1
|
Liu Y, Jia D, Li L, Wang M. Advances in Nanomedicine and Biomaterials for Endometrial Regeneration: A Comprehensive Review. Int J Nanomedicine 2024; 19:8285-8308. [PMID: 39161362 PMCID: PMC11330863 DOI: 10.2147/ijn.s473259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/30/2024] [Indexed: 08/21/2024] Open
Abstract
The endometrium is an extremely important component of the uterus and is crucial for individual health and human reproduction. However, traditional methods still struggle to ideally repair the structure and function of damaged endometrium and restore fertility. Therefore, seeking and developing innovative technologies and materials has the potential to repair and regenerate damaged or diseased endometrium. The emergence and functionalization of various nanomedicine and biomaterials, as well as the proposal and development of regenerative medicine and tissue engineering techniques, have brought great hope for solving these problems. In this review, we will summarize various nanomedicine, biomaterials, and innovative technologies that contribute to endometrial regeneration, including nanoscale exosomes, nanomaterials, stem cell-based materials, naturally sourced biomaterials, chemically synthesized biomaterials, approaches and methods for functionalizing biomaterials, as well as the application of revolutionary new technologies such as organoids, organ-on-chips, artificial intelligence, etc. The diverse design and modification of new biomaterials endow them with new functionalities, such as microstructure or nanostructure, mechanical properties, biological functions, and cellular microenvironment regulation. It will provide new options for the regeneration of endometrium, bring new hope for the reconstruction and recovery of patients' reproductive abilities.
Collapse
Affiliation(s)
- Yanhong Liu
- Center for Prenatal Diagnosis, Center for Reproductive Medicine, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Dongyun Jia
- Center for Prenatal Diagnosis, Center for Reproductive Medicine, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Lin Li
- Center for Prenatal Diagnosis, Center for Reproductive Medicine, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Meiyan Wang
- Center for Prenatal Diagnosis, Center for Reproductive Medicine, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
2
|
Wu Y, Gu S, Cobb JM, Dunn GH, Muth TA, Simchick CJ, Li B, Zhang W, Hua X. E2-Loaded Microcapsules and Bone Marrow-Derived Mesenchymal Stem Cells with Injectable Scaffolds for Endometrial Regeneration Application. Tissue Eng Part A 2024; 30:115-130. [PMID: 37930721 DOI: 10.1089/ten.tea.2023.0238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) have been recognized as new candidates for the treatment of serious endometrial injuries. However, owing to the local microenvironment of damaged endometrium, transplantation of BMSCs yielded disappointing results. In this study, Pectin-Pluronic® F-127 hydrogel as scaffolds were fabricated to provide three-dimensional architecture for the attachment, growth, and migration of BMSCs. E2 was encapsulated into the W/O/W microspheres to construct pectin-based E2-loaded microcapsules (E2 MPs), which has the potential to serve as a long-term reliable source of E2 for endometrial regeneration. Then, the BMSCs/E2 MPs/scaffolds system was injected into the uterine cavity of mouse endometrial injury model for treatment. At 4 weeks after transplantation, the system increased proliferative abilities of uterine endometrial cells, facilitated microvasculature regeneration, and restored the ability of endometrium to receive an embryo, suggesting that the BMSCs/E2 MPs/scaffolds system is a promising treatment option for endometrial regeneration. Furthermore, the mechanism of E2 in promoting the repair of endometrial injury was also investigated. Exosomes are critical paracrine mediators that act as biochemical cues to direct stem cell differentiation. In this study, it was found that the expression of endometrial epithelial cell (EEC) markers was upregulated in BMSCs treated by exosomes secreted from endometrial stromal cells (ESCs-Exos). Exosomes derived from E2-stimulated ESCs further promoted the expression level of EECs markers in BMSCs, suggesting exosomes released from ESCs by E2 stimulation could enhance the differentiation efficiency of BMSCs. Therefore, exosomes derived from ESCs play paracrine roles in endometrial regeneration stimulated by E2 and provide optimal estrogenic response.
Collapse
Affiliation(s)
- Yuelin Wu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Tongji University School of Medicine, Shanghai, China
| | - Shengyi Gu
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Shanghai Tongji University School of Medicine, Shanghai, China
| | - Jonathan M Cobb
- Chemical and Biomolecular Engineering Program, Department of Physics and Chemistry, Milwaukee School of Engineering, Milwaukee, Wisconsin, USA
| | - Griffin H Dunn
- Chemical and Biomolecular Engineering Program, Department of Physics and Chemistry, Milwaukee School of Engineering, Milwaukee, Wisconsin, USA
| | - Taylor A Muth
- Chemical and Biomolecular Engineering Program, Department of Physics and Chemistry, Milwaukee School of Engineering, Milwaukee, Wisconsin, USA
| | - Chloe J Simchick
- Chemical and Biomolecular Engineering Program, Department of Physics and Chemistry, Milwaukee School of Engineering, Milwaukee, Wisconsin, USA
| | - Baoguo Li
- School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Wujie Zhang
- Chemical and Biomolecular Engineering Program, Department of Physics and Chemistry, Milwaukee School of Engineering, Milwaukee, Wisconsin, USA
| | - Xiaolin Hua
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, Shanghai Tongji University School of Medicine, Shanghai, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Shanghai Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Duddu S, Bhattacharya A, Chakrabarti R, Chakravorty N, Shukla PC. Regeneration and Tissue Microenvironment. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
4
|
SFRP4 + stromal cell subpopulation with IGF1 signaling in human endometrial regeneration. Cell Discov 2022; 8:95. [PMID: 36163341 PMCID: PMC9512788 DOI: 10.1038/s41421-022-00438-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 06/17/2022] [Indexed: 11/08/2022] Open
Abstract
Our understanding of full-thickness endometrial regeneration after injury is limited by an incomplete molecular characterization of the cell populations responsible for the organ functions. To help fill this knowledge gap, we characterized 10,551 cells of full-thickness normal human uterine from two menstrual phases (proliferative and secretory phase) using unbiased single cell RNA-sequencing. We dissected cell heterogeneity of main cell types (epithelial, stromal, endothelial, and immune cells) of the full thickness uterine tissues, cell population architectures of human uterus cells across the menstrual cycle. We identified an SFRP4+ stromal cell subpopulation that was highly enriched in the regenerative stage of the human endometria during the menstrual cycle, and the SFRP4+ stromal cells could significantly enhance the proliferation of human endometrial epithelial organoid in vitro, and promote the regeneration of endometrial epithelial glands and full-thickness endometrial injury through IGF1 signaling pathway in vivo. Our cell atlas of full-thickness uterine tissues revealed the cellular heterogeneities, cell population architectures, and their cell-cell communications during the monthly regeneration of the human endometria, which provide insight into the biology of human endometrial regeneration and the development of regenerative medicine treatments against endometrial damage and intrauterine adhesion.
Collapse
|
5
|
Tu C, Wang Z, Xiang E, Zhang Q, Zhang Y, Wu P, Li C, Wu D. Human Umbilical Cord Mesenchymal Stem Cells Promote Macrophage PD-L1 Expression and Attenuate Acute Lung Injury in Mice. Curr Stem Cell Res Ther 2022; 17:564-575. [PMID: 35086457 DOI: 10.2174/1574888x17666220127110332] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/11/2021] [Accepted: 12/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) remains a serious clinical problem but has no approved pharmacotherapy. Mesenchymal stem cells (MSCs) represent an attractive therapeutic tool for tissue damage and inflammation owing to their unique immunomodulatory properties. The present study aims to explore the therapeutic effect and underlying mechanisms of human umbilical cord MSCs (UC-MSCs) in ALI mice. OBJECTIVE In this study, we identify a novel mechanism for human umbilical cord-derived MSCs (UC-MSCs)-mediated immunomodulation through PGE2-dependent reprogramming of host macrophages to promote their PD-L1 expression. Our study suggests that UC-MSCs or primed-UC-MSCs offer new therapeutic approaches for lung inflammatory diseases. METHODS Lipopolysaccharide (LPS)-induced ALI mice were injected with 5×105 UC-MSCs via the tail vein after 4 hours of LPS exposure. After 24 hours of UC-MSC administration, the total protein concentration and cell number in the bronchoalveolar lavage fluid (BALF), and cytokine levels in the lung tissue were measured. Lung pathological changes and macrophage infiltration after UC-MSC treatment were analyzed. Moreover, in vitro co-culture experiments were performed to analyze cytokine levels of RAW264.7 cells and Jurkat T cells. RESULTS UC-MSC treatment significantly improved LPS-induced ALI, as indicated by decreased total protein exudation concentration and cell number in BALF, and reduced pathological damage in ALI mice. UC-MSCs could inhibit pro-inflammatory cytokine levels (IL-1β, TNF-α, MCP-1, IL-2, and IFN-γ), whereas enhancing anti-inflammatory cytokine IL-10 expression, as well as reduced macrophage infiltration into the injured lung tissue. Importantly, UC-MSC administration increased programmed cell death protein ligand 1 (PD-L1) expression in the lung macrophages. Mechanistically, UC-MSCs upregulated cyclooxygenase-2 (COX2) expression and prostaglandin E2 (PGE2) secretion in response to LPS stimulation. UC-MSCs reduced the inflammatory cytokine levels in murine macrophage Raw264.7 through the COX2/PGE2 axis. Furthermore, UC-MSC-derived PGE2 enhanced PD-L1 expression in RAW264.7 cells, which in turn promoted programmed cell death protein 1 (PD-1) expression and reduced IL-2 and IFN-γ production in Jurkat T cells. CONCLUSION Our results suggest that UC-MSCs attenuate ALI via PGE2-dependent reprogramming of macrophages to promote their PD-L1 expression.
Collapse
Affiliation(s)
- Chengshu Tu
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan, China
| | | | - E Xiang
- Wuhan Hamilton Biotechnology-Co., Ltd, Wuhan, China
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Quan Zhang
- Wuhan Hamilton Biotechnology-Co., Ltd, Wuhan, China
| | - Yaqi Zhang
- Wuhan Hamilton Biotechnology-Co., Ltd, Wuhan, China
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Ping Wu
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan, China
| | - Changyong Li
- Department of Physiology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Dongcheng Wu
- Wuhan Hamilton Biotechnology-Co., Ltd, Wuhan, China
- Department of Biochemistry and Molecular Biology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Guangzhou Hamilton Biotechnology-Co., Ltd, Guangzhou, China
| |
Collapse
|
6
|
Cheng S, Lv R, Xu J, Hirman AR, Du L. IGF-1-Expressing Placenta-Derived Mesenchymal Stem Cells Promote Scalding Wound Healing. J Surg Res 2021; 265:100-113. [PMID: 33895582 DOI: 10.1016/j.jss.2021.02.057] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/01/2021] [Accepted: 02/27/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Stem cell-based regenerative therapy is a novel approach to severe damaged skin. Perinatal tissues such as placenta are considered as promising alternatives. The present study aimed to investigate the effect of insulin-like growth factor-1 (IGF-1)-expressing placenta-derived mesenchymal stem cells (hPMSCs) on healing of burn wounds. MATERIALS AND METHODS hPMSCs were isolated from human placenta, and IGF-1 was transducted into hPMSCs via lentivirus. Flow cytometry and MTT assay were performed to assess cell apoptosis and viability, respectively. Immunostaining of CK19 and ki67 was for evaluating epithelial differentiation ability and cell proliferation. For in vivo studies, we established a mouse model of scalding and performed local administration of IGF-1-expressing hPMSCs via subcutaneous injection. Wound histology was analyzed with H&E staining. The expression of fibrogenic cytokines was detected by western blot. The production of pro-inflammatory factors was measured by ELISA. RESULTS Overexpression of IGF-1 promoted cell proliferation and epithelial differentiation of hPMSCs in vitro and in vivo. Mice with burn injury displayed increased wound contraction and healing rates following treatment with IGF-1-expressing hPMSCs. There was less inflammatory infiltration and reduced collagen disposition in the presence of IGF-1 at the wound site. Administration of IGF-1-expressing hPMSCs suppressed inflammation by decreasing the levels of pro-inflammatory cytokines including tumor necrosis factor-α, interleukin-1β, and interleukin-6. Besides, IGF-1 increased VEGF expression, and decreased TGF-β1, collagen I and collagen III expressions in vivo. CONCLUSIONS IGF-1-expressing PMSCs promotes cell proliferation and epithelial differentiation, inhibits inflammation and collagen deposition, and thus contributes to wound healing.
Collapse
Affiliation(s)
- Shaohang Cheng
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Runxiao Lv
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jing Xu
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Abdul Razaq Hirman
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lili Du
- Department of Pathophysiology, College of Basic Medical Science, China Medical University, Shenyang, China.
| |
Collapse
|
7
|
Zhou Y, Shen H, Wu Y, Zhao X, Pei J, Mou Z, Dong J, Hua X. Platelet-Rich Plasma Therapy Enhances the Beneficial Effect of Bone Marrow Stem Cell Transplant on Endometrial Regeneration. Front Cell Dev Biol 2020; 8:52. [PMID: 32154246 PMCID: PMC7047166 DOI: 10.3389/fcell.2020.00052] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/21/2020] [Indexed: 01/08/2023] Open
Abstract
This study aimed to investigate the potential effect of platelet-rich plasma (PRP) therapy on treatment using bone marrow stem cell (BMSC) transplant for uterine horn damage, and to reveal the potential underlying molecular mechanism. Uterine horn damage was established in a rat model, which can be repaired by transplant using BMSCs receiving control or PRP treatment. Immunohistochemistry was conducted to evaluate thickness and expression of α-SMA and vWF in the regenerated endometrium tissues. mRNA and proteins of insulin-like growth factor-1 (IGF-1) and interleukin-10 (IL-10) were measured both in the regenerated endometrium tissues and in cultured BMSCs to evaluate the effect of PRP treatment on their expression. Enzyme-linked immunosorbent assay was employed to measure the secretory levels of IL-10 in cultured BMSCs. Multi-differentiation assays were performed to address the effect of PRP treatment on tri-lineage potential of cultured BMSCs. Chromatin immunoprecipitation and luciferase reporter assays were applied to analyze NF-κB subunit p50 binding on IL-10 promoter and the resulted regulatory effect. PRP treatment significantly improved the efficacy of BMSC transplant in repairing uterine horn damage of rats, and elevated IGF-1 and IL-10 expression in regenerated endometrium tissues and cultured BMSCs, as well as enhanced tri-lineage differentiation potential of BMSCs. On the other hand, p50 inhibition and silencing suppressed the PRP-induced expression and secretion of IL-10 without affecting IGF-1 in the BMSCs. Furthermore, p50 was able to directly bind to IL-10 promoter to promote its expression. Data in the current study propose a working model, where PRP therapy improves endometrial regeneration of uterine horn damage in rats after BMSC transplant therapy, likely mediated through the NF-κB signaling pathway subunit p50 to directly induce the expression and production of IL-10.
Collapse
Affiliation(s)
- Ying Zhou
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huaxiang Shen
- Department of Obstetrics, Jiaxing Maternity and Child Health Care Hospital, Jiaxing, China
| | - Yuelin Wu
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaobo Zhao
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jindan Pei
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhengqian Mou
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China.,Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinhua Dong
- Department of Obstetrics, Jiaxing Maternity and Child Health Care Hospital, Jiaxing, China
| | - Xiaolin Hua
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Ren X, Zhao M, Lash B, Martino MM, Julier Z. Growth Factor Engineering Strategies for Regenerative Medicine Applications. Front Bioeng Biotechnol 2020; 7:469. [PMID: 32039177 PMCID: PMC6985039 DOI: 10.3389/fbioe.2019.00469] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/23/2019] [Indexed: 01/07/2023] Open
Abstract
Growth factors are critical molecules for tissue repair and regeneration. Therefore, recombinant growth factors have raised a lot of hope for regenerative medicine applications. While using growth factors to promote tissue healing has widely shown promising results in pre-clinical settings, their success in the clinic is not a forgone conclusion. Indeed, translation of growth factors is often limited by their short half-life, rapid diffusion from the delivery site, and low cost-effectiveness. Trying to circumvent those limitations by the use of supraphysiological doses has led to serious side-effects in many cases and therefore innovative technologies are required to improve growth factor-based regenerative strategies. In this review, we present protein engineering approaches seeking to improve growth factor delivery and efficacy while reducing doses and side effects. We focus on engineering strategies seeking to improve affinity of growth factors for biomaterials or the endogenous extracellular matrix. Then, we discuss some examples of increasing growth factor stability and bioactivity, and propose new lines of research that the field of growth factor engineering for regenerative medicine may adopt in the future.
Collapse
Affiliation(s)
- Xiaochen Ren
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Moyuan Zhao
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Blake Lash
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Mikaël M. Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Ziad Julier
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Xiao B, Yang W, Lei D, Huang J, Yin Y, Zhu Y, You Z, Wang F, Sun S. PGS Scaffolds Promote the In Vivo Survival and Directional Differentiation of Bone Marrow Mesenchymal Stem Cells Restoring the Morphology and Function of Wounded Rat Uterus. Adv Healthc Mater 2019; 8:e1801455. [PMID: 30734535 DOI: 10.1002/adhm.201801455] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/26/2018] [Indexed: 01/23/2023]
Abstract
Intrauterine adhesion (IUA) causing infertility and recurrent miscarriage of reproductive female mammals usually results from endometrium injury. Nevertheless, there is no efficient therapeutic method to avoid IUA. Bone marrow derived mesenchymal stem cells (BMSCs) are an important cell source for tissue regeneration. This study designs and explores the ability of BMSC-loaded elastic poly(glycerol sebacate) (PGS) scaffold to prevent IUA and compares the effect of PGS with poly(lactic-co-glycolic acid) (PLGA) and collagen scaffolds in resumption of damaged rat uteruses. The 3D architecture provided by PGS scaffolds favors the attachment and growth of rat BMSCs. In vivo bioluminescence imaging shows that compared with direct BMSC intrauterine injection, PLGA, and collagen scaffolds, the PGS scaffold significantly prolongs the retention time of BMSCs in a wounded rat uterus model. More importantly, BMSCs can directly differentiate into endometrial stromal cells after transplantation of PGS/BMSCs constructs, but not PLGA/BMSCs and collagen/BMSCs. It is found that the level of transforming growth factor β1 (TGF-β1), basic fibroblast growth factor (bFGF), vascular endothelial growth factor, and insulin-like growth factors in the injured endometrium adjacent to PGS/BMSCs constructs is higher than those of rats receiving PLGA/BMSCs, collagen/BMSCs, or BMSCs intrauterine transplantation. Besides, transplantation of PGS/BMSCs leads to better morphology recovery of the damaged uterus than PLGA/BMSCs and collagen/BMSCs. The receptive fertility of PGS/BMSCs is 72.2 ± 6.4%, similar to the one of collagen/BMSCs, but significantly higher than 42.3 ± 3.9% in PLGA/BMSCs. Taken together, PGS/BMSCs may be a promising candidate for preventing IUA.
Collapse
Affiliation(s)
- Bang Xiao
- Department of Medical Genetics; Second Military Medical University; 800 Xiangyin Road Shanghai 200433 P. R. China
| | - Wenjun Yang
- Department of Medical Genetics; Second Military Medical University; 800 Xiangyin Road Shanghai 200433 P. R. China
| | - Dong Lei
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials; International Joint Laboratory for Advanced Fiber and Low-dimension Materials; College of Materials Science and Engineering; Donghua University; Shanghai 201620 P. R. China
| | - Jinfeng Huang
- Department of Medical Genetics; Second Military Medical University; 800 Xiangyin Road Shanghai 200433 P. R. China
| | - Yupeng Yin
- Department of Medical Genetics; Second Military Medical University; 800 Xiangyin Road Shanghai 200433 P. R. China
| | - Yiqing Zhu
- Department of Medical Genetics; Second Military Medical University; 800 Xiangyin Road Shanghai 200433 P. R. China
| | - Zhengwei You
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials; International Joint Laboratory for Advanced Fiber and Low-dimension Materials; College of Materials Science and Engineering; Donghua University; Shanghai 201620 P. R. China
| | - Fang Wang
- Department of Medical Genetics; Second Military Medical University; 800 Xiangyin Road Shanghai 200433 P. R. China
| | - Shuhan Sun
- Department of Medical Genetics; Second Military Medical University; 800 Xiangyin Road Shanghai 200433 P. R. China
| |
Collapse
|
10
|
Wang L, Yang M, Jin M, Wu Y, Zheng T, Gu S, Hua X. Transplant of insulin-like growth factor-1 expressing bone marrow stem cells improves functional regeneration of injured rat uterus by NF-κB pathway. J Cell Mol Med 2018. [PMID: 29516621 PMCID: PMC5908117 DOI: 10.1111/jcmm.13574] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
To investigate the potential beneficial effect of insulin-like growth factor-1 (IGF-1) in BMSC transplantation therapy of uterus injury and the underlying molecular mechanisms, rat BMSCs were isolated and cultured. The relative expressions of IGF-1 and IL-10 were determined by RT-PCR and immunoblotting. The secretory IL-10 and released E2 were measured using ELISA kits. The relative vWF and α-SMA expressions were determined by immunohistochemistry. The direct binding of NF-κB subunit p50 with IL-10 promoter was analysed by chromatin immunoprecipitation assay. The regulation of IL-10 expression by p50 was interrogated by luciferase reporter assay. Our data demonstrated that IGF-1 expression in BMSCs induced IL-10 expression and secretion, which was further enhanced by E2-PLGA. IGF-1 overexpression improved BMSCs transplantation therapy in rat uterus injury. We further demonstrated that both inhibition and knockdown of p50 abolished IGF-1-induced expression and secretion of IL-10 in BMSCs, which consequently compromised the IGF-1 conferred therapeutic benefits against uterus injury. Furthermore, we elucidated that p50 regulated IL-10 expression via direct association with its promoter. Our data suggested that transplantation of IGF-1 overexpressing BMSCs improved functional regeneration of injured uterus by inducing IL-10 expression and secretion via activation of NF-κB signalling.
Collapse
Affiliation(s)
- Lei Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mengnan Yang
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Minfei Jin
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuelin Wu
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Tao Zheng
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shengyi Gu
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaolin Hua
- Department of Obstetrics and Gynecology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Aydemir I, Özkut M, Fırat F, Gümürüdü A, Sal D, Erdoğan K, Göker A, Tuğlu M. Effects of stem cells applications on oxidative stress and apoptosis during implantation. ASIAN PACIFIC JOURNAL OF REPRODUCTION 2018. [DOI: 10.4103/2305-0500.241207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|