular Therapeutics University of Kansas Medical Center Kansas City Kansas USA.,The University of Kansas Cancer Center, University of Kansas Medical Center Kansas City Kansas USA
| | - Haitham Abdelhakim
- Division of Hematologic Malignancies and Cellular Therapeutics University of Kansas Medical Center Kansas City Kansas USA
| | - Mitchell W Braun
- Department of Pathology and Laboratory Medicine University of Kansas Medical Center Kansas City Kansas USA
| | - Camille V Trinidad
- Department of Microbiology Molecular Genetics and Immunology Kansas City Kansas USA
| | - Harsh B Pathak
- Department of Pathology and Laboratory Medicine University of Kansas Medical Center Kansas City Kansas USA.,The University of Kansas Cancer Center, University of Kansas Medical Center Kansas City Kansas USA
| | - Ziyan Pessetto
- Department of Pathology and Laboratory Medicine University of Kansas Medical Center Kansas City Kansas USA
| | | | - Siddhartha Ganguly
- Division of Hematologic Malignancies and Cellular Therapeutics University of Kansas Medical Center Kansas City Kansas USA
| | - Buddhadeb Dawn
- Midwest Stem Cell Therapy Center University of Kansas Medical Center Kansas City Kansas USA.,Department of Medicine University of Nevada Las Vegas Nevada USA
| | - Joseph McGuirk
- Division of Hematologic Malignancies and Cellular Therapeutics University of Kansas Medical Center Kansas City Kansas USA.,The University of Kansas Cancer Center, University of Kansas Medical Center Kansas City Kansas USA
| | - Neil Dunavin
- Division of Hematologic Malignancies and Cellular Therapeutics University of Kansas Medical Center Kansas City Kansas USA.,Division of Hematology and Blood and Marrow Transplant University of California San Francisco San Francisco California USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine University of Kansas Medical Center Kansas City Kansas USA.,The University of Kansas Cancer Center, University of Kansas Medical Center Kansas City Kansas USA.,Department of Microbiology Molecular Genetics and Immunology Kansas City Kansas USA
| |
Collapse
|
18
|
Perrichet A, Ghiringhelli F, Rébé C. Understanding Inflammasomes and PD-1/PD-L1 Crosstalk to Improve Cancer Treatment Efficiency. Cancers (Basel) 2020; 12:cancers12123550. [PMID: 33261061 PMCID: PMC7761387 DOI: 10.3390/cancers12123550] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 11/25/2020] [Accepted: 11/25/2020] [Indexed: 12/20/2022] Open
Abstract
Inflammasomes and immune checkpoints have been shown to participate in carcinogenesis, cancer growth and response to treatment. Thus, targeting cytokines resulting from inflammasome activation, such as interleukin (IL)-1β, has emerged as a new tool in the therapeutic arsenal. Moreover, the use of checkpoint inhibitors such as anti-PD-1 or anti-PD-L1 has revolutionized the treatment of some cancer patients. However, inflammasome activation and consecutive cytokine release only occurs in some chemotherapeutic treatments and immune checkpoint inhibitors only work for a restricted number of patients, thus limiting the use of therapies targeting these pathways. Expanding knowledge about the inefficiency of these therapies recently brought forward the hypothesis of targeting both pathways. In this review, we provide an overview of the crosstalk between inflammasomes and programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1) that might explain how these two pathways are mutually dependent, and perhaps why targeting only one of them leads to inefficiency of cancer treatment in some patients.
Collapse
Affiliation(s)
| | | | - Cédric Rébé
- Correspondence: ; Tel.: +33-(0)3-80-73-77-90
| |
Collapse
|