1
|
Tang X, Zhu Y, Cao Z, Wang X, Cai X, Tang Y, Zhou J, Wu M, Zhen X, Ding L, Yan G, Wang H, Sun H, Jiang R. CDC42 deficiency leads to endometrial stromal cell senescence in recurrent implantation failure. Hum Reprod 2024; 39:2768-2784. [PMID: 39487595 PMCID: PMC11630066 DOI: 10.1093/humrep/deae246] [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: 05/20/2024] [Revised: 09/22/2024] [Indexed: 11/04/2024] Open
Abstract
STUDY QUESTION Does the downregulation of cell division cycle 42 (CDC42) protein in endometrial stroma lead to endometrial senescence in patients with recurrent implantation failure (RIF), and what is the potential mechanism? SUMMARY ANSWER CDC42 deficiency causes endometrial stromal senescence and decidualization defects, impairing uterine receptivity of RIF patients, via activation of Wnt signaling pathway. WHAT IS KNOWN ALREADY Uterine aging is unique due to the cyclic remodeling and decidualization of endometrial tissue. Several transcriptomic studies have reported increased senescence in the endometrium in young patients with RIF. Our previous transcriptomic sequencing study discovered that endometrium from women with RIF showed downregulation of CDC42, which is an essential molecule affected by various senescence-related diseases. STUDY DESIGN, SIZE, DURATION The endometrial samples of a total of 71 fertile control patients and 37 RIF patients were collected to verify the association between CDC42 expression and endometrial senescence of RIF patients. Primary endometrial stromal cells (EnSCs) were isolated from endometrial biopsies taken from patients without any endometrial complications and planning to undergo IVF, then subjected to adenovirus-mediated CDC42 knockdown and decidualization induction to explore the detailed mechanism by which CDC42 governs stromal senescence and decidualization. Wnt inhibitor XAV-939 was used to correct the endometrial senescence and decidualization defect. PARTICIPANTS/MATERIALS, SETTING, METHODS Senescence was determined by cell cycle arrest markers (e.g. P16, P21, and P53), SASP molecules (e.g. IL6 and CXCL8), and SA-β-gal staining. Masson's staining and Sirius Red staining were used to detect the endometrial fibrosis. Decidualization was evaluated by the mRNA expression and protein secretion of PRL and IGFBP1, F-actin immunostaining, and the BeWo spheroids 'in vitro implantation' model. Methods used to assess cell function included adenovirus transduction, RNA-sequencing, bioinformatic analysis, western blotting, RT-qPCR, ELISA, and immunofluorescence. MAIN RESULTS AND THE ROLE OF CHANCE Here, we observed remarkably increased levels of stromal senescence and fibrosis, along with stromal CDC42 deficiency, in the endometrium of patients with RIF (P < 0.001). Knockdown of CDC42 effectively induced premature senescence in EnSCs, leading to aberrant accumulation of senescent EnSCs and collagen deposition during decidualization. CDC42 deficiency in EnSCs restrained the decidualization differentiation and receptivity to trophoblast cells. Transcriptomic analysis revealed Wnt signaling activation as a critical downstream alteration in CDC42-deficient EnSCs. Mechanistically, CDC42 interacted with AKT competitively to impede the binding of GSK3β to AKT. Knockdown of CDC42 increased AKT-mediated phosphorylation of GSK3β to inactivate the Axin-GSK3β destruction complex, leading to accumulation and nuclear translocation of β-catenin. Importantly, Wnt signaling inhibitors partially corrected the endometrial senescence caused by CDC42 deficiency, and improved both decidualization and trophoblast invasion. LARGE SCALE DATA RNA-seq data sets generated in this study have been deposited at the NCBI database with BioProject accession number PRJNA1102745. LIMITATIONS, REASONS FOR CAUTION The present study was based on in vitro cell cultures. Further studies involving CDC42-regulated endometrial senescence are needed in knockout mice model and human endometrial assembloids. WIDER IMPLICATIONS OF THE FINDINGS In addition to uncovering endometrial senescence in RIF, our findings underscore the significance of CDC42 in modulating EnSC senescence to maintain the decidualization function, and suggest Wnt signaling inhibitors as potential therapeutic agents for alleviating endometrial senescence. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by the National Natural Science Foundation of China [82271698 (R.J.), 82030040 (H.S.), 82288102 (H.W.), and 82371680 (G.Y.)]; the Natural Science Foundation of Jiangsu Province [BK20231117 (R.J.)]; and the Medical Science and Technology Development Foundation of Nanjing Department of Health [YKK23097 (Y.Z.)]. The authors declare no potential conflicts of interest.
Collapse
Affiliation(s)
- Xinyi Tang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Yingchun Zhu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Zhiwen Cao
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Xiaoying Wang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Xinyu Cai
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Yurun Tang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Jidong Zhou
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Min Wu
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Xin Zhen
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Lijun Ding
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Guijun Yan
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Haibin Wang
- Fujian Provincial Key Laboratory of Reproductive Health Research, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Haixiang Sun
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, China
| | - Ruiwei Jiang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| |
Collapse
|
2
|
Bartels HC, Hameed S, Young C, Nabhan M, Downey P, Curran KM, McCormack J, Fabre A, Kolch W, Zhernovkov V, Brennan DJ. Spatial proteomics and transcriptomics of the maternal-fetal interface in placenta accreta spectrum. Transl Res 2024; 274:67-80. [PMID: 39349165 DOI: 10.1016/j.trsl.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/21/2024] [Accepted: 09/21/2024] [Indexed: 10/02/2024]
Abstract
In severe Placenta Accreta Spectrum (PAS), trophoblasts gain deep access in the myometrium (placenta increta). This study investigated alterations at the fetal-maternal interface in PAS cases using a systems biology approach consisting of immunohistochemistry, spatial transcriptomics and proteomics. We identified spatial variation in the distribution of CD4+, CD3+ and CD8+ T-cells at the maternal-interface in placenta increta cases. Spatial transcriptomics identified transcription factors involved in promotion of trophoblast invasion such as AP-1 subunits ATF-3 and JUN, and NFKB were upregulated in regions with deep myometrial invasion. Pathway analysis of differentially expressed genes demonstrated that degradation of extracellular matrix (ECM) and class 1 MHC protein were increased in increta regions, suggesting local tissue injury and immune suppression. Spatial proteomics demonstrated that increta regions were characterised by excessive trophoblastic proliferation in an immunosuppressive environment. Expression of inhibitors of apoptosis such as BCL-2 and fibronectin were increased, while CTLA-4 was decreased and increased expression of PD-L1, PD-L2 and CD14 macrophages. Additionally, CD44, which is a ligand of fibronectin that promotes trophoblast invasion and cell adhesion was also increased in increta regions. We subsequently examined ligand receptor interactions enriched in increta regions, with interactions with ITGβ1, including with fibronectin and ADAMS, emerging as central in increta. These ITGβ1 ligand interactions are involved in activation of epithelial-mesenchymal transition and remodelling of ECM suggesting a more invasive trophoblast phenotype. In PAS, we suggest this is driven by fibronectin via AP-1 signalling, likely as a secondary response to myometrial scarring.
Collapse
Affiliation(s)
- Helena C Bartels
- Dept of UCD Obstetrics and Gynaecology, School of Medicine, University College Dublin, National Maternity Hospital, Holles Street, Dublin 2, Ireland
| | - Sodiq Hameed
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
| | - Constance Young
- Department of Histopathology, National Maternity Hospital, Dublin, Ireland
| | - Myriam Nabhan
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
| | - Paul Downey
- Department of Histopathology, National Maternity Hospital, Dublin, Ireland
| | | | - Janet McCormack
- Research Pathology Core, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Aurelie Fabre
- School of Medicine, University College Dublin, Dublin, Ireland; Research Pathology Core, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland; Histopathology, St Vincent's University Hospital, Dublin, Ireland
| | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Vadim Zhernovkov
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
| | - Donal J Brennan
- Dept of UCD Obstetrics and Gynaecology, School of Medicine, University College Dublin, National Maternity Hospital, Holles Street, Dublin 2, Ireland; Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland; University College Dublin Gynaecological Oncology Group (UCD-GOG), Mater Misericordiae University Hospital and St Vincent's University Hospital, Dublin, Ireland.
| |
Collapse
|
3
|
Wu Y, Su K, Zhang Y, Liang L, Wang F, Chen S, Gao L, Zheng Q, Li C, Su Y, Mao Y, Zhu S, Chai C, Lan Q, Zhai M, Jin X, Zhang J, Xu X, Zhang Y, Gao Y, Huang H. A spatiotemporal transcriptomic atlas of mouse placentation. Cell Discov 2024; 10:110. [PMID: 39438452 PMCID: PMC11496649 DOI: 10.1038/s41421-024-00740-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 09/10/2024] [Indexed: 10/25/2024] Open
Abstract
The placenta, a temporary but essential organ for gestational support, undergoes intricate morphological and functional transformations throughout gestation. However, the spatiotemporal patterns of gene expression underlying placentation remain poorly understood. Utilizing Stereo-seq, we constructed a Mouse Placentation Spatiotemporal Transcriptomic Atlas (MPSTA) spanning from embryonic day (E) 7.5 to E14.5, which includes the transcriptomes of large trophoblast cells that were not captured in previous single-cell atlases. We defined four distinct strata of the ectoplacental cone, an early heterogeneous trophectoderm structure, and elucidated the spatial trajectory of trophoblast differentiation during early postimplantation stages before E9.5. Focusing on the labyrinth region, the interface of nutrient exchange in the mouse placenta, our spatiotemporal ligand-receptor interaction analysis unveiled pivotal modulators essential for trophoblast development and placental angiogenesis. We also found that paternally expressed genes are exclusively enriched in the placenta rather than in the decidual regions, including a cluster of genes enriched in endothelial cells that may function in placental angiogenesis. At the invasion front, we identified interface-specific transcription factor regulons, such as Atf3, Jun, Junb, Stat6, Mxd1, Maff, Fos, and Irf7, involved in gestational maintenance. Additionally, we revealed that maternal high-fat diet exposure preferentially affects this interface, exacerbating inflammatory responses and disrupting angiogenic homeostasis. Collectively, our findings furnish a comprehensive, spatially resolved atlas that offers valuable insights and benchmarks for future explorations into placental morphogenesis and pathology.
Collapse
Affiliation(s)
- Yanting Wu
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
| | - Kaizhen Su
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Zhang
- BGI Research, Shenzhen, Guangdong, China
- Shanxi Medical University - BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Langchao Liang
- BGI Research, Qingdao, Shandong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Fei Wang
- BGI Research, Shenzhen, Guangdong, China
| | - Siyue Chen
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Ling Gao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Qiutong Zheng
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Cheng Li
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Yunfei Su
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Yiting Mao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Simeng Zhu
- Department of Cardiology, Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaochao Chai
- BGI Research, Qingdao, Shandong, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qing Lan
- BGI Research, Shenzhen, Guangdong, China
| | - Man Zhai
- BGI Research, Shenzhen, Guangdong, China
| | - Xin Jin
- BGI Research, Shenzhen, Guangdong, China
| | - Jinglan Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China
| | - Xun Xu
- BGI Research, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen, Guangdong, China
| | - Yu Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
| | - Ya Gao
- BGI Research, Shenzhen, Guangdong, China.
- Shanxi Medical University - BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, Shanxi, China.
- Shenzhen Engineering Laboratory for Birth Defects Screening, BGI Research, Shenzhen, Guangdong, China.
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China.
- Key Laboratory of Reproductive Genetics (Ministry of Education), Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China.
- Shanghai Key Laboratory of Reproduction and Development, Shanghai, China.
- Department of Reproductive Endocrinology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
4
|
Lin G, Lin L, Chen X, Chen L, Yang J, Chen Y, Qian D, Zeng Y, Xu Y. PPAR-γ/NF-kB/AQP3 axis in M2 macrophage orchestrates lung adenocarcinoma progression by upregulating IL-6. Cell Death Dis 2024; 15:532. [PMID: 39060229 PMCID: PMC11282095 DOI: 10.1038/s41419-024-06919-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024]
Abstract
Aquaporin 3 (AQP3), which is mostly expressed in pulmonary epithelial cells, was linked to lung adenocarcinoma (LUAD). However, the underlying functions and mechanisms of AQP3 in the tumor microenvironment (TME) of LUAD have not been elucidated. Single-cell RNA sequencing (scRNA-seq) was used to study the composition, lineage, and functional states of TME-infiltrating immune cells and discover AQP3-expressing subpopulations in five LUAD patients. Then the identifications of its function on TME were examined in vitro and in vivo. AQP3 was associated with TNM stages and lymph node metastasis of LUAD patients. We classified inter- and intra-tumor diversity of LUAD into twelve subpopulations using scRNA-seq analyses. The analysis showed AQP3 was mainly enriched in subpopulations of M2 macrophages. Importantly, mechanistic investigations indicated that AQP3 promoted M2 macrophage polarization by the PPAR-γ/NF-κB axis, which affected tumor growth and migration via modulating IL-6 production. Mixed subcutaneous transplanted tumor mice and Aqp3 knockout mice models were further utilized, and revealed that AQP3 played a critical role in mediating M2 macrophage polarization, modulating glucose metabolism in tumors, and regulating both upstream and downstream pathways. Overall, our study demonstrated that AQP3 could regulate the proliferation, migration, and glycometabolism of tumor cells by modulating M2 macrophages polarization through the PPAR-γ/NF-κB axis and IL-6/IL-6R signaling pathway, providing new insight into the early detection and potential therapeutic target of LUAD.
Collapse
Affiliation(s)
- Guofu Lin
- Fujian Provincial Clinical Research Center of Interventional Respirology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Ouanzhou, Fujian Province, 362000, China
| | - Lanlan Lin
- Fujian Provincial Clinical Research Center of Interventional Respirology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Ouanzhou, Fujian Province, 362000, China
| | - Xiaohui Chen
- Fujian Provincial Clinical Research Center of Interventional Respirology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Ouanzhou, Fujian Province, 362000, China
| | - Luyang Chen
- Fujian Provincial Clinical Research Center of Interventional Respirology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
- Fujian Provincial Key Laboratory of Lung Stem Cells, Ouanzhou, Fujian Province, 362000, China
| | - Jiansheng Yang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian province, 362000, China
| | - Yanling Chen
- Clinical Research Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
| | - Danwen Qian
- The Tumor Immunogenomics and Immunosurveillance (TIGI) Lab, UCL Cancer Institute, London, UK
| | - Yiming Zeng
- Fujian Provincial Clinical Research Center of Interventional Respirology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China.
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China.
- Fujian Provincial Key Laboratory of Lung Stem Cells, Ouanzhou, Fujian Province, 362000, China.
| | - Yuan Xu
- Fujian Provincial Clinical Research Center of Interventional Respirology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China.
- Clinical Research Center, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China.
| |
Collapse
|
5
|
Tian J, Zhang Z, Mei J, Kong N, Yan Y, Shen X, Zhou J, Zhang Y, Kang N, Zhen X, Ding L, Yan G, Sun H, Sheng X. Dysregulation of endometrial stromal serotonin homeostasis leading to abnormal phosphatidylcholine metabolism impairs decidualization in patients with recurrent implantation failure. Hum Reprod Open 2024; 2024:hoae042. [PMID: 39091587 PMCID: PMC11293872 DOI: 10.1093/hropen/hoae042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 05/17/2024] [Indexed: 08/04/2024] Open
Abstract
STUDY QUESTION Does abnormal serotonin homeostasis contribute to impaired endometrial decidualization in patients with recurrent implantation failure (RIF)? SUMMARY ANSWER Abnormal serotonin homeostasis in patients with RIF, which is accompanied by decreased monoamine oxidase (MAO) expression, affects the decidualization of endometrial stromal cells and leads to embryo implantation failure. WHAT IS KNOWN ALREADY Previous studies have indicated that the expression of MAO, which metabolizes serotonin, is reduced in the endometrium of patients with RIF, and serotonin can induce disruption of implantation in rats. However, whether abnormal serotonin homeostasis leads to impaired decidualization in patients with RIF and, if so, the mechanism involved, remains unclear. STUDY DESIGN SIZE DURATION Endometrial samples from 25 patients with RIF and 25 fertile patients were used to investigate the expression levels of monoamine oxidase A (MAOA), monoamine oxidase B (MAOB), and serotonin. We isolated human endometrial stromal cells to investigate the role of MAOA, MAOB, and serotonin in inducing decidualization in vitro and further explored the underlying mechanism using RNA-sequencing (RNA-seq) and liquid chromatography-mass spectrometry (LC/MS) analyses. PARTICIPANTS/MATERIALS SETTING METHODS The levels of serotonin in the endometrium of patients with RIF were detected by ELISA and immunohistofluorescence, and the key genes involved in abnormal serotonin metabolism were analyzed via combination with single-cell sequencing data. The effects of MAOA or MAOB on the decidualization of stromal cells were investigated using an in vitro human endometrial stromal cell-induced decidualization model and a mouse artificially induced decidualization model. The potential mechanisms by which MAOA and MAOB regulate decidualization were explored by RNA-seq and LC/MS analysis. MAIN RESULTS AND THE ROLE OF CHANCE We found that women with RIF have abnormal serotonin metabolism in the endometrium and attenuated MAO in endometrial stromal cells. Endometrial decidualization was accompanied by increased MAO in vivo and in vitro. However attenuated MAO caused an increased local serotonin content in the endometrium, impairing stromal cell decidualization. RNA-seq and LC/MS analyses showed that abnormal lipid metabolism, especially phosphatidylcholine metabolism, was involved in the defective decidualization caused by MAO deficiency. Furthermore, decidualization defects were rescued by phosphatidylcholine supplementation. LARGE SCALE DATA RNA-seq information and raw data can be found at NCBI Bioproject number PRJNA892255. LIMITATIONS REASONS FOR CAUTION This study revealed that impaired serotonin metabolic homeostasis and abnormally reduced MAO expression were among the reasons for RIF. However, the source and other potential functions of serotonin in the endometrium remain to be further explored. WIDER IMPLICATIONS OF THE FINDINGS This study provides new insights into the mechanisms of serotonin homeostasis in human endometrial decidualization and new biomarkers or targets for the treatment of patients with RIF. STUDY FUNDING/COMPETING INTERESTS X. Sheng is supported by grants from the National Natural Science Foundation of China (82001629), the Wenzhou Basic Public Welfare Research Project (Y20240030), the Youth Program of Natural Science Foundation of Jiangsu Province (BK20200116), and Jiangsu Province Postdoctoral Research Funding (2021K277B). H.S. is supported by grants from the National Natural Science Foundation of China (82030040). G.Y. is supported by grants from the National Natural Science Foundation of China (82171653). The authors declare no conflicts of interest.
Collapse
Affiliation(s)
- Jiao Tian
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Zhe Zhang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Jie Mei
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Na Kong
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yuan Yan
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaoyue Shen
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jidong Zhou
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Yang Zhang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Nannan Kang
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Xin Zhen
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Lijun Ding
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Guijun Yan
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
| | - Haixiang Sun
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Xiaoqiang Sheng
- Center for Reproductive Medicine and Obstetrics and Gynecology, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- Center for Reproductive Medicine, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
Huang Y, Dai F, Chen L, Li Z, Liu H, Cheng Y. BMP4 in Human Endometrial Stromal Cells Can Affect Decidualization by Regulating FOXO1 Expression. Endocrinology 2024; 165:bqae049. [PMID: 38679470 DOI: 10.1210/endocr/bqae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/06/2024] [Accepted: 04/15/2024] [Indexed: 05/01/2024]
Abstract
CONTEXT Recurrent spontaneous abortion (RSA) is defined as the loss of 2 or more consecutive intrauterine pregnancies with the same sexual partner in the first trimester. Despite its significance, the etiology and underlying mechanisms of RSA remain elusive. Defective decidualization is proposed as one of the potential causes of RSA, with abnormal decidualization leading to disturbances in trophoblast invasion function. OBJECTIVE To assess the role of bone morphogenetic protein 4 (BMP4) in decidualization and RSA. METHODS Decidual samples were collected from both RSA patients and healthy controls to assess BMP4 expression. In vitro cell experiments utilized the hESC cell line to investigate the impact of BMP4 on decidualization and associated aging, as well as its role in the maternal-fetal interface communication. Subsequently, a spontaneous abortion mouse model was established to evaluate embryo resorption rates and BMP4 expression levels. RESULTS Our study identified a significant downregulation of BMP4 expression in the decidua of RSA patients compared to the normal control group. In vitro, BMP4 knockdown resulted in inadequate decidualization and inhibited associated aging processes. Mechanistically, BMP4 was implicated in the regulation of FOXO1 expression, thereby influencing decidualization and aging. Furthermore, loss of BMP4 hindered trophoblast migration and invasion via FOXO1 modulation. Additionally, BMP4 downregulation was observed in RSA mice. CONCLUSION Our findings highlighted the downregulation of BMP4 in both RSA patients and mice. BMP4 in human endometrial stromal cells was shown to modulate decidualization by regulating FOXO1 expression. Loss of BMP4 may contribute to the pathogenesis of RSA, suggesting potential avenues for abortion prevention strategies.
Collapse
Affiliation(s)
- Yanjie Huang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| | - Liping Chen
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| | - Zhidian Li
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| | - Hua Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, People's Republic of China
| |
Collapse
|
7
|
Jin MH, Feng L, Xiang HY, Sun HN, Han YH, Kwon T. Exploring the role of Prx II in mitigating endoplasmic reticulum stress and mitochondrial dysfunction in neurodegeneration. Cell Commun Signal 2024; 22:231. [PMID: 38637880 PMCID: PMC11025193 DOI: 10.1186/s12964-024-01613-x] [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: 02/01/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Neurodegenerative diseases are increasingly recognized for their association with oxidative stress, which leads to progressive dysfunction and loss of neurons, manifesting in cognitive and motor impairments. This study aimed to elucidate the neuroprotective role of peroxiredoxin II (Prx II) in counteracting oxidative stress-induced mitochondrial damage, a key pathological feature of neurodegeneration. METHODS We investigated the impact of Prx II deficiency on endoplasmic reticulum stress and mitochondrial dysfunction using HT22 cell models with knocked down and overexpressed Prx II. We observed alcohol-treated HT22 cells using transmission electron microscopy and monitored changes in the length of mitochondria-associated endoplasmic reticulum membranes and their contact with endoplasmic reticulum mitochondria contact sites (EMCSs). Additionally, RNA sequencing and bioinformatic analysis were conducted to identify the role of Prx II in regulating mitochondrial transport and the formation of EMCSs. RESULTS Our results indicated that Prx II preserves mitochondrial integrity by facilitating the formation of EMCSs, which are essential for maintaining mitochondrial Ca2+ homeostasis and preventing mitochondria-dependent apoptosis. Further, we identified a novel regulatory axis involving Prx II, the transcription factor ATF3, and miR-181b-5p, which collectively modulate the expression of Armcx3, a protein implicated in mitochondrial transport. Our findings underscore the significance of Prx II in protecting neuronal cells from alcohol-induced oxidative damage and suggest that modulating the Prx II-ATF3-miR-181b-5p pathway may offer a promising therapeutic strategy against neurodegenerative diseases. CONCLUSIONS This study not only expands our understanding of the cytoprotective mechanisms of Prx II but also offers necessary data for developing targeted interventions to bolster mitochondrial resilience in neurodegenerative conditions.
Collapse
Affiliation(s)
- Mei-Hua Jin
- College of Life Science & Biotechnology Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, China
| | - Lin Feng
- College of Life Science & Biotechnology Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, China
| | - Hong-Yi Xiang
- College of Life Science & Biotechnology Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, China
| | - Hu-Nan Sun
- College of Life Science & Biotechnology Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, China
| | - Ying-Hao Han
- College of Life Science & Biotechnology Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, China.
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 351-33 Neongme-gil, Ibam-myeon, 56216, Jeongeup-si, Jeonbuk, Republic of Korea.
- Department of Applied Biological Engineering, KRIBB School of Biotechnology, National University of Science and Technology (UST), 34113, Daejeon, Republic of Korea.
| |
Collapse
|
8
|
Lu J, Zhang M, Liu Z, Guo L, Huang P, Xia W, Li J, Lv J, Cheung HH, Ding C, Li H, Huang B. NSUN2-Mediated m 5C Methylation Impairs Endometrial Receptivity. J Transl Med 2024; 104:100327. [PMID: 38237738 DOI: 10.1016/j.labinv.2024.100327] [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: 09/05/2023] [Revised: 12/05/2023] [Accepted: 12/23/2023] [Indexed: 02/12/2024] Open
Abstract
Impaired endometrial decidualization is the primary cause of recurrent implantation failure (RIF). RNA methylation modification, especially NSUN family mediated m5C, is crucial for various physiological events, such as maternal-to-zygotic transition, gametogenesis, embryonic development, organismal lifespan, and cell cycle. However, the regulatory mechanisms between NSUN family mediated m5C modification and RIF remain unknown. We acquired NSUN2 expression data of 15 human endometrium samples at proliferative and secretory stages from reproductive cell atlas. The overall pattern of m5C sites and genes was elucidated through m5C-BS-seq, whereas the overall m5C levels in different groups were revealed by dot blot assay. BrdU and western blotting assays were carried out to evaluate the role of NSUN2 in proliferation and autophagy. The effects of NSUN2-mediated m5C modification on embryo attachment were evaluated by an in vitro model of a confluent monolayer of Ishikawa cells cocultured with BeWo spheroids, and its downstream targets were evaluated by real-time reverse-transcription PCR and western blotting in Ishikawa cells. The molecular mechanism for NSUN2 regulating its downstream targets' expression was determined by Cut&Tag and coimmunoprecipitation assays. NSUN2 was increased in SOX9+ cells and widespread in epithelial cell type at the proliferative stage by previous single-cell RNA sequencing data. NSUN2 overexpression (NSUN2OE) in the Ishikawa cell line elevated m5C levels and promoted cell proliferation and autophagy. NSUN2OE reduced attachment efficiency of BeWo cell spheres. Overexpressed NSUN2 was found to increase STAT1 and MMP14 mRNA expressions by inducing exon skipping. NSUN2 interacted with CLDN4 through m5C modification, and NSUN2OE or NSUN2 knockdown resulted in a similar variation tendency of CLDN4. Overexpression of NSUN2 increased CLDN4 H3K9ac modification by downregulating SIRT4 expression at the protein level, leading to the upregulation of CLDN4 mRNA expression. Our results uncovered a novel intricate regulatory mechanism between NSUN2-mediated m5C and RIF and suggested a potential new therapeutic strategy for RIF.
Collapse
Affiliation(s)
- Jiafeng Lu
- State Key Laboratory of Reproductive Medicine, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Ming Zhang
- State Key Laboratory of Reproductive Medicine, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Zhenxing Liu
- State Key Laboratory of Reproductive Medicine, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Ling Guo
- State Key Laboratory of Reproductive Medicine, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Peng Huang
- State Key Laboratory of Reproductive Medicine, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Wenjuan Xia
- State Key Laboratory of Reproductive Medicine, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Jincheng Li
- State Key Laboratory of Reproductive Medicine, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Jinghuan Lv
- State Key Laboratory of Reproductive Medicine, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Hoi-Hung Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR
| | - Chenyue Ding
- State Key Laboratory of Reproductive Medicine, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.
| | - Hong Li
- State Key Laboratory of Reproductive Medicine, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.
| | - Boxian Huang
- State Key Laboratory of Reproductive Medicine, Suzhou Affiliated Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.
| |
Collapse
|
9
|
Zahir M, Tavakoli B, Zaki-Dizaji M, Hantoushzadeh S, Majidi Zolbin M. Non-coding RNAs in Recurrent implantation failure. Clin Chim Acta 2024; 553:117731. [PMID: 38128815 DOI: 10.1016/j.cca.2023.117731] [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: 11/10/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Recurrent implantation failure (RIF), defined as the inability to achieve conception following multiple consecutive in-vitro fertilization (IVF) attempts, represents a complex and multifaceted challenge in reproductive medicine. The emerging role of non-coding RNAs in RIF etiopathogenesis has only gained prominence over the last decade, illustrating a new dimension to our understanding of the intricate network underlying RIF. Successful embryo implantation demands a harmonious synchronization between an adequately decidualized endometrium, a competent blastocyst, and effective maternal-embryonic interactions. Emerging evidence has clarified the involvement of a sophisticated network of non-coding RNAs, including microRNAs, circular RNAs, and long non-coding RNAs, in orchestrating these pivotal processes. Disconcerted expression of these molecules can disrupt the delicate equilibrium required for implantation, amplifying the risk of RIF. This comprehensive review presents an in-depth investigation of the complex role played by non-coding RNAs in the pathogenesis of RIF. Furthermore, it underscores the vast potential of non-coding RNAs as diagnostic biomarkers and therapeutic targets, with the ultimate goal of enhancing implantation success rates in IVF cycles. As ongoing research continues to unravel the intercalated web of molecular interactions, exploiting the power of non-coding RNAs may offer promising avenues for mitigating the challenges posed by RIF and improving the outcomes of assisted reproduction.
Collapse
Affiliation(s)
- Mazyar Zahir
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Tavakoli
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Biology, Maragheh University, Maragheh, Iran
| | - Majid Zaki-Dizaji
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Sedigheh Hantoushzadeh
- Vali-E-Asr Reproductive Health Research Center, Family Health Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Zhao H, Lv N, Cong J, Chen G, Bao H, Liu X. Upregulated RPA2 in endometrial tissues of repeated implantation failure patients impairs the endometrial decidualization. J Assist Reprod Genet 2023; 40:2739-2750. [PMID: 37831348 PMCID: PMC10643753 DOI: 10.1007/s10815-023-02946-1] [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: 05/18/2023] [Accepted: 09/09/2023] [Indexed: 10/14/2023] Open
Abstract
PURPOSE To investigate the expression and underlying mechanism of RPA2 in endometrium of patients with repeated implantation failure (RIF). METHODS In this study, we retrieved the expression profiles from GEO databases and filtered the differentially expressed genes between RIF and the fertile control group. Ultimately, RPA2 was confirmed as a target gene. RPA2 expression in endometrial tissues of RIF patients, the control group, and different phases was detected by RT-qPCR, immunohistochemistry, and Western blotting. The role of RPA2 in endometrial decidualization was performed by in vitro decidualization inducing by 8-Br-cAMP and MPA. Furthermore, RT-qPCR was used to detect changes in the decidual biomarkers after transfection of RPA2 overexpression vector in human endometrium stromal cell (HESC). RESULTS RPA2 was significantly upregulated in the mid-secretory endometrium of patients with RIF. As a proliferation-related gene, RPA2 was obviously higher expressed at proliferative phase during the normal menstrual cycles. Moreover, the downregulation of RPA2 was discovered during decidualization of HESC. Furthermore, RPA2 overexpression impaired decidualization by inhibiting the expression of prolactin (PRL) and insulin-like growth factor-binding protein 1 (IGFBP1). CONCLUSIONS Our finding indicated that aberrant upregulation of RPA2 attenuated decidualization of HESC in RIF women and provided new potential therapeutic targets.
Collapse
Affiliation(s)
- Huishan Zhao
- Reproductive Medicine Centre, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Na Lv
- Department of Clinical Laboratory, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Jianxiang Cong
- Reproductive Medicine Centre, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Gang Chen
- Department of Breast Surgery, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Hongchu Bao
- Reproductive Medicine Centre, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China.
| | - Xuemei Liu
- Reproductive Medicine Centre, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China.
| |
Collapse
|
11
|
Zhang H, Wang Z, Zhou Q, Cao Z, Jiang Y, Xu M, Liu J, Zhou J, Yan G, Sun H. Downregulated INHBB in endometrial tissue of recurrent implantation failure patients impeded decidualization through the ADCY1/cAMP signalling pathway. J Assist Reprod Genet 2023; 40:1135-1146. [PMID: 36913138 PMCID: PMC10239411 DOI: 10.1007/s10815-023-02762-7] [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: 11/28/2022] [Accepted: 02/22/2023] [Indexed: 03/14/2023] Open
Abstract
PURPOSE This study aims to identify the mechanism of Inhibin Subunit Beta B (INHBB), a member of the transforming growth factor-β (TGF-β) family involved in the regulation of human endometrial stromal cells (HESCs) decidualization in recurrent implantation failure (RIF). METHODS RNA-seq was conducted to identify the differentially expressed genes in the endometria from control and RIF patients. RT-qPCR, WB, and immunohistochemistry were performed to analyse the expression levels of INHBB in endometrium and decidualised HESCs. RT-qPCR and immunofluorescence were used to detect changes in the decidual marker genes and cytoskeleton after knockdown INHBB. Then, RNA-seq was used to dig out the mechanism of INHBB regulating decidualization. The cAMP analogue (forskolin) and si-INHBB were used to investigate the involvement of INHBB in the cAMP signalling pathway. The correlation of INHBB and ADCY expression was analysed by Pearson's correlation analysis. RESULTS Our results showed significantly reduced expression of INHBB in endometrial stromal cells of women with RIF. In addition, INHBB was increased in the endometrium of the secretory phase and significantly induced in in-vitro decidualization of HESCs. Notably, with RNA-seq and siRNA-mediated knockdown approaches, we demonstrated that the INHBB-ADCY1-mediated cAMP signalling pathway regulates the reduction of decidualization. We found a positive association between the expression of INHBB and ADCY1 in endometria with RIF (R2 = 0.3785, P = 0.0005). CONCLUSIONS The decline of INHBB in HESCs suppressed ADCY1-induced cAMP production and cAMP-mediated signalling, which attenuated decidualization in RIF patients, indicating that INHBB is an essential component in the decidualization process.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Zhilong Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Quan Zhou
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Zhiwen Cao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Yue Jiang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Manlin Xu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Jingyu Liu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Jidong Zhou
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Guijun Yan
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
- Center for Molecular Reproductive Medicine, Nanjing University, Nanjing, China.
| | - Haixiang Sun
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China.
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
12
|
Bergsten TM, Li K, Lantvit DD, Murphy BT, Burdette JE. Kaempferol, a Phytoprogestin, Induces a Subset of Progesterone-Regulated Genes in the Uterus. Nutrients 2023; 15:1407. [PMID: 36986136 PMCID: PMC10051346 DOI: 10.3390/nu15061407] [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: 01/18/2023] [Revised: 02/27/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Progesterone functions as a steroid hormone involved in female reproductive physiology. While some reproductive disorders manifest with symptoms that can be treated by progesterone or synthetic progestins, recent data suggest that women also seek botanical supplements to alleviate these symptoms. However, botanical supplements are not regulated by the U.S. Food and Drug Administration and therefore it is important to characterize and quantify the inherent active compounds and biological targets of supplements within cellular and animal systems. In this study, we analyzed the effect of two natural products, the flavonoids, apigenin and kaempferol, to determine their relationship to progesterone treatment in vivo. According to immunohistochemical analysis of uterine tissue, kaempferol and apigenin have some progestogenic activity, but do not act in exactly the same manner as progesterone. More specifically, kaempferol treatment did not induce HAND2, did not change proliferation, and induced ZBTB16 expression. Additionally, while apigenin treatment did not appear to dramatically affect transcripts, kaempferol treatment altered some transcripts (44%) in a similar manner to progesterone treatment but had some unique effects as well. Kaempferol regulated primarily unfolded protein response, androgen response, and interferon-related transcripts in a similar manner to progesterone. However, the effects of progesterone were more significant in regulating thousands of transcripts making kaempferol a selective modifier of signaling in the mouse uterus. In summary, the phytoprogestins, apigenin and kaempferol, have progestogenic activity in vivo but also act uniquely.
Collapse
Affiliation(s)
| | | | | | | | - Joanna E. Burdette
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois Chicago, Chicago, IL 60607, USA
| |
Collapse
|
13
|
Li B, Yan YP, He YY, Liang C, Li MY, Wang Y, Yang ZM. IHH, SHH, and primary cilia mediate epithelial-stromal cross-talk during decidualization in mice. Sci Signal 2023; 16:eadd0645. [PMID: 36853961 DOI: 10.1126/scisignal.add0645] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
The establishment of pregnancy depends on interactions between the epithelial and stromal cells of the endometrium that drive the decidual reaction that remodels the stroma and enables embryo implantation. Decidualization in mice also depends on ovarian hormones and the presence of a blastocyst. Hedgehog signaling is transduced by primary cilia in many tissues and is involved in epithelial-stromal cross-talk during decidualization. We found that primary cilia on mouse uterine stromal cells increased in number and length during early pregnancy and were required for decidualization. In vitro and in vivo, progesterone promoted stromal ciliogenesis and the production of Indian hedgehog (IHH) in the epithelium and Sonic hedgehog (SHH) in the stroma. Blastocyst-derived TNF-α also induced epithelial IHH, which stimulated the production of SHH in the stroma through a mechanism that may involve the release of arachidonic acid from epithelial cells. In the stroma, SHH activated canonical Hedgehog signaling through primary cilia and promoted decidualization through a mechanism that depended on interleukin-11 (IL-11) and primary cilia. Our findings identify a primary cilia-dependent network that controls endometrial decidualization and suggest primary cilia as a candidate therapeutic target for endometrial diseases.
Collapse
Affiliation(s)
- Bo Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountain Region, College of Animal Science, Guizhou University, Guiyang 550025, China.,College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ya-Ping Yan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yu-Ying He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Chen Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Meng-Yuan Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ying Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zeng-Ming Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountain Region, College of Animal Science, Guizhou University, Guiyang 550025, China.,College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| |
Collapse
|
14
|
Lyu M, Gao W, Zhang L, Yang X, Yue F, Li H, Ma X, Liu L. Hsa_circ_0001550 impairs decidualization by regulating the proliferation and apoptosis of endometrial stromal cells. Reprod Biomed Online 2023; 46:225-233. [PMID: 36396534 DOI: 10.1016/j.rbmo.2022.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 10/01/2022] [Accepted: 10/07/2022] [Indexed: 11/09/2022]
Abstract
RESEARCH QUESTION What is the molecular function of hsa_circ_0001550 in decidualization? DESIGN Human endometrial stromal cells (HESC) were isolated from the endometrium tissues to build an in-vitro decidualization model. Different concentrations of medroxyprogesterone acetate (MPA) were used to observe whether the expression level of hsa_circ_0001550 was related to progesterone. Biological characteristics and distribution of hsa_circ_0001550 were determined by RNase R, actinomycin D (Act D) assay and cytoplasmic/nuclear fraction assay. Then the overexpression of hsa_circ_0001550 was achieved by adenovirus vector. Cell proliferation was determined by Cell Counting Kit-8 (CCK-8) assays. The cell cycle was assessed by flow cytometry analyses. Cell apoptosis was determined by annexin-V/propidium iodide double staining experiment and western blotting. RESULTS The expression of hsa_circ_0001550 was decreased in decidua and decidualized HESC (P < 0.001, P = 0.014). Hsa_circ_0001550 is a covalently closed RNA molecule that was verified by RNase R assay and Act D assay (P = 0.012). Nuclear and cytoplasmic separation experiments confirmed that hsa_circ_0001550 was mainly distributed in the cytoplasm. Overexpression of hsa_circ_0001550 inhibited decidualization of HESC (P < 0.0001). Furthermore, overexpression of hsa_circ_0001550 inhibited proliferation by decreasing the number of S phase cells (P = 0.033). Annexin-V/propidium iodide double staining experiment and western blotting revealed that overexpression of hsa_circ_0001550 promoted HESC apoptosis (P < 0.001, P = 0.0139). CONCLUSIONS Hsa_circ_0001550 impairs decidualization of HESC. Progesterone decreases the expression of hsa_circ_0001550. The results may provide new insights into the cause of decidualization.
Collapse
Affiliation(s)
- Meng Lyu
- The First Clinical Medical College of Lanzhou University, Lanzhou Gansu, China
| | - Wenxin Gao
- The First Clinical Medical College of Lanzhou University, Lanzhou Gansu, China
| | - Lili Zhang
- The Reproductive Center, The First Hospital of Lanzhou University, Lanzhou Gansu, China; Gansu Key Laboratory of Reproductive Medicine and Embryos, Lanzhou Gansu, China
| | - Xia Yang
- The Reproductive Center, The First Hospital of Lanzhou University, Lanzhou Gansu, China; Gansu Key Laboratory of Reproductive Medicine and Embryos, Lanzhou Gansu, China
| | - Feng Yue
- The Reproductive Center, The First Hospital of Lanzhou University, Lanzhou Gansu, China; Gansu Key Laboratory of Reproductive Medicine and Embryos, Lanzhou Gansu, China
| | - Hongxing Li
- The Reproductive Center, The First Hospital of Lanzhou University, Lanzhou Gansu, China; Gansu Key Laboratory of Reproductive Medicine and Embryos, Lanzhou Gansu, China
| | - Xiaoling Ma
- The Reproductive Center, The First Hospital of Lanzhou University, Lanzhou Gansu, China; Gansu Key Laboratory of Reproductive Medicine and Embryos, Lanzhou Gansu, China.
| | - Lin Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou Gansu, China; The Reproductive Center, The First Hospital of Lanzhou University, Lanzhou Gansu, China; Gansu Key Laboratory of Reproductive Medicine and Embryos, Lanzhou Gansu, China.
| |
Collapse
|
15
|
Primary Cilia Restrain PI3K-AKT Signaling to Orchestrate Human Decidualization. Int J Mol Sci 2022; 23:ijms232415573. [PMID: 36555215 PMCID: PMC9779442 DOI: 10.3390/ijms232415573] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022] Open
Abstract
Endometrial decidualization plays a pivotal role during early pregnancy. Compromised decidualization has been tightly associated with recurrent implantation failure (RIF). Primary cilium is an antenna-like sensory organelle and acts as a signaling nexus to mediate Hh, Wnt, TGFβ, BMP, FGF, and Notch signaling. However, whether primary cilium is involved in human decidualization is still unknown. In this study, we found that primary cilia are present in human endometrial stromal cells. The ciliogenesis and cilia length are increased by progesterone during in vitro and in vivo decidualization. Primary cilia are abnormal in the endometrium of RIF patients. Based on data from both assembly and disassembly of primary cilia, it has been determined that primary cilium is essential to human decidualization. Trichoplein (TCHP)-Aurora A signaling mediates cilia disassembly during human in vitro decidualization. Mechanistically, primary cilium modulates human decidualization through PTEN-PI3K-AKT-FOXO1 signaling. Our study highlights primary cilium as a novel decidualization-related signaling pathway.
Collapse
|
16
|
Endometrial stromal PRMT5 plays a crucial role in decidualization by regulating NF-κB signaling in endometriosis. Cell Death Dis 2022; 8:408. [PMID: 36195592 PMCID: PMC9532444 DOI: 10.1038/s41420-022-01196-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/08/2022]
Abstract
Decidualization is a prerequisite for successful embryo implantation, in which elongated fibroblast-like endometrial stromal cells differentiate into more rounded decidual cells. Accumulating evidence has stressed the important role of the defective eutopic endometrium in infertility in endometriosis patients. However, the role of arginine methylation in the process of physiological decidualization and pathological decidualization defects is not clear. Here, we observed that the expression level of PRMT5, the main type II PRMT, was decreased in the endometrium of endometriosis patients, predominantly in stromal cells. Compared with the undecidualized state, PRMT5 was increased in the stromal cells of normal secretory endometrium in humans and in the decidua of normal pregnant mice or mice with artificially induced decidualization. The inhibition of PRMT5 resulted in a significant decrease in uterine weight and decidualization-related regulator expression, including FOXO1, HOXA10 and WNT4, in mice and IGFBP1 and prolactin levels in human endometrial stromal cells. Transcriptome analysis showed that decreased PRMT5 activity led to NF-κB signaling activation by inducing p65 translocation to the nucleus, which was also observed in endometriosis patients. Finally, overexpression of PRMT5 rescued the defective expression of IGFBP1 and prolactin in primary endometrial stromal cells from endometriosis patients. Our results indicate that promotion of PRMT5 may provide novel therapeutic strategies for the treatment of decidualization defects in infertile women, such as those with endometriosis.
Collapse
|
17
|
Penariol LBC, Thomé CH, Tozetti PA, Paier CRK, Buono FO, Peronni KC, Orellana MD, Covas DT, Moraes MEA, Silva WA, Rosa-e-Silva JC, Ferriani RA, Faça VM, Poli-Neto OB, Tiezzi DG, Meola J. What Do the Transcriptome and Proteome of Menstrual Blood-Derived Mesenchymal Stem Cells Tell Us about Endometriosis? Int J Mol Sci 2022; 23:11515. [PMID: 36232817 PMCID: PMC9570451 DOI: 10.3390/ijms231911515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/19/2022] [Accepted: 09/22/2022] [Indexed: 11/21/2022] Open
Abstract
Given the importance of menstrual blood in the pathogenesis of endometriosis and the multifunctional roles of menstrual mesenchymal stem cells (MenSCs) in regenerative medicine, this issue has gained prominence in the scientific community. Moreover, recent reviews highlight how robust the integrated assessment of omics data are for endometriosis. To our knowledge, no study has applied the multi-omics approaches to endometriosis MenSCs. This is a case-control study at a university-affiliated hospital. MenSCs transcriptome and proteome data were obtained by RNA-seq and UHPLC-MS/MS detection. Among the differentially expressed proteins and genes, we emphasize ATF3, ID1, ID3, FOSB, SNAI1, NR4A1, EGR1, LAMC3, and ZFP36 genes and MT2A, TYMP, COL1A1, COL6A2, and NID2 proteins that were already reported in the endometriosis. Our functional enrichment analysis reveals integrated modulating signaling pathways such as epithelial-mesenchymal transition (↑) and PI3K signaling via AKT to mTORC1 (↓ in proteome), mTORC1 signaling, TGF beta signaling, TNFA signaling via NFkB, IL6 STAT3 signaling, and response to hypoxia via HIF1A targets (↑ in transcriptome). Our findings highlight primary changes in the endometriosis MenSCs, suggesting that the chronic inflammatory endometrial microenvironment can modulate these cells, providing opportunities for endometriosis etiopathogenesis. Moreover, they identify challenges for future research leveraging knowledge for regenerative and precision medicine in endometriosis.
Collapse
Affiliation(s)
- Letícia B. C. Penariol
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Carolina H. Thomé
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- Regional Blood Center, Medical School of Hemocenter Foundation of Ribeirão Preto, University of São Paulo, São Paulo 14051-140, Brazil
| | - Patrícia A. Tozetti
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Carlos R. K. Paier
- Drug Research and Development Center, Federal University of Ceara, Ceará 60430-275, Brazil
| | - Fabiana O. Buono
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Kamila C. Peronni
- Department of Genetics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Maristela D. Orellana
- Regional Blood Center, Medical School of Hemocenter Foundation of Ribeirão Preto, University of São Paulo, São Paulo 14051-140, Brazil
| | - Dimas T. Covas
- Regional Blood Center, Medical School of Hemocenter Foundation of Ribeirão Preto, University of São Paulo, São Paulo 14051-140, Brazil
| | - Maria E. A. Moraes
- Drug Research and Development Center, Federal University of Ceara, Ceará 60430-275, Brazil
| | - Wilson A. Silva
- Department of Genetics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Júlio C. Rosa-e-Silva
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- Laboratory for Translational Data Science, Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Rui A. Ferriani
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- National Institute of Hormones and Women’s Health (Hormona), CNPq, Porto Alegre 90035-003, Brazil
| | - Vitor M. Faça
- Regional Blood Center, Medical School of Hemocenter Foundation of Ribeirão Preto, University of São Paulo, São Paulo 14051-140, Brazil
- Department Biochemistry and Immunology, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Omero B. Poli-Neto
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- Laboratory for Translational Data Science, Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Daniel G. Tiezzi
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- Laboratory for Translational Data Science, Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
| | - Juliana Meola
- Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- Laboratory for Translational Data Science, Department of Gynecology and Obstetrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo 14049-900, Brazil
- National Institute of Hormones and Women’s Health (Hormona), CNPq, Porto Alegre 90035-003, Brazil
| |
Collapse
|
18
|
Tesarik J, Mendoza-Tesarik R. Molecular Clues to Understanding Causes of Human-Assisted Reproduction Treatment Failures and Possible Treatment Options. Int J Mol Sci 2022; 23:10357. [PMID: 36142268 PMCID: PMC9499616 DOI: 10.3390/ijms231810357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/01/2022] [Accepted: 09/04/2022] [Indexed: 11/16/2022] Open
Abstract
More than forty years after the first birth following in vitro fertilization (IVF), the success rates of IVF and of IVF-derived assisted reproduction techniques (ART) still remain relatively low. Interindividual differences between infertile couples and the nature of the problems underlying their infertility appear to be underestimated nowadays. Consequently, the molecular basis of each couple's reproductive function and of its disturbances is needed to offer an individualized diagnostic and therapeutic approaches to each couple, instead of applying a standard or minimally adapted protocols to everybody. Interindividual differences include sperm and oocyte function and health status, early (preimplantation) embryonic development, the optimal window of uterine receptivity for the implanting embryo, the function of the corpus luteum as the main source of progesterone production during the first days of pregnancy, the timing of the subsequent luteoplacental shift in progesterone production, and aberrant reactions of the uterine immune cells to the implanting and recently implanted embryos. In this article, the molecular basis that underlies each of these abnormalities is reviewed and discussed, with the aim to design specific treatment options to be used for each of them.
Collapse
|
19
|
James NE, Woodman M, De La Cruz P, Eurich K, Ozsoy MA, Schorl C, Hanley LC, Ribeiro JR. Adaptive transcriptomic and immune infiltrate responses in the tumor immune microenvironment following neoadjuvant chemotherapy in high grade serous ovarian cancer reveal novel prognostic associations and activation of pro-tumorigenic pathways. Front Immunol 2022; 13:965331. [PMID: 36131935 PMCID: PMC9483165 DOI: 10.3389/fimmu.2022.965331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
The high rate of ovarian cancer recurrence and chemoresistance necessitates further research into how chemotherapy affects the tumor immune microenvironment (TIME). While studies have shown that immune infiltrate increases following neoadjuvant (NACT) chemotherapy, there lacks a comprehensive understanding of chemotherapy-induced effects on immunotranscriptomics and cancer-related pathways and their relationship with immune infiltrate and patient responses. In this study, we performed NanoString nCounter® PanCancer IO360 analysis of 31 high grade serous ovarian cancer (HGSOC) patients with matched pre-treatment biopsy and post-NACT tumor. We observed increases in pro-tumorigenic and immunoregulatory pathways and immune infiltrate following NACT, with striking increases in a cohort of genes centered on the transcription factors ATF3 and EGR1. Using quantitative PCR, we analyzed several of the top upregulated genes in HGSOC cell lines, noting that two of them, ATF3 and AREG, were consistently upregulated with chemotherapy exposure and significantly increased in platinum resistant cells compared to their sensitive counterparts. Furthermore, we observed that pre-NACT immune infiltrate and pathway scores were not strikingly related to platinum free interval (PFI), but post-NACT immune infiltrate, pathway scores, and gene expression were. Finally, we found that higher levels of a cohort of proliferative and DNA damage-related genes was related to shorter PFI. This study underscores the complex alterations in the ovarian TIME following chemotherapy exposure and begins to untangle how immunologic factors are involved in mediating chemotherapy response, which will allow for the future development of novel immunologic therapies to combat chemoresistance.
Collapse
Affiliation(s)
- Nicole E. James
- Department of Obstetrics and Gynecology, Program in Women’s Oncology, Women and Infants Hospital, Providence, RI, United States
- Department of Obstetrics and Gynecology, Warren-Alpert Medical School of Brown University, Providence, RI, United States
- *Correspondence: Nicole E. James,
| | - Morgan Woodman
- Department of Obstetrics and Gynecology, Program in Women’s Oncology, Women and Infants Hospital, Providence, RI, United States
| | - Payton De La Cruz
- Pathobiology Graduate Program, Brown University, Providence, RI, United States
| | - Katrin Eurich
- Department of Obstetrics and Gynecology, Program in Women’s Oncology, Women and Infants Hospital, Providence, RI, United States
- Department of Obstetrics and Gynecology, Warren-Alpert Medical School of Brown University, Providence, RI, United States
| | - Melih Arda Ozsoy
- Department of Biochemistry, Brown University, Providence, RI, United States
| | - Christoph Schorl
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Linda C. Hanley
- Department of Pathology, Women and Infants Hospital, Providence, RI, United States
| | - Jennifer R. Ribeiro
- Department of Obstetrics and Gynecology, Program in Women’s Oncology, Women and Infants Hospital, Providence, RI, United States
- Department of Obstetrics and Gynecology, Warren-Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
20
|
Goharitaban S, Abedelahi A, Hamdi K, Khazaei M, Esmaeilivand M, Niknafs B. Role of endometrial microRNAs in repeated implantation failure (mini-review). Front Cell Dev Biol 2022; 10:936173. [PMID: 36060804 PMCID: PMC9437697 DOI: 10.3389/fcell.2022.936173] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/12/2022] [Indexed: 11/28/2022] Open
Abstract
MicroRNAs (miRNAs) play various roles in the implantation and pregnancy process. Abnormal regulation of miRNAs leads to reproductive disorders such as repeated implantation failure (RIF). During the window of implantation, different miRNAs are released from the endometrium, which can potentially reflect the status of the endometrium for in vitro fertilization (IVF). The focus of this review is to determine whether endometrial miRNAs may be utilized as noninvasive biomarkers to predict the ability of endometrium to implant and provide live birth during IVF cycles. The levels of certain miRNAs in the endometrium have been linked to implantation potential and pregnancy outcomes in previous studies. Endometrial miRNAs could be employed as non-invasive biomarkers in the assisted reproductive technology (ART) cycle to determine the optimal time for implantation. Few human studies have evaluated the association between ART outcomes and endometrial miRNAs in RIF patients. This review may pave the way for more miRNA transcriptomic studies on human endometrium and introduce a specific miRNA profile as a multivariable prediction model for choosing the optimal time in the IVF cycle.
Collapse
Affiliation(s)
- Sepide Goharitaban
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Ali Abedelahi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Kobra Hamdi
- Womens Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoumeh Esmaeilivand
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behrooz Niknafs
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- *Correspondence: Behrooz Niknafs, , 0000-0003-4438-1880
| |
Collapse
|
21
|
Shang J, Cheng YF, Li M, Wang H, Zhang JN, Guo XM, Cao DD, Yao YQ. Identification of Key Endometrial MicroRNAs and Their Target Genes Associated With Pathogenesis of Recurrent Implantation Failure by Integrated Bioinformatics Analysis. Front Genet 2022; 13:919301. [PMID: 35812749 PMCID: PMC9257071 DOI: 10.3389/fgene.2022.919301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/13/2022] [Indexed: 12/13/2022] Open
Abstract
Purpose: Recurrent implantation failure (RIF) is an enormous challenge for in vitro fertilization (IVF) clinicians. An understanding of the molecular mechanisms of RIF helps to predict prognosis and develop new therapeutic strategies. The study is designed to identify diagnostic biomarkers for RIF as well as the potential mechanisms underlying RIF by utilizing public databases together with experimental validation. Methods: Two microarray datasets of RIF patients and the healthy control endometrium were downloaded from the Gene Expression Omnibus (GEO) database. First, differentially expressed microRNAs (miRNAs) (DEMs) were identified and their target genes were predicted. Then, we identified differentially expressed genes (DEGs) and selected hub genes through protein-protein interaction (PPI) analyses. Functional enrichment analyses of DEGs and DEMs were conducted. Furthermore, the key DEMs which targeted these hub genes were selected to obtain the key miRNA–target gene network. The key genes in the miRNA-target gene network were validated by a single-cell RNA-sequencing dataset of endometrium from GEO. Finally, we selected two miRNA–target gene pairs for further experimental validation using dual-luciferase assay and quantitative polymerase chain reaction (qPCR). Results: We identified 49 DEMs between RIF patients and the fertile group and found 136,678 target genes. Then, 325 DEGs were totally used to construct the PPI network, and 33 hub genes were selected. Also, 25 DEMs targeted 16 key DEGs were obtained to establish a key miRNA–target gene network, and 16 key DEGs were validated by a single-cell RNA-sequencing dataset. Finally, the target relationship of hsa-miR-199a-5p-PDPN and hsa-miR-4306-PAX2 was verified by dual-luciferase assay, and there were significant differences in the expression of those genes between the RIF and fertile group by PCR (p < 0.05). Conclusion: We constructed miRNA–target gene regulatory networks associated with RIF which provide new insights regarding the underlying pathogenesis of RIF; hsa-miR-199a-5p-PDPN and hsa-miR-4306-PAX2 could be further explored as potential biomarkers for RIF, and their detection in the endometrium could be applied in clinics to estimate the probability of successful embryo transfer.
Collapse
Affiliation(s)
- Jin Shang
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Yan-Fei Cheng
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, China
| | - Min Li
- Department of Obstetrics and Gynecology, The Seventh Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hui Wang
- Department of Obstetrics and Gynecology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jin-Ning Zhang
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
| | - Xin-Meng Guo
- College of Medicine, Nankai University, Tianjin, China
| | - Dan-dan Cao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- *Correspondence: Dan-dan Cao, ; Yuan-Qing Yao,
| | - Yuan-Qing Yao
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Obstetrics and Gynecology, The Seventh Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Dan-dan Cao, ; Yuan-Qing Yao,
| |
Collapse
|
22
|
Shi S, Bonaccorsi-Riani E, Schurink I, van den Bosch T, Doukas M, Lila KA, Roest HP, Xhema D, Gianello P, de Jonge J, Verstegen MMA, van der Laan LJW. Liver Ischemia and Reperfusion Induce Periportal Expression of Necroptosis Executor pMLKL Which Is Associated With Early Allograft Dysfunction After Transplantation. Front Immunol 2022; 13:890353. [PMID: 35655777 PMCID: PMC9152120 DOI: 10.3389/fimmu.2022.890353] [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: 03/05/2022] [Accepted: 04/13/2022] [Indexed: 11/29/2022] Open
Abstract
Background Early allograft dysfunction (EAD) following liver transplantation (LT) remains a major threat to the survival of liver grafts and recipients. In animal models, it is shown that hepatic ischemia-reperfusion injury (IRI) triggers phosphorylation of Mixed Lineage Kinase domain-like protein (pMLKL) inducing necroptotic cell death. However, the clinical implication of pMLKL-mediated cell death in human hepatic IRI remains largely unexplored. In this study, we aimed to investigate the expression of pMLKL in human liver grafts and its association with EAD after LT. Methods The expression of pMLKL was determined by immunohistochemistry in liver biopsies obtained from both human and rat LT. Human liver biopsies were obtained at the end of preservation (T0) and ~1 hour after reperfusion (T1). The positivity of pMLKL was quantified electronically and compared in rat and human livers and post-LT outcomes. Multiplex immunofluorescence staining was performed to characterize the pMLKL-expressing cells. Results In the rat LT model, significant pMLKL expression was observed in livers after IRI as compared to livers of sham-operation animals. Similarly, the pMLKL score was highest after IRI in human liver grafts (in T1 biopsies). Both in rats and humans, the pMLKL expression is mostly observed in the portal triads. In grafts who developed EAD after LT (n=24), the pMLKL score at T1 was significantly higher as compared to non-EAD grafts (n=40). ROC curve revealed a high predictive value of pMLKL score at T1 (AUC 0.70) and the ratio of pMLKL score at T1 and T0 (pMLKL-index, AUC 0.82) for EAD. Liver grafts with a high pMLKL index (>1.64) had significantly higher levels of serum ALT, AST, and LDH 24 hours after LT compared to grafts with a low pMLKL index. Multivariate logistical regression analysis identified the pMLKL-index (Odds ratio=1.3, 95% CI 1.1-1.7) as a predictor of EAD development. Immunohistochemistry on serial sections and multiplex staining identified the periportal pMLKL-positive cells as portal fibroblasts, fibrocytes, and a minority of cholangiocytes. Conclusion Periportal pMLKL expression increased significantly after IRI in both rat and human LT. The histological score of pMLKL is predictive of post-transplant EAD and is associated with early liver injury after LT. Periportal non-parenchymal cells (i.e. fibroblasts) appear most susceptible to pMLKL-mediated cell death during hepatic IRI.
Collapse
Affiliation(s)
- Shaojun Shi
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Eliano Bonaccorsi-Riani
- Abdominal Transplant Unit, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium.,Pôle de Chirurgie Expérimentale et Transplantation Institute de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Ivo Schurink
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Thierry van den Bosch
- Department of Pathology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Michael Doukas
- Department of Pathology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Karishma A Lila
- Department of Pathology, Erasmus MC-University Medical Center, Rotterdam, Netherlands
| | - Henk P Roest
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Daela Xhema
- Pôle de Chirurgie Expérimentale et Transplantation Institute de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Pierre Gianello
- Pôle de Chirurgie Expérimentale et Transplantation Institute de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jeroen de Jonge
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Monique M A Verstegen
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
23
|
Kaminker JD, Timoshenko AV. Expression, Regulation, and Functions of the Galectin-16 Gene in Human Cells and Tissues. Biomolecules 2021; 11:1909. [PMID: 34944551 PMCID: PMC8699332 DOI: 10.3390/biom11121909] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 12/11/2022] Open
Abstract
Galectins comprise a family of soluble β-galactoside-binding proteins, which regulate a variety of key biological processes including cell growth, differentiation, survival, and death. This paper aims to address the current knowledge on the unique properties, regulation, and expression of the galectin-16 gene (LGALS16) in human cells and tissues. To date, there are limited studies on this galectin, with most focusing on its tissue specificity to the placenta. Here, we report the expression and 8-Br-cAMP-induced upregulation of LGALS16 in two placental cell lines (BeWo and JEG-3) in the context of trophoblastic differentiation. In addition, we provide the results of a bioinformatics search for LGALS16 using datasets available at GEO, Human Protein Atlas, and prediction tools for relevant transcription factors and miRNAs. Our findings indicate that LGALS16 is detected by microarrays in diverse human cells/tissues and alters expression in association with cancer, diabetes, and brain diseases. Molecular mechanisms of the transcriptional and post-transcriptional regulation of LGALS16 are also discussed based on the available bioinformatics resources.
Collapse
|