51
|
Chen J, Zheng CX, Jin Y, Hu CH. Mesenchymal stromal cell-mediated immune regulation: A promising remedy in the therapy of type 2 diabetes mellitus. STEM CELLS (DAYTON, OHIO) 2021; 39:838-852. [PMID: 33621403 DOI: 10.1002/stem.3357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/03/2021] [Indexed: 11/09/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a major threat to global public health, with increasing prevalence as well as high morbidity and mortality, to which immune dysfunction has been recognized as a crucial contributor. Mesenchymal stromal cells (MSCs), obtained from various sources and possessing potent immunomodulatory abilities, have displayed great therapeutic potential for T2DM. Interestingly, the immunomodulatory capabilities of MSCs are endowed and plastic. Among the multiple mechanisms involved in MSC-mediated immune regulation, the paracrine effects of MSCs have attracted much attention. Of note, extracellular vesicles (EVs), an important component of MSC secretome, have emerged as pivotal mediators of their immunoregulatory effects. Particularly, the necrobiology of MSCs, especially apoptosis, has recently been revealed to affect their immunomodulatory functions in vivo. In specific, a variety of preclinical studies have demonstrated the beneficial effects of MSCs on improving islet function and ameliorating insulin resistance. More importantly, clinical trials have further uncovered the therapeutic potential of MSCs for T2DM. In this review, we outline current knowledge regarding the plasticity and underlying mechanisms of MSC-mediated immune modulation, focusing on the paracrine effects. We also summarize the applications of MSC-based therapies for T2DM in both preclinical studies and clinical trials, with particular emphasis on the modulation of immune system.
Collapse
Affiliation(s)
- Ji Chen
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases,Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China.,Department of Oral Implantology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Chen-Xi Zheng
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases,Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Yan Jin
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases,Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China
| | - Cheng-Hu Hu
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases,Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi'an, People's Republic of China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, People's Republic of China
| |
Collapse
|
52
|
Mebarki M, Abadie C, Larghero J, Cras A. Human umbilical cord-derived mesenchymal stem/stromal cells: a promising candidate for the development of advanced therapy medicinal products. Stem Cell Res Ther 2021; 12:152. [PMID: 33637125 PMCID: PMC7907784 DOI: 10.1186/s13287-021-02222-y] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/11/2021] [Indexed: 02/06/2023] Open
Abstract
Umbilical cord-derived mesenchymal stem/stromal cells (UC-MSCs) emerge as a perspective for therapeutic use in immune and inflammatory diseases. Indeed, immunomodulatory and anti-inflammatory properties, associated to fewer ethical, availability, and safety issues, position UC-MSCs as a promising active substance to develop medicinal products. Since 2007, UC-MSC-based products are classified as advanced therapy medicinal products (ATMP) according to the European Regulation 1394/2007/EC. This new regulatory status required a total adaptation of stakeholders wishing to develop UC-MSC-based ATMPs. Cell production in tissue and cell banks has been replaced by the manufacturing of a medicine, in authorized establishments, according to the good manufacturing practices (GMP) specific to ATMPs. After a brief description of UC-MSCs, we described in this review their recent use in a large panel of immune and inflammatory pathologies, including early and late phase clinical trials. Despite the use of the same product, we noticed an important heterogeneity in terms of indication, posology and study design. Then, we discussed regulatory and manufacturing challenges for stakeholders, especially in terms of process harmonization and cells characterization. Our aim was to point that despite MSCs use for several decades, the development of an UC-MSC-based ATMP remains at this day a real challenge for both academic institutions and pharmaceutical companies.
Collapse
Affiliation(s)
- Miryam Mebarki
- Assistance Publique - Hôpitaux de Paris, Hôpital Saint-Louis, DMU PRISM, Unité de Thérapie Cellulaire, 1 avenue Claude Vellefaux, 75010 Paris, France
- INSERM CIC de Biothérapies CBT501, 1 avenue Claude Vellefaux, 75010 Paris, France
- Université de Paris, INSERM U976, 1 avenue Claude Vellefaux, 75010 Paris, France
- Faculté de Pharmacie, Université de Paris, 4 Avenue de l’Observatoire, 75006 Paris, France
| | - Camille Abadie
- INSERM CIC de Biothérapies CBT501, 1 avenue Claude Vellefaux, 75010 Paris, France
| | - Jérôme Larghero
- Assistance Publique - Hôpitaux de Paris, Hôpital Saint-Louis, DMU PRISM, Unité de Thérapie Cellulaire, 1 avenue Claude Vellefaux, 75010 Paris, France
- INSERM CIC de Biothérapies CBT501, 1 avenue Claude Vellefaux, 75010 Paris, France
- Université de Paris, INSERM U976, 1 avenue Claude Vellefaux, 75010 Paris, France
| | - Audrey Cras
- Assistance Publique - Hôpitaux de Paris, Hôpital Saint-Louis, DMU PRISM, Unité de Thérapie Cellulaire, 1 avenue Claude Vellefaux, 75010 Paris, France
- INSERM CIC de Biothérapies CBT501, 1 avenue Claude Vellefaux, 75010 Paris, France
- Faculté de Pharmacie, Université de Paris, 4 Avenue de l’Observatoire, 75006 Paris, France
- Université de Paris, INSERM UMR1140, 4 Avenue de l’Observatoire, 75006 Paris, France
| |
Collapse
|
53
|
Khatri R, Petry SF, Linn T. Intrapancreatic MSC transplantation facilitates pancreatic islet regeneration. Stem Cell Res Ther 2021; 12:121. [PMID: 33579357 PMCID: PMC7881671 DOI: 10.1186/s13287-021-02173-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
Background Type 1 diabetes mellitus (T1D) is characterized by the autoimmune destruction of the pancreatic β cells. The transplantation of mesenchymal stromal/stem cells (MSC) was reported to rescue the damaged pancreatic niche. However, there is an ongoing discussion on whether direct physical contact between MSC and pancreatic islets results in a superior outcome as opposed to indirect effects of soluble factors released from the MSC entrapped in the lung microvasculature after systemic administration. Hence, MSC were studied in direct contact (DC) and indirect contact (IDC) with murine pancreatic β cell line MIN6-cells damaged by nitrosourea derivative streptozotocin (STZ) in vitro. Further, the protective and antidiabetic outcome of MSC transplantation was evaluated through the intrapancreatic route (IPR) and intravenous route (IVR) in STZ-induced diabetic NMRI nude mice. Methods MSC were investigated in culture with STZ-damaged MIN6-cells, either under direct contact (DC) or separated through a semi-permeable membrane (IDC). Moreover, multiple low doses of STZ were administered to NMRI nude mice for the induction of hyperglycemia. 0.5 × 106 adipose-derived mesenchymal stem cells (ADMSC) were transferred through direct injection into the pancreas (IPR) or the tail vein (IVR), respectively. Bromodeoxyuridine (BrdU) was injected for the detection of proliferating islet cells in vivo, and real-time polymerase chain reaction (RT-PCR) was employed for the measurement of the expression of growth factor and immunomodulatory genes in the murine pancreas and human MSC. Phosphorylation of AKT and ERK was analyzed with Western blotting. Results The administration of MSC through IPR ameliorated hyperglycemia in contrast to IVR, STZ, and non-diabetic control in a 30-day window. IPR resulted in a higher number of replicating islet cells, number of islets, islet area, growth factor (EGF), and balancing of the Th1/Th2 response in vivo. Physical contact also provided a superior protection to MIN6-cells from STZ through the AKT and ERK pathway in vitro in comparison with IDC. Conclusion Our study suggests that the physical contact between MSC and pancreatic islet cells is required to fully unfold their protective potential. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02173-4.
Collapse
Affiliation(s)
- Rahul Khatri
- Clinical Research Unit, Centre of Internal Medicine, Faculty of Medicine, Justus Liebig University Giessen, Friedrichstrasse. 20/ Aulweg 123, 35392, Giessen, Germany
| | - Sebastian Friedrich Petry
- Clinical Research Unit, Centre of Internal Medicine, Faculty of Medicine, Justus Liebig University Giessen, Friedrichstrasse. 20/ Aulweg 123, 35392, Giessen, Germany
| | - Thomas Linn
- Clinical Research Unit, Centre of Internal Medicine, Faculty of Medicine, Justus Liebig University Giessen, Friedrichstrasse. 20/ Aulweg 123, 35392, Giessen, Germany.
| |
Collapse
|
54
|
Zhang J, Wang YN, Jia T, Huang H, Zhang D, Xu X. Genipin and insulin combined treatment improves implant osseointegration in type 2 diabetic rats. J Orthop Surg Res 2021; 16:59. [PMID: 33446235 PMCID: PMC7809857 DOI: 10.1186/s13018-021-02210-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 01/05/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) has a harmful effect on the stability and osseointegration of dental implants. T2DM induces mitochondrial damage by inhibiting AMPK signaling, resulting in oxidative stress and poor osteogenesis in the peri-implant bone area. Genipin is a major component of gardenia fruits with strong antioxidant, anti-inflammation, and antidiabetic actions, and it also can activate mitochondrial quality control via the AMPK pathway. The purpose of this study was to investigate the effects of genipin and insulin treatment on implant osseointegration in T2DM rats and explore the underlying mechanisms. METHODS Streptozotocin-induced diabetic rats received implant surgery in their femurs and were then assigned to five groups that were subjected to different treatments for three months: control group, T2DM group, insulin-treated T2DM group (10 IU/kg), genipin-treated T2DM group (50 mg/kg), and the genipin and insulin combination-treated T2DM group. Then, we regularly assessed the weight and glucose levels of the animals. Rats were euthanized at 3 months after the implantation procedure, and the femora were harvested for microscopic computerized tomography analysis, biomechanical tests, and different histomorphometric assessment. RESULTS The results indicated that the highest blood glucose and oxidative stress levels were measured for the T2DM group, resulting in the poorest osseointegration. The combination-treated T2DM group mitigated hyperglycemia and normalized, reactivated AMPK signaling, and alleviated oxidative stress as well as reversed the negative effect of osseointegration. There were beneficial changes observed in the T2DM-genipin and T2DM-insulin groups, but these were less in comparison to the combination treatment group. CONCLUSION Our study suggests that treatment with genipin in combination with insulin could be an effective method for promoting implant osseointegration in T2DM rats, which may be related to AMPK signaling.
Collapse
Affiliation(s)
- Jiajia Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1, Wenhua Road West, Jinan, 250012, Shandong Province, China
| | - Ya-Nan Wang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1, Wenhua Road West, Jinan, 250012, Shandong Province, China
| | - Tingting Jia
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1, Wenhua Road West, Jinan, 250012, Shandong Province, China
| | - Haiyun Huang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1, Wenhua Road West, Jinan, 250012, Shandong Province, China
| | - Dongjiao Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1, Wenhua Road West, Jinan, 250012, Shandong Province, China.
| | - Xin Xu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1, Wenhua Road West, Jinan, 250012, Shandong Province, China.
| |
Collapse
|
55
|
Wang J, Liu Y, Ding H, Shi X, Ren H. Mesenchymal stem cell-secreted prostaglandin E 2 ameliorates acute liver failure via attenuation of cell death and regulation of macrophage polarization. Stem Cell Res Ther 2021; 12:15. [PMID: 33413632 PMCID: PMC7792134 DOI: 10.1186/s13287-020-02070-2] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/02/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Acute liver failure (ALF) is an acute inflammatory liver disease with high mortality. Previous preclinical and clinical trials have confirmed that mesenchymal stem cell (MSC) is a promising therapeutic approach; however, the effect is not satisfied as the underlying molecular mechanisms of MSC in treating ALF remain unclear. METHODS MSC isolated from 4- to 6-week-old C57BL/6 mice were used to treat ALF. Histological and serological parameters were analyzed to evaluate the efficacy of MSC. We explored the molecular mechanism of MSC in the treatment of ALF by detecting liver inflammatory response and hepatocyte death. RESULTS In this study, we found that the therapeutic potential of MSC on ALF is dependent on the secretion of prostaglandin E2 (PGE2), a bioactive lipid. MSC-derived PGE2 inhibited TGF-β-activated kinase 1 (TAK1) signaling and NLRP3 inflammasome activation in liver macrophages to decrease the production of inflammatory cytokines. Meanwhile, macrophages in the liver could be induced to anti-inflammatory (M2) macrophages by MSC-derived PGE2 via STAT6 and mechanistic target of rapamycin (mTOR) signaling, which then promote inflammatory resolution and limit liver injury. Finally, administrating EP4 antagonist significantly ameliorated the therapeutic ability of MSC, which promoted liver inflammation and decreased M2 macrophages. CONCLUSIONS Our results indicate that PGE2 might be a novel important mediator of MSC in treating ALF, which is through inhibiting the liver inflammatory response and hepatocyte death.
Collapse
Affiliation(s)
- Jinglin Wang
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Department of Hepatobiliary Surgery, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Yang Liu
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
| | - Haoran Ding
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China
- Department of Hepatobiliary Surgery, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China
| | - Xiaolei Shi
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China.
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.
- Department of Hepatobiliary Surgery, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.
| | - Haozhen Ren
- Department of Hepatobiliary Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China.
- Department of Hepatobiliary Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.
- Department of Hepatobiliary Surgery, Nanjing University of Chinese Medicine, Nanjing, Jiangsu Province, China.
| |
Collapse
|
56
|
Wang Y, Shan SK, Guo B, Li F, Zheng MH, Lei LM, Xu QS, Ullah MHE, Xu F, Lin X, Yuan LQ. The Multi-Therapeutic Role of MSCs in Diabetic Nephropathy. Front Endocrinol (Lausanne) 2021; 12:671566. [PMID: 34163437 PMCID: PMC8216044 DOI: 10.3389/fendo.2021.671566] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/20/2021] [Indexed: 12/16/2022] Open
Abstract
Diabetic nephropathy (DN) is one of the most common diabetes mellitus (DM) microvascular complications, which always ends with end-stage renal disease (ESRD). Up to now, as the treatment of DN in clinic is still complicated, ESRD has become the main cause of death in diabetic patients. Mesenchymal stem cells (MSCs), with multi-differentiation potential and paracrine function, have attracted considerable attention in cell therapy recently. Increasing studies concerning the mechanisms and therapeutic effect of MSCs in DN emerged. This review summarizes several mechanisms of MSCs, especially MSCs derived exosomes in DN therapy, including hyperglycemia regulation, anti-inflammatory, anti-fibrosis, pro-angiogenesis, and renal function protection. We also emphasize the limitation of MSCs application in the clinic and the enhanced therapeutic role of pre-treated MSCs in the DN therapy. This review provides balanced and impartial views for MSC therapy as a promising strategy in diabetic kidney disease amelioration.
Collapse
Affiliation(s)
- Yi Wang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Su-Kang Shan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Guo
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Fuxingzi Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming-Hui Zheng
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Li-Min Lei
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiu-Shuang Xu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Muhammad Hasnain Ehsan Ullah
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Xu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Lin
- Department of Radiology, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Qing Yuan
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, the Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Ling-Qing Yuan,
| |
Collapse
|
57
|
Bai X, Pei Q, Pu C, Chen Y, He S, Wang B. Multifunctional Islet Transplantation Hydrogel Encapsulating A20 High-Expressing Islets. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:4021-4027. [PMID: 33061306 PMCID: PMC7532915 DOI: 10.2147/dddt.s273050] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/09/2020] [Indexed: 12/17/2022]
Abstract
Islet transplantation is regarded as the most promising treatment for type 1 diabetes (T1D). However, the function of grafted islet could be damaged on account of transplant rejection and/or hypoxia several years later after transplantation. We proposed a hypothetical functionalized hydrogel model, which encapsulates sufficient A20 high-expressing islets and supporting cells, and performs as a drug release system releasing immunosuppressants and growth factors, to improve the outcome of pancreatic islet transplantation. Once injected in vivo, the hydrogel can gel and offer a robust mechanical structure for the A20 high-expressing islets and supporting cells. The natural biomaterials (eg, heparin) added into the hydrogel provide adhesive sites for islets to promote islets’ survival. Furthermore, the hydrogel encapsulates various supporting cells, which can facilitate the vascularization and/or prevent the immune system attacking the islet graft. Based on the previous studies that generally applied one or two combined strategies to protect the function of islet graft, we designed this hypothetical multifunctional encapsulation hydrogel model with various functions. We hypothesized that the islet graft could survive and maintain its function for a longer time in vivo compared with naked islets. This hypothetical model has a limitation in terms of clinical application. Future development work will focus on verifying the function and safety of this hypothetical islet transplantation hydrogel model in vitro and in vivo.
Collapse
Affiliation(s)
- Xue Bai
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Qilin Pei
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Chunyi Pu
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Yi Chen
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Sirong He
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Basic and Translational Research of Tumor Immunology, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Bin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing 400016, People's Republic of China
| |
Collapse
|
58
|
Meng QS, Liu J, Wei L, Fan HM, Zhou XH, Liang XT. Senescent mesenchymal stem/stromal cells and restoring their cellular functions. World J Stem Cells 2020; 12:966-985. [PMID: 33033558 PMCID: PMC7524698 DOI: 10.4252/wjsc.v12.i9.966] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/23/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) have various properties that make them promising candidates for stem cell-based therapies in clinical settings. These include self-renewal, multilineage differentiation, and immunoregulation. However, recent studies have confirmed that aging is a vital factor that limits their function and therapeutic properties as standardized clinical products. Understanding the features of senescence and exploration of cell rejuvenation methods are necessary to develop effective strategies that can overcome the shortage and instability of MSCs. This review will summarize the current knowledge on characteristics and functional changes of aged MSCs. Additionally, it will highlight cell rejuvenation strategies such as molecular regulation, non-coding RNA modifications, and microenvironment controls that may enhance the therapeutic potential of MSCs in clinical settings.
Collapse
Affiliation(s)
- Qing-Shu Meng
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai 200120, China
| | - Jing Liu
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai 200120, China
| | - Lu Wei
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai 200120, China
| | - Hui-Min Fan
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai 200120, China
- Department of Heart Failure, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiao-Hui Zhou
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai 200120, China
| | - Xiao-Ting Liang
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
- Institute of Integrated Traditional Chinese and Western Medicine for Cardiovascular Chronic Diseases, Tongji University School of Medicine, Shanghai 200120, China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China.
| |
Collapse
|
59
|
Zhou DY, Mou X, Liu K, Liu WH, Xu YQ, Zhou D. In silico prediction and validation of potential therapeutic genes in pancreatic β-cells associated with type 2 diabetes. Exp Ther Med 2020; 20:60. [PMID: 32952650 PMCID: PMC7485321 DOI: 10.3892/etm.2020.9188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 03/24/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetes mellitus is becoming a major health burden worldwide. Pancreatic β-cell death is a characteristic of type 2 diabetes (T2D), but the underlying mechanisms of pancreatic β-cell death remain unknown. Therefore, the aim of the present study was to identify potential targets in the pancreatic islet of T2D. The GSE20966 dataset was obtained from the Gene Expression Omnibus (GEO) database, and differentially expressed genes (DEGs) were identified by using the GEO2R tool. The Gene Ontology terms and Kyoto Encyclopedia of Genes and Genomes Pathway enrichment analysis of DEGs were further assessed using the Database for Annotation, Visualization and Integrated Discovery. Furthermore, protein-protein interaction (PPI) networks were constructed for the up- and downregulated genes using STRING databases and were then visualized with Cytoscape. The body weight, fasting blood glucose (FBG), pancreatic index and biochemistry parameters were measured in db/db mice. Moreover, the morphology of the pancreas was detected by hematoxylin and eosin staining, and hub genes were assessed using reverse transcription-quantitative PCR (RT-qPCR) and western blot analysis. In total, 570 DEGs were screened, including 376 upregulated and 194 downregulated genes, which were associated with 'complement activation, classical pathway', 'proteolysis', 'complement activation' and 'pancreatic secretion pathway'. It was found that the body weight, FBG, alanine aminotransferase, aspartate aminotransferase, total cholesterol, triglycerides, blood urea nitrogen, creatinine, fasting serum insulin, glucagon and low-density lipoprotein cholesterol levels were significantly higher in db/db mice, while high-density lipoprotein cholesterol levels and the pancreatic index were significantly decreased. Furthermore, albumin, interleukin-8, CD44, C-C motif chemokine ligand 2, hepatocyte growth factor, cystic fibrosis transmembrane conductance regulator, histone cluster 1 H2B family member n, mitogen-activated protein kinase 11 and neurotrophic receptor tyrosine kinase 2 were identified as hub genes in PPI network. RT-qPCR and western blotting results demonstrated the same expression trend in hub genes as found by the bioinformatics analysis. Therefore, the present study identified a series of hub genes involved in the progression of pancreatic β-cell, which may help to develop effective therapeutic strategy for T2D.
Collapse
Affiliation(s)
- Di Yi Zhou
- Department of Endocrinology, Zhejiang Integrated Traditional and Western Medicine Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Xin Mou
- Department of Endocrinology, Zhejiang Integrated Traditional and Western Medicine Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Kaiyuan Liu
- Department of Endocrinology, Zhejiang Integrated Traditional and Western Medicine Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Wen Hong Liu
- College of The Second Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
| | - Ya Qing Xu
- Department of Endocrinology, Zhejiang Integrated Traditional and Western Medicine Hospital, Hangzhou, Zhejiang 310003, P.R. China
| | - Danyang Zhou
- Department of Endocrinology, Zhejiang Integrated Traditional and Western Medicine Hospital, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
60
|
Deng M, Tan J, Hu C, Hou T, Peng W, Liu J, Yu B, Dai Q, Zhou J, Yang Y, Dong R, Ruan C, Dong S, Xu J. Modification of PLGA Scaffold by MSC-Derived Extracellular Matrix Combats Macrophage Inflammation to Initiate Bone Regeneration via TGF-β-Induced Protein. Adv Healthc Mater 2020; 9:e2000353. [PMID: 32424991 DOI: 10.1002/adhm.202000353] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/25/2020] [Indexed: 12/20/2022]
Abstract
The immunologic response toward chronic inflammation or bone regeneration via the accumulation of M1 or M2 macrophages after injury could determine the fate of biomaterial. Human umbilical cord mesenchymal stem cells (hUCMSCs) have a pivotal immunomodulatory property on directing macrophage behaviors. Herein, for the first time, 3D-printed poly(lactide-co-glycolide) (PLGA) scaffolds modified with hUCMSC-derived extracellular matrix (PLGA-ECM) are prepared by a facile tissue engineering technique with physical decellularization and 2.44 ± 0.29 mg cm-3 proteins immobilized on the PLGA-ECM contain multiple soluble cytokines with a sustainable release profile. The PLGA-ECM not only attenuates the foreign body response, but also improves bone regeneration by increasing the accumulation of M2 macrophages in an improved heterotopic transplantation model of SCID mice. Furthermore, the PLGA-ECM scaffolds with the knockdown of transforming growth factor-β-induced protein (TGFβI/βig-H3) demonstrate that M2 macrophage accumulation improved by the PLGA-ECM could be attributed to increasing the migration of M2 macrophages and the repolarization of M1 macrophages to M2 phenotype, which are mediated by multiple integrin signaling pathways involving in integrin β7, integrin α9, and integrin β1 in a TGFβI-dependent manner. This study presents an effective surface modification strategy of polymeric scaffolds to initiate tissue regeneration and combat inflammatory response by increasing M2 macrophage accumulation.
Collapse
Affiliation(s)
- Moyuan Deng
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Jiulin Tan
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Chengshen Hu
- Research Center for Human Tissue and Organs Degeneration, Institute Biomedical and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Tianyong Hou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Wei Peng
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Juan Liu
- Research Center for Human Tissue and Organs Degeneration, Institute Biomedical and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Bo Yu
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Qijie Dai
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Jiangling Zhou
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Yusheng Yang
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| | - Rui Dong
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University, Chongqing, 400038, China
| | - Changshun Ruan
- Research Center for Human Tissue and Organs Degeneration, Institute Biomedical and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, College of Biomedical Engineering, Third Military Medical University, Chongqing, 400038, China
| | - Jianzhong Xu
- National & Regional United Engineering Lab of Tissue Engineering, Department of Orthopaedics, Southwest Hospital, the Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
61
|
Liu W, Yu M, Xie D, Wang L, Ye C, Zhu Q, Liu F, Yang L. Melatonin-stimulated MSC-derived exosomes improve diabetic wound healing through regulating macrophage M1 and M2 polarization by targeting the PTEN/AKT pathway. Stem Cell Res Ther 2020; 11:259. [PMID: 32600435 PMCID: PMC7322868 DOI: 10.1186/s13287-020-01756-x] [Citation(s) in RCA: 323] [Impact Index Per Article: 64.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/27/2020] [Accepted: 06/01/2020] [Indexed: 12/11/2022] Open
Abstract
Background After surgery, wound recovery in diabetic patients may be disrupted due to delayed inflammation, which can lead to undesired consequences, and there is currently a lack of effective measures to address this issue. Mesenchymal stem cell (MSC)-derived exosomes (Exo) have been proven to be appropriate candidates for diabetic wound healing through the anti-inflammatory effects. In this study, we investigated whether melatonin (MT)-pretreated MSCs-derived exosomes (MT-Exo) could exert superior effects on diabetic wound healing, and we attempted to elucidate the underlying mechanism. Methods For the evaluation of the anti-inflammatory effect of MT-Exo, in vitro and in vivo studies were performed. For in vitro research, we detected the secreted levels of inflammation-related factors, such as IL-1β, TNF-α and IL-10 via ELISA and the relative gene expression of the IL-1β, TNF-α, IL-10, Arg-1 and iNOS via qRT-PCR and investigated the expression of PTEN, AKT and p-AKT by Western blotting. For in vivo study, we established air pouch model and streptozotocin (STZ)-treated diabetic wound model, and evaluated the effect of MT-Exo by flow cytometry, optical imaging, H&E staining, Masson trichrome staining, immunohistochemical staining, immunofluorescence, and qRT-PCR (α-SMA, collagen I and III). Results MT-Exo significantly suppressed the pro-inflammatory factors IL-1β and TNF-α and reduced the relative gene expression of IL-1β, TNF-α and iNOS, while promoting the anti-inflammatory factor IL-10 along with increasing the relative expression of IL-10 and Arg-1, compared with that of the PBS, LPS and the Exo groups in vitro. This effect was mediated by the increased ratio of M2 polarization to M1 polarization through upregulating the expression of PTEN and inhibiting the phosphorylation of AKT. Similarly, MT-Exo significantly promoted the healing of diabetic wounds by inhibiting inflammation, thereby further facilitating angiogenesis and collagen synthesis in vivo. Conclusions MT-Exo could promote diabetic wound healing by suppressing the inflammatory response, which was achieved by increasing the ratio of M2 polarization to M1 polarization through activating the PTEN/AKT signalling pathway, and the pretreatment of MT was proved to be a promising method for treating diabetic wound healing. Graphical abstract: MT-Exo promotes diabetic wound healing by regulating M1 and M2 macrophage polarization. ![]()
Collapse
Affiliation(s)
- Wei Liu
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Muyu Yu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Dong Xie
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Longqing Wang
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Cheng Ye
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Qi Zhu
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Fang Liu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| | - Lili Yang
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China.
| |
Collapse
|
62
|
Gao C, Wang X, Lu J, Li Z, Jia H, Chen M, Chang Y, Liu Y, Li P, Zhang B, Du X, Qi F. Mesenchymal stem cells transfected with sFgl2 inhibit the acute rejection of heart transplantation in mice by regulating macrophage activation. Stem Cell Res Ther 2020; 11:241. [PMID: 32552823 PMCID: PMC7301524 DOI: 10.1186/s13287-020-01752-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/19/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have become a promising candidate for cell-based immune therapy for acute rejection (AR) after heart transplantation due to possessing immunomodulatory properties. In this study, we evaluated the efficacy of soluble fibronectin-like protein 2 (sFgl2) overexpressing mesenchymal stem cells (sFgl2-MSCs) in inhibiting AR of heart transplantation in mice by regulating immune tolerance through inducing M2 phenotype macrophage polarization. METHODS AND RESULTS The sFgl2, a novel immunomodulatory factor secreted by regulatory T cells, was transfected into MSCs to enhance their immunosuppressive functions. After being co-cultured for 72 h, the sFgl2-MSCs inhibited M1 polarization whereas promoted M2 of polarization macrophages through STAT1 and NF-κB pathways in vitro. Besides, the sFgl2-MSCs significantly enhanced the migration and phagocytosis ability of macrophages stimulated with interferon-γ (IFN-γ) and lipopolysaccharide (LPS). Further, the application potential of sFgl2-MSCs in AR treatment was demonstrated by heterotopic cardiac transplantation in mice. The tissue damage and macrophage infiltration were evaluated by H&E and immunohistochemistry staining, and the secretion of inflammatory cytokines was analyzed by ELISA. The results showed that sFgl2-MSCs injected intravenously were able to locate in the graft, promote the M2 polarization of macrophages in vivo, regulate the local and systemic immune response, significantly protect tissues from damaging, and finally prolonged the survival time of mice heart grafts. CONCLUSION sFgl2-MSCs ameliorate AR of heart transplantation by regulating macrophages, which provides a new idea for the development of anti-AR treatment methods after heart transplantation.
Collapse
Affiliation(s)
- Chao Gao
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Xiaodong Wang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, 310003, Zhejiang province, China
| | - Jian Lu
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Zhilin Li
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Haowen Jia
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Minghao Chen
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Yuchen Chang
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Yanhong Liu
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Peiyuan Li
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Baotong Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
- Tianjin General Surgery Institute, Tianjin, 300052, China
| | - Xuezhi Du
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Feng Qi
- Department of General Surgery, Tianjin Medical University General Hospital, No. 154 Anshan Road, Heping District, Tianjin, 300052, China.
- Tianjin General Surgery Institute, Tianjin, 300052, China.
| |
Collapse
|
63
|
Low-Dose Decitabine Assists Human Umbilical Cord-Derived Mesenchymal Stem Cells in Protecting β Cells via the Modulation of the Macrophage Phenotype in Type 2 Diabetic Mice. Stem Cells Int 2020; 2020:4689798. [PMID: 32322278 PMCID: PMC7157805 DOI: 10.1155/2020/4689798] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/22/2020] [Accepted: 03/10/2020] [Indexed: 01/09/2023] Open
Abstract
Background Progressive β-cell dysfunction, a major characteristic of type 2 diabetes (T2D), is closely related to the infiltration of inflammatory macrophages within islets. Mesenchymal stem cells (MSCs) have been identified to alleviate β-cell dysfunction by modulating macrophage phenotype in T2D, but the restoration of β-cells by a single MSC infusion is relatively transient. Decitabine (DAC) has been reported to polarize macrophages towards the anti-inflammatory phenotype at low doses. We therefore investigated whether low-dose decitabine could enhance the antidiabetic effect of MSCs and further promote the restoration of β-cell function. Methods We induced a T2D mice model by high-fat diets and streptozotocin (STZ) injection. Mice were divided into five groups: the normal group, the T2D group, the DAC group, the MSC group, and the MSC plus DAC group (MD group). We examined the blood glucose and serum insulin levels of mice 1, 2, and 4 weeks after MSC and/or DAC treatment. Dynamic changes in islets and the phenotype of intraislet macrophages were detected via immunofluorescence. In vitro, we explored the effect of MSCs and DAC on macrophage polarization. Results The blood glucose and serum insulin levels revealed that DAC prolonged the antidiabetic effect of MSCs to 4 weeks in T2D mice. Immunofluorescence staining demonstrated more sustainable morphological and structural amelioration in islets of the MD group than in the MSC group. Interestingly, further analysis showed more alternatively activated macrophages (M2, anti-inflammatory) and fewer classically activated macrophages (M1, proinflammatory) in islets of the MD group 4 weeks after treatment. An in vitro study demonstrated that DAC together with MSCs further polarized macrophages from the M1 to M2 phenotype via the PI3K/AKT pathway. Conclusion These data unveiled that DAC prolonged the antidiabetic effect of MSCs and promoted sustainable β-cell restoration, possibly by modulating the macrophage phenotype. Our results offer a preferable therapeutic strategy for T2D.
Collapse
|
64
|
Yuan Y, Li L, Zhu L, Liu F, Tang X, Liao G, Liu J, Cheng J, Chen Y, Lu Y. Mesenchymal stem cells elicit macrophages into M2 phenotype via improving transcription factor EB-mediated autophagy to alleviate diabetic nephropathy. Stem Cells 2020; 38:639-652. [PMID: 31904160 DOI: 10.1002/stem.3144] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/15/2019] [Indexed: 02/05/2023]
Abstract
Diabetic nephropathy (DN) is a leading cause of end-stage renal disease. Chronic inflammation is recognized as a key causal factor in the development and progression of DN, and the imbalance of M1/M2 macrophages (Mφ) contributes to this process. Mesenchymal stem cells (MSCs) have been reported to prevent renal injuries via immune regulation in diabetic models, but whether these benefits are owing to the regulation of Mφ, and the underlying signaling pathways are unknown. Here, we showed that MSCs elicited Mφ into M2 phenotype and prevented renal injuries in DN mice, but these effects were abolished when the Mφ were depleted by clodronate liposomes (Lipo-Clod), suggesting that Mφ were necessary for renal protection of MSCs in DN mice. Moreover, the MSCs promoted M2 polarization was attributable to the activation of transcription factor EB (TFEB) and subsequent restore of lysosomal function and autophagy activity in Mφ. Furthermore, in vivo adoptive transfer of Mφin vivo (Mφ from DN + MSCs mice) or MφMSCs (Mφ cocultured with MSCs in vitro) to DN mice improved renal function. While, TFEB knockdown in Mφ significantly abolished the protective role of MφMSCs . Altogether, these findings revealed that MSCs suppress inflammatory response and alleviate renal injuries in DN mice via TFEB-dependent Mφ switch.
Collapse
Affiliation(s)
- Yujia Yuan
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Lan Li
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Lingling Zhu
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Fei Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xi Tang
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Guangneng Liao
- Animal Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jingping Liu
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Jingqiu Cheng
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Younan Chen
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yanrong Lu
- NHC Key Laboratory of Transplant Engineering and Immunology, Department of Nephrology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
65
|
Wang L, Liu T, Liang R, Wang G, Liu Y, Zou J, Liu N, Zhang B, Liu Y, Ding X, Cai X, Wang Z, Xu X, Ricordi C, Wang S, Shen Z. Mesenchymal stem cells ameliorate β cell dysfunction of human type 2 diabetic islets by reversing β cell dedifferentiation. EBioMedicine 2020; 51:102615. [PMID: 31918404 PMCID: PMC7000334 DOI: 10.1016/j.ebiom.2019.102615] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 12/05/2019] [Accepted: 12/17/2019] [Indexed: 02/07/2023] Open
Abstract
Background A physiological hallmark of patients with type 2 diabetes mellitus (T2DM) is β cell dysfunction. Despite adequate treatment, it is an irreversible process that follows disease progression. Therefore, the development of novel therapies that restore β cell function is of utmost importance. Methods This study aims to unveil the mechanistic action of mesenchymal stem cells (MSCs) by investigating its impact on isolated human T2DM islets ex vivo and in vivo. Findings We propose that MSCs can attenuate β cell dysfunction by reversing β cell dedifferentiation in an IL-1Ra-mediated manner. In response to the elevated expression of proinflammatory cytokines in human T2DM islet cells, we observed that MSCs was activated to secret IL-1R antagonist (IL-1Ra) which acted on the inflammed islets and reversed β cell dedifferentiation, suggesting a crosstalk between MSCs and human T2DM islets. The co-transplantation of MSCs with human T2DM islets in diabetic SCID mice and intravenous infusion of MSCs in db/db mice revealed the reversal of β cell dedifferentiation and improved glycaemic control in the latter. Interpretation This evidence highlights the potential of MSCs in future cell-based therapies regarding the amelioration of β cell dysfunction.
Collapse
Affiliation(s)
- Le Wang
- Organ Transplant Centre, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China; NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China
| | - Tengli Liu
- NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China; Diabetes Research Institute Federation, Hollywood, FL 33021, USA
| | - Rui Liang
- NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China; Diabetes Research Institute Federation, Hollywood, FL 33021, USA
| | - Guanqiao Wang
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Yaojuan Liu
- NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China
| | - Jiaqi Zou
- NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China
| | - Na Liu
- NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China
| | - Boya Zhang
- Organ Transplant Centre, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Yan Liu
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Xuejie Ding
- Organ Transplant Centre, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Xiangheng Cai
- The First Central Clinical College, Tianjin Medical University, Tianjin, 300192, China
| | - Zhiping Wang
- Organ Transplant Centre, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Xiumin Xu
- Diabetes Research Institute, Cell Transplant Centre; Department of Surgery; Department Medicine; Miller School of Medicine, University of Miami, Miami, FL 33136, USA; The Cure Alliance, Miami, FL 33137, USA; Diabetes Research Institute Federation, Hollywood, FL 33021, USA
| | - Camillo Ricordi
- Diabetes Research Institute, Cell Transplant Centre; Department of Surgery; Department Medicine; Miller School of Medicine, University of Miami, Miami, FL 33136, USA; The Cure Alliance, Miami, FL 33137, USA; Diabetes Research Institute Federation, Hollywood, FL 33021, USA
| | - Shusen Wang
- Organ Transplant Centre, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China; NHC Key Laboratory for Critical Care Medicine, Tianjin 300384, China; Diabetes Research Institute Federation, Hollywood, FL 33021, USA.
| | - Zhongyang Shen
- Organ Transplant Centre, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China; Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China.
| |
Collapse
|
66
|
Lu X, Li N, Zhao L, Guo D, Yi H, Yang L, Liu X, Sun D, Nian H, Wei R. Human umbilical cord mesenchymal stem cells alleviate ongoing autoimmune dacryoadenitis in rabbits via polarizing macrophages into an anti-inflammatory phenotype. Exp Eye Res 2019; 191:107905. [PMID: 31891674 DOI: 10.1016/j.exer.2019.107905] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 12/08/2019] [Accepted: 12/27/2019] [Indexed: 02/06/2023]
Abstract
Mesenchymal stem cells (MSCs) exhibit beneficial effects on autoimmune dacryoadenitis. However, the underlying mechanisms are not fully understood. In this study, we investigated the therapeutic effect of human umbilical cord mesenchymal stem cells (hUC-MSCs) on rabbit autoimmune dacryoadenitis, an animal model of Sjögren's syndrome (SS) dry eye, and explored whether the effects of MSCs were related to their modulation on macrophage polarization. We have showed that systemic infusion of hUC-MSCs after disease onset efficiently diminished the chronic inflammation in diseased LGs and improved the clinical symptoms. Further analysis revealed that hUC-MSC treatment significantly inhibited the expression of pro-inflammatory M1 macrophage markers iNOS, TNF-α and IL-6, and promoted the expression of anti-inflammatory M2 macrophage markers Arg1, CD206, IL-10, IL-4 and TGF-β in LGs. Mechanistically, hUC-MSCs activated AKT pathway in macrophages, resulting in upregulation of M2-associated molecule Arg1, which was partly abolished by PI3K inhibitor, LY294002. Together, our data indicated that hUC-MSCs can skew macrophages into an M2 phenotype via affecting AKT pathway. These data may provide a new insight into the mechanisms of hUC-MSCs in the therapy of SS dry eye.
Collapse
Affiliation(s)
- Xiaoxiao Lu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Na Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Lu Zhao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Di Guo
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Huanfa Yi
- Central Laboratory of the Eastern Division, The First Hospital, Jilin University, Changchun, China
| | - Liyuan Yang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Xun Liu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China
| | - Deming Sun
- Doheny Eye Institute, And Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, USA
| | - Hong Nian
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China.
| | - Ruihua Wei
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China.
| |
Collapse
|
67
|
Kim J, Kim WJ, Ha KS, Han ET, Park WS, Yang SR, Hong SH. Perivascular Stem Cells Suppress Inflammasome Activation during Inflammatory Responses in Macrophages. Int J Stem Cells 2019; 12:419-429. [PMID: 31658511 PMCID: PMC6881042 DOI: 10.15283/ijsc19115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/06/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022] Open
Abstract
Background and Objectives Perivascular stem cells (PVCs) have been identified as precursors of mesenchymal stem cells (MSCs) that offer promising prospects for application in the development of cellular therapies. Although PVCs have been demonstrated to have greater therapeutic potential compared to bone marrow and adipose tissue-derived MSCs in various diseases, the regulatory role of PVCs on inflammasome activation during macrophage-mediated inflammatory responses has not been investigated. Methods and Results In this study, we found that the PVC secretome effectively alleviates secretion of both caspase-1 and interleukin-1β in lipopolysaccharide-primed and activated human and murine macrophages by blocking inflammasome activation and attenuating the production of mitochondrial reactive oxygen species (ROS). We further showed that the PVC secretome significantly reduces inflammatory responses and endoplasmic reticulum stress in peritoneal macrophages in a mouse model of monosodium urate-induced peritonitis. A cytokine antibody array analysis revealed that the PVC secretome contains high levels of serpin E1 and angiogenin, which may be responsible for the inhibitory effects on mitochondrial ROS generation as well as on inflammasome activation. Conclusions Our results suggest that PVCs may be therapeutically useful for the treatment of macrophage- and inflammation-mediated diseases by paracrine action via the secretion of various biological factors.
Collapse
Affiliation(s)
- Jeeyoung Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Woo Jin Kim
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Kwon-Soo Ha
- Scripps Korea Antibody Institute and Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Won Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Se-Ran Yang
- Department of Thoracic and Cardiovascular Surgery, School of Medicine, Kangwon National University, Chuncheon, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
68
|
Treatment with adipose tissue-derived mesenchymal stem cells exerts anti-diabetic effects, improves long-term complications, and attenuates inflammation in type 2 diabetic rats. Stem Cell Res Ther 2019; 10:333. [PMID: 31747961 PMCID: PMC6868748 DOI: 10.1186/s13287-019-1474-8] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/03/2019] [Accepted: 10/30/2019] [Indexed: 02/08/2023] Open
Abstract
Background Long-term diabetes-associated complications are the major causes of morbidity and mortality in individuals with diabetes. These diabetic complications are closely linked to immune system activation along with chronic, non-resolving inflammation, but therapies to directly reverse these complications are still not available. Our previous study demonstrated that mesenchymal stem cells (MSCs) attenuated chronic inflammation in type 2 diabetes mellitus (T2DM), resulting in improved insulin sensitivity and islet function. Therefore, we speculated that MSCs might exert anti-inflammatory effects and promote the reversal of diabetes-induced kidney, liver, lung, heart, and lens diseases in T2DM rats. Methods We induced a long-term T2DM complication rat model by using a combination of a low dose of streptozotocin (STZ) with a high-fat diet (HFD) for 32 weeks. Adipose-derived mesenchymal stem cells (ADSCs) were systemically administered once a week for 24 weeks. Then, we investigated the role of ADSCs in modulating the progress of long-term diabetic complications. Results Multiple infusions of ADSCs attenuated chronic kidney disease (CKD), nonalcoholic steatohepatitis (NASH), lung fibrosis, and cataracts; improved cardiac function; and lowered serum lipid levels in T2DM rats. Moreover, the levels of inflammatory cytokines in the serum of each animal group revealed that ADSC infusions were able to not only inhibit pro-inflammatory cytokines IL-6, IL-1β, and TNF-α expression but also increase anti-inflammatory cytokine IL-10 systematically. Additionally, MSCs reduced the number of iNOS(+) M1 macrophages and restored the number of CD163(+) M2 macrophages. Conclusions Multiple intravenous infusions of ADSCs produced significant protective effects against long-term T2DM complications by alleviating inflammation and promoting tissue repair. The present study suggests ADSCs may be a novel, alternative cell therapy for long-term diabetic complications.
Collapse
|
69
|
Human Umbilical Cord-Derived Mesenchymal Stem Cell Therapy Ameliorates Nonalcoholic Fatty Liver Disease in Obese Type 2 Diabetic Mice. Stem Cells Int 2019; 2019:8628027. [PMID: 31781248 PMCID: PMC6875176 DOI: 10.1155/2019/8628027] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 08/09/2019] [Accepted: 09/09/2019] [Indexed: 12/16/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is increasingly common among patients with type 2 diabetes mellitus (T2DM). The two conditions can act synergistically to produce adverse outcomes. However, the therapeutic options for patients with NAFLD and T2DM are currently limited. Human umbilical cord-derived mesenchymal stem cells (UC-MSCs) have shown therapeutic potential for diabetes and hepatic disorders such as liver cirrhosis and fulminant hepatic failure. The present study is aimed at investigating the effect of human UC-MSCs on a mouse model of NAFLD and T2DM, characterized by obesity-induced hyperglycaemia, dyslipidaemia, hepatic steatosis, and liver dysfunction. Thirty-week-old male C57BL/6 db/db mice were infused with human UC-MSCs or phosphate-buffered saline (PBS) via the tail vein once a week for six weeks. Age-matched male C57BL/6 wild-type db/+ mice were used as controls. Body weight and random blood glucose were measured every week. One week after the sixth infusion, intraperitoneal glucose tolerance tests and insulin tolerance tests were performed and the blood and liver were harvested for biochemical and histopathological examinations. Quantitative real-time reverse transcriptase polymerase chain reaction (qRT-PCR), immunofluorescence staining, and western blot were performed to monitor the expression of the lipid metabolism- and regulatory pathway-related genes. UC-MSC infusions significantly ameliorated hyperglycaemia, attenuated the elevation of hepatic transaminases, and decreased lipid contents, including triglyceride, total cholesterol, and low-density lipoprotein cholesterol. Moreover, histological lesions in the liver diminished markedly, as evidenced by reduced lipid accumulation and attenuated hepatic steatosis. Mechanistically, UC-MSCs were found to regulate lipid metabolism by increasing the expression of fatty acid oxidation-related genes and inhibiting the expression of lipogenesis-related genes, which were associated with the upregulation of the HNF4α-CES2 pathway. Our results demonstrate that human UC-MSCs can ameliorate NAFLD and reverse metabolic syndrome in db/db mice. Thus, UC-MSCs may serve as a novel therapeutic agent for T2DM patients with NAFLD.
Collapse
|
70
|
Wang YN, Jia T, Zhang J, Lan J, Zhang D, Xu X. PTPN2 improves implant osseointegration in T2DM via inducing the dephosphorylation of ERK. Exp Biol Med (Maywood) 2019; 244:1493-1503. [PMID: 31615285 DOI: 10.1177/1535370219883419] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is considered to compromise implant osseointegration. Protein tyrosine phosphatase non-receptor type 2 (PTPN2) regulates glucose metabolism, systemic inflammation, and bone regeneration. This study aimed to investigate the role of PTPN2 in implant osseointegration in T2DM and explore the potential mechanisms. Streptozotocin-induced diabetic rats received implant surgery, with or without local overexpression of PTPN2 for three months, and implant osseointegration was examined by histological evaluation, micro-CT analysis, pull-out test, and scanning electron microscope. Rat bone marrow stem cells (RBMSCs) were isolated and exposed to high glucose, and osteogenic differentiation was evaluated by alizarin red staining, ALP assay, and Western blot analysis. Overexpression of PTPN2 could improve impaired implant osseointegration in T2DM rats and promote osteogenic differentiation of RBMSCs in high glucose. In addition, p-ERK level in RBMSCs was increased in high glucose and decreased after PTPN2 overexpression. These results suggest that PTPN2 promotes implant osseointegration in T2DM rats and enhances osteogenesis of RBMSCs in high glucose medium via inducing the dephosphorylation of ERK. PTPN2 may be a novel target for the therapy of impaired implant osseointegration in T2DM patients. Impact statement Using both in vivo and in vitro approaches, we made important findings that PTPN2 promoted implant osseointegration in T2DM rats and enhanced osteogenesis of RBMSCs in high glucose medium. The positive effects of PTPN2 on osteogenesis are related to the dephosphorylation of ERK and the inhibition of MAPK/ERK pathway. PTPN2 may be a novel target for the therapy of impaired implant osseointegration in T2DM patients.
Collapse
Affiliation(s)
- Ya-Nan Wang
- Department of Implantology, School and Hospital of Stomatology, Shandong University, Shandong 250012, China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong 250012, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong 250012, China
| | - Tingting Jia
- Department of Implantology, School and Hospital of Stomatology, Shandong University, Shandong 250012, China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong 250012, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong 250012, China
| | - Jiajia Zhang
- Department of Implantology, School and Hospital of Stomatology, Shandong University, Shandong 250012, China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong 250012, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong 250012, China
| | - Jing Lan
- Department of Implantology, School and Hospital of Stomatology, Shandong University, Shandong 250012, China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong 250012, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong 250012, China
| | - Dongjiao Zhang
- Department of Implantology, School and Hospital of Stomatology, Shandong University, Shandong 250012, China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong 250012, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong 250012, China
| | - Xin Xu
- Department of Implantology, School and Hospital of Stomatology, Shandong University, Shandong 250012, China.,Shandong Provincial Key Laboratory of Oral Tissue Regeneration, Shandong 250012, China.,Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Shandong 250012, China
| |
Collapse
|
71
|
Mesenchymal Stem Cells Alleviate DHEA-Induced Polycystic Ovary Syndrome (PCOS) by Inhibiting Inflammation in Mice. Stem Cells Int 2019; 2019:9782373. [PMID: 31611920 PMCID: PMC6757294 DOI: 10.1155/2019/9782373] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/12/2019] [Accepted: 07/22/2019] [Indexed: 12/29/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common cause of anovulatory infertility in women of reproductive age. Chronic inflammation is considered to be the cause of ovarian dysfunction. Increasing evidence in animal studies and in preliminary clinical trials has demonstrated that MSCs possess immunomodulatory effects via their interaction with immune cells. However, their contribution to PCOS remains unclear. In this study, we showed that the administration of hUC-MSCs could efficiently improve the pathological changes of PCOS mice induced by dehydroepiandrosterone (DHEA), including ovarian histopathology and function. Moreover, we found that the administration of MSCs significantly downregulated the expression of proinflammatory factors (TNF-α, IL-1β, and IFN-γ) and fibrosis-related genes (CTGF) in ovarian and uterus tissues and affected the systemic inflammatory response. The percentage of peripheral neutrophils, M1 macrophages, and B cells was significantly reduced, while M2 macrophages and regulatory T cells (Tregs) were increased in hUC-MSC-treated mice. In the spleen, the percentage of neutrophils, M1 macrophages, IFN-γ+CD19+B cell, IFN-γ+CD4+T cells (Th1), and IL-17+CD4+T cells (Th17) was significantly decreased in hUC-MSC-treated mice. These results suggested that hUC-MSC treatment could alleviate ovarian dysfunction by inhibiting ovarian local and systemic inflammatory responses.
Collapse
|
72
|
Qi Y, Ma J, Li S, Liu W. Applicability of adipose-derived mesenchymal stem cells in treatment of patients with type 2 diabetes. Stem Cell Res Ther 2019; 10:274. [PMID: 31455405 PMCID: PMC6712852 DOI: 10.1186/s13287-019-1362-2] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is mainly characterized by insulin resistance (IR) and impaired insulin secretion. The chronic inflammatory process contributed to IR and could also hamper pancreatic β cell function. However, currently applied treatment cannot reverse β cell damage or alleviate inflammation. Mesenchymal stem cells (MSCs), the cell-based therapy for their self-renewable, differentiation potential, and immunosuppressive properties, have been demonstrated in displaying therapeutic effects in T2DM. Adipose-derived MSCs (AD-MSCs) attracted more attention due to less harvested inconvenience and ethical issues commonly accompany with bone marrow-derived MSCs (BM-MSCs) and fetal annex-derived MSCs. Both AD-MSC therapy studies and mechanism explorations in T2DM animals presented that AD-MSCs could translate to clinical application. However, hyperglycemia, hyperinsulinemia, and metabolic disturbance in T2DM are crucial for impairment of AD-MSC function, which may limit the therapeutical effects of MSCs. This review focuses on the outcomes and the molecular mechanisms of MSC therapies in T2DM which light up the hope of AD-MSCs as an innovative strategy to cure T2DM.
Collapse
Affiliation(s)
- Yicheng Qi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, RenJi Hospital, School of Medicine, Shanghai Jiaotong University, 160# Pujian Road, Pudong, Shanghai, 200127, China
| | - Jing Ma
- Division of Endocrinology and Metabolism, Department of Internal Medicine, RenJi Hospital, School of Medicine, Shanghai Jiaotong University, 160# Pujian Road, Pudong, Shanghai, 200127, China
| | - Shengxian Li
- Division of Endocrinology and Metabolism, Department of Internal Medicine, RenJi Hospital, School of Medicine, Shanghai Jiaotong University, 160# Pujian Road, Pudong, Shanghai, 200127, China
| | - Wei Liu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, RenJi Hospital, School of Medicine, Shanghai Jiaotong University, 160# Pujian Road, Pudong, Shanghai, 200127, China.
| |
Collapse
|
73
|
Gao J, Cheng Y, Hao H, Yin Y, Xue J, Zhang Q, Li L, Liu J, Xie Z, Yu S, Li B, Han W, Mu Y. Decitabine assists umbilical cord-derived mesenchymal stem cells in improving glucose homeostasis by modulating macrophage polarization in type 2 diabetic mice. Stem Cell Res Ther 2019; 10:259. [PMID: 31426846 PMCID: PMC6700792 DOI: 10.1186/s13287-019-1338-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/04/2019] [Accepted: 07/14/2019] [Indexed: 01/10/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) have emerged as a promising therapy for type 2 diabetes (T2D). Mechanistic researches demonstrate that the anti-diabetic effect of MSCs is partially mediated by eliciting macrophages into an anti-inflammatory phenotype thus alleviating insulin resistance. However, single MSC infusion is insufficient to ameliorate sustained hyperglycemia or normalize blood glucose levels. In this study, we used decitabine (DAC), which is involved in the regulation of macrophage polarization, to test whether MSCs combined with decitabine can prolong and enhance the anti-diabetic effect in T2D mice. Methods High-fat diet (HFD) and streptozocin (STZ) were given to induce T2D mouse model. Successfully induced T2D mice were randomly divided into four groups: T2D group, MSC group, DAC group, and MSC + DAC group. Blood glucose was monitored, and glucose tolerance and insulin sensitivity were evaluated during the entire analysis period. Epididymal fat was extracted for analysis of macrophage phenotype and inflammation in adipose tissue. In vitro, we examined the effect of MSC + DAC on macrophage polarization in bone marrow-derived macrophages (BMDMs) and explore the possible mechanism. Results MSC infusion effectively improved insulin sensitivity and glucose homeostasis in T2D mice within 1 week, whereas combination therapy of MSCs + DAC extended the anti-diabetic effects of MSCs from 1 to 4 weeks (the end of the observation). Correspondingly, more M2 macrophages in adipose tissue were observed in the combination therapy group over the entire study period. In vitro, compared with the MSC group, MSCs combined with decitabine more effectively polarized M1 macrophages to M2 macrophages. Further analysis showed that the effect of MSC + DAC on macrophage polarization was largely abrogated by the peroxisome proliferator-activated receptor gamma (PPARγ) antagonist GW9662. Conclusions Our data suggest that MSCs combined with decitabine can more effectively alleviate insulin resistance and prolong and enhance the anti-diabetic effect of MSCs in T2D mice in part by prompting M2 polarization in a PPARγ-dependent manner. Thus, decitabine may be an applicable addition to MSCs for diabetes therapy. Graphic Abstract UC-MSCs combined with decitabine activate the IL4R/STAT6/STAT3/PPARγ axis to further promote M2 macrophage polarization in adipose tissue, reduce inflammation, improve insulin sensitivity, and lead to better glucose metabolism and long-term hypoglycemic effects
![]() Electronic supplementary material The online version of this article (10.1186/s13287-019-1338-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jieqing Gao
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China.,Department of Endocrinology, Beijing Rehabilitation Hospital of Capital Medical University, Beijing, China
| | - Yu Cheng
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Haojie Hao
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Yaqi Yin
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Jing Xue
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Qi Zhang
- Department of Endocrinology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lin Li
- Department of Endocrinology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Jiejie Liu
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Zongyan Xie
- Department of Geriatrics, China-Japan Friendship Hospital, Beijing, China
| | - Songyan Yu
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Bing Li
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Weidong Han
- Department of Molecular Biology, Institute of Basic Medicine, School of Life Science, Chinese PLA General Hospital, Beijing, China
| | - Yiming Mu
- Department of Endocrinology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China.
| |
Collapse
|
74
|
Zhao Q, Wang X, Hu Q, Zhang R, Yin Y. Suppression of TLR4 by miR-448 is involved in Diabetic development via regulating Macrophage polarization. ACTA ACUST UNITED AC 2018; 71:806-815. [PMID: 30536833 DOI: 10.1111/jphp.13048] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/10/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Lipopolysaccharide (LPS) contributed to the development and progression of type 2 diabetes mellitus (T2D), while TLR4 is reported to mediate the LPS-induced inflammation in macrophages. However, the potential molecular mechanisms for TLR4-mediated macrophages activation in T2D have not yet to be fully clarified. METHODS Type 2 diabetes models in C57BL/6J mice were generated by a combination administration of streptozotocin (STZ) and a high-fat diet (HFD). Cell proportions of M1 and M2 macrophages were analyzed using flow cytometry. Expression profiles of miR-448 and TLR 4 were determined by qRT-PCR and Western blot. KEY FINDINGS LPS/IFN-γ significantly induced M1 polarization in macrophages characterized by the increased levels of TNF-α, IL-6, IL-12, iNOS and decreased levels of TNF-β, CCL-22, IL-10 and Arg-1, with a higher expression of toll-like receptor 4 (TLR4) in vitro. Consistently, T2D mice-derived macrophages had a significantly elevated expression of TLR4 mRNA and decreased expression of miR-448. We further confirmed that miR-448 could inhibit TLR4 expression by targeting the 3'-UTR of TLR4, rescuing the LPS/IFN-γ-induced M1 macrophage polarization. CONCLUSIONS Taken together, our results indicated that decreased miR-448 in diabetic macrophages may contribute to LPS-induced M1 polarization by targeting TLR4, thereby modulating T2D development.
Collapse
Affiliation(s)
- Qing Zhao
- Department of Endocrinology, Lianshui County People's Hospital, Huai'an, China
| | - Xin Wang
- Department of Endocrinology, Lianshui County People's Hospital, Huai'an, China
| | - Qiaosheng Hu
- Department of Endocrinology, Lianshui County People's Hospital, Huai'an, China
| | - Ridong Zhang
- Department of endocrinology, Huai'an First People's Hospital of Nanjing Media University, Huai'an, China
| | - Yong Yin
- Department of Endocrinology, Changzhou Wujin People's Hospital, Changzhou, China
| |
Collapse
|