1
|
He J, Yao Y, Wang R, Liu Y, Wan X, Wang H, Zhou Y, Wang W, Ma Y, Lv X. Enhanced renal ischemia/reperfusion injury repair potential of exosomes derived from B7-H1 high mesenchymal stem cells. Front Genet 2025; 16:1516626. [PMID: 40242472 PMCID: PMC12000007 DOI: 10.3389/fgene.2025.1516626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/11/2025] [Indexed: 04/18/2025] Open
Abstract
Two subgroups with high expression of B7-H1 and low expression of B7-H1 were successfully isolated from primitive human umbilical cord mesenchymal stem cells. And exosomes with high B7-H1 expression and low B7-H1 expression were successfully isolated. In comparison to the sham-operated group, mice in the IRI group demonstrated elevated serum levels of blood urea nitrogen (BUN) and serum creatinine (Scr), accompanied by a more pronounced degree of renal tissue damage. The administration of exosomes via the tail vein markedly accelerated the recovery of renal function in IRI mice, with the therapeutic effect beingmore pronounced in those treated with B7-H1high-Exo. Moreover RNA sequencing of mouse kidney treated with B7-H1high-Exo and B7-H1low-Exo showed that eight genes (C3, IRF7, AREG, CXCL10, Aldh1l2, Fnip2, Vcam1, St6galnac3) were involved in the pathophysiological process of ischemia-reperfusion injury. The in vitro and in vivo experiments showed that the expression level of C3 protein was significantly decreased, which indicated that B7-H1high-Exo played a therapeutic role by down-regulating C3.
Collapse
Affiliation(s)
- Jiahui He
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yawei Yao
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Ruiyan Wang
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yujia Liu
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xingyu Wan
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Hao Wang
- Department of Day Surgery Center, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yuqiang Zhou
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Wenjing Wang
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yan Ma
- Department of Anaesthesia, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xinghua Lv
- Department of Day Surgery Center, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
2
|
Dai L, Wang Q. Targeting ferroptosis: opportunities and challenges of mesenchymal stem cell therapy for type 1 diabetes mellitus. Stem Cell Res Ther 2025; 16:47. [PMID: 39901210 PMCID: PMC11792594 DOI: 10.1186/s13287-025-04188-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/24/2025] [Indexed: 02/05/2025] Open
Abstract
Type 1 diabetes mellitus (T1DM) is characterized by progressive β-cell death, leading to β-cell loss and insufficient insulin secretion. Mesenchymal stem cells (MSCs) transplantation is currently one of the most promising methods for β-cell replacement therapy. However, recent studies have shown that ferroptosis is not only one of the key mechanisms of β-cell death, but also one of the reasons for extensive cell death within a short period of time after MSCs transplantation. Ferroptosis is a new type of regulated cell death (RCD) characterized by iron-dependent accumulation of lipid peroxides. Due to the weak antioxidant capacity of β-cells, they are susceptible to cytotoxic stimuli such as oxidative stress (OS), and are therefore susceptible to ferroptosis. Transplanted MSCs are also extremely susceptible to perturbations in their microenvironment, especially OS, which can weaken their antioxidant capacity and induce MSCs death through ferroptosis. In the pathophysiological process of T1DM, a large amount of reactive oxygen species (ROS) are produced, causing OS. Therefore, targeting ferroptosis may be a key way to protect β-cells and improve the therapeutic effect of MSCs transplantation. This review reviews the research related to ferroptosis of β-cells and MSCs, and summarizes the currently developed strategies that help inhibit cell ferroptosis. This study aims to help understand the ferroptosis mechanism of β-cell death and MSCs death after transplantation, emphasize the importance of targeting ferroptosis for protecting β-cells and improving the survival and function of transplanted MSCs, and provide a new research direction for stem cells transplantation therapy of T1DM in the future.
Collapse
Affiliation(s)
- Le Dai
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, 126 Xiantai Avenue, Changchun City, Jilin Province, China
| | - Qing Wang
- Department of Endocrinology, China-Japan Union Hospital of Jilin University, 126 Xiantai Avenue, Changchun City, Jilin Province, China.
| |
Collapse
|
3
|
Lin S, Luo Y, Mao X, He W, Xu C, Zeng M. Homeobox B4 optimizes the therapeutic effect of bone marrow mesenchymal stem cells on endotoxin-associated acute lung injury in rats. Am J Med Sci 2024; 368:242-252. [PMID: 38795966 DOI: 10.1016/j.amjms.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: 05/03/2023] [Revised: 04/05/2024] [Accepted: 05/20/2024] [Indexed: 05/28/2024]
Abstract
BACKGROUND Alveolar capillary endothelial cell (EC) injury has a pivotal role in driving acute respiratory distress syndrome (ARDS) progression and maintaining endothelial homeostasis. A previous ex vivo study revealed that overexpression of homeobox B4 (HOXB4) in bone marrow mesenchymal stem cells (BMSCs) enhanced protection against lipopolysaccharide (LPS)-induced EC injury by activating the Wnt/β-catenin pathway. This in vivo study was performed to verify whether BMSCs overexpressing HOXB4 exert similar protective effects on LPS-induced acute lung injury (ALI) in an animal model. METHODS The ALI rat model was established by intraperitoneal injection of LPS. Wildtype BMSCs or BMSCs overexpressing HOXB4 were then injected via the tail vein. The lung characteristics of rats were visualized by computed tomography. Lung histopathological characteristics and collagen deposition were assessed by hematoxylin-eosin and Masson's staining, respectively, which were combined with the lung wet/dry ratio and proinflammatory factor levels in bronchoalveolar lavage fluid to further evaluate therapeutic effects. Expression of β-catenin and VE-cadherin was assessed by western blotting and immunofluorescence. RESULTS Compared with wildtype BMSCs, overexpression of HOXB4 optimized the therapeutic effects of BMSCs, which manifested as improvements in lung exudation and histopathological features, reduced lung collagen deposition, amelioration of lung permeability, attenuation of lung inflammation, and enhanced expression of β-catenin and VE-cadherin proteins. CONCLUSIONS HOXB4-overexpressing BMSCs optimized the protective effect against LPS-induced ALI by partially activating Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Shan Lin
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, PR China; Department of Respiratory and Critical Care Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, PR China
| | - Yuling Luo
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, PR China
| | - Xueyan Mao
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, PR China
| | - Wanmei He
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, PR China
| | - Caixia Xu
- Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Mian Zeng
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, PR China.
| |
Collapse
|
4
|
Sun Y, Zhao J, Liu Q, Xu Y, Qin Y, He R, Zheng L, Xie Y, Li C, Wu T, Cao Y, Duan C, Lu H, Hu J. Intranasal delivery of small extracellular vesicles from specific subpopulation of mesenchymal stem cells mitigates traumatic spinal cord injury. J Control Release 2024; 369:335-350. [PMID: 38519036 DOI: 10.1016/j.jconrel.2024.03.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Vascular injury following spinal cord injury (SCI) can significantly exacerbate secondary SCI and result in neurological dysfunction. Strategies targeting angiogenesis have demonstrated potential in enhancing functional recovery post-SCI. In the context of angiogenesis, the CD146+ and CD271+ subpopulations of mesenchymal stem cells (MSCs) have been recognized for their angiogenic capabilities in tissue repair. Small extracellular vesicles (sEVs) derived from MSCs are nanoscale vesicles containing rich bioactive components that play a crucial role in tissue regeneration. However, the precise role of sEVs derived from CD146+CD271+ UCMSCs (CD146+CD271+ UCMSC-sEVs) in SCI remain unclear. In this study, CD146+CD271+ UCMSC-sEVs were non-invasively administered via intranasal delivery, demonstrating a significant capacity to stimulate angiogenesis and improve functional recovery in mice following SCI. Furthermore, in vitro assessments revealed the effective enhancement of migration and tube formation capabilities of the murine brain microvascular endothelial cell line (bEnd.3) by CD146+CD271+UCMSC-sEVs. MicroRNA array analysis confirmed significant enrichment of multiple microRNAs within CD146+CD271+ UCMSC-sEVs. Subsequent in vivo and in vitro experiments demonstrated that CD146+CD271+ UCMSC-sEVs promote enhanced angiogenesis and improved functional recovery mediated by miR-27a-3p. Further mechanistic studies revealed that miR-27a-3p sourced from CD146+CD271+ UCMSC-sEVs enhances migration and tube formation of bEnd.3 cells in vitro by suppressing the expression of Delta Like Canonical Notch Ligand 4 (DLL4), thereby promoting angiogenesis in vivo. Collectively, our results demonstrate that a crucial role of CD146+CD271+ UCMSC-sEVs in inhibiting DLL4 through the transfer of miR-27a-3p, which leads to the promotion of angiogenesis and improved functional recovery after SCI.
Collapse
Affiliation(s)
- Yi Sun
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Jinyun Zhao
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Quanbo Liu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Yan Xu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Yiming Qin
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Rundong He
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Lifu Zheng
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Yong Xie
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Chengjun Li
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Tianding Wu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Yong Cao
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Chunyue Duan
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China
| | - Hongbin Lu
- Department of Sports Medicine, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China.
| | - Jianzhong Hu
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Road 87, Changsha 410008, China; Hunan Engineering Research Center of Sports and Health, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, China.
| |
Collapse
|
5
|
Wang Q, Li Y, Yuan H, Peng L, Dai Z, Sun Y, Liu R, Li W, Li J, Zhu C. Hypoxia preconditioning of human amniotic mesenchymal stem cells enhances proliferation and migration and promotes their homing via the HGF/C-MET signaling axis to augment the repair of acute liver failure. Tissue Cell 2024; 87:102326. [PMID: 38442547 DOI: 10.1016/j.tice.2024.102326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/17/2024] [Accepted: 02/06/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Transplantation of mesenchymal stem cells (MSCs) is a newly developed strategy for treating acute liver failure (ALF). Nonetheless, the low survival rate of MSCs after transplantation and their poor homing to damaged tissues limit the clinical application of MSCs. The research assessed whether hypoxic preconditioning (HPC) can improve the biological activity of human amniotic mesenchymal stem cells (hA-MSCs), promote their homing ability to the liver of mice with ALF, and influence liver tissue repair. METHODS Flow cytometry, CCK8, Transwell, and Western blotting assays were conducted to assess the effects of hypoxic preconditioning on the phenotype, proliferation, and migration of hA-MSCs and the changes in the c-Met and CXCR4 gene expression levels were studied. To evaluate the effects of the transplantation of hypoxic preconditioning of hA-MSCs on the homing and repair of D-galactosamine (D-GalN)/LPS-induced ALF, the mechanism was elucidated by adding c-Met, CXCR4-specific blockers (SU11274 and AMD3100). RESULTS After hypoxia pretreatment (1% oxygen volume fraction), hA-MSCs maintained the morphological characteristics of adherence and vortex colony growth and showed high CD44, CD90, and CD105 and low CD31, CD34, and CD45 expression levels. Hypoxic preconditioning of hA-MSCs significantly increased their proliferation and migration and highly expressed the c-Met and CXCR4 genes. In vivo and in vitro, this migration-promoting effect was suppressed by the c-Met specific blocker SU11274. In the acute liver failure mouse model, the HGF expression level was considerably elevated in the liver than that in the serum, lungs and kidneys. The transplantation of hypoxic preconditioned hA-MSCs introduced a remarkable improvement in the liver function and survival rate of mice with ALF and enhanced the anti-apoptosis ability of liver cells. The anti-apoptotic enhancing effect of hypoxic preconditioning was suppressed by the c-Met specific blocker SU11274. Hypoxic hA-MSCs administration was observed to have considerably increased the fluorescent cells in the liver than that recorded after administering normal oxygen-hA-MSCs. The number of hepatic fluorescent cells decreased remarkably after adding the c-Met inhibitor SU11274, compared to that recorded after hypoxic pretreatment, whereas the effect of c-Met inhibitor SU11274 on normal oxygen-hA-MSCs was not significant. CONCLUSIONS Hypoxic preconditioning depicted no impact on the morphology and phenotype features of the human amniotic mesenchymal stem cells, but it can promote their proliferation, migration, anti-apoptotic effect, and homing rate and improve the repair of acute liver failure, which might be mediated by the HGF/c-Met signaling axis.
Collapse
Affiliation(s)
- Qian Wang
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuwen Li
- Department of Pediatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hui Yuan
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Linya Peng
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zixing Dai
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ye Sun
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Rui Liu
- Department of Infectious and Tropical Diseases, The Second Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Wenting Li
- Department of Infectious and Tropical Diseases, The Second Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Jun Li
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Chuanlong Zhu
- Department of Infectious Disease, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Infectious and Tropical Diseases, The Second Affiliated Hospital of Hainan Medical University, Hainan, China.
| |
Collapse
|
6
|
Long R, Wang S. Exosomes from preconditioned mesenchymal stem cells: Tissue repair and regeneration. Regen Ther 2024; 25:355-366. [PMID: 38374989 PMCID: PMC10875222 DOI: 10.1016/j.reth.2024.01.009] [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: 12/14/2023] [Revised: 01/14/2024] [Accepted: 01/25/2024] [Indexed: 02/21/2024] Open
Abstract
As a prominent research area in tissue repair and regeneration, mesenchymal stem cells (MSCs) have garnered substantial attention for their potential in the treatment of various diseases. It is now widely recognized that the therapeutic effects of MSCs primarily occur through paracrine mechanisms. Among these mechanisms, exosomes play a crucial role by exerting a series of regulatory effects on surrounding cells and tissues. While exosomes have shown promise in treating various diseases, they do have some limitations, such as limited secretion, poor targeting, and single functionality. However, MSC preconditioning can enhance the production of exosomes, lead to more stable functionality and improve therapeutic effects. Moreover, exosomes could also serve as carriers for specific drugs or genes, enabling more precise treatments of diseases. This review summarizes the most recent literatures on how preconditioning of MSCs influences the regenerative potential of their exosomes in tissue repair and provides new insights into the therapeutic application of exosomes derived from MSCs.
Collapse
Affiliation(s)
- Ruili Long
- School and Hospital of Stomatology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Shuai Wang
- School and Hospital of Stomatology, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
7
|
Kim SW, Lim KM, Cho SG, Ryu B, Kim CY, Park SY, Jang K, Jung JH, Park C, Choi C, Kim JH. Efficacy of Allogeneic and Xenogeneic Exosomes for the Treatment of Canine Atopic Dermatitis: A Pilot Study. Animals (Basel) 2024; 14:282. [PMID: 38254451 PMCID: PMC10812568 DOI: 10.3390/ani14020282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Canine atopic dermatitis (CAD) is a genetically predisposed inflammatory pruritic skin disease. The available treatments for CAD have several adverse effects and vary in efficacy, indicating the need for the development of improved treatments. In this study, we aimed to elucidate the therapeutic effects of allogeneic and xenogeneic exosomes on CAD. Six laboratory beagle dogs with CAD were randomly assigned to three treatment groups: control, canine exosome (cExos), or human exosome (hExos) groups. Dogs in the cExos and hExos groups were intravenously administered 1.5 mL of cExos (5 × 1010) and hExos (7.5 × 1011) solutions, respectively, while those in the control group were administered 1.5 mL of normal saline three times per week for 4 weeks. Skin lesion score and transepidermal water loss decreased in cExos and hExos groups compared with those in the control group. The exosome treatments decreased the serum levels of inflammatory cytokines (interferon-γ, interleukin-2, interleukin-4, interleukin-12, interleukin-13, and interleukin-31) but increased those of anti-inflammatory cytokines (interleukin-10 and transforming growth factor-β), indicating the immunomodulatory effect of exosomes. Skin microbiome analysis revealed that the exosome treatments alleviated skin bacterial dysbiosis. These results suggest that allogeneic and xenogeneic exosome therapy may alleviate CAD in dogs.
Collapse
Affiliation(s)
- Sang-Won Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea;
| | - Kyung-Min Lim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029, Republic of Korea; (K.-M.L.); (S.-G.C.)
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, Seoul 05029, Republic of Korea; (K.-M.L.); (S.-G.C.)
| | - Bokyeong Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (B.R.); (C.-Y.K.)
- Department of Biomedical Informatics, College of Applied Life Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - C-Yoon Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea; (B.R.); (C.-Y.K.)
| | - Seon Young Park
- ILIAS Biologics Inc., Daejeon 34014, Republic of Korea; (S.Y.P.); (K.J.); (J.H.J.); (C.P.); (C.C.)
| | - Kyungmin Jang
- ILIAS Biologics Inc., Daejeon 34014, Republic of Korea; (S.Y.P.); (K.J.); (J.H.J.); (C.P.); (C.C.)
| | - Jae Heon Jung
- ILIAS Biologics Inc., Daejeon 34014, Republic of Korea; (S.Y.P.); (K.J.); (J.H.J.); (C.P.); (C.C.)
| | - Cheolhyoung Park
- ILIAS Biologics Inc., Daejeon 34014, Republic of Korea; (S.Y.P.); (K.J.); (J.H.J.); (C.P.); (C.C.)
| | - Chulhee Choi
- ILIAS Biologics Inc., Daejeon 34014, Republic of Korea; (S.Y.P.); (K.J.); (J.H.J.); (C.P.); (C.C.)
| | - Jung-Hyun Kim
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea;
| |
Collapse
|
8
|
Li J, He X, Liu F, Zheng X, Jiang J. Tumor Necrosis Factor-α-Induced Protein-8-like 2 Transfected Adipose-Derived Stem Cells Regulated the Dysfunction of Monocrotaline Pyrrole-Induced Pulmonary Arterial Smooth Muscle Cells and Pulmonary Arterial Endothelial Cells. J Cardiovasc Pharmacol 2024; 83:73-85. [PMID: 38180455 DOI: 10.1097/fjc.0000000000001505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 10/12/2023] [Indexed: 01/06/2024]
Abstract
ABSTRACT Pulmonary arterial hypertension (PAH) is characterized by pulmonary arterial endothelial cell (PAEC) dysfunction and pulmonary arterial smooth muscle cell (PASMC) activation. For decades, the therapies for PAH based on stem cells have been shown to be effective. Meanwhile, tumor necrosis factor-α-induced protein-8-like 2 (TIPE2) promote the viability of human amniotic mesenchymal stem cells. Therefore, we aimed to explore the role of TIPE2 in adipose-derived stem cells (ADSCs) and the function of TIPE2-transfected ADSCs in the regulation of PAH. We first explored the role and underlying molecular mechanism of TIPE2 in viability and migration of ADSCs. Moreover, the ADSCs transfected with TIPE2 were cocultured with monocrotaline pyrrole (MCTP)-stimulated PASMCs or PAECs. The effects and mechanisms of TIPE2-transfected ADSCs on MCTP-induced PASMCs and PAECs were further investigated. The results showed that TIPE2 overexpression promoted viability and migration of ADSCs by activating the TLR4-ERK1/2 pathway. In addition, TIPE2-transfected ADSCs inhibited the abnormal proliferation and the impaired apoptosis of PASMCs via NF-κB signaling and promoted the conversion of PASMCs from synthetic to contractile. Meanwhile, TIPE2-transfected ADSCs reduced the apoptosis, endothelial-to-mesenchymal transition, and migration of PAECs via PI3K/AKT signaling after MCTP treatment. MCTP-induced oxidative stress and inflammation of PAECs were significantly decreased by TIPE2-transfected ADSCs. In rat model, TIPE2-ADSCs administration further decreased the monocrotaline-induced increase in the right ventricular systolic pressure and ratio of right ventricle weight/left ventricle and septa weight (L + S) and right ventricle weight/body weight compared with the ADSCs group. In conclusion, TIPE2-transfected ADSCs dramatically attenuated the PAH via inhibiting the dysfunction of PASMCs and PAECs.
Collapse
Affiliation(s)
- Jing Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; and
| | - Xin He
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; and
| | - Feng Liu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; and
| | - Xinglong Zheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China; and
| | - Jing Jiang
- Department of Pediatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
9
|
WEN JY, PENG HX, WANG D, WEN ZM, LIU YT, QU J, CUI HX, WANG YY, DU YL, WANG T, GENG C, XU B. Lipopolysaccharides protect mesenchymal stem cell against cardiac ischemia-reperfusion injury by HMGB1/STAT3 signaling. J Geriatr Cardiol 2023; 20:801-812. [PMID: 38098470 PMCID: PMC10716610 DOI: 10.26599/1671-5411.2023.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Myocardial ischemia-reperfusion (I/R) is a serious and irreversible injury. Bone marrow-derived mesenchymal stem cells (MSCs) is considered to be a potential therapy for I/R injury due to the paracrine effects. High-mobility group box 1 (HMGB1) is a novel mediator in MSC and regulates the response of inflammation injury. Signal Transduction and Transcription Activator 3 (STAT3) is a critical transcription factor and important for release of paracrine factors. However, the relationship between HMGB1 and STAT3 in paracrine effect of MSC remains unknown. METHODS In vitro, hypoxia/reoxygenation injury model was established by AnaeroPack System and examined by Annexin V flow cytometry, CCK8 assay and morphology observation. Detection of apoptotic proteins and protein expression of HMGB1 and STAT3 by Western blot. RESULTS The conditioned medium of MSCs with or without LPS pretreatment was cocultured with H9C2 cells for 24 h before hypoxia treatment and MSC showed obvious cardiomyocytes protect role, as evidence by decreased apoptosis rate and improved cells viability, and LPS pretreated MSC exhibited better protect role than untreated MSC. However, such effect was abolished in HMGB1 deficiency group, silencing HMGB1 decreased the secretion of vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF), insulin growth factor (IGF), cell viability, and the expression of STAT3. Furthermore, STAT3 silence attenuated the protective effect of LPS in MSC. CONCLUSIONS These findings suggested that LPS improved MSC-mediated cardiomyocytes protection by HMGB1/STAT3 signaling.
Collapse
Affiliation(s)
- Jing-Yi WEN
- Department of Clinical Pharmacy, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Pharmacy, the Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Hui-Xi PENG
- Department of Clinical Pharmacy, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Dan WANG
- Department of Pharmacy, Ordos Central Hospital, Ordos, Inner Mongolia, China
| | - Zhi-Min WEN
- Department of Clinical Laboratory, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yu-Tong LIU
- Department of Clinical Laboratory, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jian QU
- Department of Clinical Laboratory, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hong-Xuan CUI
- Department of Clinical Pharmacy, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yu-Ying WANG
- Department of Clinical Pharmacy, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yan-Lin DU
- Department of Clinical Pharmacy, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ting WANG
- Department of Clinical Laboratory, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Cong GENG
- Department of Clinical Laboratory, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bing XU
- Department of Clinical Pharmacy, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
10
|
Miceli V, Zito G, Bulati M, Gallo A, Busà R, Iannolo G, Conaldi PG. Different priming strategies improve distinct therapeutic capabilities of mesenchymal stromal/stem cells: Potential implications for their clinical use. World J Stem Cells 2023; 15:400-420. [PMID: 37342218 PMCID: PMC10277962 DOI: 10.4252/wjsc.v15.i5.400] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/07/2023] [Accepted: 04/17/2023] [Indexed: 05/26/2023] Open
Abstract
Mesenchymal stromal/stem cells (MSCs) have shown significant therapeutic potential, and have therefore been extensively investigated in preclinical studies of regenerative medicine. However, while MSCs have been shown to be safe as a cellular treatment, they have usually been therapeutically ineffective in human diseases. In fact, in many clinical trials it has been shown that MSCs have moderate or poor efficacy. This inefficacy appears to be ascribable primarily to the heterogeneity of MSCs. Recently, specific priming strategies have been used to improve the therapeutic properties of MSCs. In this review, we explore the literature on the principal priming approaches used to enhance the preclinical inefficacy of MSCs. We found that different priming strategies have been used to direct the therapeutic effects of MSCs toward specific pathological processes. Particularly, while hypoxic priming can be used primarily for the treatment of acute diseases, inflammatory cytokines can be used mainly to prime MSCs in order to treat chronic immune-related disorders. The shift in approach from regeneration to inflammation implies, in MSCs, a shift in the production of functional factors that stimulate regenerative or anti-inflammatory pathways. The opportunity to fine-tune the therapeutic properties of MSCs through different priming strategies could conceivably pave the way for optimizing their therapeutic potential.
Collapse
Affiliation(s)
- Vitale Miceli
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy.
| | - Giovanni Zito
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Matteo Bulati
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Alessia Gallo
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Rosalia Busà
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Gioacchin Iannolo
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| | - Pier Giulio Conaldi
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad alta Specializzazione), Palermo 90127, Italy
| |
Collapse
|
11
|
Kang MJ, Cho YW, Kim TH. Progress in Nano-Biosensors for Non-Invasive Monitoring of Stem Cell Differentiation. BIOSENSORS 2023; 13:bios13050501. [PMID: 37232862 DOI: 10.3390/bios13050501] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/20/2023] [Accepted: 04/22/2023] [Indexed: 05/27/2023]
Abstract
Non-invasive, non-destructive, and label-free sensing techniques are required to monitor real-time stem cell differentiation. However, conventional analysis methods, such as immunocytochemistry, polymerase chain reaction, and Western blot, involve invasive processes and are complicated and time-consuming. Unlike traditional cellular sensing methods, electrochemical and optical sensing techniques allow non-invasive qualitative identification of cellular phenotypes and quantitative analysis of stem cell differentiation. In addition, various nano- and micromaterials with cell-friendly properties can greatly improve the performance of existing sensors. This review focuses on nano- and micromaterials that have been reported to improve sensing capabilities, including sensitivity and selectivity, of biosensors towards target analytes associated with specific stem cell differentiation. The information presented aims to motivate further research into nano-and micromaterials with advantageous properties for developing or improving existing nano-biosensors to achieve the practical evaluation of stem cell differentiation and efficient stem cell-based therapies.
Collapse
Affiliation(s)
- Min-Ji Kang
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Yeon-Woo Cho
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| | - Tae-Hyung Kim
- School of Integrative Engineering, Chung-Ang University, 84 Heukseuk-ro, Dongjak-gu, Seoul 06974, Republic of Korea
| |
Collapse
|
12
|
Chen P, Tang S, Li M, Wang D, Chen C, Qiu Y, Fang Z, Zhang H, Gao H, Weng H, Hu K, Lin J, Lin Q, Tan Y, Li S, Chen J, Chen L, Chen X. Single-Cell and Spatial Transcriptomics Decodes Wharton's Jelly-Derived Mesenchymal Stem Cells Heterogeneity and a Subpopulation with Wound Repair Signatures. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204786. [PMID: 36504438 PMCID: PMC9896049 DOI: 10.1002/advs.202204786] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/18/2022] [Indexed: 06/17/2023]
Abstract
The highly heterogeneous characteristics of Wharton's jelly mesenchymal stem cells (WJ-MSCs) may be responsible for the poor clinical outcomes and poor reproducibility of treatments based on WJ-MSCs. Exploration of WJ-MSC heterogeneity with multimodal single-cell technologies will aid in establishing accurate MSC subtyping and developing screening protocols for dominant functional subpopulations. Here, the characteristics of WJ-MSCs are systematically analyzed by single cell and spatial transcriptome sequencing. Single-cell transcriptomics analysis identifies four WJ-MSC subpopulations, namely proliferative_MSCs, niche-supporting_MSCs, metabolism-related_MSCs and biofunctional-type_MSCs. Furthermore, the transcriptome, cellular heterogeneity, and cell-state trajectories of these subpopulations are characterized. Intriguingly, the biofunctional-type MSCs (marked by S100A9, CD29, and CD142) selected in this study exhibit promising wound repair properties in vitro and in vivo. Finally, by integrating omics data, it has been found that the S100A9+ CD29+ CD142+ subpopulation is more enriched in the fetal segment of the umbilical cord, suggesting that this subpopulation deriving from the fetal segment may have potential for developing into an ideal therapeutic agent for wound healing. Overall, the presented study comprehensively maps the heterogeneity of WJ-MSCs and provides an essential resource for future development of WJ-MSC-based drugs.
Collapse
|
13
|
Yeung CK, Yan Y, Yan L, Duan Y, Li E, Huang B, Lu K, Li K, Zhou M, Zhang L, Wu Y, Luo KQ, Ji W, Xu RH, Si W. Preclinical safety evaluation and tracing of human mesenchymal stromal cell spheroids following intravenous injection into cynomolgus monkeys. Biomaterials 2022; 289:121759. [DOI: 10.1016/j.biomaterials.2022.121759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 06/30/2022] [Accepted: 08/19/2022] [Indexed: 11/02/2022]
|
14
|
Lin Z, Rao Z, Chen J, Chu H, Zhou J, Yang L, Quan D, Bai Y. Bioactive Decellularized Extracellular Matrix Hydrogel Microspheres Fabricated Using a Temperature-Controlling Microfluidic System. ACS Biomater Sci Eng 2022; 8:1644-1655. [PMID: 35357124 DOI: 10.1021/acsbiomaterials.1c01474] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hydrogel microspheres have drawn great attention as functional three-dimensional (3D) microcarriers for cell attachment and growth, which have shown great potential in cell-based therapies and biomedical research. Hydrogels derived from a decellularized extracellular matrix (dECM) retain the intrinsic physical and biological cues from the native tissues, which often exhibit high bioactivity and tissue-specificity in promoting tissue regeneration. Herein, a novel two-stage temperature-controlling microfluidic system was developed which enabled production of pristine dECM hydrogel microspheres in a high-throughput manner. Porcine decellularized peripheral nerve matrix (pDNM) was used as the model raw dECM material for continuous generation of pDNM microgels without additional supporting materials or chemical crosslinking. The sizes of the microspheres were well-controlled by tuning the feed ratios of water/oil phases into the microfluidic device. The resulting pDNM microspheres (pDNM-MSs) were relatively stable, which maintained a spherical shape and a nanofibrous ultrastructure for at least 14 days. Schwann cells and PC12 cells preseeded on the pDNM-MSs not only showed excellent viability and an adhesive property, but also promoted cell extension compared to the commercially available gelatin microspheres. Moreover, primary neural stem/progenitor cells attached well to the pDNM-MSs, which further facilitated their proliferation. The successfully fabricated dECM hydrogel microspheres provided a highly bioactive microenvironment for 3D cell culture and functionalization, which showed promising potential in versatile biomedical applications.
Collapse
Affiliation(s)
- Zudong Lin
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Zilong Rao
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Jiaxin Chen
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Hanyu Chu
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Jing Zhou
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Liqun Yang
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Daping Quan
- PCFM Lab, GD HPPC Lab, School of Chemistry, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China.,Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| | - Ying Bai
- Guangdong Engineering Technology Research Centre for Functional Biomaterials, School of Materials Science and Engineering, Sun Yat-sen University, 132 Waihuan West Road, HEMC, Guangzhou 510006, China
| |
Collapse
|
15
|
Zhang Q, Wan XX, Hu XM, Zhao WJ, Ban XX, Huang YX, Yan WT, Xiong K. Targeting Programmed Cell Death to Improve Stem Cell Therapy: Implications for Treating Diabetes and Diabetes-Related Diseases. Front Cell Dev Biol 2021; 9:809656. [PMID: 34977045 PMCID: PMC8717932 DOI: 10.3389/fcell.2021.809656] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
Stem cell therapies have shown promising therapeutic effects in restoring damaged tissue and promoting functional repair in a wide range of human diseases. Generations of insulin-producing cells and pancreatic progenitors from stem cells are potential therapeutic methods for treating diabetes and diabetes-related diseases. However, accumulated evidence has demonstrated that multiple types of programmed cell death (PCD) existed in stem cells post-transplantation and compromise their therapeutic efficiency, including apoptosis, autophagy, necroptosis, pyroptosis, and ferroptosis. Understanding the molecular mechanisms in PCD during stem cell transplantation and targeting cell death signaling pathways are vital to successful stem cell therapies. In this review, we highlight the research advances in PCD mechanisms that guide the development of multiple strategies to prevent the loss of stem cells and discuss promising implications for improving stem cell therapy in diabetes and diabetes-related diseases.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Xin-xing Wan
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha, China
| | - Xi-min Hu
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Wen-juan Zhao
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Xiao-xia Ban
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yan-xia Huang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Wei-tao Yan
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
16
|
Kotikalapudi N, Sampath SJP, Sukesh Narayan S, R B, Nemani H, Mungamuri SK, Venkatesan V. The promise(s) of mesenchymal stem cell therapy in averting preclinical diabetes: lessons from in vivo and in vitro model systems. Sci Rep 2021; 11:16983. [PMID: 34417511 PMCID: PMC8379204 DOI: 10.1038/s41598-021-96121-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity (Ob) poses a significant risk factor for the onset of metabolic syndrome with associated complications, wherein the Mesenchymal Stem Cell (MSC) therapy shows pre-clinical success. Here, we explore the therapeutic applications of human Placental MSCs (P-MSCs) to address Ob-associated Insulin Resistance (IR) and its complications. In the present study, we show that intramuscular injection of P-MSCs homed more towards the visceral site, restored HOMA-IR and glucose homeostasis in the WNIN/GR-Ob (Ob-T2D) rats. P-MSC therapy was effective in re-establishing the dysregulated cytokines. We report that the P-MSCs activates PI3K-Akt signaling and regulates the Glut4-dependant glucose uptake and its utilization in WNIN/GR-Ob (Ob-T2D) rats compared to its control. Our data reinstates P-MSC treatment's potent application to alleviate IR and restores peripheral blood glucose clearance evidenced in stromal vascular fraction (SVF) derived from white adipose tissue (WAT) of the WNIN/GR-Ob rats. Gaining insights, we show the activation of the PI3K-Akt pathway by P-MSCs both in vivo and in vitro (palmitate primed 3T3-L1 cells) to restore the insulin sensitivity dysregulated adipocytes. Our findings suggest a potent application of P-MSCs in pre-clinical/Ob-T2D management.
Collapse
Affiliation(s)
- Nagasuryaprasad Kotikalapudi
- Division of Cell and Molecular Biology, ICMR-National Institute of Nutrition, Jamai-Osmania P.O., Tarnaka, Hyderabad, 500007, India
| | - Samuel Joshua Pragasam Sampath
- Division of Cell and Molecular Biology, ICMR-National Institute of Nutrition, Jamai-Osmania P.O., Tarnaka, Hyderabad, 500007, India
| | - Sinha Sukesh Narayan
- Division of Food Safety, ICMR-National Institute of Nutrition, Jamai-Osmania P.O., Tarnaka, Hyderabad, 500007, India
| | - Bhonde R
- Department of Regenerative Medicine, Manipal Institute of Regenerative Medicine, GKVK Post, Bellary Road, Allalasandra, Yelahanka, Bangalore, 560065, India
- Dr. D. Y. Patil Vidyapeeth, Pune, 411018, India
| | - Harishankar Nemani
- Division of Animal Facility, ICMR-National Institute of Nutrition, Jamai-Osmania P.O., Tarnaka, Hyderabad, 500007, India
| | - Sathish Kumar Mungamuri
- Division of Food Safety, ICMR-National Institute of Nutrition, Jamai-Osmania P.O., Tarnaka, Hyderabad, 500007, India
| | - Vijayalakshmi Venkatesan
- Division of Cell and Molecular Biology, ICMR-National Institute of Nutrition, Jamai-Osmania P.O., Tarnaka, Hyderabad, 500007, India.
| |
Collapse
|
17
|
Duan A, Shen K, Li B, Li C, Zhou H, Kong R, Shao Y, Qin J, Yuan T, Ji J, Guo W, Wang X, Xue T, Li L, Huang X, Sun Y, Cai Z, Liu W, Liu F. Extracellular vesicles derived from LPS-preconditioned human synovial mesenchymal stem cells inhibit extracellular matrix degradation and prevent osteoarthritis of the knee in a mouse model. Stem Cell Res Ther 2021; 12:427. [PMID: 34321073 PMCID: PMC8317426 DOI: 10.1186/s13287-021-02507-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 07/11/2021] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Previous studies report that lipopolysaccharide (LPS)-preconditioned mesenchymal stem cells have enhanced trophic support and improved regenerative and repair properties. Extracellular vesicles secreted by synovial mesenchymal stem cells (EVs) can reduce cartilage damage caused by osteoarthritis (OA). Previous studies show that extracellular vesicles secreted by LPS-preconditioned synovial mesenchymal stem cells (LPS-pre EVs) can improve the response to treatment of osteoarthritis (OA). This study sought to explore effects of LPS-pre EVs on chondrocyte proliferation, migration, and chondrocyte apoptosis, as well as the protective effect of LPS-pre EVs on mouse articular cartilage. METHODS Chondrocytes were extracted to explore the effect of LPS-pre EVs on proliferation, migration, and apoptosis of chondrocytes. In addition, the effect of LPS-pre EVs on expression level of important proteins of chondrocytes was explored suing in vitro experiments. Further, intraarticular injection of LPS-pre EVs was performed on the destabilization of the medial meniscus (DMM)-induced mouse models of OA to explore the therapeutic effect of LPS-pre EVs on osteoarthritis in vivo. RESULTS Analysis showed that LPS-pre EVs significantly promoted proliferation and migration of chondrocytes and inhibited the apoptosis of chondrocytes compared with PBS and EVs. Moreover, LPS-pre EVs inhibited decrease of aggrecan and COL2A1 and increase of ADAMTS5 caused by IL-1β through let-7b. Furthermore, LPS-pre EVs significantly prevented development of OA in DMM-induced mouse models of OA. CONCLUSIONS LPS pretreatment is an effective and promising method to improve therapeutic effect of extracellular vesicles secreted from SMSCs on OA.
Collapse
Affiliation(s)
- Ao Duan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Kai Shen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Beichen Li
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210001, Jiangsu, China
| | - Cong Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Hao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Renyi Kong
- Department of Orthopedics, Xincheng Hospital of Traditional Chinese Medicine, Maanshan, 243131, Anhui, China
| | - Yuqi Shao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jian Qin
- Department of Orthopedics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, China
| | - Tangbo Yuan
- Department of Orthopedics, Sir Run Run Hospital, Nanjing Medical University, Nanjing, 211100, China
| | - Juan Ji
- Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211100, China
| | - Wei Guo
- Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211100, China
| | - Xipeng Wang
- Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211100, China
| | - Tengfei Xue
- Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211100, China
| | - Lei Li
- Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Center for Global Health, Nanjing Medical University, Nanjing, 211100, China
| | - Xinxin Huang
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yuqin Sun
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Zhenyu Cai
- Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Wei Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Feng Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
18
|
Lin S, Chen Q, Zhang L, Ge S, Luo Y, He W, Xu C, Zeng M. Overexpression of HOXB4 Promotes Protection of Bone Marrow Mesenchymal Stem Cells Against Lipopolysaccharide-Induced Acute Lung Injury Partially Through the Activation of Wnt/β-Catenin Signaling. J Inflamm Res 2021; 14:3637-3649. [PMID: 34349541 PMCID: PMC8326777 DOI: 10.2147/jir.s319416] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Pulmonary vascular endothelial cell (EC) injury is recognized as one of the pathological factors of acute lung injury/acute respiratory distress syndrome (ALI/ARDS). Bone marrow mesenchymal stem cell (BMSC)-based cytotherapy has attracted substantial attention over recent years as a promising therapeutic approach for ALI/ARDS; however, its use remains limited due to inconsistent efficacy. Currently, gene modification techniques are widely applied to MSCs. In the present study, we aimed to investigate the effect of BMSCs overexpressing Homeobox B4 (HOXB4) on lipopolysaccharide (LPS)-induced EC injury. METHODS We used LPS to induce EC injury and established EC-BMSC coculture system using transwell chambers. The effect of BMSCs on ECs was explored by detecting EC proliferation, apoptosis, migration, tube formation, and permeability, and determining whether the Wnt/β-catenin pathway is involved in the regulatory mechanism using XAV-939, inhibitor of Wnt/ β-catenin. RESULTS As compared to BMSCWT, BMSCHOXB4 coculture promoted EC proliferation, migration, and tube formation after LPS stimulation and attenuated LPS-induced EC apoptosis and vascular permeability. Mechanistically, BMSCHOXB4 coculture prevented LPS-induced EC injury by activating the Wnt/β-catenin pathway, which is partially reversible by XAV-939. When cocultured with BMSCHOXB4, pro-inflammatory factors were dramatically decreased and anti-inflammatory factors were greatly increased in the EC medium compared to those in the LPS group (P<0.05). Additionally, when compared to BMSCWT coculture, the BMSCHOXB4 coculture showed an enhanced modulation of IL-6, TNF-α, and IL-10, but there was no statistically significant effect on IL-1β and IL-4. CONCLUSION Coculturing of BMSCHOXB4 prevented LPS-induced EC injury by reversing the inactivation of the Wnt/β-catenin signaling pathway. An in vivo study remains warranted to ascertain whether engraftment of BMSCHOXB4 can be an attractive strategy for the treatment of ALI/ARDS.
Collapse
Affiliation(s)
- Shan Lin
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Qingui Chen
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Lishan Zhang
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Shanhui Ge
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Yuling Luo
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Wanmei He
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Caixia Xu
- Research Center of Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, People’s Republic of China
| | - Mian Zeng
- Department of Medical Intensive Care Unit, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510080, People’s Republic of China
- Institute of Pulmonary Diseases, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
19
|
Hu XM, Zhang Q, Zhou RX, Wu YL, Li ZX, Zhang DY, Yang YC, Yang RH, Hu YJ, Xiong K. Programmed cell death in stem cell-based therapy: Mechanisms and clinical applications. World J Stem Cells 2021; 13:386-415. [PMID: 34136072 PMCID: PMC8176847 DOI: 10.4252/wjsc.v13.i5.386] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/26/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
Stem cell-based therapy raises hopes for a better approach to promoting tissue repair and functional recovery. However, transplanted stem cells show a high death percentage, creating challenges to successful transplantation and prognosis. Thus, it is necessary to investigate the mechanisms underlying stem cell death, such as apoptotic cascade activation, excessive autophagy, inflammatory response, reactive oxygen species, excitotoxicity, and ischemia/hypoxia. Targeting the molecular pathways involved may be an efficient strategy to enhance stem cell viability and maximize transplantation success. Notably, a more complex network of cell death receives more attention than one crucial pathway in determining stem cell fate, highlighting the challenges in exploring mechanisms and therapeutic targets. In this review, we focus on programmed cell death in transplanted stem cells. We also discuss some promising strategies and challenges in promoting survival for further study.
Collapse
Affiliation(s)
- Xi-Min Hu
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Rui-Xin Zhou
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Yan-Lin Wu
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Zhi-Xin Li
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Dan-Yi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Yi-Chao Yang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Rong-Hua Yang
- Department of Burns, Fo Shan Hospital of Sun Yat-Sen University, Foshan 528000, Guangdong Province, China
| | - Yong-Jun Hu
- Department of Cardiovascular Medicine, Hunan People's Hospital (the First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan Province, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China.
| |
Collapse
|
20
|
Dogan F, Aljumaily RMK, Kitchen M, Forsyth NR. DNMT3B Is an Oxygen-Sensitive De Novo Methylase in Human Mesenchymal Stem Cells. Cells 2021; 10:1032. [PMID: 33925659 PMCID: PMC8145390 DOI: 10.3390/cells10051032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/19/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022] Open
Abstract
The application of physiological oxygen (physoxia) concentrations is becoming increasingly commonplace within a mammalian stem cell culture. Human mesenchymal stem cells (hMSCs) attract widespread interest for clinical application due to their unique immunomodulatory, multi-lineage potential, and regenerative capacities. Descriptions of the impact of physoxia on global DNA methylation patterns in hMSCs and the activity of enzymatic machinery responsible for its regulation remain limited. Human bone marrow-derived mesenchymal stem cells (BM-hMSCs, passage 1) isolated in reduced oxygen conditions displayed an upregulation of SOX2 in reduced oxygen conditions vs. air oxygen (21% O2, AO), while no change was noted for either OCT-4 or NANOG. DNA methylation marks 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) showed decreases in 2% O2 environment (workstation) (2% WKS). DNMT3B (DNA methyltransferase 3B) and TET1 (Ten-eleven translocation enzyme 1) displayed reduced transcription in physoxia. Consistent with transcriptional downregulation, we noted increased promoter methylation levels of DNMT3B in 2% WKS accompanied by reduced DNMT3B and TET1 protein expression. Finally, a decrease in HIF1A (Hypoxia-inducible factor 1A) gene expression in 2% WKS environment correlated with protein levels, while HIF2A was significantly higher in physoxia correlated with protein expression levels vs. AO. Together, these data have demonstrated, for the first time, that global 5mC, 5hmC, and DNMT3B are oxygen-sensitive in hMSCs. Further insights into the appropriate epigenetic regulation within hMSCs may enable increased safety and efficacy development within the therapeutic ambitions.
Collapse
Affiliation(s)
- Fatma Dogan
- The Guy Hilton Research Laboratories, Faculty of Medicine and Health Sciences, School of Pharmacy and Bioengineering, Keele University, Stoke on Trent ST5 5BG, UK; (F.D.); (M.K.)
| | - Rakad M Kh Aljumaily
- Department of Biology, College of Science, University of Baghdad, Baghdad 17635, Iraq;
| | - Mark Kitchen
- The Guy Hilton Research Laboratories, Faculty of Medicine and Health Sciences, School of Pharmacy and Bioengineering, Keele University, Stoke on Trent ST5 5BG, UK; (F.D.); (M.K.)
| | - Nicholas R. Forsyth
- The Guy Hilton Research Laboratories, Faculty of Medicine and Health Sciences, School of Pharmacy and Bioengineering, Keele University, Stoke on Trent ST5 5BG, UK; (F.D.); (M.K.)
| |
Collapse
|
21
|
Hong YT, Teo JY, Jeon H, Kong H. Shear-Resistant, Biological Tethering of Nanostimulators for Enhanced Therapeutic Cell Paracrine Factor Secretion. ACS APPLIED MATERIALS & INTERFACES 2021; 13:17276-17288. [PMID: 33830733 PMCID: PMC10440850 DOI: 10.1021/acsami.1c01520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Mesenchymal stromal cells (MSCs) secreting multiple growth factors and immunomodulatory cytokines are promising for regenerative medicine. To further enhance their secretory activity, efforts have emerged to tether nanosized carriers of secretory stimuli, named nanostimulators, to the MSC surface by forming nonchemical bonds. Despite some successes, there is a great need to improve the retention of nanostimulators during transport through a syringe needle, where high shear stress exerted on the cell surface separates them. To this end, we hypothesize that poly(lactic-co-glycolic acid)-block-hyaluronic acid (PLGA-HA) conjugated with integrin-binding RGD peptides, denoted PLGA-HA-RGD, can form nanostimulators that remain on the cell surface stably during the injection. The resulting HA-CD44 and RGD-integrin bonds would synergistically increase the adhesion strength of nanostimulators. Interestingly, nanostimulators prepared with PLGA-HA-RGD show 3- to 6-fold higher retention than those made with PLGA-HA. Therefore, the PLGA-HA-RGD nanostimulators induced MSCs to secrete 1.5-fold higher vascular endothelial growth factors and a 1.2-fold higher tissue inhibitor of matrix metalloproteinase-1 as compared to PLGA-HA nanostimulators. Consequently, MSCs tethered with PLGA-HA-RGD nanostimulators served to stimulate endothelial cell activities to form a blood vessel-like endothelial lumen with increased length and number of junctions. The nanostimulator design strategy would also be broadly applicable to regulate, protect, and home a broad array of therapeutic or immune cells by tethering carriers with bioactive molecules of interest.
Collapse
Affiliation(s)
- Yu-Tong Hong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Jye Yng Teo
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore 138669, Singapore
| | - Hojeong Jeon
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 136-701, Republic of Korea
| | - Hyunjoon Kong
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
22
|
Liu WZ, Ma ZJ, Li JR, Kang XW. Mesenchymal stem cell-derived exosomes: therapeutic opportunities and challenges for spinal cord injury. Stem Cell Res Ther 2021; 12:102. [PMID: 33536064 PMCID: PMC7860030 DOI: 10.1186/s13287-021-02153-8] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/07/2021] [Indexed: 12/31/2022] Open
Abstract
Spinal cord injury (SCI) often leads to serious motor and sensory dysfunction of the limbs below the injured segment. SCI not only results in physical and psychological harm to patients but can also cause a huge economic burden on their families and society. As there is no effective treatment method, the prevention, treatment, and rehabilitation of patients with SCI have become urgent problems to be solved. In recent years, mesenchymal stem cells (MSCs) have attracted more attention in the treatment of SCI. Although MSC therapy can reduce injured volume and promote axonal regeneration, its application is limited by tumorigenicity, a low survival rate, and immune rejection. Accumulating literature shows that exosomes have great potential in the treatment of SCI. In this review, we summarize the existing MSC-derived exosome studies on SCI and discuss the advantages and challenges of treating SCI based on exosomes derived from MSCs.
Collapse
Affiliation(s)
- Wen-Zhao Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, China
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, China
| | - Zhan-Jun Ma
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, China
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, China
| | - Jie-Ru Li
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Xue-Wen Kang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, 730030, Gansu, China.
- Department of Orthopedics, Lanzhou University Second Hospital, No.82 Cuiyingmen Street, Lanzhou, 730030, Gansu, China.
- The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
23
|
Shabbir A, Suzuki G, Lee T. Influence of Organismal Aging in Mesenchymal Stem Cell Therapy. ACTA ACUST UNITED AC 2020; 9:105-106. [PMID: 33225310 PMCID: PMC7676743 DOI: 10.34297/ajbsr.2020.09.001363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
| | - Gen Suzuki
- Department of Biochemistry and Department of Medicine
| | - Techung Lee
- Department of Biochemistry and Department of Medicine
| |
Collapse
|
24
|
Xing D, Liu W, Li JJ, Liu L, Guo A, Wang B, Yu H, Zhao Y, Chen Y, You Z, Lyu C, Li W, Liu A, Du Y, Lin J. Engineering 3D functional tissue constructs using self-assembling cell-laden microniches. Acta Biomater 2020; 114:170-182. [PMID: 32771588 DOI: 10.1016/j.actbio.2020.07.058] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 12/21/2022]
Abstract
Tissue engineering using traditional size fixed scaffolds and injectable biomaterials are faced with many limitations due to the difficulties of producing macroscopic functional tissues. In this study, 3D functional tissue constructs were developed by inducing self-assembly of microniches, which were cell-laden gelatin microcryogels. During self-assembly, the accumulation of extracellular matrix (ECM) components was found to strengthen cell-cell and cell-ECM interactions, leading to the construction of a 'native' microenvironment that better preserved cell viability and functions. MSCs grown in self-assembled constructs showed increased maintenance of stemness, reduced senescence and improved paracrine activity compared with cells grown in individual microniches without self-assembly. As an example of applying the self-assembled constructs in tissue regeneration, the constructs were used to induce in vivo articular cartilage repair and successfully regenerated hyaline-like cartilage tissue in the absence of other extrinsic factors. This unique approach of developing self-assembled 3D functional constructs holds great promise for the generation of tissue engineered organoids and repair of challenging tissue defects. STATEMENT OF SIGNIFICANCE: We developed 3D functional tissue constructs using a unique gelatin-based microscopic hydrogel (microcryogels). Mesenchymal stem cells (MSCs) were loaded into gelatin microcryogels to form microscopic cell-laden units (microniches), which were induced to undergo self-assembly using a specially designed 3D printed frame. Extracellular matrix accumulation among the microniches resulted in self-assembled macroscopic constructs with superior ability to maintain the phenotypic characteristics and stemness of MSCs, together with the suppression of senescence and enhanced paracrine function. As an example of application in tissue regeneration, the self-assembled constructs were shown to successfully repair articular cartilage defects without any other supplements. This unique strategy for developing 3D functional tissue constructs allows the optimisation of stem cell functions and construction of biomimetic tissue organoids.
Collapse
Affiliation(s)
- Dan Xing
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing 100044, China; Arthritis Institute, Peking University, Beijing 100044, China
| | - Wei Liu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Jiao Jiao Li
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney (UTS), Ultimo, NSW 2007, Australia
| | - Longwei Liu
- Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Anqi Guo
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Bin Wang
- Department of Sports Medicine and Adult Reconstruction Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing 210009, China
| | - Hongsheng Yu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Yu Zhao
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing 100044, China; Arthritis Institute, Peking University, Beijing 100044, China
| | - Yuling Chen
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing 100084, China
| | - Zhifeng You
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Cheng Lyu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Wenjing Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China
| | - Aifeng Liu
- Department of Orthopedics, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing 100084, China.
| | - Jianhao Lin
- Arthritis Clinic & Research Center, Peking University People's Hospital, Peking University, Beijing 100044, China; Arthritis Institute, Peking University, Beijing 100044, China.
| |
Collapse
|
25
|
Garcia-Arranz M, Alonso-Gregorio S, Fontana-Portella P, Bravo E, Diez Sebastian J, Fernandez-Santos ME, Garcia-Olmo D. Two phase I/II clinical trials for the treatment of urinary incontinence with autologous mesenchymal stem cells. Stem Cells Transl Med 2020; 9:1500-1508. [PMID: 32864818 PMCID: PMC7695632 DOI: 10.1002/sctm.19-0431] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 06/06/2020] [Accepted: 07/19/2020] [Indexed: 12/21/2022] Open
Abstract
We evaluated the safety and feasibility of adipose‐derived mesenchymal stem cells to treat endoscopically urinary incontinence after radical prostatectomy in men or female stress urinary. We designed two prospective, nonrandomized phase I‐IIa clinical trials of urinary incontinence involving 9 men (8 treated) and 10 women to test the feasibility and safety of autologous mesenchymal stem cells for this use. Cells were obtained from liposuction containing 150 to 200 g of fat performed on every patient. After 4 to 6 weeks and under sedation, endoscopic intraurethral injection of the cells was performed. On each visit (baseline, 1, 3, 6, and 12 months), clinical parameters were measured, and blood samples, urine culture, and uroflowmetry were performed. Every patient underwent an urethrocystoscopy and urodynamic studies on the first and last visit. Data from pad test, quality‐of‐life and incontinence questionnaires, and pads used per day were collected at every visit. Statistical analysis was done by Wilcoxon signed‐rank test. No adverse effects were observed. Three men (37.5%) and five women (50%) showed an objective improvement of >50% (P < .05) and a subjective improvement of 70% to 80% from baseline. In conclusion, intraurethral application of stem cells derived from adipose tissue is a safe and feasible procedure to treat urinary incontinence after radical prostatectomy or in female stress urinary incontinence. A statistically significant difference was obtained for pad‐test improvement in 3/8 men and 5/10 women. Our results encourage studies to confirm safety and to analyze efficacy.
Collapse
Affiliation(s)
- Mariano Garcia-Arranz
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Madrid, Spain.,Surgery Department, Autonoma University of Madrid, Madrid, Spain
| | | | | | - Elena Bravo
- Department of Plastic Surgery, La Paz University Hospital, Madrid, Spain
| | | | | | - Damian Garcia-Olmo
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Madrid, Spain.,Surgery Department, Autonoma University of Madrid, Madrid, Spain
| |
Collapse
|
26
|
Magnetic targeting enhances the cutaneous wound healing effects of human mesenchymal stem cell-derived iron oxide exosomes. J Nanobiotechnology 2020; 18:113. [PMID: 32799868 PMCID: PMC7429707 DOI: 10.1186/s12951-020-00670-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022] Open
Abstract
Human mesenchymal stem cell (MSC)-derived exosomes (Exos) are a promising therapeutic agent for cell-free regenerative medicine. However, their poor organ-targeting ability and therapeutic efficacy have been found to critically limit their clinical applications. In the present study, we fabricated iron oxide nanoparticle (NP)-labeled exosomes (Exo + NPs) from NP-treated MSCs and evaluated their therapeutic efficacy in a clinically relevant model of skin injury. We found that the Exos could be readily internalized by human umbilical vein endothelial cells (HUVECs), and could significantly promote their proliferation, migration, and angiogenesis both in vitro and in vivo. Moreover, the protein expression of proliferative markers (Cyclin D1 and Cyclin A2), growth factors (VEGFA), and migration-related chemokines (CXCL12) was significantly upregulated after Exo treatment. Unlike the Exos prepared from untreated MSCs, the Exo + NPs contained NPs that acted as a magnet-guided navigation tool. The in vivo systemic injection of Exo + NPs with magnetic guidance significantly increased the number of Exo + NPs that accumulated at the injury site. Furthermore, these accumulated Exo + NPs significantly enhanced endothelial cell proliferation, migration, and angiogenic tubule formation in vivo; moreover, they reduced scar formation and increased CK19, PCNA, and collagen expression in vivo. Collectively, these findings confirm the development of therapeutically efficacious extracellular nanovesicles and demonstrate their feasibility in cutaneous wound repair.
Collapse
|
27
|
Jin C, Zhou F, Zhang L, Shen J. Overexpression of heat shock protein 70 enhanced mesenchymal stem cell treatment efficacy in phosgene-induced acute lung injury. J Biochem Mol Toxicol 2020; 34:e22515. [PMID: 32394537 DOI: 10.1002/jbt.22515] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/22/2020] [Indexed: 12/19/2022]
Abstract
In our previous study, we have confirmed that in phosgene-induced acute lung injury (ALI) rats, mesenchymal stem cells (MSCs) can treat the disease. Moreover, heat shock protein 70 (Hsp70) can be used as a protective protein, and Hsp70 upregulated drastically when exposed to stressful conditions. We aimed to assess that MSCs overexpressed Hsp70 could enhance the capacity of MSCs and have a good therapeutic effect on phosgene-induced ALI. We transduced MSCs with Hsp70 and then we tested the function of the transduced MSCs. Sprague Dawley rats inhaled phosgene in a closed container for 5 minutes. The transduced MSCs and MSCs were administered via the trachea immediately. Rats in each group were killed at 6, 24, and 48 hours after exposure. Compared to MSCs, MSCs overexpressed Hsp70 enhanced MSCs viability, antiapoptotic ability, and migration ability, and these effects disappeared when using the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathway inhibitor. Furthermore, the results of pathological alterations improved. The lung wet-to-dry ratio declined. The lung injury index total protein content and total cells in bronchoalveolar lavage fluid (BALF) also declined. The level of tumor necrosis factor α declined and the level of interleukin-10 improved in BALF and serum. MSCs overexpressed Hsp70 can enhance the capacity and efficacy of MSCs in the treatment of phosgene-induced ALI and may be mediated through the PI3k/AKT signaling pathway. This article introduces a new approach to stem cell therapy for improving the efficacy of phosgene-induced ALI.
Collapse
Affiliation(s)
- Chaoyuan Jin
- Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Fangqing Zhou
- Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Lin Zhang
- Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jie Shen
- Center of Emergency and Intensive Care Unit, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Center of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China.,Medical Research Center for Chemical Injury, Emergency and Critical Care of Chemical Injury, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
28
|
Kamaldinov T, Erndt-Marino J, Levin M, Kaplan DL, Hahn MS. Assessment of Enrichment of Human Mesenchymal Stem Cells Based on Plasma and Mitochondrial Membrane Potentials. Bioelectricity 2020; 2:21-32. [PMID: 32292894 DOI: 10.1089/bioe.2019.0024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background: Human mesenchymal stem cells (hMSCs) are utilized preclinically and clinically as a candidate cell therapy for a wide range of inflammatory and degenerative diseases. Despite promising results in early clinical trials, consistent outcomes with hMSC-based therapies have proven elusive in many of these applications. In this work, we attempt to address this limitation through the design of a stem cell therapy to enrich hMSCs for desired electrical and ionic properties with enhanced stemness and immunomodulatory/regenerative capacity. Materials and Methods: In this study, we sought to develop initial protocols to achieve electrically enriched hMSCs (EE-hMSCs) with distinct electrical states and assess the potential relationship with respect to hMSC state and function. We sorted hMSCs based on fluorescence intensity of tetramethylrhodamine ethyl ester (TMRE) and investigated phenotypic differences between the sorted populations. Results: Subpopulations of EE-hMSCs exhibit differential expression of genes associated with senescence, stemness, immunomodulation, and autophagy. EE-hMSCs with low levels of TMRE, indicative of depolarized membrane potential, have reduced mRNA expression of senescence-associated markers, and increased mRNA expression of autophagy and immunomodulatory markers relative to EE-hMSCs with high levels of TMRE (hyperpolarized). Conclusions : This work suggests that the utilization of EE-hMSCs may provide a novel strategy for cell therapies, enabling live cell enrichment for distinct phenotypes that can be exploited for different therapeutic outcomes.
Collapse
Affiliation(s)
- Timothy Kamaldinov
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Josh Erndt-Marino
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York.,Department of Biomedical Engineering, Tufts University, Medford, Massachusetts.,Allen Discovery Center at Tufts University, Department of Biology, Tufts University, Medford, Massachusetts
| | - Michael Levin
- Allen Discovery Center at Tufts University, Department of Biology, Tufts University, Medford, Massachusetts
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts.,Allen Discovery Center at Tufts University, Department of Biology, Tufts University, Medford, Massachusetts
| | - Mariah S Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
29
|
Yin L, Yang Z, Wu Y, Denslin V, Yu CC, Tee CA, Lim CT, Han J, Lee EH. Label-free separation of mesenchymal stem cell subpopulations with distinct differentiation potencies and paracrine effects. Biomaterials 2020; 240:119881. [PMID: 32092592 DOI: 10.1016/j.biomaterials.2020.119881] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 01/23/2020] [Accepted: 02/11/2020] [Indexed: 12/23/2022]
Abstract
Mesenchymal stem cells (MSCs) have the capability to differentiate into multiple cell lineages, and produce trophic factors to facilitate tissue repair and regeneration, and disease regression. However, the heterogeneity of MSCs, whether inherent or developed during culture expansion, has a significant impact on their therapeutic efficacy. Therefore, the ability to identify and select an efficacious subpopulation of MSCs targeting specific tissue damage or disease holds great clinical significance. In this study, we separated three subpopulations from culture expanded human bone marrow derived MSCs according to cell size, using a high-throughput label-free microfluidic cell sorting technology. The size-sorted MSC subpopulations varied in tri-lineage differentiation potencies. The large MSCs showed the strongest osteogenesis, medium-size MSCs were advantageous in chondrogenesis and adipogenesis, and the small MSCs showed the weakest tri-lineage differentiation. The size-sorted MSC subpopulations also exhibited different secretome profiles. The large MSC secretome possessed highest levels of osteogenic promotor proteins and senescence-associated factors, but lower levels of osteogenic inhibitor proteins compared to the medium-size MSC secretome. The medium-size MSC secretome had high levels of chondrogenic promotor proteins, and contained lower levels of chondrogenic inhibitor proteins compared to the large MSC secretome. The secretome of size-sorted MSC subpopulations showed differences in paracrine effects. We found that the secretome of large MSCs enhanced osteogenic and adipogenic potencies during MSC culture expansion, but also induced cell senescence; and the secretome of medium-size MSCs promoted chondrogenesis. This study demonstrates size-dependent differentiation potency and secretome profile of MSC subpopulations, and provides an effective and practical technology to isolate the respective subpopulations, which may be used for more targeted tissue repair and regeneration.
Collapse
Affiliation(s)
- Lu Yin
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore
| | - Zheng Yang
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore 27 Medical Drive1, DSO (Kent Bridge) Building, Level 4, Singapore, 11751, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore
| | - Yingnan Wu
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore 27 Medical Drive1, DSO (Kent Bridge) Building, Level 4, Singapore, 11751, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore
| | - Vinitha Denslin
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore 27 Medical Drive1, DSO (Kent Bridge) Building, Level 4, Singapore, 11751, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore
| | - Chia Chen Yu
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA 02139, USA
| | - Ching Ann Tee
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore
| | - Chwee Teck Lim
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, T-Lab, #10-01, Singapore, 117411, Singapore; Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Engineering Block 4, #04-08, Singapore, 117583, Singapore; Institute for Health Innovation and Technology, National University of Singapore, MD6, 14 Medical Drive, #14-01, Singapore, 117599, Singapore
| | - Jongyoon Han
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; Department of Electrical Engineering and Computer Science, Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA.
| | - Eng Hin Lee
- Critical Analytics for Manufacturing of Personalised Medicine Interdisciplinary Research Group, Singapore-MIT Alliance in Research and Technology, 1 Create Way, #04-13/14, Singapore, 138602, Singapore; NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore 27 Medical Drive1, DSO (Kent Bridge) Building, Level 4, Singapore, 11751, Singapore; Department of Orthopaedic Surgery, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block 11, Singapore, 119288, Singapore.
| |
Collapse
|
30
|
Pinto DS, Ahsan T, Serra J, Fernandes-Platzgummer A, Cabral JMS, da Silva CL. Modulation of the in vitro angiogenic potential of human mesenchymal stromal cells from different tissue sources. J Cell Physiol 2020; 235:7224-7238. [PMID: 32037550 DOI: 10.1002/jcp.29622] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 01/08/2020] [Indexed: 12/14/2022]
Abstract
Mesenchymal stromal cells (MSCs) have been widely exploited for the treatment of several conditions due to their intrinsic regenerative and immunomodulatory properties. MSC have demonstrated to be particularly relevant for the treatment of ischemic diseases, where MSC-based therapies can stimulate angiogenesis and induce tissue regeneration. Regardless of the condition targeted, recent analyses of MSC-based clinical trials have demonstrated limited benefits indicating a need to improve the efficacy of this cell product. Preconditioning MSC ex vivo through microenvironment modulation was found to improve MSC survival rate and thus prolong their therapeutic effect. This workstudy aims at enhancing the in vitro angiogenic capacity of a potential MSC-based medicinal product by comparing different sources of MSC and culture conditions. MSC from three different sources (bone marrow [BM], adipose tissue [AT], and umbilical cord matrix [UCM]) were cultured with xenogeneic-/serum-free culture medium under static conditions and their angiogenic potential was studied. Results indicated a higher in vitro angiogenic capacity of UCM MSC, compared with cells derived from BM and AT. Physicochemical preconditioning of UCM MSC through a microcarrier-based culture platform and low oxygen concentration (2% O2 , compared with atmospheric air) increased the in vitro angiogenic potential of the cultured cells. Envisaging the clinical manufacturing of an allogeneic, off-the-shelf MSC-based product, preconditioned UCM MSC maintain the angiogenic gene expression profile upon cryopreservation and delivery processes in the conditions of our study. These results are expected to contribute to the development of MSC-based therapies in the context of angiogenesis.
Collapse
Affiliation(s)
- Diogo S Pinto
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| | - Tabassum Ahsan
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana
| | - Joana Serra
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Fernandes-Platzgummer
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB, Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
31
|
Kabat M, Bobkov I, Kumar S, Grumet M. Trends in mesenchymal stem cell clinical trials 2004-2018: Is efficacy optimal in a narrow dose range? Stem Cells Transl Med 2019; 9:17-27. [PMID: 31804767 PMCID: PMC6954709 DOI: 10.1002/sctm.19-0202] [Citation(s) in RCA: 303] [Impact Index Per Article: 50.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 08/26/2019] [Indexed: 12/19/2022] Open
Abstract
The number of clinical trials using mesenchymal stem cells (MSCs) has increased since 2008, but this trend slowed in the past several years and dropped precipitously in 2018. Previous reports have analyzed MSC clinical trials by disease, phase, cell source, country of origin, and trial initiation date, all of which can be downloaded directly from http://clinicaltrials.gov. We have extended analyses to a larger group of 914 MSC trials reported through 2018. To search for potential factors that may influence the design of new trials, we extracted data on routes of administration and dosing from individual http://clinicaltrials.gov records as this information cannot be downloaded directly from the database. Intravenous (IV) injection is the most common, least invasive and most reproducible method, accounting for 43% of all trials. The median dose for IV delivery is 100 million MSCs/patient/dose. Analysis of all trials using IV injection that reported positive outcomes indicated minimal effective doses (MEDs) ranging from 70 to 190 million MSCs/patient/dose in 14/16 trials with the other two trials administering much higher doses of at least 900 million cells. Dose‐response data showing differential efficacy for improved outcomes were reported in only four trials, which indicated a narrower MED range of 100‐150 million MSCs/patient with lower and higher IV doses being less effective. The results suggest that it may be critical to determine MEDs in early trials before proceeding with large clinical trials.
Collapse
Affiliation(s)
- Maciej Kabat
- W. M. Keck Center for Collaborative Neuroscience, Rutgers Stem Cell Research Center, Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Ivan Bobkov
- W. M. Keck Center for Collaborative Neuroscience, Rutgers Stem Cell Research Center, Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Martin Grumet
- W. M. Keck Center for Collaborative Neuroscience, Rutgers Stem Cell Research Center, Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, New Jersey
| |
Collapse
|
32
|
Central metabolism of functionally heterogeneous mesenchymal stromal cells. Sci Rep 2019; 9:15420. [PMID: 31659213 PMCID: PMC6817850 DOI: 10.1038/s41598-019-51937-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 10/08/2019] [Indexed: 12/16/2022] Open
Abstract
Metabolism and mitochondrial biology have gained a prominent role as determinants of stem cell fate and function. In the context of regenerative medicine, innovative parameters predictive of therapeutic efficacy could be drawn from the association of metabolic or mitochondrial parameters to different degrees of stemness and differentiation potentials. Herein, this possibility was addressed in human mesenchymal stromal/stem cells (hMSC) previously shown to differ in lifespan and telomere length. First, these hMSC were shown to possess significantly distinct proliferation rate, senescence status and differentiation capacity. More potential hMSC were associated to higher mitochondrial (mt) DNA copy number and lower mtDNA methylation. In addition, they showed higher expression levels of oxidative phosphorylation subunits. Consistently, they exhibited higher coupled oxygen consumption rate and lower transcription of glycolysis-related genes, glucose consumption and lactate production. All these data pointed at oxidative phosphorylation-based central metabolism as a feature of higher stemness-associated hMSC phenotypes. Consistently, reduction of mitochondrial activity by complex I and III inhibitors in higher stemness-associated hMSC triggered senescence. Finally, functionally higher stemness-associated hMSC showed metabolic plasticity when challenged by glucose or glutamine shortage, which mimic bioenergetics switches that hMSC must undergo after transplantation or during self-renewal and differentiation. Altogether, these results hint at metabolic and mitochondrial parameters that could be implemented to identify stem cells endowed with superior growth and differentiation potential.
Collapse
|
33
|
Xu R, Zhang F, Chai R, Zhou W, Hu M, Liu B, Chen X, Liu M, Xu Q, Liu N, Liu S. Exosomes derived from pro-inflammatory bone marrow-derived mesenchymal stem cells reduce inflammation and myocardial injury via mediating macrophage polarization. J Cell Mol Med 2019; 23:7617-7631. [PMID: 31557396 PMCID: PMC6815833 DOI: 10.1111/jcmm.14635] [Citation(s) in RCA: 181] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/16/2019] [Accepted: 07/30/2019] [Indexed: 12/15/2022] Open
Abstract
Exosomes are served as substitutes for stem cell therapy, playing important roles in mediating heart repair during myocardial infarction injury. Evidence have indicated that lipopolysaccharide (LPS) pre-conditioning bone marrow-derived mesenchymal stem cells (BMSCs) and their secreted exosomes promote macrophage polarization and tissue repair in several inflammation diseases; however, it has not been fully elucidated in myocardial infarction (MI). This study aimed to investigate whether LPS-primed BMSC-derived exosomes could mediate inflammation and myocardial injury via macrophage polarization after MI. Here, we found that exosomes derived from BMSCs, in both Exo and L-Exo groups, increased M2 macrophage polarization and decreased M1 macrophage polarization under LPS stimulation, which strongly depressed LPS-dependent NF-κB signalling pathway and partly activated the AKT1/AKT2 signalling pathway. Compared with Exo, L-Exo had superior therapeutic effects on polarizing M2 macrophage in vitro and attenuated the post-infarction inflammation and cardiomyocyte apoptosis by mediating macrophage polarization in mice MI model. Consequently, we have confidence in the perspective that low concentration of LPS pre-conditioning BMSC-derived exosomes may develop into a promising cell-free treatment strategy for clinical treatment of MI.
Collapse
Affiliation(s)
- Ruqin Xu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Fangcheng Zhang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Renjie Chai
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Wenyi Zhou
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ming Hu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Bin Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xuke Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Mingke Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qiong Xu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ningning Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shiming Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
34
|
Shojaei F, Rahmati S, Banitalebi Dehkordi M. A review on different methods to increase the efficiency of mesenchymal stem cell-based wound therapy. Wound Repair Regen 2019; 27:661-671. [PMID: 31298446 DOI: 10.1111/wrr.12749] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/04/2019] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) accelerate wound healing but the harsh environment of wound site limits the engraftment, retention, and survival rate of transplanted cells. There are multiple approaches that amplify the therapeutic potential of MSCs. The MSCs derived from medical waste material, provide comparable regenerative abilities compared to traditional sources. The application of different scaffolds increases MSC delivery and migration into the wound. The spheroid culture of MSC increases the paracrine effects of the entrapped cells and the secretion of pro-angiogenic and anti-inflammatory cytokines. The MSC pretreating and preconditioning enhances the cell migration, proliferation, and survival rate, which lead to higher angiogenesis, re-epithelialization, wound closure, and granulation tissue formation. Moreover, genetic modification has been performed in order to increase MSC angiogenesis, differentiation potential, as well as the cell life span. Herein, we review the results of aforementioned approaches and provide information accommodating to the continued development of MSC-based wound therapy in the future.
Collapse
Affiliation(s)
- Fereshteh Shojaei
- Department of Medical biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Shima Rahmati
- Cellular and Molecular Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mehdi Banitalebi Dehkordi
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
35
|
Özdemir RBÖ, Özdemir AT, Sarıboyacı AE, Uysal O, Tuğlu Mİ, Kırmaz C. The investigation of immunomodulatory effects of adipose tissue mesenchymal stem cell educated macrophages on the CD4 T cells. Immunobiology 2019; 224:585-594. [PMID: 31072631 PMCID: PMC7124282 DOI: 10.1016/j.imbio.2019.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/27/2019] [Accepted: 04/01/2019] [Indexed: 12/11/2022]
Abstract
Mesenchymal stem cells (MSCs) are strong immunomodulatory cells investigated in numerous clinical studies on fatal pathologies, such as graft versus host disease and autoimmune diseases; e.g., systemic lupus erythematosus, Crohn's disease, and ulcerative colitis. Macrophages are one of the critical cells linking the innate and adaptive immune system, and it has been shown that MSCs can differentiate between pro-inflammatory M1 phenotype and anti-inflammatory M2 phenotype of macrophages. However, it has not yet been fully clarified whether these differentiated macrophages are functional. In this study, we compared the immunomodulatory effects on the CD4 T cells of M1, M2a and M2c macrophages with the macrophages that directly and indirectly cultured with MSCs. We analyzed the changes in CD14, CD64, CD80, CD163 and CD200R expression to evaluate macrophage phenotypes, and the changes in CD4, IFN-g, IL-4, IL-17a and FoxP3 expression to evaluate T helper subsets using the FACS method. The changes in IL-1b, IL-4, IL-10, IL-12p70, IL-17a and IFN-g in the media supernatants were analyzed using the Luminex method. We also performed WST-1 and Caspase-3 ELISA analyses to observe the proliferation and apoptosis status of the T cells. MSCs were found to differentiate macrophages into a distinctive phenotype, which was close to the M2c phenotype, but was not considered as an M2c cell due to the low expression of CD163, a characteristic marker for M2c. While MEM-D, MEM-ID and MSCs showed similar inhibitory effects on the Th2 and Th17 cells, the most significant increase in Treg cell frequencies was seen in MEM-D cells. Macrophages can alter their phenotypes and functions according to the stimuli from the environment. The fact that macrophages educated with MSCs suppressed the production of all the cytokines we evaluated even after the removal of MSCs suggests that these cells may be differentiated by MSCs into a suppressive macrophage subgroup. However, the Treg cell activation caused by direct interactions between MSCs and macrophage cells may be the most prominent observation of this study compared to previous work. As a result, according to our data, the interactions between MSCs and macrophages may lead to differentiation of macrophage cells into an immunosuppressive phenotype, and these macrophages may suppress the T lymphocyte subgroups at least as effectively as MSCs. However, our data obtained from in vitro experiments should be supported by future in vivo studies.
Collapse
Affiliation(s)
| | - Alper Tunga Özdemir
- Ege University, Institute of Health Sciences, Department of Stem Cell, Izmir, Turkey.
| | - Ayla Eker Sarıboyacı
- Eskişehir Osmangazi University, Cellular Therapy and Stem Cell Production Application and Research Center, Eskisehir, Turkey
| | - Onur Uysal
- Eskişehir Osmangazi University, Cellular Therapy and Stem Cell Production Application and Research Center, Eskisehir, Turkey
| | - Mehmet İbrahim Tuğlu
- Manisa Celal Bayar University, Faculty of Medicine, Department of Histology and Embryology, Manisa, Turkey
| | - Cengiz Kırmaz
- Manisa Celal Bayar University, Medical School, Department of Internal Medicine, Division of Allergy and Clinical Immunology, Manisa, Turkey
| |
Collapse
|
36
|
Hu C, Zhao L, Wu D, Li L. Modulating autophagy in mesenchymal stem cells effectively protects against hypoxia- or ischemia-induced injury. Stem Cell Res Ther 2019; 10:120. [PMID: 30995935 PMCID: PMC6471960 DOI: 10.1186/s13287-019-1225-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In mammals, a basal level of autophagy, a self-eating cellular process, degrades cytosolic proteins and subcellular organelles in lysosomes to provide energy, recycles the cytoplasmic components, and regenerates cellular building blocks; thus, autophagy maintains cellular and tissue homeostasis in all eukaryotic cells. In general, adaptive autophagy increases when cells confront stressful conditions to improve the survival rate of the cells, while destructive autophagy is activated when the cellular stress is not manageable and elicits the regenerative capacity. Hypoxia-reoxygenation (H/R) injury and ischemia-reperfusion (I/R) injury initiate excessive autophagy and endoplasmic reticulum (ER) stress and consequently induce a string of damage in mammalian tissues or organs. Mesenchymal stem cell (MSC)-based therapy has yielded promising results in repairing H/R- or I/R-induced injury in various tissues. However, MSC transplantation in vivo must overcome the barriers including the low survival rate of transplanted stem cells, limited targeting capacity, and low grafting potency; therefore, much effort is needed to increase the survival and activity of MSCs in vivo. Modulating autophagy regulates the stemness and the anti-oxidative stress, anti-apoptosis, and pro-survival capacity of MSCs and can be applied to MSC-based therapy for repairing H/R- or I/R-induced cellular or tissue injury.
Collapse
Affiliation(s)
- Chenxia Hu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lingfei Zhao
- Kidney Disease Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.,Key Laboratory of Kidney Disease Prevention and Control Technology, Hangzhou, Zhejiang, People's Republic of China.,Institute of Nephrology, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Daxian Wu
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Lanjuan Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, School of Medicine, First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
37
|
Lin CH, Lin W, Su YC, Cheng-Yo Hsuan Y, Chen YC, Chang CP, Chou W, Lin KC. Modulation of parietal cytokine and chemokine gene profiles by mesenchymal stem cell as a basis for neurotrauma recovery. J Formos Med Assoc 2019; 118:1661-1673. [PMID: 30709695 DOI: 10.1016/j.jfma.2019.01.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/07/2019] [Accepted: 01/11/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND & PURPOSE Following traumatic brain injury (TBI), primary mechanical injury to the brain may cause blood-brain-barrier damage followed by secondary injury, ultimately culminating in cell death. We aimed to test whether one injection of mesenchymal stem cells (MSC) derived from the human umbilical cord can modulate brain cytokine and chemokine gene profiles and attenuate neurological injury in rats with TBI. METHODS One-day post-TBI, the injured rats were treated with one injection of MSC (4 × 106/rat, i.v.). Three days later, immediately after assessment of neurobehavioral function, animals were sacrificed for analysis of neurological injury (evidenced by both brain contusion volume and neurological deficits) and parietal genes encoding 84 cytokines and chemokines in the injured brain by qPCR methods. RESULTS Three days post-TBI, rats displayed both neurological injury and upgrade of 11 parietal genes in the ipsilateral brain. One set of 8 parietal genes (e.g., chemokine [C-X-C motif] ligand 12, platelet factor 4, interleukin-7, chemokine [C-C motif] ligand (CCL)19, CCL 22, secreted phosphoprotein 1, pro-platelet basic protein 1, and CCL 2) differentially upgraded by TBI was related to pro-inflammatory and/or neurodegenerative processes. Another set of 3 parietal genes up-graded by TBI (e.g., glucose-6-phosphate isomerase, bone morphogenetic protein (BMP) 2, and BMP 4) was related to anti-inflammatory/neuroregenerative events. Administration of MSC attenuated neurological injury, down-regulated these 8 parietal pro-inflammatory genes, and up-regulated these 3 parietal anti-inflammatory genes in the rats with TBI. CONCLUSION Our data suggest that modulation of parietal cytokines and chemokines gene profiles by MSC as a basis for neurotrauma recovery.
Collapse
Affiliation(s)
- Cheng-Hsien Lin
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan.
| | - Willie Lin
- Meridigen Biotech Co. Ltd., Neihu, Taipei 11493, Taiwan.
| | - Yu-Chin Su
- Meridigen Biotech Co. Ltd., Neihu, Taipei 11493, Taiwan.
| | | | - Yu-Chien Chen
- Department of Medical Research, Chi Mei Medical Center, Tainan 710, Taiwan.
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, Tainan 710, Taiwan.
| | - Willy Chou
- Department of Physical Medicine and Rehabilitation, Chi Mei Medical Center, Tainan 710, Taiwan; Department of Recreation and Healthcare Management, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan.
| | - Kao-Chang Lin
- Department of Neurology, Chi Mei Medical Center, Tainan 710, Taiwan.
| |
Collapse
|
38
|
Maumus M, Pers YM, Ruiz M, Jorgensen C, Noël D. [Mesenchymal stem cells and regenerative medicine: future perspectives in osteoarthritis]. Med Sci (Paris) 2019; 34:1092-1099. [PMID: 30623767 DOI: 10.1051/medsci/2018294] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stromal or stem cells (MSCs) are multipotent adult cells that can be isolated from a variety of adult or neonatal tissues, such as bone marrow, fat tissue, placenta or umbilical cord. A therapy based on MSCs can be justified in osteoarthritis (OA) thanks to their differentiation abilities but mostly, to their paracrine and immunosuppressive properties. Possible therapeutic strategies therefore rely on the articular injection of MSCs suspensions for trophic activity or the implantation of MSCs combined with biodegradable materials for tissue engineering applications. Depending on the mode of administration and behavior after implantation, they can decrease local inflammation, prevent chondrocyte hypertrophy and apoptosis as well as differentiate into cartilage-forming chondrocytes. In this review, we summarize pathophysiological and mechanistic data and discuss perspectives confirming the interest of MSCs as a potential therapeutic strategy in OA.
Collapse
Affiliation(s)
- Marie Maumus
- IRMB, université de Montpellier, Inserm U1183, 80, avenue Augustin Fliche, 34295 Montpellier, France
| | - Yves-Marie Pers
- IRMB, université de Montpellier, Inserm U1183, 80, avenue Augustin Fliche, 34295 Montpellier, France - Unité d'immunologie clinique et de thérapeutique des maladies ostéoarticulaires, département de rhumatologie, hôpital Lapeyronie, 34000 Montpellier, France
| | - Maxime Ruiz
- IRMB, université de Montpellier, Inserm U1183, 80, avenue Augustin Fliche, 34295 Montpellier, France
| | - Christian Jorgensen
- IRMB, université de Montpellier, Inserm U1183, 80, avenue Augustin Fliche, 34295 Montpellier, France - Unité d'immunologie clinique et de thérapeutique des maladies ostéoarticulaires, département de rhumatologie, hôpital Lapeyronie, 34000 Montpellier, France
| | - Danièle Noël
- IRMB, université de Montpellier, Inserm U1183, 80, avenue Augustin Fliche, 34295 Montpellier, France - Unité d'immunologie clinique et de thérapeutique des maladies ostéoarticulaires, département de rhumatologie, hôpital Lapeyronie, 34000 Montpellier, France
| |
Collapse
|
39
|
Proksch S, Galler KM. Scaffold Materials and Dental Stem Cells in Dental Tissue Regeneration. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/s40496-018-0197-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
40
|
Kim HY, Kumar H, Jo MJ, Kim J, Yoon JK, Lee JR, Kang M, Choo YW, Song SY, Kwon SP, Hyeon T, Han IB, Kim BS. Therapeutic Efficacy-Potentiated and Diseased Organ-Targeting Nanovesicles Derived from Mesenchymal Stem Cells for Spinal Cord Injury Treatment. NANO LETTERS 2018; 18:4965-4975. [PMID: 29995418 DOI: 10.1021/acs.nanolett.8b01816] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Human mesenchymal stem cell (hMSC)-derived exosomes have been spotlighted as a promising therapeutic agent for cell-free regenerative medicine. However, poor organ-targeting ability and insufficient therapeutic efficacy of systemically injected hMSC-exosomes were identified as critical limitations for their further applications. Therefore, in this study we fabricated iron oxide nanoparticle (IONP)-incorporated exosome-mimetic nanovesicles (NV-IONP) from IONP-treated hMSCs and evaluated their therapeutic efficacy in a clinically relevant model for spinal cord injury. Compared to exosome-mimetic nanovesicles (NV) prepared from untreated hMSCs, NV-IONP not only contained IONPs which act as a magnet-guided navigation tool but also carried greater amounts of therapeutic growth factors that can be delivered to the target cells. The increased amounts of therapeutic growth factors inside NV-IONP were attributed to IONPs that are slowly ionized to iron ions which activate the JNK and c-Jun signaling cascades in hMSCs. In vivo systemic injection of NV-IONP with magnetic guidance significantly increased the amount of NV-IONP accumulating in the injured spinal cord. Accumulated NV-IONP enhanced blood vessel formation, attenuated inflammation and apoptosis in the injured spinal cord, and consequently improved spinal cord function. Taken together, these findings highlight the development of therapeutic efficacy-potentiated extracellular nanovesicles and demonstrate their feasibility for repairing injured spinal cord.
Collapse
Affiliation(s)
- Han Young Kim
- School of Chemical and Biological Engineering , Seoul National University , Seoul , 08826 , Republic of Korea
| | - Hemant Kumar
- Department of Neurosurgery , CHA University , CHA Bundang Medical Center, Seongnam-si , 13488 , Republic of Korea
| | - Min-Jae Jo
- Department of Neurosurgery , CHA University , CHA Bundang Medical Center, Seongnam-si , 13488 , Republic of Korea
| | - Jonghoon Kim
- School of Chemical and Biological Engineering , Seoul National University , Seoul , 08826 , Republic of Korea
- Center for Nanoparticle Research , Institute for Basic Science (IBS) , Seoul 151-744 , Republic of Korea
| | - Jeong-Kee Yoon
- School of Chemical and Biological Engineering , Seoul National University , Seoul , 08826 , Republic of Korea
| | - Ju-Ro Lee
- School of Chemical and Biological Engineering , Seoul National University , Seoul , 08826 , Republic of Korea
| | - Mikyung Kang
- Interdisciplinary Program of Bioengineering , Seoul National University , Seoul , 08826 , Republic of Korea
| | - Yeon Woong Choo
- School of Chemical and Biological Engineering , Seoul National University , Seoul , 08826 , Republic of Korea
| | - Seuk Young Song
- School of Chemical and Biological Engineering , Seoul National University , Seoul , 08826 , Republic of Korea
| | - Sung Pil Kwon
- School of Chemical and Biological Engineering , Seoul National University , Seoul , 08826 , Republic of Korea
| | - Taeghwan Hyeon
- School of Chemical and Biological Engineering , Seoul National University , Seoul , 08826 , Republic of Korea
- Center for Nanoparticle Research , Institute for Basic Science (IBS) , Seoul 151-744 , Republic of Korea
| | - In-Bo Han
- Department of Neurosurgery , CHA University , CHA Bundang Medical Center, Seongnam-si , 13488 , Republic of Korea
| | - Byung-Soo Kim
- School of Chemical and Biological Engineering , Seoul National University , Seoul , 08826 , Republic of Korea
- Interdisciplinary Program of Bioengineering , Seoul National University , Seoul , 08826 , Republic of Korea
- Institute of Chemical Processes , Seoul National University , Seoul , 08826 , Republic of Korea
| |
Collapse
|
41
|
Low-affinity Nerve Growth Factor Receptor (CD271) Heterogeneous Expression in Adult and Fetal Mesenchymal Stromal Cells. Sci Rep 2018; 8:9321. [PMID: 29915318 PMCID: PMC6006357 DOI: 10.1038/s41598-018-27587-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 06/01/2018] [Indexed: 12/28/2022] Open
Abstract
Human multipotent mesenchymal stromal cells (MSC) are isolated from a plethora of tissue sources for cell therapy purposes. In 2006, the International Society for Cellular Therapy (ISCT) published minimal guidelines to define MSC identity. Nevertheless, many independent studies demonstrated that cells meeting the ISCT criteria possessed heterogeneous phenotypes and functionalities, heavily influenced by culture conditions. In this study, human MSC derived from many adult (bone marrow and adipose tissue) or fetal (cord blood, Wharton's jelly, umbilical cord perivascular compartment and amniotic fluid) tissues were investigated. Their immunophenotype was analyzed to define consistent source-specific markers by extensive flow cytometry analysis and real-time qRT-PCR. CD271+ subpopulations were detected in adult MSC, whereas NG2 was significantly more expressed in fetal MSC but failed validation on independent samples coming from an external laboratory. The highest number of CD271+ adult MSC were detected soon after isolation in serum-based culture conditions. Furthermore, heterogeneous percentages of CD271 expression were found in platelet lysate-based or serum-free culture conditions. Finally, CD271+ adult MSC showed high clonogenic and osteogenic properties as compared to CD271- cells. To conclude, in this phenotype-function correlation study CD271+ subpopulation confers heterogeneity on adult MSC, confirming the need of more specific markers to address MSC properties.
Collapse
|
42
|
Fas-L promotes the stem cell potency of adipose-derived mesenchymal cells. Cell Death Dis 2018; 9:695. [PMID: 29891848 PMCID: PMC5995957 DOI: 10.1038/s41419-018-0702-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 04/26/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022]
Abstract
Fas-L is a TNF family member known to trigger cell death. It has recently become evident that Fas-L can transduce also non-apoptotic signals. Mesenchymal stem cells (MSCs) are multipotent cells that are derived from various adult tissues. Although MSCs from different tissues display common properties they also display tissue-specific characteristics. Previous works have demonstrated massive apoptosis following Fas-L treatment of bone marrow-derived MSCs both in vitro and following their administration in vivo. We therefore set to examine Fas-L-induced responses in adipose-derived stem cells (ASCs). Human ASCs were isolated from lipoaspirates and their reactivity to Fas-L treatment was examined. ASCs responded to Fas-L by simultaneous apoptosis and proliferation, which yielded a net doubling of cell quantities and a phenotypic shift, including reduced expression of CD105 and increased expression of CD73, in association with increased bone differentiation potential. Treatment of freshly isolated ASCs led to an increase in large colony forming unit fibroblasts, likely produced by early stem cell progenitor cells. Fas-L-induced apoptosis and proliferation signaling were found to be independent as caspase inhibition attenuated Fas-L-induced apoptosis without impacting proliferation, whereas inhibition of PI3K and MEK, but not of JNK, attenuated Fas-L-dependent proliferation, but not apoptosis. Thus, Fas-L signaling in ASCs leads to their expansion and phenotypic shift toward a more potent stem cell state. We speculate that these reactions ensure the survival of ASC progenitor cells encountering Fas-L-enriched environments during tissue damage and inflammation and may also enhance ASC survival following their administration in vivo.
Collapse
|
43
|
Mak WC, Magne B, Cheung KY, Atanasova D, Griffith M. Thermo-rheological responsive microcapsules for time-dependent controlled release of human mesenchymal stromal cells. Biomater Sci 2018; 5:2241-2250. [PMID: 28972602 DOI: 10.1039/c7bm00663b] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Human mesenchymal stromal cells (hMSCs) are adult-source cells that have been extensively evaluated for cell-based therapies. hMSCs delivered by intravascular injection have been reported to accumulate at the sites of injury to promote tissue repair and can also be employed as vectors for the delivery of therapeutic genes. However, the full potential of hMSCs remains limited as the cells are lost after injection due to anoikis and the adverse pathologic environment. Encapsulation of cells has been proposed as a means of increasing cell viability. However, controlling the release of therapeutic cells over time to target tissue still remains a challenge today. Here, we report the design and development of thermo-rheological responsive hydrogels that allow for precise, time dependent controlled-release of hMSCs. The encapsulated hMSCs retained good viability from 76% to 87% dependent upon the hydrogel compositions. We demonstrated the design of different blended hydrogel composites with modulated strength (S parameter) and looseness of hydrogel networks (N parameter) to control the release of hMSCs from thermo-responsive hydrogel capsules. We further showed the feasibility for controlled-release of encapsulated hMSCs within 3D matrix scaffolds. We reported for the first time by a systematic analysis that there is a direct correlation between the thermo-rheological properties associated with the degradation of the hydrogel composite and the cell release kinetics. This work therefore provides new insights into the further development of smart carrier systems for stem cell therapy.
Collapse
Affiliation(s)
- W C Mak
- Department of Clinical and Experimental Medicine, Linköping University, SE58185, Linköping, Sweden.
| | | | | | | | | |
Collapse
|
44
|
Jakovljevic J, Harrell CR, Fellabaum C, Arsenijevic A, Jovicic N, Volarevic V. Modulation of autophagy as new approach in mesenchymal stem cell-based therapy. Biomed Pharmacother 2018; 104:404-410. [PMID: 29787987 DOI: 10.1016/j.biopha.2018.05.061] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 05/08/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Due to their trophic and immunoregulatory characteristics mesenchymal stem cells (MSCs) have tremendous potential for use in a variety of clinical applications. Challenges in MSCs' clinical applications include low survival of transplanted cells and low grafting efficiency requiring use of a high number of MSCs to achieve therapeutic benefits. Accordingly, new approaches are urgently needed in order to overcome these limitations. Recent evidence indicates that modulation of autophagy in MSCs prior to their transplantation enhances survival and viability of engrafted MSCs and promotes their pro-angiogenic and immunomodulatory characteristics. Here, we review the current literature describing mechanisms by which modulation of autophagy strengthens pro-angiogenic and immunosuppressive characteristics of MSCs in animal models of multiple sclerosis, osteoporosis, diabetic limb ischemia, myocardial infarction, acute graft-versus-host disease, kidney and liver diseases. Obtained results suggest that modulation of autophagy in MSCs may represent a new therapeutic approach that could enhance efficacy of MSCs in the treatment of ischemic and autoimmune diseases.
Collapse
Affiliation(s)
- Jelena Jakovljevic
- University of Kragujevac Serbia, Faculty of Medical Sciences, Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, 69 Svetozar Markovic Street, 34000, Kragujevac, Serbia
| | - C Randall Harrell
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N Palm Harbor, Palm Harbor, Florida, United States
| | - Crissy Fellabaum
- Regenerative Processing Plant, LLC, 34176 US Highway 19 N Palm Harbor, Palm Harbor, Florida, United States
| | - Aleksandar Arsenijevic
- University of Kragujevac Serbia, Faculty of Medical Sciences, Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, 69 Svetozar Markovic Street, 34000, Kragujevac, Serbia
| | - Nemanja Jovicic
- University of Kragujevac Serbia, Faculty of Medical Sciences, Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, 69 Svetozar Markovic Street, 34000, Kragujevac, Serbia
| | - Vladislav Volarevic
- University of Kragujevac Serbia, Faculty of Medical Sciences, Department of Microbiology and immunology, Center for Molecular Medicine and Stem Cell Research, 69 Svetozar Markovic Street, 34000, Kragujevac, Serbia.
| |
Collapse
|
45
|
Talloj SK, Cheng B, Weng JP, Lin HC. Glucosamine-Based Supramolecular Nanotubes for Human Mesenchymal Cell Therapy. ACS APPLIED MATERIALS & INTERFACES 2018; 10:15079-15087. [PMID: 29651840 DOI: 10.1021/acsami.8b03226] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Herein, we demonstrate an example of glucosamine-based supramolecular hydrogels that can be used for human mesenchymal cell therapy. We designed and synthesized a series of amino acid derivatives based on a strategy of capping d-glucosamine moiety at the C-terminus and fluorinated benzyl group at the N-terminus. From a systematic study on chemical structures, we discovered that the glucosamine-based supramolecular hydrogel [pentafluorobenzyl (PFB)-F-Glu] self-assembled with one-dimensional nanotubular structures at physiological pH. The self-assembly of a newly discovered PFB-F-Glu motif is attributed to the synergistic effect of π-π stacking and extensive intermolecular hydrogen bonding network in aqueous medium. Notably, PFB-F-Glu nanotubes are proven to be nontoxic to human mesenchymal stem cells (hMSCs) and have been shown to enhance hMSC proliferation while maintaining their pluripotency. Retaining of pluripotency capabilities provides potentially unlimited source of undifferentiated cells for the treatment of future cell therapies. Furthermore, hMSCs cultured on PFB-F-Glu are able to secrete paracrine factors that downregulate profibrotic gene expression in lipopolysaccharide-treated human skin fibroblasts, which demonstrates that PFB-F-Glu nanotubes have the potential to be used for wound healing applications. Overall, this article addresses the importance of chemical design to generate supramolecular biomaterials for stem cell therapy.
Collapse
Affiliation(s)
- Satish Kumar Talloj
- Department of Materials Science and Engineering , National Chiao Tung University , Hsinchu 30010 , Taiwan , Republic of China
| | - Bill Cheng
- Department of Materials Science and Engineering , National Chiao Tung University , Hsinchu 30010 , Taiwan , Republic of China
| | - Jen-Po Weng
- Department of Materials Science and Engineering , National Chiao Tung University , Hsinchu 30010 , Taiwan , Republic of China
| | - Hsin-Chieh Lin
- Department of Materials Science and Engineering , National Chiao Tung University , Hsinchu 30010 , Taiwan , Republic of China
| |
Collapse
|
46
|
Zuo W, Xie B, Li C, Yan Y, Zhang Y, Liu W, Huang J, Chen D. The Clinical Applications of Endometrial Mesenchymal Stem Cells. Biopreserv Biobank 2018; 16:158-164. [PMID: 29265881 PMCID: PMC5906727 DOI: 10.1089/bio.2017.0057] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Endometrial mesenchymal stem cells (enMSCs) are a class of novel adult stem cells with self-renewal capacity, differentiation potential, low immunogenicity, low tumorigenicity, and other biological characteristics. Since the discovery of enMSCs, they have become a hot research topic. In recent years, research on enMSC isolation and application have made great progress. In this review, we focus on the clinical applications of this cell type. The latest research on the applications of enMSCs in the immune, gynecological, cardiovascular, digestive, nervous systems and metabolic diseases, as well as biobanking of enMSCs will be reviewed.
Collapse
Affiliation(s)
- Wanyun Zuo
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Bingyu Xie
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Chenglong Li
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yuhan Yan
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yangyi Zhang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Liu
- Institute of Human Reproductive and Stem Cell Engineering, Xiangya School of Medicine, Central South University, Changsha, China
| | - Jufang Huang
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Dan Chen
- Department of Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
47
|
Devetzi M, Goulielmaki M, Khoury N, Spandidos DA, Sotiropoulou G, Christodoulou I, Zoumpourlis V. Genetically‑modified stem cells in treatment of human diseases: Tissue kallikrein (KLK1)‑based targeted therapy (Review). Int J Mol Med 2018; 41:1177-1186. [PMID: 29328364 PMCID: PMC5819898 DOI: 10.3892/ijmm.2018.3361] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/02/2018] [Indexed: 12/12/2022] Open
Abstract
The tissue kallikrein-kinin system (KKS) is an endogenous multiprotein metabolic cascade which is implicated in the homeostasis of the cardiovascular, renal and central nervous system. Human tissue kallikrein (KLK1) is a serine protease, component of the KKS that has been demonstrated to exert pleiotropic beneficial effects in protection from tissue injury through its anti-inflammatory, anti-apoptotic, anti-fibrotic and anti-oxidative actions. Mesenchymal stem cells (MSCs) or endothelial progenitor cells (EPCs) constitute populations of well-characterized, readily obtainable multipotent cells with special immunomodulatory, migratory and paracrine properties rendering them appealing potential therapeutics in experimental animal models of various diseases. Genetic modification enhances their inherent properties. MSCs or EPCs are competent cellular vehicles for drug and/or gene delivery in the targeted treatment of diseases. KLK1 gene delivery using adenoviral vectors or KLK1 protein infusion into injured tissues of animal models has provided particularly encouraging results in attenuating or reversing myocardial, renal and cerebrovascular ischemic phenotype and tissue damage, thus paving the way for the administration of genetically modified MSCs or EPCs with the human tissue KLK1 gene. Engraftment of KLK1-modified MSCs and/or KLK1-modified EPCs resulted in advanced beneficial outcome regarding heart and kidney protection and recovery from ischemic insults. Collectively, findings from pre-clinical studies raise the possibility that tissue KLK1 may be a novel future therapeutic target in the treatment of a wide range of cardiovascular, cerebrovascular and renal disorders.
Collapse
Affiliation(s)
- Marina Devetzi
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Maria Goulielmaki
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Nicolas Khoury
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, 71003 Heraklion, Greece
| | | | - Ioannis Christodoulou
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 11635 Athens, Greece
| |
Collapse
|
48
|
Bartlett RS, Guille JT, Chen X, Christensen MB, Wang SF, Thibeault SL. Mesenchymal stromal cell injection promotes vocal fold scar repair without long-term engraftment. Cytotherapy 2017; 18:1284-96. [PMID: 27637759 DOI: 10.1016/j.jcyt.2016.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 07/18/2016] [Accepted: 07/24/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Regenerative medicine holds promise for restoring voice in patients with vocal fold scarring. As experimental treatments approach clinical translation, several considerations remain. Our objective was to evaluate efficacy and biocompatibility of four bone marrow mesenchymal stromal cell (BM-MSC) and tunable hyaluronic acid based hydrogel (HyStem-VF) treatments for vocal fold scar using clinically acceptable materials, a preclinical sample size and a dosing comparison. METHODS Vocal folds of 84 rabbits were injured and injected with four treatment variations (BM-MSC, HyStem-VF, and BM-MSC in HyStem-VF at two concentrations) 6 weeks later. Efficacy was assessed with rheometry, real-time polymerase chain reaction (RT-PCR) and histology at 2, 4 and 10 weeks following treatment. Lung, liver, kidney, spleen and vocal folds were screened for biocompatibility by a pathologist. RESULTS AND DISCUSSION Persistent inflammation was identified in all hydrogel-injected groups. The BM-MSC alone treatment appeared to be the most efficacious and safe, providing an early resolution of viscoelasticity, gene expression consistent with desirable extracellular matrix remodeling (less fibronectin, collagen 1α2, collagen 3, procollagen, transforming growth factor [TGF]β1, alpha smooth muscle actin, interleukin-1β, interleukin-17β and tumor necrosis factor [TNF] than injured controls) and minimal inflammation. Human beta actin expression in BM-MSC-treated vocal folds was minimal after 2 weeks, suggesting that paracrine signaling from the BM-MSCs may have facilitated tissue repair.
Collapse
Affiliation(s)
- R S Bartlett
- Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - J T Guille
- Department of ENT and Head and Neck Surgery, University Hospital of Pointe à Pitre, Guadeloupe, French West Indies
| | - X Chen
- Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - M B Christensen
- National Center for Voice and Speech, University of Utah, Salt Lake City, Utah, USA
| | - S F Wang
- Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - S L Thibeault
- Department of Surgery, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
49
|
Zhu L, Liu YJ, Shen H, Gu PQ, Zhang L. Astragalus and Baicalein Regulate Inflammation of Mesenchymal Stem Cells (MSCs) by the Mitogen-Activated Protein Kinase (MAPK)/ERK Pathway. Med Sci Monit 2017; 23:3209-3216. [PMID: 28667247 PMCID: PMC5507801 DOI: 10.12659/msm.902441] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) have emerged as an attractive alternative to modulating immune response after transplantation. Recent studies have shown that systemically administered MSCs enter the inflamed intestine. In the present study, we propose a strategy to improve the efficacy of MSC-based cellular therapy for inflammation using Astragaloside and Baicalein to enhance cell survival, inhibit apoptosis, and modulate inflammatory response in vitro. Material/Methods MSCs were induced with lipopolysaccharide (LPS) as an inflammatory model before being treated for 48 h with Astragaloside, Baicalein, and the combination of both. MSCs proliferation was determined using the MTT method. The cell cycle situation was monitored using flow cytometry, and the apoptosis ability of MSCs was detected with Annexin-V flow cytometry. The levels of cytokine IL-1β, IL-8, and TNF-α, and their relations with the ERK pathway were measured using ELISA, RT-PCR, and Western blot. Results Compared to the control groups (containing no drug), each drug-treated group showed the ability to promote epithelial differentiation and cell growth and to inhibit apoptosis. The combination group had reduced levels of IL-1β, IL-8, and TNF-α in LPS-induced MSCs, much more than in the other 2 groups. Compared with the other groups, the combination of Astragaloside and Baicalin more efficiently reduced IL-1β, IL-8, and TNF-α levels in the LPS-induced MSCs model, and ERK inhibitor was capable of recovering the inflammatory effect. Conclusions The results demonstrated that Astragaloside and Baicalin can promote epithelial differentiation and proliferation, inhibit apoptosis, and reduce inflammatory effects.
Collapse
Affiliation(s)
- Lei Zhu
- Jiangsu Province Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (TCM), Nanjing, Jiangsu, China (mainland)
| | - Ya-Jun Liu
- Jiangsu Province Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (TCM), Nanjing, Jiangsu, China (mainland)
| | - Hong Shen
- Jiangsu Province Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (TCM), Nanjing, Jiangsu, China (mainland)
| | - Pei-Qing Gu
- Jiangsu Province Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Nanjing University of TCM, Nanjing, Jiangsu, China (mainland)
| | - Lu Zhang
- Jiangsu Province Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Nanjing University of Traditional Chinese Medicine (TCM), Nanjing, Jiangsu, China (mainland)
| |
Collapse
|
50
|
Rammal H, Dubus M, Aubert L, Reffuveille F, Laurent-Maquin D, Terryn C, Schaaf P, Alem H, Francius G, Quilès F, Gangloff SC, Boulmedais F, Kerdjoudj H. Bioinspired Nanofeatured Substrates: Suitable Environment for Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2017; 9:12791-12801. [PMID: 28301131 DOI: 10.1021/acsami.7b01665] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Bone mimicking coatings provide a complex microenvironment in which material, through its inherent properties (such as nanostructure and composition), affects the commitment of stem cells into bone lineage and the production of bone tissue regulating factors required for bone healing and regeneration. Herein, a bioactive mineral/biopolymer composite made of calcium phosphate/chitosan and hyaluronic acid (CaP-CHI-HA) was elaborated using a versatile simultaneous spray coating of interacting species. The resulting CaP-CHI-HA coating was mainly constituted of bioactive, carbonated and crystalline hydroxyapatite with 277 ± 98 nm of roughness, 1 μm of thickness, and 2.3 ± 1 GPa of stiffness. After five days of culture, CaP-CHI-HA suggested a synergistic effect of intrinsic biophysical features and biopolymers on stem cell mechanobiology and nuclear organization, leading to the expression of an early osteoblast-like phenotype and the production of bone tissue regulating factors such as osteoprotegerin and vascular endothelial growth factor. More interestingly, amalgamation with biopolymers conferred to the mineral a bacterial antiadhesive property. These significant data shed light on the potential regenerative application of CaP-CHI-HA bioinspired coating in providing a suitable environment for stem cell bone regeneration and an ideal strategy to prevent implant-associated infections.
Collapse
Affiliation(s)
- H Rammal
- EA 4691, Biomatériaux et Inflammation en Site Osseux (BIOS), SFR-CAP Santé (FED 4231), Université de Reims Champagne Ardenne , 51100 Reims, France
- UFR d'Odontologie, Université de Reims Champagne Ardenne , 51100 Reims, France
| | - M Dubus
- EA 4691, Biomatériaux et Inflammation en Site Osseux (BIOS), SFR-CAP Santé (FED 4231), Université de Reims Champagne Ardenne , 51100 Reims, France
- UFR d'Odontologie, Université de Reims Champagne Ardenne , 51100 Reims, France
| | - L Aubert
- EA 4691, Biomatériaux et Inflammation en Site Osseux (BIOS), SFR-CAP Santé (FED 4231), Université de Reims Champagne Ardenne , 51100 Reims, France
- UFR de Pharmacie, Université de Reims Champagne Ardenne , 51100 Reims, France
| | - F Reffuveille
- EA 4691, Biomatériaux et Inflammation en Site Osseux (BIOS), SFR-CAP Santé (FED 4231), Université de Reims Champagne Ardenne , 51100 Reims, France
- UFR de Pharmacie, Université de Reims Champagne Ardenne , 51100 Reims, France
| | - D Laurent-Maquin
- EA 4691, Biomatériaux et Inflammation en Site Osseux (BIOS), SFR-CAP Santé (FED 4231), Université de Reims Champagne Ardenne , 51100 Reims, France
- UFR d'Odontologie, Université de Reims Champagne Ardenne , 51100 Reims, France
| | - C Terryn
- Plateforme d'Imagerie Cellulaire et Tissulaire (PICT), Université de Reims Champagne Ardenne , 51100 Reims, France
| | - P Schaaf
- INSERM, UMR-S 1121, "Biomatériaux et Bioingénierie", Fédération de médecine translationnelle de Strasbourg, Faculté de Chirurgie Dentaire, Université de Strasbourg , 67000 Strasbourg, France
- CNRS, Institut Charles Sadron UPR 22, Université de Strasbourg , 23 rue du Loess, 67034 Strasbourg Cedex, France
| | - H Alem
- CNRS, UMR 7198, Institut Jean Lamour (IJL), Université de Lorraine , 54500 Vandoeuvre Lès Nancy, France
| | - G Francius
- CNRS, UMR 7564, Laboratoire de Chimie Physique et Microbiologie pour l'Environnement (LCPME), Université de Lorraine , 54500 Vandoeuvre Lès Nancy, France
| | - F Quilès
- CNRS, UMR 7564, Laboratoire de Chimie Physique et Microbiologie pour l'Environnement (LCPME), Université de Lorraine , 54500 Vandoeuvre Lès Nancy, France
| | - S C Gangloff
- EA 4691, Biomatériaux et Inflammation en Site Osseux (BIOS), SFR-CAP Santé (FED 4231), Université de Reims Champagne Ardenne , 51100 Reims, France
- UFR de Pharmacie, Université de Reims Champagne Ardenne , 51100 Reims, France
| | - F Boulmedais
- CNRS, Institut Charles Sadron UPR 22, Université de Strasbourg , 23 rue du Loess, 67034 Strasbourg Cedex, France
| | - H Kerdjoudj
- EA 4691, Biomatériaux et Inflammation en Site Osseux (BIOS), SFR-CAP Santé (FED 4231), Université de Reims Champagne Ardenne , 51100 Reims, France
- UFR d'Odontologie, Université de Reims Champagne Ardenne , 51100 Reims, France
| |
Collapse
|