1
|
Liang Y, Meng J, Yu Z, Guo Y, Zhang X, Yan Y, Du S, Jin S, Li J, Yang H, Zhang X, Liu Z, Li L, Xie J. Ru single-atom nanozymes targeting ROS-ferroptosis pathways for enhanced endometrial regeneration in intrauterine adhesion therapy. Biomaterials 2025; 315:122923. [PMID: 39489016 DOI: 10.1016/j.biomaterials.2024.122923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/15/2024] [Accepted: 10/26/2024] [Indexed: 11/05/2024]
Abstract
Intrauterine adhesion (IUA) presents a significant challenge in gynecology, characterized by excessive fibrosis and compromised reproductive function, leading to severe infertility. Although biocompatible hydrogels integrated with stem cells offer a promising approach for IUA therapy, clinical applications remain limited. Recent studies have highlighted the role of ferroptosis and reactive oxygen species (ROS) in IUA pathogenesis, yet strategies targeting ferroptosis through antioxidant stress are underexplored. This study investigates the therapeutic effects and mechanisms of a Ru-Single-Atom Nanozyme (Ru-SAN) incorporated into chitosan hydrogel for treating IUA. Ru-SAN, which mimics the enzyme activities of catalase, superoxide dismutase, and glutathione peroxidase, effectively clears excess ROS and shows promise in treating oxidative stress-induced diseases. The results demonstrate the superior antioxidative capabilities of Ru-SAN, significantly suppressing the ROS-ferroptosis cycle at the injury site. This creates a favorable microenvironment for post-injury repair by inhibiting inflammation, enhancing mesenchymal-to-epithelial transformation, promoting angiogenesis, and polarizing M2 macrophages. Importantly, it mitigates adverse repair outcomes from inflammation and excessive collagen fiber deposition, ultimately restoring uterine glandular structures and thickness, thereby achieving the ultimate goal of restoring fertility and live birth rates. In conclusion, our study delineates a pioneering therapeutic approach leveraging the antioxidant properties of Ru-SAN to target ferroptosis, thereby offering an efficacious treatment for IUA.
Collapse
Affiliation(s)
- Yuxiang Liang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China; Shanxi Key Laboratory of Human Disease and Animal Models, Experimental Animal Center of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jian Meng
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China; Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Zhaowei Yu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Yuqian Guo
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Xiao Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Yujia Yan
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Shaobo Du
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Shanshan Jin
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Jing Li
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Hailan Yang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Xiaozheng Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Zhizhen Liu
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Liping Li
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Key Laboratory of Birth Defect and Cell Regeneration, MOE Key Laboratory of Coal Environmental Pathogenicity and Prevention, Department of Obstetrics and Gynecology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
2
|
Xypolita ME, Goolam M, Bikoff EK, Robertson EJ, Mould AW. The zinc-finger transcription factor Blimp1/Prdm1 is required for uterine remodelling and repair in the mouse. Nat Commun 2025; 16:1220. [PMID: 39890816 PMCID: PMC11785775 DOI: 10.1038/s41467-025-56511-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 01/17/2025] [Indexed: 02/03/2025] Open
Abstract
The zinc finger transcription factor Blimp1/PRDM1 regulates gene expression in diverse cell types. Its activity controls the maternal decidual response at early post-implantation stages of development. The present experiments demonstrate surprisingly that Blimp1 activity in the uterus is required for tissue remodelling at sites of embryonic failure. Moreover Blimp1 mutant females are refractory to RU486 induced decidual shedding. RNA-seq together with immunostaining experiments strongly suggest that the failure to up-regulate expression of the matrix metalloprotease Mmp10 in combination with insufficient suppression of BMP signalling, likely explain Blimp1-dependent phenotypic changes. In the post-partum uterus Blimp1 together with Mmp10 are highly upregulated at sites of tissue repair following placental detachment. Conditional Blimp1 removal significantly impairs the re-epithelization process and severely impacts involution of the endometrium and luminal epithelium. Overall these results identify Blimp1 as a master regulator of uterine tissue remodelling and repair.
Collapse
Affiliation(s)
- Maria-Eleni Xypolita
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Mubeen Goolam
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
- Department of Human Biology and Neuroscience Institute, University of Cape Town, Cape Town, 7925, South Africa
| | - Elizabeth K Bikoff
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| | - Elizabeth J Robertson
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK.
| | - Arne W Mould
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
- Department of Psychiatry, University of Oxford, Warneford Hospital, Oxford, OX3 7JX, UK
| |
Collapse
|
3
|
Xu X, Zhu P, Wang H, Chen K, Liu L, Du L, Jiang L, Hu Y, Zhou X, Zhang B, Pu X, Hu X, Xu Q, Zhang L, Li W. CD34 + PI16 + fibroblast progenitors aggravate neointimal lesions of allograft arteries via CCL11/CCR3-PI3K/AKT pathway. Theranostics 2025; 15:2523-2543. [PMID: 39990233 PMCID: PMC11840720 DOI: 10.7150/thno.104650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/01/2025] [Indexed: 02/25/2025] Open
Abstract
Rationale: Transplant-accelerated arteriosclerosis is a common complication that limits the long-term survival of organ transplant recipients. While previous studies have indicated the involvement of CD34+ stem/progenitor cells (SPCs) in this process, their heterogeneity and potential adverse effects remains incompletely understood. Methods: To investigate the role of CD34+ SPCs in transplant arteriosclerosis, we used various genetically modified mouse models, including BALB/c, C57BL/6J, CD34-CreERT2, Rosa26-tdTomato, Rosa26-iDTR, CD34-Dre, PI16-CreERT2, and CAG-LSL-RSR-tdTomato-2A-DTR mice. Single-cell RNA sequencing (scRNA-seq), chemokine antibody microarrays, ELISA assays, and immunohistochemistry were employed to identify fibroblast progenitors and their interactions with smooth muscle cells. Furthermore, in vivo and in vitro experiments targeting the CCL11/CCR3-PI3K/AKT signaling pathway were conducted to assess its role in the pathogenesis of transplant arteriosclerosis. Results: Single-cell RNA-seq and genetic lineage tracing revealed a subpopulation of fibroblast progenitors, characterized by high CD34 and PI16 expression, which differentiated into a distinct chemotactic fibroblast subset. Proteomic and scRNA analysis revealed that this CD34+ PI16- subgroup released CCL11 (Eotaxin-1), which promoted intimal hyperplasia through the paracrine activation of smooth muscle cells. Binding of CCL11 to its receptor CCR3 activated the PI3K/AKT signaling pathway in smooth muscle cells, driving their proliferation and migration. In vivo, overexpression of CCL11 promoted neointimal hyperplasia, while neutralizing CCL11 or inhibiting CCR3 alleviated neointimal formation. Conclusions: These findings identified CD34+ PI16+ fibroblast progenitors that differentiate into specific chemotactic fibroblasts, releasing chemokines pivotal for neointima formation, suggesting a therapeutic strategy targeting their chemotactic activity.
Collapse
Affiliation(s)
- Xiaodong Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pengwei Zhu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Han Wang
- School of Engineering and Materials Science, Queen Mary University of London, United Kingdom
| | - Kai Chen
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Liu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Luping Du
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liujun Jiang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanhua Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuhao Zhou
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bohuan Zhang
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangyuan Pu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaosheng Hu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Zhang
- Department of Cardiology, and Institute for Cardiovascular Development and Regenerative Medicine, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weidong Li
- Department of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Wang J, Zhan H, Wang Y, Zhao L, Huang Y, Wu R. Current advances in understanding endometrial epithelial cell biology and therapeutic applications for intrauterine adhesion. Stem Cell Res Ther 2024; 15:379. [PMID: 39456113 PMCID: PMC11515228 DOI: 10.1186/s13287-024-03989-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The human endometrium is a highly regenerative tissue capable of undergoing scarless repair during the menstruation and postpartum phases. This process is mediated by endometrial adult stem/progenitor cells. During the healing of endometrial injuries, swift reepithelization results in the rapid covering of the wound surface and facilitates subsequent endometrial restoration. The involvement of endogenous endometrial epithelial stem cells, stromal cells, and bone marrow-derived cells in the regeneration of the endometrial epithelium has been a subject of prolonged debate. Increasing evidence suggests that the regeneration of the endometrial epithelium mainly relies on epithelial stem cells rather than stromal cells and bone marrow-derived cells. Currently, no consensus has been established on the identity of epithelial stem cells in the epithelial compartment. Several markers, including stage-specific embryonic antigen-1 (SSEA-1), sex-determining region Y-box 9 (SOX9), neural-cadherin (N-cadherin), leucine-rich-repeat-containing G-protein-coupled receptor 5 (LGR5), CD44, axis inhibition protein 2 (Axin2), and aldehyde dehydrogenase 1A1 (ALDH1A1), have been suggested as potential candidate markers for endometrial epithelial stem cells. The identification of endometrial epithelial stem cells contributes to our understanding of endometrial regeneration and offers new therapeutic insights into diseases characterized by regenerative defects in the endometrium, such as intrauterine adhesion. This review explores different perspectives on the origins of human and mouse endometrial epithelial cells. It summarizes the potential markers, locations, and hierarchies of epithelial stem cells in both human and mouse endometrium. It also discusses epithelial cell-based treatments for intrauterine adhesion, hoping to inspire further research and clinical application of endometrial epithelial stem cells.
Collapse
Affiliation(s)
- Jia Wang
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, 310006, Zhejiang, People's Republic of China
- Zhejiang Key Laboratory of Maternal and Infant Health, Hangzhou, People's Republic of China
| | - Hong Zhan
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, 310006, Zhejiang, People's Republic of China
- Zhejiang Key Laboratory of Maternal and Infant Health, Hangzhou, People's Republic of China
| | - Yinfeng Wang
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, 310006, Zhejiang, People's Republic of China
- Zhejiang Key Laboratory of Maternal and Infant Health, Hangzhou, People's Republic of China
| | - Li Zhao
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, 310006, Zhejiang, People's Republic of China
- Zhejiang Key Laboratory of Maternal and Infant Health, Hangzhou, People's Republic of China
| | - Yunke Huang
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, 310006, Zhejiang, People's Republic of China
- Zhejiang Key Laboratory of Maternal and Infant Health, Hangzhou, People's Republic of China
| | - Ruijin Wu
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, 310006, Zhejiang, People's Republic of China.
- Zhejiang Key Laboratory of Maternal and Infant Health, Hangzhou, People's Republic of China.
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, People's Republic of China.
| |
Collapse
|
5
|
Xu Y, Zhang H, Chen Y, Pober JS, Zhou M, Zhou JH, Min W. SRF SUMOylation modulates smooth muscle phenotypic switch and vascular remodeling. Nat Commun 2024; 15:6919. [PMID: 39134547 PMCID: PMC11319592 DOI: 10.1038/s41467-024-51350-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 08/05/2024] [Indexed: 08/15/2024] Open
Abstract
Serum response factor (SRF) controls gene transcription in vascular smooth muscle cells (VSMCs) and regulates VSMC phenotypic switch from a contractile to a synthetic state, which plays a key role in the pathogenesis of cardiovascular diseases (CVD). It is not known how post-translational SUMOylation regulates the SRF activity in CVD. Here we show that Senp1 deficiency in VSMCs increased SUMOylated SRF and the SRF-ELK complex, leading to augmented vascular remodeling and neointimal formation in mice. Mechanistically, SENP1 deficiency in VSMCs increases SRF SUMOylation at lysine 143, reducing SRF lysosomal localization concomitant with increased nuclear accumulation and switching a contractile phenotype-responsive SRF-myocardin complex to a synthetic phenotype-responsive SRF-ELK1 complex. SUMOylated SRF and phospho-ELK1 are increased in VSMCs from coronary arteries of CVD patients. Importantly, ELK inhibitor AZD6244 prevents the shift from SRF-myocardin to SRF-ELK complex, attenuating VSMC synthetic phenotypes and neointimal formation in Senp1-deficient mice. Therefore, targeting the SRF complex may have a therapeutic potential for the treatment of CVD.
Collapse
Affiliation(s)
- Yue Xu
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- State Key Laboratory of Ophthalmology and Guangdong Province Key Laboratory of Ophthalmology and Visual Science, Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Haifeng Zhang
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Yuxin Chen
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Jordan S Pober
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Min Zhou
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Jenny Huanjiao Zhou
- Interdepartmental Program in Vascular Biology and Therapeutics, Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| | - Wang Min
- Cardiovascular Medical Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
6
|
Zhou Z, Wu X, Chen T, Zhang B, Li W, Zhou M, Zhao J, Dong E, Li T. Restoration of functional endometrium in an intrauterine adhesion rat model with endometrial stromal cells transplantation. Stem Cell Res Ther 2024; 15:181. [PMID: 38902788 PMCID: PMC11191336 DOI: 10.1186/s13287-024-03788-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 06/09/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Intrauterine adhesion (IUA) as a prevalent gynecological disease is developed from infection or trauma. However, therapeutic strategies to repair damaged endometrium are relatively limited. Emerging studies have shed light on the crucial role of endometrial stromal cells (EnSCs) in the process of uterine endometrial regeneration. EnSCs isolated from the uterine endometrium have similar characteristics to mesenchymal stem cells (MSCs). However, it is still unknown whether EnSCs could be used as donor cells to treat IUA. The aim of this study was to evaluate the potential efficacy of EnSCs in treating rat IUA. METHODS Human EnSCs were isolated from the endometrial tissue of healthy female donors and subjected to extensive expansion and culture in vitro. Immunofluorescence, flow cytometry, cell proliferation assay, trilineage differentiation experiment, and decidualization assay were used to characterize the biological properties of EnSCs. We evaluated the immunoregulatory potential of EnSCs by analyzing their secreted cytokines and conducting bulk RNA sequencing after IFN-γ treatment. After EnSCs were transplanted into the uterine muscle layer in IUA rats, their therapeutic effects and underlying mechanisms were analyzed using histological analysis, Q-PCR, fertility and pregnancy outcome assay, and transcriptome analysis. RESULTS We successfully isolated EnSCs from the endometrium of human donors and largely expanded in vitro. EnSCs exhibited characteristics of mesenchymal stem cells and retained responsiveness to sex hormones. Following IFN-γ stimulation, EnSCs upregulated the anti-inflammatory cytokines and activated immunosuppressive molecules. Xenogeneic transplantation of EnSCs successfully repaired injured endometrium and significantly restored the pregnancy rate in IUA rats. Mechanistically, the therapeutic effects of EnSCs on IUA endometrium functioned through anti-inflammation, anti-fibrosis and the secretion of regeneration factor. CONCLUSIONS Due to their large expansion ability, immunoregulatory properties, and great potential in treating IUA, EnSCs, as a valuable source of donor cells, could offer a potential treatment avenue for injury-induced IUA.
Collapse
Affiliation(s)
- Zhengli Zhou
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, 650500, China
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650031, China
| | - Xiaomei Wu
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650031, China
| | - Tingwei Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, 650500, China
| | - Bo Zhang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, 650500, China
| | - Wenxin Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, 650500, China
| | - Min Zhou
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, 650500, China
| | - Jingxue Zhao
- The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650031, China
| | - E Dong
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, 650500, China.
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, 650500, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, 650500, China.
| |
Collapse
|
7
|
Balough JL, Moalli P. Regenerative Medicine in Gynecology. Obstet Gynecol 2024; 143:767-773. [PMID: 38663014 PMCID: PMC11216342 DOI: 10.1097/aog.0000000000005590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/21/2024] [Indexed: 05/18/2024]
Abstract
The female reproductive tract undergoes dynamic changes across the life span. Congenital abnormalities, life events, and medical interventions can negatively affect the structure and function of reproductive tract organs, resulting in lifelong sequelae. The objective of regenerative gynecology is to discover and promote endogenous mechanisms by which a healthy tissue maintains overall tissue integrity after injury, after disease, or with age. In this review, we discuss some of the key state-of-the-art cell-based and scaffolding therapies that have been applied to regenerate gynecologic tissues and organs primarily in animal and tissue culture models. We further discuss the limitations of current technologies, problems of implementation and scalability, and future outlook of the field.
Collapse
Affiliation(s)
- Julia L. Balough
- Department of Obstetrics, Gynecology & Reproductive Science, University of Pittsburgh, Pittsburgh, PA
- Magee-Women’s Research Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Pamela Moalli
- Department of Obstetrics, Gynecology & Reproductive Science, University of Pittsburgh, Pittsburgh, PA
- Magee-Women’s Research Institute, University of Pittsburgh Medical Center, Pittsburgh, PA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA
- The McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA
- Division of Urogynecology & Reconstructive Pelvic Surgery, University of Pittsburgh Medical Center Magee-Women’s Hospital, Pittsburgh, PA, USA
| |
Collapse
|
8
|
Flesken-Nikitin A, Pirtz MG, Ashe CS, Ellenson LH, Cosgrove BD, Nikitin AY. Dysregulation of cell state dynamics during early stages of serous endometrial carcinogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585274. [PMID: 38562813 PMCID: PMC10983873 DOI: 10.1101/2024.03.15.585274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Serous endometrial carcinoma (SEC) constitutes about 10% of endometrial carcinomas and is one of the most aggressive and lethal types of uterine cancer. Due to the rapid progression of SEC, early detection of this disease is of utmost importance. However, molecular and cellular dynamics during the pre-dysplastic stage of this disease remain largely unknown. Here, we provide a comprehensive census of cell types and their states for normal, pre-dysplastic, and dysplastic endometrium in a mouse model of SEC. This model is associated with inactivation of tumor suppressor genes Trp53 and Rb1 , whose pathways are altered frequently in SEC. We report that pre-dysplastic changes are characterized by an expanded and increasingly diverse immature luminal epithelial cell populations. Consistent with transcriptome changes, cells expressing the luminal epithelial marker TROP2 begin to substitute FOXA2+ cells in the glandular epithelium. These changes are associated with a reduction in number and strength of predicted interactions between epithelial and stromal endometrial cells. By using a multi-level approach combining single-cell and spatial transcriptomics paired with screening for clinically relevant genes in human endometrial carcinoma, we identified a panel of 44 genes suitable for further testing of their validity as early diagnostic and prognostic markers. Among these genes are known markers of human SEC, such as C DKN2A, and novel markers, such as OAS2 and OASL, members of 2-5A synthetase family that is essential for the innate immune response. In summary, our results suggest an important role of the luminal epithelium in SEC pathogenesis, highlight aberrant cell-cell interactions in pre-dysplastic stages, and provide a new platform for comparative identification and characterization of novel, clinically relevant prognostic and diagnostic markers and potential therapeutic modalities.
Collapse
|
9
|
Ya X, Ma L, Liu C, Ge P, Xu Y, Zheng Z, Mou S, Wang R, Zhang Q, Ye X, Zhang D, Zhang Y, Wang W, Li H, Zhao J. Metabolic alterations of peripheral blood immune cells and heterogeneity of neutrophil in intracranial aneurysms patients. Clin Transl Med 2024; 14:e1572. [PMID: 38314932 PMCID: PMC10840020 DOI: 10.1002/ctm2.1572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/15/2024] [Accepted: 01/21/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Intracranial aneurysms (IAs) represent a severe cerebrovascular disease that can potentially lead to subarachnoid haemorrhage. Previous studies have demonstrated the involvement of peripheral immune cells in the formation and progression of IAs. Nevertheless, the impact of metabolic alterations in peripheral immune cells and changes in neutrophil heterogeneity on the occurrence and progression of IAs remains uncertain. METHODS Single-cell Cytometry by Time-of-Flight (CyTOF) technology was employed to profile the single-cell atlas of peripheral blood mononuclear cells (PBMCs) and polymorphonuclear cells (PMNs) in 72 patients with IAs. In a matched cohort, metabolic shifts in PBMC subsets of IA patients were investigated by contrasting the expression levels of key metabolic enzymes with their respective counterparts in the healthy control group. Simultaneously, compositional differences in peripheral blood PMNs subsets between the two groups were analysed to explore the impact of altered heterogeneity in neutrophils on the initiation and progression of IAs. Furthermore, integrating immune features based on CyTOF analysis and clinical characteristics, we constructed an aneurysm occurrence model and an aneurysm growth model using the random forest method in conjunction with LASSO regression. RESULTS Different subsets exhibited distinct metabolic characteristics. Overall, PBMCs from patients elevated CD98 expression and increased proliferation. Conversely, CD36 was up-regulated in T cells, B cells and monocytes from the controls but down-regulated in NK and NKT cells. The comparison also revealed differences in the metabolism and function of specific subsets between the two groups. In terms of PMNs, the neutrophil landscape within patients group revealed a pronounced shift towards heightened complexity. Various neutrophil subsets from the IA group generally exhibited lower expression levels of anti-inflammatory functional molecules (IL-4 and IL-10). By integrating clinical and immune features, the constructed aneurysm occurrence model could precisely identify patients with IAs with high prediction accuracy (AUC = 0.987). Furthermore, the aneurysm growth model also exhibited superiority over ELAPSS scores in predicting aneurysm growth (lower prediction errors and out-of-bag errors). CONCLUSION These findings enhanced our understanding of peripheral immune cell participation in aneurysm formation and growth from the perspectives of immune metabolism and neutrophil heterogeneity. Moreover, the predictive model based on CyTOF features holds the potential to aid in diagnosing and monitoring the progression of human IAs.
Collapse
Affiliation(s)
- Xiaolong Ya
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Long Ma
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Chenglong Liu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Peicong Ge
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Yiqiao Xu
- School of Clinical MedicineCapital Medical UniversityBeijingChina
| | - Zhiyao Zheng
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Department of NeurosurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Siqi Mou
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Medical SchoolUniversity of Chinese Academy of SciencesBeijingChina
| | - Rong Wang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Qian Zhang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Xun Ye
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Dong Zhang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- Department of NeurosurgeryBeijing HospitalBeijingChina
| | - Yan Zhang
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Wenjing Wang
- Beijing Institute of HepatologyBeijing YouAn HospitalCapital Medical UniversityBeijingChina
| | - Hao Li
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Jizong Zhao
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| |
Collapse
|
10
|
Hong IS. Endometrial Stem Cells: Orchestrating Dynamic Regeneration of Endometrium and Their Implications in Diverse Endometrial Disorders. Int J Biol Sci 2024; 20:864-879. [PMID: 38250149 PMCID: PMC10797688 DOI: 10.7150/ijbs.89795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 12/24/2023] [Indexed: 01/23/2024] Open
Abstract
The human endometrium, a vital component of the uterus, undergoes dynamic changes during the menstrual cycle to create a receptive environment for embryo implantation. Its remarkable regenerative capacity can be attributed to the presence of tissue-resident stem cell populations within the endometrium. Despite variations in characteristics among different subtypes, endometrial stem cells exhibit notably robust self-renewal capacity and the ability to differentiate into multiple lineages. This review offers a comprehensive insight into the current literature and recent advancements regarding the roles of various endometrial stem cell types during dynamic regeneration of the endometrium during the menstrual cycle. In addition, emerging evidence suggests that dysfunction or depletion of endometrial stem cells may play critical roles in the development and progression of various endometrial disorders, such as endometriosis, uterine fibroids, adenomyosis, infertility, and endometrial cancer. Therefore, we also highlight potential roles of endometrial stem cells in the development and progression of these endometrial diseases, including their ability to accumulate genetic mutations and express genes associated with endometrial diseases. Understanding the dynamic properties of the endometrium and the roles of endometrial stem cells in various endometrial disorders will shed light on potential therapeutic strategies for managing these conditions and improving women's fertility outcomes.
Collapse
Affiliation(s)
- In-Sun Hong
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea
- Department of Molecular Medicine, School of Medicine, Gachon University, Incheon 406-840, Republic of Korea
| |
Collapse
|
11
|
Abstract
The uterine lining (endometrium) regenerates repeatedly over the life span as part of its normal physiology. Substantial portions of the endometrium are shed during childbirth (parturition) and, in some species, menstruation, but the tissue is rapidly rebuilt without scarring, rendering it a powerful model of regeneration in mammals. Nonetheless, following some assaults, including medical procedures and infections, the endometrium fails to regenerate and instead forms scars that may interfere with normal endometrial function and contribute to infertility. Thus, the endometrium provides an exceptional platform to answer a central question of regenerative medicine: Why do some systems regenerate while others scar? Here, we review our current understanding of diverse endometrial disruption events in humans, nonhuman primates, and rodents, and the associated mechanisms of regenerative success and failure. Elucidating the determinants of these disparate repair processes promises insights into fundamental mechanisms of mammalian regeneration with substantial implications for reproductive health.
Collapse
Affiliation(s)
- Claire J Ang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA;
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Taylor D Skokan
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA;
| | - Kara L McKinley
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA;
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
12
|
Xu Y, Hu J, Lv Q, Shi C, Qiu M, Xie L, Liu W, Yang B, Shan W, Cheng Y, Zhao B, Chen X. Endometrium-derived mesenchymal stem cells suppress progression of endometrial cancer via the DKK1-Wnt/β-catenin signaling pathway. Stem Cell Res Ther 2023; 14:159. [PMID: 37287079 DOI: 10.1186/s13287-023-03387-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 05/25/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) therapy is an attractive treatment option for various cancers. Whether MSCs can be used to treat well-differentiated endometrial cancer (EC) remains unclear. The aim of this study is to explore the potential therapeutic effects of MSCs on EC and the underlying mechanisms. METHODS The effects of adipose-derived MSCs (AD-MSCs), umbilical-cord-derived MSCs (UC-MSCs), and endometrium-derived MSCs (eMSCs) on the malignant behaviors of EC cells were explored via in vitro and in vivo experiments. Three EC models, including patient-derived EC organoid lines, EC cell lines, and EC xenograft model in female BALB/C nude mice, were used for this study. The effects of MSCs on EC cell proliferation, apoptosis, migration, and the growth of xenograft tumors were evaluated. The potential mechanisms by which eMSCs inhibit EC cell proliferation and stemness were explored by regulating DKK1 expression in eMSCs or Wnt signaling in EC cells. RESULTS Our results showed that eMSCs had the highest inhibitory effect on EC cell viability, and EC xenograft tumor growth in mice compared to AD-MSCs and UC-MSCs. Conditioned medium (CM) obtained from eMSCs significantly suppressed the sphere-forming ability and stemness-related gene expression of EC cells. In comparison to AD-MSCs and UC-MSCs, eMSCs had the highest level of Dickkopf-related protein 1 (DKK1) secretion. Mechanistically, eMSCs inhibited Wnt/β-catenin signaling in EC cells via secretion of DKK1, and eMSCs suppressed EC cell viability and stemness through DKK1-Wnt/β-catenin signaling. Additionally, the combination of eMSCs and medroxyprogesterone acetate (MPA) significantly inhibited the viability of EC organoids and EC cells compared with eMSCs or MPA alone. CONCLUSIONS The eMSCs, but not AD-MSCs or UC-MSCs, could suppress the malignant behaviors of EC both in vivo and in vitro via inhibiting the Wnt/β-catenin signaling pathway by secreting DKK1. The combination of eMSCs and MPA effectively inhibited EC growth, indicating that eMSCs may potentially be a new therapeutic strategy for young EC patients desiring for fertility preservation.
Collapse
Affiliation(s)
- Yuhui Xu
- Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, People's Republic of China
| | - Jiali Hu
- Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, People's Republic of China
| | - Qiaoying Lv
- Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, People's Republic of China
| | - Chenyi Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, People's Republic of China
| | - Mengdi Qiu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, People's Republic of China
| | - Liying Xie
- Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, People's Republic of China
| | - Wei Liu
- Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, People's Republic of China
| | - Bingyi Yang
- Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, People's Republic of China
| | - Weiwei Shan
- Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, People's Republic of China
| | - Yali Cheng
- Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, People's Republic of China
| | - Bing Zhao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, People's Republic of China.
| | - Xiaojun Chen
- Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
13
|
Li X, Duan H, Wang S, Lv CX. Umbilical cord mesenchymal stem cell-derived exosomes reverse endometrial fibrosis by the miR-145-5p/ZEB2 axis in intrauterine adhesions. Reprod Biomed Online 2023; 46:234-243. [PMID: 36567149 DOI: 10.1016/j.rbmo.2022.05.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/21/2022] [Accepted: 05/24/2022] [Indexed: 02/07/2023]
Abstract
RESEARCH QUESTION What is the specific mechanism of umbilical cord mesenchymal stem cell-derived exosomes (UCMSC-exos) in regulating endometrial repair and regeneration? DESIGN In this study, UCMSC-exos were harvested by differential ultracentrifugation from umbilical cord mesenchymal stem cell culture supernatant and identified with western blotting, transmission electron microscopy and nanoparticle tracking analysis. Transforming growth factor-β1 (TGFβ1) at different concentrations was used to construct the intrauterine adhesions cell model. The fibrotic markers were assessed by quantitative reverse transcription-polymerase chain reaction and western blotting. The effects of miR-145-5p over-expression on endometrial fibrosis were assessed. Dual luciferase assay was performed to verify the relationship between miR-145-5p and zinc finger E-box binding homeobox 2 (ZEB2). RESULTS The isolated UCMSC-exos had a typical cup-shaped morphology, expressed the specific exosomal markers Alix, CD63 and TSG101, and were approximately 50-150 nm in diameter. TGFβ1 at 10 ng/ml significantly promoted endometrial fibrosis, which was reversed by 20 µg/ml UCMSC-exos. Exosomal miR-145-5p ameliorated TGFβ1-induced endometrial fibrosis. ZEB2 was inversely regulated by exosomal miR-145-5p as a direct target. CONCLUSIONS UCMSC-exos might reverse endometrial stromal cell fibrosis by regulating the miR-145-5p/ZEB2 axis, representing a potential novel strategy to promote endometrial repair.
Collapse
Affiliation(s)
- Xiao Li
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Hua Duan
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China.
| | - Sha Wang
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| | - Cheng-Xiao Lv
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing 100006, China
| |
Collapse
|
14
|
Rosa-E-Silva ACJDS, Mamillapalli R, Rosa-E-Silva JC, Ucar A, Schwartz J, Taylor HS. Uterine administration of C-X-C motif chemokine ligand 12 increases the pregnancy rates in mice with induced endometriosis. F&S SCIENCE 2023; 4:65-73. [PMID: 36252793 DOI: 10.1016/j.xfss.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To study the effect of intrauterine injection of C-X-C motif chemokine ligand 12 (CXCL12), also known as a stem cell chemoattractant (stromal cell-derived factor 1), on fertility and endometrial receptivity in mice with endometriosis. DESIGN Laboratory study. SETTING Academic Medical Center. ANIMAL(S) Fifty-six mice underwent chemotherapy and bone marrow transplantation. Thirty-six of these mice underwent either surgery to induce endometriosis (n = 20) or sham surgery (n = 16). INTERVENTION(S) Injection of CXCL12 as a potential therapeutic agent to improve fertility in endometriosis. MAIN OUTCOME MEASURE(S) Pregnancy rate, bone marrow-derived cell (BMDC) recruitment and endometrial receptivity markers. RESULT(S) The mice with or without endometriosis received a single uterine injection of either CXCL12 or placebo. Uterine injection of CXCL12 increased the pregnancy rates in a mouse model of endometriosis. Mice were euthanized after delivery, and implantation markers homeobox A11, alpha-v beta-3 integrin, and progesterone receptor were analyzed by immunohistochemistry, whereas green fluorescent protein positive BMDC recruitment was quantified by immunohistochemistry and immunofluorescence. The sham surgery groups without endometriosis had the highest cumulative pregnancy rate (100%) regardless of CXCL12 treatment. The endometriosis group treated with placebo had the lowest pregnancy rate. An increased pregnancy rate was noted in the endometriosis group after treatment with CXCL12. There was also an increase in BMDC recruitment and endometrial expression of progesterone receptor and alpha-v beta-3 integrin in the endometriosis group that received CXCL12 compared with that in the endometriosis group that received placebo. CONCLUSION(S) Uterine injection of CXCL12 increased the pregnancy rates in a mouse model of endometriosis. These results suggest that CXCL12 has a potential role as a therapeutic agent in women with infertility related to endometriosis and potentially other endometrial receptivity defects.
Collapse
Affiliation(s)
- Ana Carolina Japur de Sá Rosa-E-Silva
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut; Department of Gynecology and Obstetrics-Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brasil
| | - Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut.
| | - Julio Cesar Rosa-E-Silva
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut; Department of Gynecology and Obstetrics-Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brasil
| | - Abdullah Ucar
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Joshua Schwartz
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
15
|
Chen K, Zheng S, Fang F. Endometrial Stem Cells and Their Applications in Intrauterine Adhesion. Cell Transplant 2023; 32:9636897231159561. [PMID: 36891869 PMCID: PMC9998408 DOI: 10.1177/09636897231159561] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2023] Open
Abstract
Intrauterine adhesion (IUA), resulting from pregnancy or nonpregnant uterine trauma, is one of the major causes of abnormal menstruation, infertility, or repeated pregnancy loss. Although a few methods, including hysteroscopy and hormone therapy, are routinely used for its diagnosis and treatment, they cannot restore tissue regeneration. Stem cells, which have self-renewal and tissue regeneration abilities, have been proposed as a promising therapy for patients with severe IUAs. In this review, we summarize the origin and features of endometrium-associated stem cells and their applications in the treatment of IUAs based on animal models and human clinical trials. We expect that this information will help to elucidate the underlying mechanism for tissue regeneration and to improve the design of stem cell-based therapies for IUAs.
Collapse
Affiliation(s)
- Kai Chen
- Reproductive Medicine Center & Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Wannan Medical College, Wuhu, China
| | - Shengxia Zheng
- Reproductive Medicine Center & Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fang Fang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
16
|
Endometriosis Stem Cells as a Possible Main Target for Carcinogenesis of Endometriosis-Associated Ovarian Cancer (EAOC). Cancers (Basel) 2022; 15:cancers15010111. [PMID: 36612107 PMCID: PMC9817684 DOI: 10.3390/cancers15010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Endometriosis is a serious recurrent disease impairing the quality of life and fertility, and being a risk for some histologic types of ovarian cancer defined as endometriosis-associated ovarian cancers (EAOC). The presence of stem cells in the endometriotic foci could account for the proliferative, migrative and angiogenic activity of the lesions. Their phenotype and sources have been described. The similarly disturbed expression of several genes, miRNAs, galectins and chaperones has been observed both in endometriotic lesions and in ovarian or endometrial cancer. The importance of stem cells for nascence and sustain of malignant tumors is commonly appreciated. Although the proposed mechanisms promoting carcinogenesis leading from endometriosis into the EAOC are not completely known, they have been discussed in several articles. However, the role of endometriosis stem cells (ESCs) has not been discussed in this context. Here, we postulate that ESCs may be a main target for the carcinogenesis of EAOC and present the possible sequence of events resulting finally in the development of EAOC.
Collapse
|
17
|
Dufour D, Dumontet T, Sahut-Barnola I, Carusi A, Onzon M, Pussard E, Wilmouth JJ, Olabe J, Lucas C, Levasseur A, Damon-Soubeyrand C, Pointud JC, Roucher-Boulez F, Tauveron I, Bossis G, Yeh ET, Breault DT, Val P, Lefrançois-Martinez AM, Martinez A. Loss of SUMO-specific protease 2 causes isolated glucocorticoid deficiency by blocking adrenal cortex zonal transdifferentiation in mice. Nat Commun 2022; 13:7858. [PMID: 36543805 PMCID: PMC9772323 DOI: 10.1038/s41467-022-35526-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
SUMOylation is a dynamic posttranslational modification, that provides fine-tuning of protein function involved in the cellular response to stress, differentiation, and tissue development. In the adrenal cortex, an emblematic endocrine organ that mediates adaptation to physiological demands, the SUMOylation gradient is inversely correlated with the gradient of cellular differentiation raising important questions about its role in functional zonation and the response to stress. Considering that SUMO-specific protease 2 (SENP2), a deSUMOylating enzyme, is upregulated by Adrenocorticotropic Hormone (ACTH)/cAMP-dependent Protein Kinase (PKA) signalling within the zona fasciculata, we generated mice with adrenal-specific Senp2 loss to address these questions. Disruption of SENP2 activity in steroidogenic cells leads to specific hypoplasia of the zona fasciculata, a blunted reponse to ACTH and isolated glucocorticoid deficiency. Mechanistically, overSUMOylation resulting from SENP2 loss shifts the balance between ACTH/PKA and WNT/β-catenin signalling leading to repression of PKA activity and ectopic activation of β-catenin. At the cellular level, this blocks transdifferentiation of β-catenin-positive zona glomerulosa cells into fasciculata cells and sensitises them to premature apoptosis. Our findings indicate that the SUMO pathway is critical for adrenal homeostasis and stress responsiveness.
Collapse
Affiliation(s)
- Damien Dufour
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
| | - Typhanie Dumontet
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
- Department of Internal Medicine, Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI, USA
- Training Program in Organogenesis, Center for Cell Plasticity and Organ Design, University of Michigan, Ann Arbor, MI, USA
| | - Isabelle Sahut-Barnola
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
| | - Aude Carusi
- IGMM, Université de Montpellier, CNRS, Montpellier, France
| | - Méline Onzon
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
| | - Eric Pussard
- Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpital de Bicêtre, Assistance Publique-Hôpitaux de Paris (APHP), Physiologie et Physiopathologie Endocriniennes, INSERM, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - James Jr Wilmouth
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
| | - Julie Olabe
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
| | - Cécily Lucas
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
- Endocrinologie Moléculaire et Maladies Rares, Centre Hospitalier Universitaire, Université Claude Bernard Lyon 1, Bron, France
| | - Adrien Levasseur
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
| | - Christelle Damon-Soubeyrand
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
| | - Jean-Christophe Pointud
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
| | - Florence Roucher-Boulez
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
- Endocrinologie Moléculaire et Maladies Rares, Centre Hospitalier Universitaire, Université Claude Bernard Lyon 1, Bron, France
| | - Igor Tauveron
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
- Service d'Endocrinologie, Centre Hospitalier Universitaire Gabriel Montpied, Université Clermont Auvergne, Clermont-Ferrand, France
| | | | - Edward T Yeh
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Pierre Val
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
| | - Anne-Marie Lefrançois-Martinez
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France
| | - Antoine Martinez
- institut Génétique, Reproduction & Développement (iGReD), CNRS, INSERM, Université Clermont Auvergne, Clermont-Ferrand, F-63000, France.
| |
Collapse
|
18
|
Zhu Q, Liang P, Chu C, Zhang A, Zhou W. Protein sumoylation in normal and cancer stem cells. Front Mol Biosci 2022; 9:1095142. [PMID: 36601585 PMCID: PMC9806136 DOI: 10.3389/fmolb.2022.1095142] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
Stem cells with the capacity of self-renewal and differentiation play pivotal roles in normal tissues and malignant tumors. Whereas stem cells are supposed to be genetically identical to their non-stem cell counterparts, cell stemness is deliberately regulated by a dynamic network of molecular mechanisms. Reversible post-translational protein modifications (PTMs) are rapid and reversible non-genetic processes that regulate essentially all physiological and pathological process. Numerous studies have reported the involvement of post-translational protein modifications in the acquirement and maintenance of cell stemness. Recent studies underscore the importance of protein sumoylation, i.e., the covalent attachment of the small ubiquitin-like modifiers (SUMO), as a critical post-translational protein modification in the stem cell populations in development and tumorigenesis. In this review, we summarize the functions of protein sumoylation in different kinds of normal and cancer stem cells. In addition, we describe the upstream regulators and the downstream effectors of protein sumoylation associated with cell stemness. We also introduce the translational studies aiming at sumoylation to target stem cells for disease treatment. Finally, we propose future directions for sumoylation studies in stem cells.
Collapse
Affiliation(s)
- Qiuhong Zhu
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Panpan Liang
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Cuiying Chu
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Aili Zhang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States,*Correspondence: Aili Zhang, ; Wenchao Zhou,
| | - Wenchao Zhou
- Intelligent Pathology Institute, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China,*Correspondence: Aili Zhang, ; Wenchao Zhou,
| |
Collapse
|
19
|
Kirkwood PM, Gibson DA, Shaw I, Dobie R, Kelepouri O, Henderson NC, Saunders PTK. Single-cell RNA sequencing and lineage tracing confirm mesenchyme to epithelial transformation (MET) contributes to repair of the endometrium at menstruation. eLife 2022; 11:e77663. [PMID: 36524724 PMCID: PMC9873258 DOI: 10.7554/elife.77663] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
The human endometrium experiences repetitive cycles of tissue wounding characterised by piecemeal shedding of the surface epithelium and rapid restoration of tissue homeostasis. In this study, we used a mouse model of endometrial repair and three transgenic lines of mice to investigate whether epithelial cells that become incorporated into the newly formed luminal epithelium have their origins in one or more of the mesenchymal cell types present in the stromal compartment of the endometrium. Using scRNAseq, we identified a novel population of PDGFRb + mesenchymal stromal cells that developed a unique transcriptomic signature in response to endometrial breakdown/repair. These cells expressed genes usually considered specific to epithelial cells and in silico trajectory analysis suggested they were stromal fibroblasts in transition to becoming epithelial cells. To confirm our hypothesis we used a lineage tracing strategy to compare the fate of stromal fibroblasts (PDGFRa+) and stromal perivascular cells (NG2/CSPG4+). We demonstrated that stromal fibroblasts can undergo a mesenchyme to epithelial transformation and become incorporated into the re-epithelialised luminal surface of the repaired tissue. This study is the first to discover a novel population of wound-responsive, plastic endometrial stromal fibroblasts that contribute to the rapid restoration of an intact luminal epithelium during endometrial repair. These findings form a platform for comparisons both to endometrial pathologies which involve a fibrotic response (Asherman's syndrome, endometriosis) as well as other mucosal tissues which have a variable response to wounding.
Collapse
Affiliation(s)
- Phoebe M Kirkwood
- Centre for Inflammation Research, University of EdinburghEdinburghUnited Kingdom
| | - Douglas A Gibson
- Centre for Inflammation Research, University of EdinburghEdinburghUnited Kingdom
| | - Isaac Shaw
- Centre for Inflammation Research, University of EdinburghEdinburghUnited Kingdom
| | - Ross Dobie
- Centre for Inflammation Research, University of EdinburghEdinburghUnited Kingdom
| | - Olympia Kelepouri
- Centre for Inflammation Research, University of EdinburghEdinburghUnited Kingdom
| | - Neil C Henderson
- Centre for Inflammation Research, University of EdinburghEdinburghUnited Kingdom
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of EdinburghEdinburghUnited Kingdom
| | - Philippa TK Saunders
- Centre for Inflammation Research, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
20
|
Hu M, Sun D, Yu J, Fu Y, Qin Z, Huang B, Zhang Q, Chen X, Wei Y, Zhu H, Wang Y, Feng Y, Zheng W, Liao H, Li J, Wu S, Zhang Z. Brusatol sensitizes endometrial hyperplasia and cancer to progestin by suppressing NRF2-TET1-AKR1C1-mediated progestin metabolism. J Transl Med 2022; 102:1335-1345. [PMID: 36038734 DOI: 10.1038/s41374-022-00816-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 05/28/2022] [Accepted: 05/31/2022] [Indexed: 11/09/2022] Open
Abstract
Progestin resistance is the main obstacle for the conservative therapy to maintain fertility in women with endometrial cancer. Brusatol was identified as an inhibitor of the NRF2 pathway; however, its impact on progestin resistance and the underlying mechanism remains unclear. Here, we found that brusatol sensitized endometrial cancer to progestin by suppressing NRF2-TET1-AKR1C1-mediated progestin metabolism. Brusatol transcriptionally suppressed AKR1C1 via modifying the hydroxymethylation status in its promoter region through TET1 inhibition. Suppression of AKR1C1 by brusatol resulted in decreased progesterone catabolism and maintained potent progesterone to inhibit endometrial cancer growth. This inhibition pattern has also been found in the established xenograft mouse and organoid models. Aberrant overexpression of AKR1C1 was found in paired endometrial hyperplasia and cancer samples from the same individuals with progestin resistance, whereas attenuated or loss of AKR1C1 was observed in post-treatment samples with well progestin response as compared with paired pre-treatment tissues. Our findings suggest that AKR1C1 expression pattern may serve as an important biomarker of progestin resistance in endometrial cancer.
Collapse
Affiliation(s)
- Meiyan Hu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
- Department of Obstetrics and Gynecology, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, 201900, China
| | - Di Sun
- Center for Reproductive Medicine and Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Jing Yu
- Department of Pathology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Yue Fu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Zuoshu Qin
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Baozhu Huang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Qiuju Zhang
- Department of Obstetrics and Gynecology, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, 201900, China
| | - Xiong Chen
- Department of Obstetrics and Gynecology, Zhongshan Hospital Wusong Branch, Fudan University, Shanghai, 201900, China
| | - Youheng Wei
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, China
| | - Huiting Zhu
- Department of Pathology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Yue Wang
- Department of Obstetrics and Gynecology, Henan Province People's Hospital, Zhengzhou, China
| | - Youji Feng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Wenxin Zheng
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Hong Liao
- Department of Lab Medicine, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 200040, China
| | - Jingjie Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China.
| | - Sufang Wu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China.
| | - Zhenbo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China.
| |
Collapse
|
21
|
Co-expression of a PD-L1-specific chimeric switch receptor augments the efficacy and persistence of CAR T cells via the CD70-CD27 axis. Nat Commun 2022; 13:6051. [PMID: 36229619 PMCID: PMC9561169 DOI: 10.1038/s41467-022-33793-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/29/2022] [Indexed: 12/24/2022] Open
Abstract
Co-expression of chimeric switch receptors (CSRs) specific for PD-L1 improves the antitumor effects of chimeric antigen receptor (CAR) T cells. However, the effects of trans-recognition between CSRs and PD-L1 expressed by activated CAR T cells remain unclear. Here, we design a CSR specific for PD-L1 (CARP), containing the transmembrane and cytoplasmic signaling domains of CD28 but not the CD3 ζ chain. We show that CARP T cells enhance the antitumor activity of anti-mesothelin CAR (CARMz) T cells in vitro and in vivo. In addition, confocal microscopy indicates that PD-L1 molecules on CARMz T cells accumulate at cell-cell contacts with CARP T cells. Using single-cell RNA-sequencing analysis, we reveal that CARP T cells promote CARMz T cells differentiation into central memory-like T cells, upregulate genes related to Th1 cells, and downregulate Th2-associated cytokines through the CD70-CD27 axis. Moreover, these effects are not restricted to PD-L1, as CAR19 T cells expressing anti-CD19 CSR exhibit similar effects on anti-PSCA CAR T cells with truncated CD19 expression. These findings suggest that target trans-recognition by CSRs on CAR T cells may improve the efficacy and persistence of CAR T cells via the CD70-CD27 axis.
Collapse
|
22
|
Crosstalk between Extracellular Matrix Stiffness and ROS Drives Endometrial Repair via the HIF-1α/YAP Axis during Menstruation. Cells 2022; 11:cells11193162. [PMID: 36231126 PMCID: PMC9562179 DOI: 10.3390/cells11193162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
Although the menstrual cycle driven by sex steroid hormones is an uncomplicated physiological process, it is important for female health, fertility and regenerative biology. However, our understanding of this unique type of tissue homeostasis remains unclear. Here, we examined the biological effects of mechanical force by evaluating the changing trend of extracellular matrix (ECM) stiffness, and the results suggested that ECM stiffness was reduced and that breaking of mechanotransduction delayed endometrium repair in a mouse model of simulated menses. We constructed an ECM stiffness interference model in vitro to explain the mechanical force conduction mechanism during endometrial regeneration. We discovered that ECM stiffness increased the expression and nuclear transfer of YAP, which improved the creation of a microenvironment, in a manner that induced proliferation and angiogenesis for endometrial repair by activating YAP. In addition, we observed that physiological endometrial hypoxia occurs during the menstrual cycle and that the expression of HIF-1α was increased. Mechanistically, in addition to the classical F-actin/YAP pathway, we also found that the ROS/HIF-1α/YAP axis was involved in the transmission of mechanical signals. This study provides novel insights into the essential menstrual cycle and presents an effective, nonhormonal treatment for menstrual disorders.
Collapse
|
23
|
Vannuccini S, Jain V, Critchley H, Petraglia F. From menarche to menopause, heavy menstrual bleeding is the underrated compass in reproductive health. Fertil Steril 2022; 118:625-636. [PMID: 36075746 DOI: 10.1016/j.fertnstert.2022.07.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/14/2022] [Accepted: 07/19/2022] [Indexed: 01/13/2023]
Abstract
Menstruation is defined as monthly uterine bleeding, regarded as a sign of reproductive health. When characterized by excessive bleeding (heavy menstrual bleeding [HMB]), it may act as a useful clinical marker for diagnosis of reproductive diseases. Endometrial and myometrial mechanisms underlying abnormal uterine bleeding (AUB), which includes HMB, have hormonal, cellular, and molecular aspects. Structural and nonstructural causes of AUB, presenting with HMB as the major symptom, result in iron depletion and consequent anemia. Heavy menstrual bleeding can be considered as a single entity to identify the possible underlying causes, which may be different to some extent to those of AUB, as a whole. Furthermore, the difficulties in defining HMB through objective methods do not allow the current epidemiological scenario on the prevalence of the symptom among reproductive-age women to be outlined. Moreover, the introduction of new diagnostic methods, including imaging technologies, entails a revision of the available figures on HMB in different age groups from menarche to menopause. In addition, a proper diagnostic algorithm for HMB should be implemented to adapt recommendations for clinical investigation when HMB is present.
Collapse
Affiliation(s)
- Silvia Vannuccini
- Obstetrics and Gynecology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Careggi University Hospital, Florence, Italy
| | - Varsha Jain
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, Scotland
| | - Hilary Critchley
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, Scotland
| | - Felice Petraglia
- Obstetrics and Gynecology, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Careggi University Hospital, Florence, Italy.
| |
Collapse
|
24
|
Dong R, Ma S, Zhao X, Wang B, Roy M, Yao L, Xia T, Liu Y. Recent progress of Bioinspired Hydrogel-based delivery system for endometrial repair. Front Bioeng Biotechnol 2022; 10:1013217. [PMID: 36159661 PMCID: PMC9503822 DOI: 10.3389/fbioe.2022.1013217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022] Open
Abstract
Endometrial injury is the main fact leading to infertility. Current treatments of endometrial injury present many problems, such as unable to achieve desired effects due to low retention and the inherent potential risk of injury. Besides, it is important to the development of bioinspired material that can mimic the natural tissue and possess native tissue topography. Hydrogel is a kind of bioinspired superhydrophilic materials with unique characteristics, such as excellent biocompatibility, biodegradability, porosity, swelling, and cross-linkage. These unique physiochemical properties of bioinspired hydrogels enable their promising application as novel delivery platform and alternative therapies for endometrial injury. In this mini review, we summarize the recent advances in bioinispred hydrogel-based delivery system for endometrial repair, including as a post-operative physical barrier and therapeutic delivery system. In addition, present status, limitations, and future perspectives are also discussed.
Collapse
Affiliation(s)
- Rong Dong
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Saihua Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xiaoli Zhao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Baojuan Wang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Mridul Roy
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Lu Yao
- Hemay Zhihui Science and Technology Co. Ltd, Tianjin, China
| | - Tian Xia
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Yanting Liu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| |
Collapse
|
25
|
Park SR, Kim SK, Kim SR, Yu WJ, Lee SJ, Lee HY. Effects of smoking on the tissue regeneration-associated functions of human endometrial stem cells via a novel target gene SERPINB2. Stem Cell Res Ther 2022; 13:404. [PMID: 35932085 PMCID: PMC9356492 DOI: 10.1186/s13287-022-03061-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/19/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Smokers directly inhale mainstream cigarette smoke, which contains numerous known and potential toxic substances, and thus, smoking is expected to have broad harmful effects that cause tissue injury and dysfunction. Interestingly, many studies have suggested that the recent decline in female fertility and increased rate of spontaneous abortion could be associated with increased smoking rates. Indeed, women that smoked for 10 years or more were reported to have a ~ 20% higher infertility rate than women that had never smoked. However, the reasons for the underlying harmful aspects of smoking on female fertility remain a matter of debate. Importantly, a previous study revealed that resident endometrial stem cell deficiency significantly limits the cyclic regeneration potential of endometrium, which, in turn, decreases successful pregnancy outcomes. In this context, we postulated that exposure to mainstream cigarette smoke extracts might decrease female fertility by inhibiting the functions of resident endometrial stem cells. METHODS We investigated whether cigarette mainstream smoke exposure directly inhibits various tissue regeneration-associated functions of endometrial stem cells, such as self-renewal, migration, pluripotency, and differentiation capacity in vitro. Next, we determined whether SERPINB2 mediates cigarette smoke-induced suppressive effects on various tissue regeneration-associated functions by depleting SERPINB2 expression with specific shRNA targeting SERPINB2. Mice were injected intraperitoneally with low (0.5 mg/kg) or high (1 mg/kg) doses of cigarette smoke extract (10 times for two weeks), and endometrial stem cells were then isolated from mice uterine tissues. RESULTS We found that exposure to cigarette smoke extracts remarkably suppressed various tissue regeneration-associated functions of endometrial stem cells, such as self-renewal, migration, multilineage differentiation ability, and pluripotency in vitro and in vivo by activating the SERPINB2 gene. Indeed, cigarette smoke-induced inhibitory effects on various endometrial stem cell functions were significantly abolished by SERPINB2 knockdown. CONCLUSIONS These findings provide valuable information on the harmful effects of cigarette smoking on resident endometrial stem cells and hopefully will facilitate the developments of promising therapeutic strategies for subfertile or infertile women that smoke cigarettes.
Collapse
Affiliation(s)
- Se-Ra Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 406-840, Republic of Korea
| | - Seong-Kwan Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 406-840, Republic of Korea
| | - Soo-Rim Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, 21999, Republic of Korea.,Department of Molecular Medicine, School of Medicine, Gachon University, Incheon, 406-840, Republic of Korea
| | - Wook-Joon Yu
- Developmental and Reproductivoxicology Research Group, Korea Institute of Toxicology, Deajeon, 34114, Republic of Korea
| | - Seung-Jin Lee
- Developmental and Reproductivoxicology Research Group, Korea Institute of Toxicology, Deajeon, 34114, Republic of Korea
| | - Hwa-Yong Lee
- Division of Science Education, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
26
|
Endometrial stem/progenitor cells: Properties, origins, and functions. Genes Dis 2022. [DOI: 10.1016/j.gendis.2022.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
27
|
GCN5 participates in KLF4-VEGFA feedback to promote endometrial angiogenesis. iScience 2022; 25:104509. [PMID: 35733790 PMCID: PMC9207667 DOI: 10.1016/j.isci.2022.104509] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/01/2022] [Accepted: 05/27/2022] [Indexed: 11/22/2022] Open
Abstract
Endometrial angiogenesis is necessary for good endometrial receptivity. Krüppel-like factor 4 (KLF4) is a transcription factor that is essential for regulating angiogenesis. Here we found that vascular endothelial growth factor A (VEGFA) can form a positive feedback loop with KLF4 to promote the proliferation and migration of human endometrial microvascular endothelial cells (HEMECs) and inhibit cell apoptosis. General control non-derepressible 5 (GCN5) is also time-dependent on VEGFA and participates in the KLF4-VEGFA loop. In addition, we found that GCN5 is a succinyltransferase that modulates the succinylation of histones and nonhistones. GCN5 interacts with KLF4 and is recruited to the KLF4-binding site of the VEGFA promoter to succinylate H3K79, which initiates gene transcription epigenetically. For nonhistones, GCN5 succinylates KLF4 that is activated by ERK signaling in HEMECs treated with VEGFA to increase its transcription activity. These results demonstrate KLF4-VEGFA positive feedback loop is regulated by epigenetics, which contributes to endometrial angiogenesis. KLF4 mediates VEGFA-induced endometrial angiogenesis VEGFA increases the interaction between KLF4 and GCN5 VEGFA promotes H3K79 succinylation by upregulating KLF4 and GCN5 VEGFA succinylates KLF4 and promotes interaction of KLF4 and GCN5 via ERK pathway
Collapse
|
28
|
Louwen F, Kreis NN, Ritter A, Friemel A, Solbach C, Yuan J. BCL6, a key oncogene, in the placenta, pre-eclampsia and endometriosis. Hum Reprod Update 2022; 28:890-909. [PMID: 35640966 PMCID: PMC9629482 DOI: 10.1093/humupd/dmac027] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/02/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The key oncogene B-cell lymphoma 6 (BCL6) drives malignant progression by promoting proliferation, overriding DNA damage checkpoints and blocking cell terminal differentiation. However, its functions in the placenta and the endometrium remain to be defined. OBJECTIVE AND RATIONALE Recent studies provide evidence that BCL6 may play various roles in the human placenta and the endometrium. Deregulated BCL6 might be related to the pathogenesis of pre-eclampsia (PE) as well as endometriosis. In this narrative review, we aimed to summarize the current knowledge regarding the pathophysiological role of BCL6 in these two reproductive organs, discuss related molecular mechanisms, and underline associated research perspectives. SEARCH METHODS We conducted a comprehensive literature search using PubMed for human, animal and cellular studies published until October 2021 in the following areas: BCL6 in the placenta, in PE and in endometriosis, in combination with its functions in proliferation, fusion, migration, invasion, differentiation, stem/progenitor cell maintenance and lineage commitment. OUTCOMES The data demonstrate that BCL6 is important in cell proliferation, survival, differentiation, migration and invasion of trophoblastic cells. BCL6 may have critical roles in stem/progenitor cell survival and differentiation in the placenta and the endometrium. BCL6 is aberrantly upregulated in pre-eclamptic placentas and endometriotic lesions through various mechanisms, including changes in gene transcription and mRNA translation as well as post-transcriptional/translational modifications. Importantly, increased endometrial BCL6 is considered to be a non-invasive diagnostic marker for endometriosis and a predictor for poor outcomes of IVF. These data highlight that BCL6 is crucial for placental development and endometrium homeostasis, and its upregulation is associated with the pathogenesis of PE, endometriosis and infertility. WIDER IMPLICATIONS The lesson learned from studies of the key oncogene BCL6 reinforces the notion that numerous signaling pathways and regulators are shared by tumors and reproductive organs. Their alteration may promote the progression of malignancies as well as the development of gestational and reproductive disorders.
Collapse
Affiliation(s)
- Frank Louwen
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Nina-Naomi Kreis
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Andreas Ritter
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Alexandra Friemel
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Christine Solbach
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Juping Yuan
- Division of Obstetrics and Prenatal Medicine, Department of Gynecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| |
Collapse
|
29
|
Zhang L, Long W, Xu W, Chen X, Zhao X, Wu B. Digital Cell Atlas of Mouse Uterus: From Regenerative Stage to Maturational Stage. Front Genet 2022; 13:847646. [PMID: 35669188 PMCID: PMC9163836 DOI: 10.3389/fgene.2022.847646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/03/2022] [Indexed: 11/23/2022] Open
Abstract
Endometrium undergoes repeated repair and regeneration during the menstrual cycle. Previous attempts using gene expression data to define the menstrual cycle failed to come to an agreement. Here we used single-cell RNA sequencing data of C57BL/6J mice uteri to construct a novel integrated cell atlas of mice uteri from the regenerative endometrium to the maturational endometrium at the single-cell level, providing a more accurate cytological-based elucidation for the changes that occurred in the endometrium during the estrus cycle. Based on the expression levels of proliferating cell nuclear antigen, differentially expressed genes, and gene ontology terms, we delineated in detail the transitions of epithelial cells, stromal cells, and immune cells that happened during the estrus cycle. The transcription factors that shaped the differentiation of the mononuclear phagocyte system had been proposed, being Mafb, Irf7, and Nr4a1. The amounts and functions of immune cells varied sharply in two stages, especially NK cells and macrophages. We also found putative uterus tissue-resident macrophages and identified potential endometrial mesenchymal stem cells (high expression of Cd34, Pdgfrb, Aldh1a2) in vivo. The cell atlas of mice uteri presented here would improve our understanding of the transitions that occurred in the endometrium from the regenerative endometrium to the maturational endometrium. With the assistance of a normal cell atlas as a reference, we may identify morphologically unaffected abnormalities in future clinical practice. Cautions would be needed when adopting our conclusions, for the limited number of mice that participated in this study may affect the strength of our conclusions.
Collapse
Affiliation(s)
- Leyi Zhang
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Institute (Key Laboratory of Cancer Prevention & Intervention, National Ministry of Education), Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Breast Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenying Long
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Wanwan Xu
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Xiuying Chen
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Xiaofeng Zhao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Bingbing Wu
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- *Correspondence: Bingbing Wu,
| |
Collapse
|
30
|
Cousins FL, Filby CE, Gargett CE. Endometrial Stem/Progenitor Cells–Their Role in Endometrial Repair and Regeneration. FRONTIERS IN REPRODUCTIVE HEALTH 2022; 3:811537. [PMID: 36304009 PMCID: PMC9580754 DOI: 10.3389/frph.2021.811537] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022] Open
Abstract
The human endometrium is a remarkable tissue, undergoing ~450 cycles of proliferation, differentiation, shedding (menstruation), repair, and regeneration over a woman's reproductive lifespan. Post-menstrual repair is an extremely rapid and scar-free process, with re-epithelialization of the luminal epithelium completed within 48 h of initiation of shedding. Following menstruation, the functionalis grows from the residual basalis layer during the proliferative phase under the influence of rising circulating estrogen levels. The regenerative capacity of the endometrium is attributed to stem/progenitor cells which reside in both the epithelial and stromal cell compartments of the basalis layer. Finding a definitive marker for endometrial epithelial progenitors (eEPCs) has proven difficult. A number of different markers have been suggested as putative progenitor markers including, N-cadherin, SSEA-1, AXIN2, SOX-9 and ALDH1A1, some of which show functional stem cell activity in in vitro assays. Each marker has a unique location(s) in the glandular epithelium, which has led to the suggestion that a differentiation hierarchy exists, from the base of epithelial glands in the basalis to the luminal epithelium lining the functionalis, where epithelial cells express different combinations of markers as they differentiate and move up the gland into the functionalis away from the basalis niche. Perivascular endometrial mesenchymal stem cells (eMSCs) can be identified by co-expression of PDGFRβ and CD146 or by a single marker, SUSD2. This review will detail the known endometrial stem/progenitor markers; their identity, location and known interactions and hierarchy across the menstrual cycle, in particular post-menstrual repair and estrogen-driven regeneration, as well as their possible contributions to menstruation-related disorders such as endometriosis and regeneration-related disorder Asherman's syndrome. We will also highlight new techniques that allow for a greater understanding of stem/progenitor cells' role in repair and regeneration, including 3D organoids, 3D slice cultures and gene sequencing at the single cell level. Since mouse models are commonly used to study menstruation, repair and regeneration we will also detail the mouse stem/progenitor markers that have been investigated in vivo.
Collapse
Affiliation(s)
- Fiona L. Cousins
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynecology, Monash University, Clayton, VIC, Australia
- *Correspondence: Fiona L. Cousins
| | - Caitlin E. Filby
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynecology, Monash University, Clayton, VIC, Australia
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Obstetrics and Gynecology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
31
|
Mao M, Cheng Y, Yang J, Chen Y, Xu L, Zhang X, Li Z, Chen C, Ju S, Zhou J, Wang L. Multifaced roles of PLAC8 in cancer. Biomark Res 2021; 9:73. [PMID: 34627411 PMCID: PMC8501656 DOI: 10.1186/s40364-021-00329-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/20/2021] [Indexed: 12/21/2022] Open
Abstract
The role of PLAC8 in tumorigenesis has been gradually elucidated with the development of research. Although there are common molecular mechanisms that enforce cell growth, the impact of PLAC8 is varied and can, in some instances, have opposite effects on tumorigenesis. To systematically understand the role of PLAC8 in tumors, the molecular functions of PLAC8 in cancer will be discussed by focusing on how PLAC8 impacts tumorigenesis when it arises within tumor cells and how these roles can change in different stages of cancer progression with the ultimate goal of suppressing PLAC8-relevant cancer behavior and related pathologies. In addition, we highlight the diversity of PLAC8 in different tumors and its functional output beyond cancer cell growth. The comprehension of PLAC8's molecular function might provide new target and lead to the development of novel anticancer therapies.
Collapse
Affiliation(s)
- Misha Mao
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, 310000, Hangzhou, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Zhejiang, 310000, Hangzhou, China
| | - Yifan Cheng
- Department of Gastrointestinal Surgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, 318000, People's Republic of China
| | - Jingjing Yang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, 310000, Hangzhou, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Zhejiang, 310000, Hangzhou, China
| | - Yongxia Chen
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, 310000, Hangzhou, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Zhejiang, 310000, Hangzhou, China
| | - Ling Xu
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, 310000, Hangzhou, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Zhejiang, 310000, Hangzhou, China
| | - Xun Zhang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, 310000, Hangzhou, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Zhejiang, 310000, Hangzhou, China
| | - Zhaoqing Li
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, 310000, Hangzhou, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Zhejiang, 310000, Hangzhou, China
| | - Cong Chen
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, 310000, Hangzhou, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Zhejiang, 310000, Hangzhou, China
| | - Siwei Ju
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, 310000, Hangzhou, China.,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Zhejiang, 310000, Hangzhou, China
| | - Jichun Zhou
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, 310000, Hangzhou, China. .,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Zhejiang, 310000, Hangzhou, China.
| | - Linbo Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Zhejiang, 310000, Hangzhou, China. .,Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Zhejiang, 310000, Hangzhou, China.
| |
Collapse
|
32
|
Advancements in endometrial epithelial stem cell research. SCIENCE CHINA-LIFE SCIENCES 2021; 65:215-218. [PMID: 34586574 DOI: 10.1007/s11427-021-1988-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
|
33
|
Mamillapalli R, Mutlu L, Taylor HS. Characterization of Bone Marrow Progenitor Cell Uterine Engraftment and Transdifferentiation. Reprod Sci 2021; 29:2382-2390. [PMID: 34515983 DOI: 10.1007/s43032-021-00738-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 09/07/2021] [Indexed: 10/20/2022]
Abstract
Regeneration of uterine tissue is an important physiological process that allows for maintenance of fertility after menstruation or pregnancy. Stem cells, especially bone marrow-derived progenitors, play a crucial role in this regeneration. Here, we describe the conversion of DsRed-labeled bone marrow-derived stem cells (BMDSCs) into specific uterine cell types with both differentiated and stem cell properties in a murine model. Irradiated recipient mice underwent bone marrow transplant with DsRed-expressing BMDSCs and were analyzed for engraftment and differentiation of BMDSCs in the uterus after 2, 6, and 16 weeks. Microarray and qRT-PCR analysis of bone marrow-derived cells obtained from the uterus identified upregulation of markers indicating a contribution to the population of stromal, epithelial, endothelial, and muscle cells, followed by a late expansion of epithelial cells. Other engrafted BMDSCs in the uterus were characterized by the continued expression of specific stem cell markers such as Sca1, CD44, CD146, and CD133, indicating the some BMDSCs remain as progenitor cells. BMDSCs established in recipient mice by the 16th week were sorted by flow cytometry using DsRed and progenitor cell surface markers. In vitro cell culture studies showed that single sorted cells had clonogenic properties. These results suggest that engrafted BMDSCs in the uterus had both a stem cell component and were able to differentiate into several differentiated cell types. The pool of progenitor cells likely continues to supply differentiated uterine cells in the process of uterine repair and remodeling.
Collapse
Affiliation(s)
- Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
| | - Levent Mutlu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| |
Collapse
|
34
|
Lv Q, Wang L, Luo X, Chen X. Adult stem cells in endometrial regeneration: Molecular insights and clinical applications. Mol Reprod Dev 2021; 88:379-394. [PMID: 34014590 PMCID: PMC8362170 DOI: 10.1002/mrd.23476] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 03/23/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023]
Abstract
Endometrial damage is an important cause of female reproductive problems, manifested as menstrual abnormalities, infertility, recurrent pregnancy loss, and other complications. These conditions are collectively termed "Asherman syndrome" (AS) and are typically associated with recurrent induced pregnancy terminations, repeated diagnostic curettage and intrauterine infections. Cancer treatment also has unexpected detrimental side effects on endometrial function in survivors independently of ovarian effects. Endometrial stem cells act in the regeneration of the endometrium and in repair through direct differentiation or paracrine effects. Nonendometrial adult stem cells, such as bone marrow-derived mesenchymal stem cells and umbilical cord-derived mesenchymal stem cells, with autologous and allogenic applications, can also repair injured endometrial tissue in animal models of AS and in human studies. However, there remains a lack of research on the repair of the damaged endometrium after the reversal of tumors, especially endometrial cancers. Here, we review the biological mechanisms of endometrial regeneration, and research progress and challenges for adult stem cell therapy for damaged endometrium, and discuss the potential applications of their use for endometrial repair after cancer remission, especially in endometrial cancers. Successful application of such cells will improve reproductive parameters in patients with AS or cancer. Significance: The endometrium is the fertile ground for embryos, but damage to the endometrium will greatly impair female fertility. Adult stem cells combined with tissue engineering scaffold materials or not have made great progress in repairing the injured endometrium due to benign lesions. However, due to the lack of research on the repair of the damaged endometrium caused by malignant tumors or tumor therapies, the safety and effectiveness of such stem cell-based therapies need to be further explored. This review focuses on the molecular insights and clinical application potential of adult stem cells in endometrial regeneration and discusses the possible challenges or difficulties that need to be overcome in stem cell-based therapies for tumor survivors. The development of adult stem cell-related new programs will help repair damaged endometrium safely and effectively and meet fertility needs in tumor survivors.
Collapse
Affiliation(s)
- Qiaoying Lv
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| | - Lulu Wang
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| | - Xuezhen Luo
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| | - Xiaojun Chen
- Department of GynecologyObstetrics and Gynecology Hospital of Fudan UniversityShanghaiChina
| |
Collapse
|
35
|
Abuwala N, Tal R. Endometrial stem cells: origin, biological function, and therapeutic applications for reproductive disorders. Curr Opin Obstet Gynecol 2021; 33:232-240. [PMID: 33896919 PMCID: PMC9313610 DOI: 10.1097/gco.0000000000000702] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Endometrial stem cells (ESCs) are multipotent cells that are thought to originate locally in the endometrium as well as in the bone marrow (BM). They have remarkable plasticity and hold promise as an autologous source for regenerative medicine. This review focuses on recent studies that have advanced our understanding of the biology and function of ESCs and BM-derived stem cells (BMDSCs) as related to physiological reproductive processes and pathologies. Moreover, it reviews recent data on potential therapeutic applications of stem cells to endometrial disorders that lead to reproductive failure. RECENT FINDINGS Growing evidence from basic and preclinical studies suggests that ESCs participate in endometrial tissue regeneration and repair. Recent evidence also suggests that ESCs and BMDSCs play important roles in physiological reproductive functions including decidualization, implantation, pregnancy maintenance, and postpartum uterine remodeling. Initial preclinical and clinical studies with ESCs and BMDSCs suggest they have the potential to provide new therapies for various endometrial disorders associated with reproductive failure. SUMMARY Uterine ESCs and BMDSCs appear to play an important biological role in reproductive success and failure, and have the potential to become treatment targets for reproductive diseases including recurrent implantation failure, thin endometrium, Asherman, and recurrent pregnancy loss.
Collapse
Affiliation(s)
- Nafeesa Abuwala
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Reshef Tal
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
36
|
Li S, Ding L. Endometrial Perivascular Progenitor Cells and Uterus Regeneration. J Pers Med 2021; 11:477. [PMID: 34071743 PMCID: PMC8230145 DOI: 10.3390/jpm11060477] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/23/2022] Open
Abstract
Ovarian steroid-regulated cyclical regeneration of the endometrium is crucial for endometrial receptivity and embryo implantation, and it is dependent on the dynamic remodeling of the endometrial vasculature. Perivascular cells, including pericytes surrounding capillaries and microvessels and adventitial cells located in the outermost layer of large vessels, show properties of mesenchymal stem cells, and they are thus promising candidates for uterine regeneration. In this review, we discuss the structure and functions of the endometrial blood vasculature and their roles in endometrial regeneration, the main biomarkers and characteristics of perivascular cells in the endometrium, and stem cell-based angiogenetic therapy for Asherman's syndrome.
Collapse
Affiliation(s)
- Shiyuan Li
- Center for Reproductive Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China;
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, China
- Center for Clinical Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Lijun Ding
- Center for Reproductive Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China;
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing 210008, China
- Center for Clinical Stem Cell Research, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| |
Collapse
|
37
|
Ghosh A, Syed SM, Kumar M, Carpenter TJ, Teixeira JM, Houairia N, Negi S, Tanwar PS. In Vivo Cell Fate Tracing Provides No Evidence for Mesenchymal to Epithelial Transition in Adult Fallopian Tube and Uterus. Cell Rep 2021; 31:107631. [PMID: 32402291 PMCID: PMC8094408 DOI: 10.1016/j.celrep.2020.107631] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/10/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023] Open
Abstract
The mesenchymal to epithelial transition (MET) is thought to be involved in the maintenance, repair, and carcinogenesis of the fallopian tube (oviduct) and uterine epithelium. However, conclusive evidence for the conversion of mesenchymal cells to epithelial cells in these organs is lacking. Using embryonal cell lineage tracing with reporters driven by mesenchymal cell marker genes of the female reproductive tract (AMHR2, CSPG4, and PDGFRβ), we show that these reporters are also expressed by some oviductal and uterine epithelial cells at birth. These mesenchymal reporter-positive epithelial cells are maintained in adult mice across multiple pregnancies, respond to ovarian hormones, and form organoids. However, no labeled epithelial cells are present in any oviductal or uterine epithelia when mesenchymal cell labeling was induced in adult mice. Organoids developed from mice labeled in adulthood were also negative for mesenchymal reporters. Collectively, our work found no definitive evidence of MET in the adult fallopian tube and uterine epithelium. Mesenchymal to epithelial transition (MET) is postulated to be involved in the maintenance and regeneration of the epithelium of female reproductive organs. Here, Ghosh et al. report no definitive evidence of MET in the adult epithelium of oviduct and uterus using in vivo cell lineage tracing and organoids.
Collapse
Affiliation(s)
- Arnab Ghosh
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Shafiq M Syed
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Manish Kumar
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Tyler J Carpenter
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Jose M Teixeira
- Department of Obstetrics, Gynecology and Reproductive Biology, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Nathaniel Houairia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Sumedha Negi
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Pradeep S Tanwar
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia; Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia.
| |
Collapse
|
38
|
Cheung VC, Peng CY, Marinić M, Sakabe NJ, Aneas I, Lynch VJ, Ober C, Nobrega MA, Kessler JA. Pluripotent stem cell-derived endometrial stromal fibroblasts in a cyclic, hormone-responsive, coculture model of human decidua. Cell Rep 2021; 35:109138. [PMID: 34010658 DOI: 10.1016/j.celrep.2021.109138] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/22/2021] [Accepted: 04/26/2021] [Indexed: 12/16/2022] Open
Abstract
Various human diseases and pregnancy-related disorders reflect endometrial dysfunction. However, rodent models do not share fundamental biological processes with the human endometrium, such as spontaneous decidualization, and no existing human cell cultures recapitulate the cyclic interactions between endometrial stromal and epithelial compartments necessary for decidualization and implantation. Here we report a protocol differentiating human pluripotent stem cells into endometrial stromal fibroblasts (PSC-ESFs) that are highly pure and able to decidualize. Coculture of PSC-ESFs with placenta-derived endometrial epithelial cells generated organoids used to examine stromal-epithelial interactions. Cocultures exhibited specific endometrial markers in the appropriate compartments, organization with cell polarity, and hormone responsiveness of both cell types. Furthermore, cocultures recapitulate a central feature of the human decidua by cyclically responding to hormone withdrawal followed by hormone retreatment. This advance enables mechanistic studies of the cyclic responses that characterize the human endometrium.
Collapse
Affiliation(s)
| | - Chian-Yu Peng
- Department of Neurology, Northwestern University, Chicago, IL 60611, USA
| | - Mirna Marinić
- Department of Human Genetics, University of Chicago, Chicago, IL, USA; Current address: Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL, USA
| | - Noboru J Sakabe
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Ivy Aneas
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Vincent J Lynch
- Department of Human Genetics, University of Chicago, Chicago, IL, USA; Department of Biological Sciences, University of Buffalo, Buffalo, NY, USA
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Marcelo A Nobrega
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - John A Kessler
- Department of Neurology, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
39
|
Jiang X, Li X, Fei X, Shen J, Chen J, Guo M, Li Y. Endometrial membrane organoids from human embryonic stem cell combined with the 3D Matrigel for endometrium regeneration in asherman syndrome. Bioact Mater 2021; 6:3935-3946. [PMID: 33937593 PMCID: PMC8079828 DOI: 10.1016/j.bioactmat.2021.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 03/29/2021] [Accepted: 04/04/2021] [Indexed: 12/18/2022] Open
Abstract
Asherman's syndrome (AS), a leading cause of uterine infertility worldwide, is characterized by scarring of the uterine surfaces lacking endometrial epithelial cells, which prevents endometrial regeneration. Current research on cell therapy for AS focuses on mesenchymal and adult stem cells from the endometrium. However, insufficient number, lack of purity, and rapid senescence of endometrial epithelial progenitor cells (EEPCs) during experimental processes restrict their use in cell therapies. In this study, we induced human embryonic stem cells-9 (H9-ESC) into EEPCs by optimizing the induction factors from the definitive endoderm. EEPCs, which act as endometrial epithelial cells, accompanied by human endometrial stromal cells provide a niche environment for the development of endometrial membrane organoids (EMOs) in an in vitro 3D culture model. To investigate the function of EMOs, we transplanted tissue-engineered constructs with EMOs into an in vivo rat AS model. The implantation of EMOs into the damaged endometrium facilitates endometrial regeneration and angiogenesis. Implanting EMOs developed from human embryonic stem cells into the endometrium might prove useful for "endometrial re-engineering" in the treatment of Asherman's syndrome.
Collapse
Affiliation(s)
- Xiuxiu Jiang
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310006, China
| | - Xingmiao Li
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310006, China
| | - Xiangwei Fei
- Key Laboratory of Women′s Reproductive Health Research of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310006, China
| | - Jiajie Shen
- Key Laboratory of Women′s Reproductive Health Research of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310006, China
| | - Jianhua Chen
- Department of Pathology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310006, China
| | - Meijun Guo
- Department of Gynecology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310006, China
| | - Yangyang Li
- Key Laboratory of Women′s Reproductive Health Research of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, 310006, China
- Corresponding author.
| |
Collapse
|
40
|
Koo HS, Yoon MJ, Hong SH, Ahn J, Cha H, Lee D, Ko JE, Kwon H, Choi DH, Lee KA, Ko JJ, Kang YJ. CXCL12 enhances pregnancy outcome via improvement of endometrial receptivity in mice. Sci Rep 2021; 11:7397. [PMID: 33795831 PMCID: PMC8016928 DOI: 10.1038/s41598-021-86956-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 03/22/2021] [Indexed: 02/08/2023] Open
Abstract
Successful pregnancy inevitably depends on the implantation of a competent embryo into a receptive endometrium. Although many substances have been suggested to improve the rate of embryo implantation targeting enhancement of endometrial receptivity, currently there rarely are effective evidence-based treatments to prevent or cure this condition. Here we strongly suggest minimally-invasive intra-uterine administration of embryo-secreted chemokine CXCL12 as an effective therapeutic intervention. Chemokine CXCL12 derived from pre- and peri-implanting embryos significantly enhances the rates of embryo attachment and promoted endothelial vessel formation and sprouting in vitro. Consistently, intra-uterine CXCL12 administration in C57BL/6 mice improved endometrial receptivity showing increased integrin β3 and its ligand osteopontin, and induced endometrial angiogenesis displaying increased numbers of vessel formation near the lining of endometrial epithelial layer with higher CD31 and CD34 expression. Furthermore, intra-uterine CXCL12 application dramatically promoted the rates of embryo implantation with no morphologically retarded embryos. Thus, our present study provides a novel evidence that improved uterine endometrial receptivity and enhanced angiogenesis induced by embryo-derived chemokine CXCL12 may aid to develop a minimally-invasive therapeutic strategy for clinical treatment or supplement for the patients with repeated implantation failure with less risk.
Collapse
Affiliation(s)
- Hwa Seon Koo
- CHA Fertility Center Bundang, CHA University, Seongnam-si, Gyunggi-do, South Korea
| | - Min-Ji Yoon
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, Gyunggi-do, South Korea
| | - Seon-Hwa Hong
- CHA Fertility Center Bundang, CHA University, Seongnam-si, Gyunggi-do, South Korea
| | - Jungho Ahn
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, Gyunggi-do, South Korea
| | - Hwijae Cha
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, Gyunggi-do, South Korea
| | - Danbi Lee
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, Gyunggi-do, South Korea
| | - Ji-Eun Ko
- CHA Fertility Center Bundang, CHA University, Seongnam-si, Gyunggi-do, South Korea
| | - Hwang Kwon
- CHA Fertility Center Bundang, CHA University, Seongnam-si, Gyunggi-do, South Korea
| | - Dong Hee Choi
- CHA Fertility Center Bundang, CHA University, Seongnam-si, Gyunggi-do, South Korea
| | - Kyung-Ah Lee
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, Gyunggi-do, South Korea
| | - Jung-Jae Ko
- Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, Gyunggi-do, South Korea
| | - Youn-Jung Kang
- CHA Fertility Center Bundang, CHA University, Seongnam-si, Gyunggi-do, South Korea. .,Department of Biomedical Science, School of Life Science, CHA University, Seongnam-si, Gyunggi-do, South Korea. .,Department of Biochemistry, School of Medicine, CHA University, Seongnam-si, Gyunggi-do, South Korea.
| |
Collapse
|
41
|
Eremichev R, Kulebyakina M, Alexandrushkina N, Nimiritsky P, Basalova N, Grigorieva O, Egiazaryan M, Dyikanov D, Tkachuk V, Makarevich P. Scar-Free Healing of Endometrium: Tissue-Specific Program of Stromal Cells and Its Induction by Soluble Factors Produced After Damage. Front Cell Dev Biol 2021; 9:616893. [PMID: 33718358 PMCID: PMC7947248 DOI: 10.3389/fcell.2021.616893] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Besides certain exceptions, healing of most tissues in the human body occurs via formation of scar tissue, rather than restoration of lost structures. After extensive acute injuries, this phenomenon substantially limits the possibility of lost function recovery and, in case of chronic injury, it leads to pathological remodeling of organs affected. Managing outcomes of damaged tissue repair is one of the main objectives of regenerative medicine. The first priority for reaching it is comparative investigation of mechanisms responsible for complete restoration of damaged tissues and mechanisms of scarring. However, human body tissues that undergo complete scar-free healing are scarce. The endometrium is a unique mucous membrane in the human body that heals without scarring after various injuries, as well as during each menstrual cycle (i.e., up to 400 times during a woman's life). We hypothesized that absence of scarring during endometrial healing may be associated with tissue-specific features of its stromal cells (SCs) or their microenvironment, since SCs transform into myofibroblasts-the main effector link of scarring. We found that during healing of the endometrium, soluble factors are formed that inhibit the transition of SCs into myofibroblasts. Without influence of these factors, the SCs of the endometrium undergo transformation into myofibroblasts after transforming growth factor β1 (TGF-β1) treatment as well as the SCs from tissues that heal by scarring-skin or fat. However, unlike the latter, endometrial SCs organize extracellular matrix (ECM) in a specific way and are not prone to formation of bulky connective tissue structures. Thus, we may suggest that tissue-specific features of endometrial SCs along with effects of soluble factors secreted in utero during menstruation ensure scar-free healing of human endometrium.
Collapse
Affiliation(s)
- Roman Eremichev
- Medical Research and Education Center, Institute for Regenerative Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Maria Kulebyakina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Nataliya Alexandrushkina
- Medical Research and Education Center, Institute for Regenerative Medicine, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Peter Nimiritsky
- Medical Research and Education Center, Institute for Regenerative Medicine, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Nataliya Basalova
- Medical Research and Education Center, Institute for Regenerative Medicine, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Olga Grigorieva
- Medical Research and Education Center, Institute for Regenerative Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Mane Egiazaryan
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Daniyar Dyikanov
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Vsevolod Tkachuk
- Medical Research and Education Center, Institute for Regenerative Medicine, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,Laboratory of Molecular Endocrinology, National Medical Research Center of Cardiology, Moscow, Russia
| | - Pavel Makarevich
- Medical Research and Education Center, Institute for Regenerative Medicine, Lomonosov Moscow State University, Moscow, Russia.,Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
42
|
Endometriosis stromal cells induce bone marrow mesenchymal stem cell differentiation and PD-1 expression through paracrine signaling. Mol Cell Biochem 2021; 476:1717-1727. [PMID: 33428059 DOI: 10.1007/s11010-020-04012-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 12/02/2020] [Indexed: 12/28/2022]
Abstract
Endometriosis is an estrogen-dependent, inflammatory gynecological disorder characterized by the growth of endometrial cells in lesions outside the uterus. Bone marrow-derived cells (BMDCs) engraft lesions and increase lesion size. Do endometriosis cells regulate differentiation of engrafted BMDCs in the pathogenesis and growth of endometriosis? Here, we report endometriosis derived stromal cells promote the differentiation of BMDCs to stromal, epithelial and leukocyte cell fates through paracrine signaling. In-vitro studies demonstrated that both mRNA and protein levels of vimentin, cytokeratin and PD-1 were significantly increased in BMDCs cocultured with stromal cells from endometriosis (ENDO) patients compared to stromal cells from normal endometrium (CNTL). Increased expression of PD-1 has been reported in malignancy where it promotes T cell quiescence and immune tolerance. Increased PD-1 was also confirmed in-vivo where we showed that PD-1 expression was induced in BMDCs engrafted into endometriotic lesions in a murine model of endometriosis. AMD3100, an antagonist for CXCR4 receptor inhibited PD-1 expression in BMDCs suggesting that PD-1 induction requires CXCL12. These results suggest that endometriosis stimulated BMDC differentiation through paracrine signaling and increased T cell PD-1 expression. Increased PD-1 expression may be one mechanism by which endometriosis avoids immune surveillance.
Collapse
|
43
|
Zhu X, Yu F, Yan G, Hu Y, Sun H, Ding L. Human endometrial perivascular stem cells exhibit a limited potential to regenerate endometrium after xenotransplantation. Hum Reprod 2021; 36:145-159. [PMID: 33283858 DOI: 10.1093/humrep/deaa261] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/02/2020] [Indexed: 12/15/2022] Open
Abstract
STUDY QUESTION What are the localization, characteristics and potential for tissue regeneration of two perivascular stem cells, namely CD34+ adventitial cells and CD146+ pericytes, in human endometrium? SUMMARY ANSWER Human endometrial CD34+ adventitial cells (located in the outermost layer of blood vessels and mainly in the basal layer) and CD146+ pericytes showed mesenchymal stem cell (MSC) phenotypes in in vitro culture, but presented limited potential to regenerate endometrium. WHAT IS KNOWN ALREADY Periodic endometrial regeneration is considered to be maintained by MSCs. Blood vessel wall, regarded as stem cell niche, harbors a large reserve of progenitor cells that may be integral to the origin of MSCs. However, a lack of validated markers has hampered the isolation of putative endometrial MSCs. Currently, CD146+ pericytes and Sushi Domain Containing 2 (SUSD2) positive cells have been identified in the endometrial perivascular region as sharing MSCs characteristics. STUDY DESIGN, SIZE, DURATION The locations of adventitial cells and pericytes in the human endometrium were identified by immunofluorescence staining (n = 4). After CD34+CD146-CD45-CD56-CD144- adventitial cells and CD146+CD34-CD45-CD56-CD144- pericytes were isolated from the endometrium of normal women (n = 6) by fluorescence-activated cell sorting, their characteristics were investigated in culture. Adventitial cells and pericytes were induced to differentiate, respectively, into vascular endothelial-like cells or endometrial stromal-like cells in vitro, with their potential explored by in vivo xenotransplantation (n = 2 in each group) and eutopic transplantation (n = 2 in each group). PARTICIPANTS/MATERIALS, SETTING, METHODS CD34+ adventitial cells and CD146+ pericytes were cultured in the inducing medium to differentiate into endothelial-like cells in vitro, and then analyzed for CD31, von Willebrand factor immunofluorescent staining and tube formation. They were also cultured to differentiate into endometrial stromal cells in vitro, with the expression of vimentin and CD13 being detected by western blot before and after induction, and the expression of prolactin and insulin-like growth factor-binding protein 1 being determined as well. Single dispersed CD34+ adventitial cells and CD146+ pericytes were respectively transplanted under the kidney capsule of NOG mice to investigate their differentiation potential in vivo. A eutopic transplantation model was constructed by grafting recellularized uterine matrix loaded up with CM-Dil labeled adventitial cells or pericytes into the injury region of nude rat's uterus. MAIN RESULTS AND THE ROLE OF CHANCE CD34+ adventitial cells were mainly located at the outmost layer of endometrial large vessels, while CD146+ pericytes were found surrounding the inner endothelial cells of microvessels. A small proportion of CD34+ adventitial cells expressed SUSD2. The number of adventitial cells was ∼40 times higher than that of pericytes in the endometrium. Both adventitial cells and pericytes showed MSC phenotypes after in vitro culture. After in vitro induction into endometrial endothelial-like cells and stromal-like cells, adventitial cells showed higher plasticity than pericytes and a closer correlation with stromal-like cells. In the mouse xenotransplantation model, vimentin+ cells, CD31+ endothelial-like cells and CD146+ pericyte-like cells could be observed after adventitial cells were transplanted. CM-Dil-labeled adventitial cells or pericytes could survive in the immunocompromised nude rats after eutopic transplantation, and vimentin+ cells were detected. In addition, CM-Dil-labeled adventitial cells or pericytes did not express α-smooth muscle actin or E-cadherin after transplantation. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION CD34 was chosen as a novel marker to isolate adventitial cells from human endometrium according to previous literature. The association of endometrial CD34+ adventitial cells and SUSD2+ MSCs should be further investigated. WIDER IMPLICATIONS OF THE FINDINGS The decellularized uterine matrix model might be useful in endometrial stem cell therapy. STUDY FUNDING/COMPETING INTEREST(S) L.D. is supported by grants from National Key Research and Development Program of China (2018YFC1004700), Nature Science Foundation of China (81871128, 81571391) and Nanjing Medical Science Development Project (ZKX16042). H.S. is supported by a grant from Jiangsu Province Social Development Project (BE2018602). X.Z. was supported by grants from the Postgraduate Innovative Project of Jiangsu Province (KYCX19-1177). The authors declare no conflict of interest.
Collapse
Affiliation(s)
- Xinxin Zhu
- Center for Reproductive Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fei Yu
- Center for Experimental Animal, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Guijun Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yali Hu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Haixiang Sun
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Lijun Ding
- Center for Reproductive Medicine, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu, China
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- Clinical Center for Stem Cell Research, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- MRC Center for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
44
|
Lin Y, Dong S, Zhao W, Hu KL, Liu J, Wang S, Tu M, Du B, Zhang D. Application of Hydrogel-Based Delivery System in Endometrial Repair. ACS APPLIED BIO MATERIALS 2020; 3:7278-7290. [PMID: 35019471 DOI: 10.1021/acsabm.0c00971] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A receptive endometrium with proper thickness is essential for successful embryo implantation. However, endometrial injury caused by intrauterine procedures often leads to pathophysiological changes in its environment, resulting in subsequent female infertility. Among diverse treatment methods of endometrial injury, hydrogels are a class of hydrophilic three-dimensional polymeric network with biocompatibility as well as the capability of absorbing water and encapsulation, which have potential applications as a promising intrauterine controlled-release delivery system. This review summarizes recent advances in the approaches of endometrial repair and further focuses on the application of a hydrogel-based delivery system in endometrial repair, including its preparation, therapeutic loading considerations, clinical applications, as well as working mechanisms.
Collapse
Affiliation(s)
- Yifeng Lin
- Key Laboratory of Re/productive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, PR China
| | - Shunni Dong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, PR China
| | - Wei Zhao
- Key Laboratory of Women Reproductive Health of Zhejiang Province, and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, PR China
| | - Kai-Lun Hu
- Key Laboratory of Re/productive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, PR China
| | - Juan Liu
- Key Laboratory of Re/productive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, PR China
| | - Siwen Wang
- Key Laboratory of Re/productive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, PR China
| | - Mixue Tu
- Key Laboratory of Re/productive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, PR China
| | - Binyang Du
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science & Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, PR China
| | - Dan Zhang
- Key Laboratory of Re/productive Genetics (Ministry of Education) and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, PR China.,Key Laboratory of Women Reproductive Health of Zhejiang Province, and Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, PR China
| |
Collapse
|
45
|
Critchley HOD, Babayev E, Bulun SE, Clark S, Garcia-Grau I, Gregersen PK, Kilcoyne A, Kim JYJ, Lavender M, Marsh EE, Matteson KA, Maybin JA, Metz CN, Moreno I, Silk K, Sommer M, Simon C, Tariyal R, Taylor HS, Wagner GP, Griffith LG. Menstruation: science and society. Am J Obstet Gynecol 2020; 223:624-664. [PMID: 32707266 PMCID: PMC7661839 DOI: 10.1016/j.ajog.2020.06.004] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 05/13/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022]
Abstract
Women's health concerns are generally underrepresented in basic and translational research, but reproductive health in particular has been hampered by a lack of understanding of basic uterine and menstrual physiology. Menstrual health is an integral part of overall health because between menarche and menopause, most women menstruate. Yet for tens of millions of women around the world, menstruation regularly and often catastrophically disrupts their physical, mental, and social well-being. Enhancing our understanding of the underlying phenomena involved in menstruation, abnormal uterine bleeding, and other menstruation-related disorders will move us closer to the goal of personalized care. Furthermore, a deeper mechanistic understanding of menstruation-a fast, scarless healing process in healthy individuals-will likely yield insights into a myriad of other diseases involving regulation of vascular function locally and systemically. We also recognize that many women now delay pregnancy and that there is an increasing desire for fertility and uterine preservation. In September 2018, the Gynecologic Health and Disease Branch of the Eunice Kennedy Shriver National Institute of Child Health and Human Development convened a 2-day meeting, "Menstruation: Science and Society" with an aim to "identify gaps and opportunities in menstruation science and to raise awareness of the need for more research in this field." Experts in fields ranging from the evolutionary role of menstruation to basic endometrial biology (including omic analysis of the endometrium, stem cells and tissue engineering of the endometrium, endometrial microbiome, and abnormal uterine bleeding and fibroids) and translational medicine (imaging and sampling modalities, patient-focused analysis of menstrual disorders including abnormal uterine bleeding, smart technologies or applications and mobile health platforms) to societal challenges in health literacy and dissemination frameworks across different economic and cultural landscapes shared current state-of-the-art and future vision, incorporating the patient voice at the launch of the meeting. Here, we provide an enhanced meeting report with extensive up-to-date (as of submission) context, capturing the spectrum from how the basic processes of menstruation commence in response to progesterone withdrawal, through the role of tissue-resident and circulating stem and progenitor cells in monthly regeneration-and current gaps in knowledge on how dysregulation leads to abnormal uterine bleeding and other menstruation-related disorders such as adenomyosis, endometriosis, and fibroids-to the clinical challenges in diagnostics, treatment, and patient and societal education. We conclude with an overview of how the global agenda concerning menstruation, and specifically menstrual health and hygiene, are gaining momentum, ranging from increasing investment in addressing menstruation-related barriers facing girls in schools in low- to middle-income countries to the more recent "menstrual equity" and "period poverty" movements spreading across high-income countries.
Collapse
Affiliation(s)
- Hilary O D Critchley
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, United Kingdom.
| | - Elnur Babayev
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Serdar E Bulun
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | - Iolanda Garcia-Grau
- Igenomix Foundation-Instituto de Investigación Sanitaria Hospital Clínico, INCLIVA, Valencia, Spain; Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain
| | - Peter K Gregersen
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY
| | | | | | | | - Erica E Marsh
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI
| | - Kristen A Matteson
- Division of Research, Department of Obstetrics and Gynecology, Women and Infants Hospital, Warren Alpert Medical School of Brown University, Providence, RI
| | - Jacqueline A Maybin
- Medical Research Council Centre for Reproductive Health, The University of Edinburgh, United Kingdom
| | - Christine N Metz
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY
| | - Inmaculada Moreno
- Igenomix Foundation-Instituto de Investigación Sanitaria Hospital Clínico, INCLIVA, Valencia, Spain
| | - Kami Silk
- Department of Communication, University of Delaware, Newark, DE
| | - Marni Sommer
- Department of Sociomedical Sciences, Columbia University Mailman School of Public Health, New York, NY
| | - Carlos Simon
- Igenomix Foundation-Instituto de Investigación Sanitaria Hospital Clínico, INCLIVA, Valencia, Spain; Department of Pediatrics, Obstetrics and Gynecology, School of Medicine, University of Valencia, Valencia, Spain; Beth Israel Deaconess Medical Center, Harvard University, Boston, MA; Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX
| | | | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT
| | - Günter P Wagner
- Department of Ecology and Evolutionary Biology, Department of Obstetrics, Gynecology and Reproductive Sciences, Systems Biology Institute, Yale University, New Haven, CT; Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Linda G Griffith
- Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
46
|
Bozorgmehr M, Gurung S, Darzi S, Nikoo S, Kazemnejad S, Zarnani AH, Gargett CE. Endometrial and Menstrual Blood Mesenchymal Stem/Stromal Cells: Biological Properties and Clinical Application. Front Cell Dev Biol 2020; 8:497. [PMID: 32742977 PMCID: PMC7364758 DOI: 10.3389/fcell.2020.00497] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
A highly proliferative mesenchymal stem/stromal cell (MSC) population was recently discovered in the dynamic, cyclically regenerating human endometrium as clonogenic stromal cells that fulfilled the International Society for Cellular Therapy (ISCT) criteria. Specific surface markers enriching for clonogenic endometrial MSC (eMSC), CD140b and CD146 co-expression, and the single marker SUSD2, showed their perivascular identity in the endometrium, including the layer which sheds during menstruation. Indeed, cells with MSC properties have been identified in menstrual fluid and commonly termed menstrual blood stem/stromal cells (MenSC). MenSC are generally retrieved from menstrual fluid as plastic adherent cells, similar to bone marrow MSC (bmMSC). While eMSC and MenSC share several biological features with bmMSC, they also show some differences in immunophenotype, proliferation and differentiation capacities. Here we review the phenotype and functions of eMSC and MenSC, with a focus on recent studies. Similar to other MSC, eMSC and MenSC exert immunomodulatory and anti-inflammatory impacts on key cells of the innate and adaptive immune system. These include macrophages, T cells and NK cells, both in vitro and in small and large animal models. These properties suggest eMSC and MenSC as additional sources of MSC for cell therapies in regenerative medicine as well as immune-mediated disorders and inflammatory diseases. Their easy acquisition via an office-based biopsy or collected from menstrual effluent makes eMSC and MenSC attractive sources of MSC for clinical applications. In preparation for clinical translation, a serum-free culture protocol was established for eMSC which includes a small molecule TGFβ receptor inhibitor that prevents spontaneous differentiation, apoptosis, senescence, maintains the clonogenic SUSD2+ population and enhances their potency, suggesting potential for cell-therapies and regenerative medicine. However, standardization of MenSC isolation protocols and culture conditions are major issues requiring further research to maximize their potential for clinical application. Future research will also address crucial safety aspects of eMSC and MenSC to ensure these protocols produce cell products free from tumorigenicity and toxicity. Although a wealth of data on the biological properties of eMSC and MenSC has recently been published, it will be important to address their mechanism of action in preclinical models of human disease.
Collapse
Affiliation(s)
- Mahmood Bozorgmehr
- Reproductive Immunology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shanti Gurung
- Centre for Reproductive Health, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Saeedeh Darzi
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Shohreh Nikoo
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Somaieh Kazemnejad
- Nanobitechnology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
| | - Amir-Hassan Zarnani
- Reproductive Immunology Research Center, Avicenna Research Institute, Academic Center for Education, Culture and Research (ACECR), Tehran, Iran
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Caroline E. Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, Melbourne, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
47
|
Lv CX, Duan H, Wang S, Gan L, Xu Q. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Promote Proliferation of Allogeneic Endometrial Stromal Cells. Reprod Sci 2020; 27:1372-1381. [PMID: 32006246 DOI: 10.1007/s43032-020-00165-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/01/2019] [Indexed: 12/13/2022]
Abstract
Umbilical cord mesenchymal stem cells (UCMSCs) have been proposed as an ideal source for cell-based therapy to promote endometrial repair and regeneration. Furthermore, increasing evidence has indicated that UCMSC-derived exosomes (UCMSC-exos) act as important paracrine mediators to recapitulate the features of MSCs and may play a vital role in this process. UCMSCs and human endometrial stromal cells (ESCs) were isolated and characterized. ESCs were cocultured with UCMSCs and further assessed by flow cytometry and EdU incorporation assays. UCMSC-exos were extracted by differential ultracentrifugation and identified by western blots, transmission electron microscopy, and nanoparticle tracking analysis. The internalization of UCMSC-exos by ESCs was observed under a confocal microscope. ESCs were treated with UCMSC-exos at different concentrations and for different durations, with cell viability evaluated by CCK-8 assays. The cell cycle analysis showed that the percentage of ESCs in S phase significantly increased after coculture with UCMSCs, whereas it significantly decreased after inhibition of UCMSC-exo secretions. EdU incorporation assays also showed a similar trend. The isolated UCMSC-exos had a typical cup-shaped morphology with a monolayer membrane, expressed the specific exosomal markers Alix, CD63, and TSG101 and were approximately 60 to 200 nm in diameter. The PKH26-labeled UCMSC-exos were incorporated into ESCs. Moreover, UCMSC-exos enhanced the cell growth and viability of ESCs in a dose-dependent manner, and the effects occurred in a short period of time. UCMSC-exos promote the proliferation of ESCs in a dose-dependent manner; thus, they could be used as a potential treatment to promote endometrial repair.
Collapse
Affiliation(s)
- Cheng-Xiao Lv
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Hua Duan
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China.
| | - Sha Wang
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Lu Gan
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Qian Xu
- Department of Minimally Invasive Gynecologic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
48
|
Muruganandan S, Fan X, Dhal S, Nayak NR. Development of A 3D Tissue Slice Culture Model for the Study of Human Endometrial Repair and Regeneration. Biomolecules 2020; 10:biom10010136. [PMID: 31947662 PMCID: PMC7022976 DOI: 10.3390/biom10010136] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 12/28/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023] Open
Abstract
The human endometrium undergoes sequential phases of shedding of the upper functionalis zone during menstruation, followed by regeneration of the functionalis zone from the remaining basalis zone cells, and secretory differentiation under the influence of the ovarian steroid hormones estradiol (E2) and progesterone (P4). This massive tissue regeneration after menstruation is believed to arise from endometrial stromal and epithelial stem cells residing in the basal layer of the endometrium. Although many endometrial pathologies are thought to be associated with defects in these stem cells, studies on their identification and regulation are limited, primarily due to lack of easily accessible animal models, as these processes are unique to primates. Here we describe a robust new method to study endometrial regeneration and differentiation processes using human endometrial tissue slice cultures incorporating an air-liquid interface into a 3D matrix scaffold of type I collagen gel, allowing sustained tissue viability over three weeks. The 3D collagen gel-embedded endometrial tissue slices in a double-dish culture system responded to ovarian steroid hormones, mimicking the endometrial changes that occur in vivo during the menstrual cycle. These changes included the E2-induced upregulation of Ki-67, estrogen receptor (ER), and progesterone receptor (PR) in all endometrial compartments and were markedly suppressed by both P4 and E2 plus P4 treatments. There were also distinct changes in endometrial morphology after E2 and P4 treatments, including subnuclear vacuolation and luminal secretions in glands as well as decidualization of stromal cells, typical characteristics of a progestational endometrium in vivo. This long-term slice culture method provides a unique in vivo-like microenvironment for the study of human endometrial functions and remodeling during early pregnancy and experiments on stem cell populations involved in endometrial regeneration and remodeling. Furthermore, this model has the potential to enable studies on several endometrial diseases, including endometrial cancers and pregnancy complications associated with defects in endometrial remodeling.
Collapse
Affiliation(s)
- Shanmugam Muruganandan
- Perinatal Research Initiative, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.M.); (S.D.); (N.R.N.)
- Department of Developmental Biology, Harvard School of Dental Medicine, 188 Longwood Avenue, Harvard University, Boston, MA 02115, USA
| | - Xiujun Fan
- Laboratory of Reproductive Health, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Correspondence:
| | - Sabita Dhal
- Perinatal Research Initiative, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.M.); (S.D.); (N.R.N.)
| | - Nihar R. Nayak
- Perinatal Research Initiative, Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.M.); (S.D.); (N.R.N.)
| |
Collapse
|
49
|
Saatcioglu HD, Kano M, Horn H, Zhang L, Samore W, Nagykery N, Meinsohn MC, Hyun M, Suliman R, Poulo J, Hsu J, Sacha C, Wang D, Gao G, Lage K, Oliva E, Morris Sabatini ME, Donahoe PK, Pépin D. Single-cell sequencing of neonatal uterus reveals an Misr2+ endometrial progenitor indispensable for fertility. eLife 2019; 8:46349. [PMID: 31232694 PMCID: PMC6650247 DOI: 10.7554/elife.46349] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 06/24/2019] [Indexed: 12/16/2022] Open
Abstract
The Mullerian ducts are the anlagen of the female reproductive tract, which regress in the male fetus in response to MIS. This process is driven by subluminal mesenchymal cells expressing Misr2, which trigger the regression of the adjacent Mullerian ductal epithelium. In females, these Misr2+ cells are retained, yet their contribution to the development of the uterus remains unknown. Here, we report that subluminal Misr2+ cells persist postnatally in the uterus of rodents, but recede by week 37 of gestation in humans. Using single-cell RNA sequencing, we demonstrate that ectopic postnatal MIS administration inhibits these cells and prevents the formation of endometrial stroma in rodents, suggesting a progenitor function. Exposure to MIS during the first six days of life, by inhibiting specification of the stroma, dysregulates paracrine signals necessary for uterine development, eventually resulting in apoptosis of the Misr2+ cells, uterine hypoplasia, and complete infertility in the adult female. In the womb, mammals possess all of the preliminary sexual structures necessary to become either male or female. This includes the Mullerian duct, which develops into the Fallopian tubes, uterus, cervix, and vagina in female fetuses. In male fetuses, the testis secretes a hormone called Mullerian inhibiting substance (MIS). This triggers the activity of a small group of cells, known as Misr2+ cells, that cause the Mullerian duct to degenerate, preventing males from developing female sexual organs. It was not clear what happens to Misr2+ cells in female fetuses or if they affect how the uterus develops. Saatcioglu et al. now show that in newborn female mice and rats, a type of Misr2+ cell that sits within a thin inner layer of the developing uterus still responds to MIS. At this time, the uterus is in a critical early period of development. Treating the mice and rats with MIS protein during their first six days of life eventually caused the Misr2+ cells to die. The treatment also prevented a layer of connective tissue, known as the endometrial stroma, from forming in the uterus. As a result, the mice and rats were infertile and had severely underdeveloped uteri. While the Misr2+ cells are present in newborn rats and mice, Saatcioglu et al. found that they disappeared before birth in humans. However, the overall results suggest that Misr2+ cells act as progenitor cells that develop into the cells of the endometrial stroma. Future work could investigate the roles these cells play in causing uterine developmental disorders and infertility disorders. Furthermore, the finding that MIS inhibits the Misr2+ cells could help researchers to develop treatments for uterine cancer and other conditions where the cells of the uterus grow and divide too much.
Collapse
Affiliation(s)
- Hatice Duygu Saatcioglu
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, United States.,Department of Surgery, Harvard Medical School, Boston, United States
| | - Motohiro Kano
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, United States.,Department of Surgery, Harvard Medical School, Boston, United States
| | - Heiko Horn
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, United States.,Department of Surgery, Harvard Medical School, Boston, United States.,Stanley Center, Broad Institute of MIT and Harvard, Cambridge, United States
| | - Lihua Zhang
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, United States.,Department of Surgery, Harvard Medical School, Boston, United States
| | - Wesley Samore
- Department of Pathology, Massachusetts General Hospital, Boston, United States
| | - Nicholas Nagykery
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, United States.,Department of Surgery, Harvard Medical School, Boston, United States
| | - Marie-Charlotte Meinsohn
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, United States.,Department of Surgery, Harvard Medical School, Boston, United States
| | - Minsuk Hyun
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Rana Suliman
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, United States.,Department of Surgery, Harvard Medical School, Boston, United States
| | - Joy Poulo
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, United States.,Department of Surgery, Harvard Medical School, Boston, United States.,Stanley Center, Broad Institute of MIT and Harvard, Cambridge, United States
| | - Jennifer Hsu
- Department of Gynecology and Reproductive Biology, Massachussets General Hospital, Boston, United States
| | - Caitlin Sacha
- Department of Gynecology and Reproductive Biology, Massachussets General Hospital, Boston, United States
| | - Dan Wang
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, United States
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, United States
| | - Kasper Lage
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, United States.,Department of Surgery, Harvard Medical School, Boston, United States.,Stanley Center, Broad Institute of MIT and Harvard, Cambridge, United States
| | - Esther Oliva
- Department of Pathology, Massachusetts General Hospital, Boston, United States
| | - Mary E Morris Sabatini
- Department of Gynecology and Reproductive Biology, Massachussets General Hospital, Boston, United States
| | - Patricia K Donahoe
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, United States.,Department of Surgery, Harvard Medical School, Boston, United States
| | - David Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, United States.,Department of Surgery, Harvard Medical School, Boston, United States
| |
Collapse
|