1
|
Wang G, Shen WB, Chen AW, Reece EA, Yang P. Diabetes and Early Development: Epigenetics, Biological Stress, and Aging. Am J Perinatol 2025; 42:977-987. [PMID: 39209306 DOI: 10.1055/a-2405-1493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Pregestational diabetes, either type 1 or type 2 diabetes, induces structural birth defects including neural tube defects and congenital heart defects in human fetuses. Rodent models of type 1 and type 2 diabetic embryopathy have been established and faithfully mimic human conditions. Hyperglycemia of maternal diabetes triggers oxidative stress in the developing neuroepithelium and the embryonic heart leading to the activation of proapoptotic kinases and excessive cell death. Oxidative stress also activates the unfolded protein response and endoplasmic reticulum stress. Hyperglycemia alters epigenetic landscapes by suppressing histone deacetylation, perturbing microRNA (miRNA) expression, and increasing DNA methylation. At cellular levels, besides the induction of cell apoptosis, hyperglycemia suppresses cell proliferation and induces premature senescence. Stress signaling elicited by maternal diabetes disrupts cellular organelle homeostasis leading to mitochondrial dysfunction, mitochondrial dynamic alteration, and autophagy impairment. Blocking oxidative stress, kinase activation, and cellular senescence ameliorates diabetic embryopathy. Deleting the mir200c gene or restoring mir322 expression abolishes maternal diabetes hyperglycemia-induced senescence and cellular stress, respectively. Both the autophagy activator trehalose and the senomorphic rapamycin can alleviate diabetic embryopathy. Thus, targeting cellular stress, miRNAs, senescence, or restoring autophagy or mitochondrial fusion is a promising approach to prevent poorly controlled maternal diabetes-induced structural birth defects. In this review, we summarize the causal events in diabetic embryopathy and propose preventions for this pathological condition. · Maternal diabetes induces structural birth defects.. · Kinase signaling and cellular organelle stress are critically involved in neural tube defects.. · Maternal diabetes increases DNA methylation and suppresses developmental gene expression.. · Cellular apoptosis and senescence are induced by maternal diabetes in the neuroepithelium.. · microRNAs disrupt mitochondrial fusion leading to congenital heart diseases in diabetic pregnancy..
Collapse
Affiliation(s)
- Guanglei Wang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Anna Wu Chen
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - E Albert Reece
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
2
|
Dong X, Wang H, Cai J, Wang Y, Chai D, Sun Z, Chen J, Li M, Xiao T, Shan C, Zhang JV, Yu M. ST6GALNAC1-mediated sialylation in uterine endometrial epithelium facilitates the epithelium-embryo attachment. J Adv Res 2025; 72:197-212. [PMID: 39111624 DOI: 10.1016/j.jare.2024.07.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/13/2024] Open
Abstract
INTRODUCTION Embryo implantation requires synergistic interaction between the embryo and the receptive endometrium. Glycoproteins and glycan-binding proteins are involved in endometrium-embryo attachment. Sialyl Tn (sTn), a truncated O-glycan, is catalyzed by ST6 N-Acetylgalactosaminide Alpha-2,6-Sialyltransferase 1 (ST6GALNAC1) and can be detected by specific Sialic-acid-binding immunoglobulin-like lectins (Siglecs). Whether the sTn-Siglecs axis supports embryo implantation remains unknown. OBJECTIVES This paper aims to study the role of ST6GALNAC1/sTn-Siglecs axis in embryo implantation. METHODS ST6GALNAC1 and sTn in human endometrium were analyzed by immunohistochemistry. An in vitro implantation model was conducted to evaluate the effects of ST6GALNAC1/sTn on the receptivity of human endometrial AN3CA cells to JAR spheroids. Immunoprecipitation combined with mass spectrometry analysis was carried out to identify the key proteins modified by sTn in endometrial cells. Siglec-6 in human embryos was analyzed by published single-cell RNA sequencing (scRNA-seq) datasets. Protein interaction assay was applied to verify the bond between the Siglec-6 with sTn-modified CD44. St6galnac1 siRNAs and anti-sTn antibodies were injected into the uterine horn of the mouse at the pre-implantation stage to evaluate the role of endometrial St6galnac1/sTn in embryo implantation. Siglec-G in murine embryos was analyzed by immunofluorescence staining. The function of Siglec-G is evidenced by uterine horn injection and protein interaction assay. RESULTS Both human and murine endometrium at the receptive stage exhibit higher ST6GALNAC1 and sTn levels compared to the non-receptive stage. Overexpression of ST6GALNAC1 significantly enhanced the receptivity of AN3CA cells to JAR spheroids. Inhibition of endometrial ST6GALNAC1/sTn substantially impaired embryo implantation in vivo. CD44 was identified as a carrier for sTn in the endometrial cells of both species. Siglec-6 and Siglec-G, expressed in the embryonic trophectoderm, were found to promote embryo attachment, which may be achieved through binding with sTn-modified CD44. CONCLUSION ST6GALNAC1-regulated sTn in the endometrium aids in embryo attachment through interaction with trophoblastic Siglecs.
Collapse
Affiliation(s)
- Xinyue Dong
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, China; College of Life Science, Northeast Forestry University, Harbin, China
| | - Hao Wang
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, China
| | - Jinxuan Cai
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, China
| | - Yichun Wang
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, China; Department of Medical Oncology, The Fourth Hospital of China Medical University, Shenyang, Liaoning, China
| | - Dezhi Chai
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, China
| | - Zichen Sun
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, China
| | - Jie Chen
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, China
| | - Mengxia Li
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, China
| | - Tianxia Xiao
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, China
| | - Chunhua Shan
- College of Life Science, Northeast Forestry University, Harbin, China
| | - Jian V Zhang
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, China; Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, China.
| | - Ming Yu
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, China.
| |
Collapse
|
3
|
Wu B, Neupane J, Zhou Y, Zhang J, Chen Y, Surani MA, Zhang Y, Bao S, Li X. Stem cell-based embryo models: a tool to study early human development. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1626-1645. [PMID: 39969747 DOI: 10.1007/s11427-024-2741-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/23/2024] [Indexed: 02/20/2025]
Abstract
How a mammalian fertilized egg acquires totipotency and develops into a full-term offspring is a fundamental scientific question. Human embryonic development is difficult to study due to limited resources, technical challenges and ethics. Moreover, the precise regulatory mechanism underlying early human embryonic development remains unknown. In recent years, the emergence of stem cell-based embryo models (SCBEM) provides the opportunity to reconstitute pre- to post-implantation development in vitro. These models to some extent mimic the embryo morphologically and transcriptionally, and thus may be used to study key events in mammalian pre- and post-implantation development. Many groups have successfully generated SCBEM of the mouse and human. Here, we provide a comparative review of the mouse and human SCBEM, discuss the capability of these models to mimic natural embryos and give a perspective on their potential future applications.
Collapse
Affiliation(s)
- Baojiang Wu
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - Jitesh Neupane
- The Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Yang Zhou
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - Jingcheng Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Yanglin Chen
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China
| | - M Azim Surani
- The Gurdon Institute, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Yong Zhang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China.
| | - Siqin Bao
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
| | - Xihe Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
- Research Center for Animal Genetic Resources of Mongolia Plateau, College of Life Sciences, Inner Mongolia University, Hohhot, 010020, China.
- Inner Mongolia Saikexing Institute of Breeding and Reproductive Biotechnology in Domestic Animals, Hohhot, 011517, China.
| |
Collapse
|
4
|
Zhao Z, Chen X, Wang S, Fu M, Shen H, Li J, Xu J, Qin J, Shi C. Chemically defined and xeno-free media enables the derivation of human extended pluripotent stem cell lines from discarded blastocysts with a high efficiency. Cell Biosci 2025; 15:72. [PMID: 40448189 PMCID: PMC12123846 DOI: 10.1186/s13578-025-01410-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 05/13/2025] [Indexed: 06/02/2025] Open
Abstract
Human extended pluripotent stem cells (hEPS) had been reported to be derived from discarded blastocysts, whereas the derivation method of hEPS cells was extremely complex in this protocol with feeder and animal serum conditions, which also limited the safety and homogeneity of hEPS. Here, we report an optimized, highly efficient protocol by utilizing chemically defined and xeno-free media for the derivation of human extended pluripotent stem cell lines from discarded blastocysts. With this method, we successfully isolated hEPS cell lines from discarded blastocysts with an efficiency of 46%. Chemically defined and xeno-free media simplified the process of hEPS cell isolation with a higher survival rate of cell aggregation passaging from outgrowth. To our knowledge, this is the first report of hEPS cells being efficiently derived from discarded blastocysts under chemically defined and xeno-free conditions.
Collapse
Affiliation(s)
- Zhuran Zhao
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Department of Pediatrics, Peking University People's Hospital, Beijing, 100044, China
| | - Xi Chen
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Shan Wang
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, 100044, China
- Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Min Fu
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Huan Shen
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, 100044, China
| | - Jiayu Li
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Stem Cell Research Center, Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Jun Xu
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Stem Cell Research Center, Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Jiong Qin
- Department of Pediatrics, Peking University People's Hospital, Beijing, 100044, China.
- Epilepsy Center, Peking University People's Hospital, Beijing, China#, China.
| | - Cheng Shi
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Peking University People's Hospital, Peking University, Beijing, 100044, China.
| |
Collapse
|
5
|
Tang X, Zhao H, Ding Y, Qin Y, Yang X, Jiang X, Zhou H, Liu B. Enhancement of endometrial receptivity by Bushen Zhuyun Decoction via cryptotanshinone-mediated TRIM28 induction and HIF-1α suppression. JOURNAL OF ETHNOPHARMACOLOGY 2025:119943. [PMID: 40449692 DOI: 10.1016/j.jep.2025.119943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 06/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Bushen Zhuyun Decoction (BSZYD), a traditional Chinese remedy, has demonstrated clinical efficacy in the treatment of luteal phase deficiency (LPD), though its mechanistic pathways remain largely undefined. AIM OF THE STUDY This study aims to elucidate the mechanism by which BSZYD enhances endometrial receptivity. MATERIALS AND METHODS In an LPD rat model induced by RU-486 and treated with BSZYD, molecular markers of endometrial receptivity were evaluated using scanning electron microscopy (SEM). Furthermore, these markers were analyzed in the RL95-2 human adenocarcinoma cell line following knockdown of Tripartite motif containing 28 (TRIM28). Network pharmacology and UPLC-MS/MS were utilized to identify bioactive components that modulate Hypoxia-inducible factor-1 (HIF-1) signaling, followed by validation through molecular docking. The interaction between HIF-1α and TRIM28 was assessed using co-immunoprecipitation (Co-IP) and confocal microscopy. The effect of cryptotanshinone on TRIM28 expression was also examined in RL95-2 cells. RESULTS BSZYD significantly increased the number of embryo implantation sites and reduced endometrial reactive oxygen species (ROS) levels in LPD rats. TRIM28 was found to be crucial for BSZYD's enhancement of endometrial receptivity. Cryptotanshinone, a key component of BSZYD, downregulated HIF-1α expression in RL95-2 cells. The interaction between HIF-1α and TRIM28 was confirmed both in vivo and in vitro. In vitro, cryptotanshinone mitigated H2O2-induced oxidative stress. Furthermore, HIF-1α agonist administration attenuated BSZYD's ability to induce TRIM28 expression. CONCLUSIONS BSZYD and its bioactive constituent, cryptotanshinone, promote endometrial receptivity by inhibiting HIF-1α and upregulating TRIM28. These findings offer novel molecular targets and pharmacological insights for the prevention and treatment of LPD.
Collapse
Affiliation(s)
- Xingran Tang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Huijin Zhao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yinyin Ding
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yajie Qin
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaotian Yang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xiaoyue Jiang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China; Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Huifang Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Bei Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| |
Collapse
|
6
|
Lou J, Li W, Chen P, Chen H, Shakoor A, Chen Y, Hua J, Wang Y, Zhang S. Application of induced pluripotent stem cells in the conservation of endangered animals. Stem Cell Res Ther 2025; 16:261. [PMID: 40437543 PMCID: PMC12121184 DOI: 10.1186/s13287-025-04392-5] [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: 11/25/2024] [Accepted: 05/14/2025] [Indexed: 06/01/2025] Open
Abstract
The accelerating biodiversity crisis urgently demands innovative approaches that transcend traditional conservation strategies, which are often constrained by genetic bottlenecks and disease risks. Induced pluripotent stem cells (iPSCs) technology emerges as a transformative solution, enabling non-invasive genetic preservation and multi-pathway species recovery. This review synthesizes advances in reprogramming somatic cells from endangered species into iPSCs through integration-free strategies, such as mRNA, Sendai virus, episomal systems, adenoviruses and chemical induction, thereby reducing genomic instability. We highlight breakthroughs in differentiating iPSCs into functional gametes for assisted reproduction and blastoids formation for embryonic reconstruction, circumventing donor oocyte dependency and genetic homogeneity risks. Despite challenges in lineage specification and epigenetic fidelity, combining iPSC biobanking with ecosystem management enables large-scale genetic rescue. By combining these technologies with ethical frameworks and habitat restoration, the plasticity of cells may be transformed into population resilience, potentially redefining biodiversity conservation.
Collapse
Affiliation(s)
- Jiao Lou
- College of Veterinary Medicine, Shaanxi Stem Cell Engineering Research Center, Northwest A&F University, Yangling, 712100, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, China
| | - Weina Li
- College of Veterinary Medicine, Shaanxi Stem Cell Engineering Research Center, Northwest A&F University, Yangling, 712100, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, China
| | - Panlong Chen
- College of Veterinary Medicine, Shaanxi Stem Cell Engineering Research Center, Northwest A&F University, Yangling, 712100, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, China
| | - Haiyan Chen
- College of Veterinary Medicine, Shaanxi Stem Cell Engineering Research Center, Northwest A&F University, Yangling, 712100, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, China
| | - Amna Shakoor
- College of Veterinary Medicine, Shaanxi Stem Cell Engineering Research Center, Northwest A&F University, Yangling, 712100, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, China
| | - Yunlong Chen
- College of Veterinary Medicine, Shaanxi Stem Cell Engineering Research Center, Northwest A&F University, Yangling, 712100, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Stem Cell Engineering Research Center, Northwest A&F University, Yangling, 712100, China
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, China
| | - Yan Wang
- College of Veterinary Medicine, Shaanxi Stem Cell Engineering Research Center, Northwest A&F University, Yangling, 712100, China.
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, China.
| | - Shiqiang Zhang
- College of Veterinary Medicine, Shaanxi Stem Cell Engineering Research Center, Northwest A&F University, Yangling, 712100, China.
- Key Laboratory of Livestock Biology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
7
|
Ziyafati Kafi F, Eslami N, Shekari F, Bazrgar M. Theranostic potential of extracellular vesicles in reproductive tracts: implications for recurrent implantation failure. Mol Biol Rep 2025; 52:502. [PMID: 40411714 DOI: 10.1007/s11033-025-10619-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 05/15/2025] [Indexed: 05/26/2025]
Abstract
Embryo implantation is a critical step at the beginning of pregnancy, occurring during a specific and limited period known as the "implantation window". Successful implantation involves various signaling pathways and molecular interactions. Recent studies have highlighted the importance of extracellular vesicles (EVs) in mediating these complex interactions. Different cell types release EVs to transfer signals to other cells or tissues. Additionally, emerging evidence suggests that EVs regulate signaling between the developing embryo and endometrium. In this review, we summarize current findings that highlight the role of EVs in the reproductive tract, gamete production, and their potential roles in embryo development and implantation. We then examine studies emphasizing the role of EVs in embryo-maternal interactions and implantation. Finally, we will explore the theranostic potential of EVs in various aspects of assisted reproductive technology (ART), including modulation of embryo-maternal interactions, enhancement of embryo quality, and improvement of endometrial receptivity. A more comprehensive understanding of EVs in the pathology of recurrent implantation failure could support the development of personalized treatments.
Collapse
Affiliation(s)
- Fatemeh Ziyafati Kafi
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Nasim Eslami
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Advanced Therapy Medicinal Product Technology Development Center (ATMP-TDC), Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Masood Bazrgar
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
8
|
Tesarik J. Noninvasive Biomarkers of Human Embryo Developmental Potential. Int J Mol Sci 2025; 26:4928. [PMID: 40430065 PMCID: PMC12112732 DOI: 10.3390/ijms26104928] [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: 04/17/2025] [Revised: 05/15/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
There are two types of noninvasive biomarkers of human embryo developmental potential: those based on a direct assessment of embryo morphology over time and those using spent media after embryo in vitro culture as source of information. Both are derived from previously acquired knowledge on different aspects of pre-implantation embryo development. These aspects include embryo morphology and kinetics, chromosomal ploidy status, metabolism, and embryonic gene transcription, translation, and expression. As to the direct assessment of morphology and kinetics, pertinent data can be obtained by analyzing sequential microscopic images of in vitro cultured embryos. Spent media can serve a source of genomic, metabolomic, transcriptomic and proteomic markers. Methods used in the early pioneering studies, such as microscopy, fluorescence in situ hybridization, autoradiography, electrophoresis and immunoblotting, or enzyme-linked immunosorbent assay, are too subjective, invasive, and/or time-consuming. As such, they are unsuitable for the current in vitro fertilization (IVF) practice, which needs objective, rapid, and noninvasive selection of the best embryo for uterine transfer or cryopreservation. This has been made possible by the use of high-throughput techniques such as time-lapse (for direct embryo evaluation), next-generation sequencing, quantitative real-time polymerase chain reaction, high-performance liquid chromatography, nanoparticle tracking analysis, flow cytometry, mass spectroscopy, Raman spectroscopy, near-infrared spectroscopy, and nuclear magnetic resonance spectroscopy (for spent culture media analysis). In this review, individual markers are presented systematically, with each marker's history and current status, including available methodologies, strengths, and limitations, so as to make the essential information accessible to all health professionals, even those whose expertise in the matter is limited.
Collapse
Affiliation(s)
- Jan Tesarik
- MARGen (Molecular Assisted Reproduction and Genetics) Clinic, Calle Gracia, 36, 18002 Granada, Spain
| |
Collapse
|
9
|
Zhang X, Zhu Z, Shen C, Tang G. The causal effects of systemic antioxidant capacity on male infertility: A two-sample mendelian randomization analysis. Sci Rep 2025; 15:17009. [PMID: 40379801 PMCID: PMC12084361 DOI: 10.1038/s41598-025-02243-0] [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: 05/11/2024] [Accepted: 05/12/2025] [Indexed: 05/19/2025] Open
Abstract
The present research aimed to assess the potential causal relationship between systemic antioxidant capacity and male infertility using a two-sample Mendelian randomization approach. The primary MR analysis utilized the inverse variance weighted (IVW)method, supplemented by complementary analyses including MR-Egger, weighted mode, simple mode, and weighted median methods. For male infertility, the available summarized data were gained from the open database (IEU OPEN GWAS PROJECT), which includes a total of 680 male patients with infertility and 72,799 controls of European population.10 biomarkers related to systemic antioxidant capacity were examined to investigate their potential association with male infertility, including glutathione S-transferase (GST), superoxide dismutase(SOD), glutathione peroxidase(GPX), catalase (CAT), total bilirubin, albumin, α-tocopherol, ascorbate, retinol, and uric acid. MR analyses using IVW mode revealed that genetically determined systemic antioxidant capacity biomarkers had no causal effects on male infertility risk, including GST(OR = 1.08, 95%CI: 0.91-1.29, P = 0.35), SOD(OR = 0.83, 95%CI: 0.66-1.04, P = 0.11), GPX(OR = 1.12, 95%CI: 0.92-1.36,P = 0.26), CAT(OR = 1.04, 95%CI: 0.83-1.29, P = 0.75), total bilirubin(OR = 0.98, 95%CI: 0.94-1.01, P = 0.18), albumin(OR = 1.14, 95%CI: 0.73-1.76, P = 0.57), α-tocopherol(OR = 0.56, 95%CI: 0.03-9.38, P = 0.69), ascorbate(OR = 1.06, 95%CI: 0.24-4.60, P = 0.94), retinol(OR = 1.29, 95%CI: 0.34-4.96, P = 0.71), and uric acid (OR = 0.88, 95% CI : 0.67-1.17, P = 0.39). The current study found no significantly causal link between systemic antioxidant capacity and male infertility. Further research with larger sample sizes and data from different ethnicities is needed.
Collapse
Affiliation(s)
- Xiaolong Zhang
- Department of Urology, Shaoxing People's Hospital(The First Affiliated Hospital, Shaoxing University), 568 Zhongxing North Road, 312000, Shaoxing, Zhejiang, China
| | - Zhirong Zhu
- Department of Urology, Shaoxing People's Hospital(The First Affiliated Hospital, Shaoxing University), 568 Zhongxing North Road, 312000, Shaoxing, Zhejiang, China
| | - Chaodong Shen
- Department of Urology, Shaoxing People's Hospital(The First Affiliated Hospital, Shaoxing University), 568 Zhongxing North Road, 312000, Shaoxing, Zhejiang, China
| | - Guiliang Tang
- Department of Urology, Shaoxing People's Hospital(The First Affiliated Hospital, Shaoxing University), 568 Zhongxing North Road, 312000, Shaoxing, Zhejiang, China.
| |
Collapse
|
10
|
Li J, Liu T, Xian M, Wei J. Therapeutic applications of exercise in neurodegenerative diseases: focusing on the mechanism of SIRT1. Mol Cell Biochem 2025:10.1007/s11010-025-05299-8. [PMID: 40358811 DOI: 10.1007/s11010-025-05299-8] [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: 02/06/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025]
Abstract
Neurodegenerative diseases comprise a group of central nervous system disorders marked by progressive neuronal degeneration and dysfunction. Their pathogenesis is multifactorial, involving oxidative stress, mitochondrial dysfunction, excitotoxicity, and neuroinflammation. Recent research has highlighted the potential of exercise as a non-pharmacological intervention for both the prevention and treatment of these disorders. In particular, exercise has received growing attention for its capacity to upregulate the expression and activity of SIRT1, a critical mediator of neuroprotection via downstream signaling pathways. SIRT1, a key member of the Sirtuin family, is a nicotinamide adenine dinucleotide (NAD +)-dependent class III histone deacetylase. It plays an essential role in regulating cellular metabolism, energy homeostasis, gene expression, and cellular longevity. In the context of neurodegenerative diseases, SIRT1 confers neuroprotection by modulating multiple signaling cascades through deacetylation, suppressing neuronal apoptosis, and promoting neural repair and regeneration. Exercise enhances SIRT1 expression and activity by increasing NAD + synthesis and utilization, improving intracellular redox balance, alleviating oxidative stress-induced inhibition of SIRT1, and thereby promoting its activation. Moreover, exercise may indirectly modulate SIRT1 function by influencing interacting molecular networks. This review summarizes recent advances in the therapeutic application of exercise for neurodegenerative diseases, with a focus on SIRT1 as a central mechanism. It examines how exercise mediates neuroprotection through the regulation of SIRT1 and its associated molecular mechanisms and signaling pathways. Finally, the paper discusses the potential applications and challenges of integrating exercise and SIRT1-targeted strategies in the management of neurodegenerative diseases, offering novel perspectives for the development of innovative treatments and improvements in patients' quality of life.
Collapse
Affiliation(s)
- Jingwen Li
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, 475004, Henan, China
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Tingting Liu
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Meiyan Xian
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Jianshe Wei
- Institute for Sports and Brain Health, School of Physical Education, Henan University, Kaifeng, 475004, Henan, China.
- Institute for Brain Sciences Research, School of Life Sciences, Henan University, Kaifeng, 475004, China.
| |
Collapse
|
11
|
Sun X, Feng Y, Xie L, Wu Y, Mao J, Zhang Y, Zhang L, Yuan X, Ni J, Xiao X, Liu H, Dai L, Ma F. Maternal plasma extracellular vesicles tsRNA as potential biomarkers for assessing preterm labor risk. BMC Pregnancy Childbirth 2025; 25:553. [PMID: 40348952 PMCID: PMC12065249 DOI: 10.1186/s12884-025-07672-3] [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: 01/15/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Spontaneous preterm labor (PTL) accounts for approximately 70% of preterm births, posing significant risks to both maternal and neonatal health. Current predictive biomarkers lack sufficient reliability, underscoring the need for non-invasive and dependable indicators. Emerging research indicates that tRNA-derived small RNAs (tsRNAs) are involved in various diseases; however, their potential association with PTL remains underexplored. METHODS Bioinformatics analyses of public GEO datasets (PRJNA415953 and PRJNA428989) were conducted to identify tsRNAs associated with PTL. Validation was performed using plasma extracellular vesicles samples collected at 12 weeks of gestation from PTL patients (n = 45) and healthy controls (n = 38). Functional assays were used to assess the impact of tsRNA1 (tRNA-Gly-GCC-5p-tRF-921) on extravillous trophoblast (EVT) function, including apoptosis, migration, invasion, and endothelial-like tube formation in HTR8/SVneo cells. Transcriptomic sequencing was conducted to identify tsRNA1-mediated pathways, and DNA methylation patterns were predicted based on the transcriptomic data. Statistical significance was determined using Student's t-test. RESULTS Two tsRNAs, tsRNA1 and tsRNA3 (tRNA-Gly-GCC-5p-tR-half-368), were significantly upregulated in PTL patient samples compared to controls. Overexpression of tsRNA1 impaired EVT function, increased apoptosis, and altered DNA methylation profiles, implicating its critical role in PTL mechanisms. CONCLUSIONS This study identifies tsRNA1 as a key regulator of EVT dysfunction and placental pathology in PTL. The findings provide novel insights into the mechanistic role of tsRNAs in PTL and highlight tsRNA1 as a promising biomarker for early risk stratification and prediction of the condition. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Xinrui Sun
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ying Feng
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Liang Xie
- The Joint Laboratory for Pulmonary Development and Related Diseases, West China Second University Hospital, West China Institute of Women and Children's Health, Sichuan University, Chengdu, Sichuan, China
| | - Yilun Wu
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jia Mao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, State Key Laboratory of Biotherapy and Cancer Center, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Yi Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Linyu Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xin Yuan
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiangping Ni
- College of Horticulture, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Xue Xiao
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China.
| | - Hanmin Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China.
- Department of Pediatric Pulmonology and Immunology, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Li Dai
- National Center for Birth Defects Monitoring, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- National Health Commission Key Laboratory of Chronobiology (Sichuan University), Chengdu, Sichuan, China.
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China.
| | - Fang Ma
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), West China Second University Hospital, Ministry of Education, Sichuan University, Chengdu, Sichuan, 610041, P. R. China.
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
12
|
Shi Y, Wang H, Chai M, Ji M, Zhao W, Xu Q, Yan T, Liu Z, Weng X. The analysis of X chromosome activity of porcine embryonic stem Cells: Study based on parthenogenetic embryonic stem cells with LCDM medium. Theriogenology 2025; 244:117479. [PMID: 40367543 DOI: 10.1016/j.theriogenology.2025.117479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 05/08/2025] [Accepted: 05/08/2025] [Indexed: 05/16/2025]
Abstract
The derivation of porcine embryonic stem cell (pESC) lines remains a major challenge in this field. To date, the porcine naïve ESCs have yet to be successfully established, and standardized criteria for their characterization and evaluation are still lacking. The regulation of X-chromosome activity integrates information from embryonic development and the dosage of sex chromosomes, which is closely associated with the pluripotent state of embryonic stem cells. In this study, we aimed to establish pESC lines in LCDM medium from porcine blastocyst-stage embryos, and analyzed the features of ESCs from the sight of X chromosome activity. We assessed molecular markers and epigenetic characteristics to confirm pluripotency and X chromosome activity in porcine parthenogenetic ESCs (named as ppLCDM) using XIST RNA-FISH, immunofluorescence staining, single-cell RNA sequencing (scRNA-seq), and other techniques. Results showed that ppLCDM cells expressed most pluripotent markers. The percentage of ppLCDM cells exhibiting H3K27me3 and XIST aggregation signals increased with passage, indicating the progressive establishment of X-chromosome inactivation (XCI). Meanwhile, the pluripotency of most ppLCDM cells gradually declined during extended passaging. However, two distinct patterns of ppLCDM cells were observed from passage 35 (type I cells, P35-I) displayed normal XCI states, while type II cells (P35-II) exhibited X-chromosome erosion-like state, characterized by the loss of aggregation signals, abnormal X-linked gene ratios. Particularly, the pluripotency of ppLCDM cells with an X-chromosome erosion-like state undergoes unusual changes compared to normal cells. These findings indicate that X chromosome activity is closely associated with the pluripotent state of porcine ESCs and that heterogeneity in X chromosome activity arises during passaging. Our research provides crucial insights into X chromosome dynamics in large-animal ESC models and contribute to ongoing efforts to establish stable naïve pESC lines.
Collapse
Affiliation(s)
- Yu Shi
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China; Engineering Research Center of Intelligent Breeding and Farming of Pig in Northern Cold Region, Ministry of Education, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China
| | - Hongxing Wang
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China; Engineering Research Center of Intelligent Breeding and Farming of Pig in Northern Cold Region, Ministry of Education, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China
| | - Mengjia Chai
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China; Engineering Research Center of Intelligent Breeding and Farming of Pig in Northern Cold Region, Ministry of Education, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China
| | - Mengru Ji
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China; Engineering Research Center of Intelligent Breeding and Farming of Pig in Northern Cold Region, Ministry of Education, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China
| | - Wenqian Zhao
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China; Engineering Research Center of Intelligent Breeding and Farming of Pig in Northern Cold Region, Ministry of Education, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China
| | - Qianqian Xu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China; Engineering Research Center of Intelligent Breeding and Farming of Pig in Northern Cold Region, Ministry of Education, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China
| | - Tingsheng Yan
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China; Engineering Research Center of Intelligent Breeding and Farming of Pig in Northern Cold Region, Ministry of Education, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China
| | - Zhonghua Liu
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China; Engineering Research Center of Intelligent Breeding and Farming of Pig in Northern Cold Region, Ministry of Education, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China.
| | - Xiaogang Weng
- Key Laboratory of Animal Cellular and Genetics Engineering of Heilongjiang Province, College of Life Science, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China; Engineering Research Center of Intelligent Breeding and Farming of Pig in Northern Cold Region, Ministry of Education, Northeast Agricultural University, Harbin, 150030, Heilongjiang, PR China.
| |
Collapse
|
13
|
Zimmerlin L, Angarita A, Park TS, Evans-Moses R, Thomas J, Yan S, Uribe I, Vegas I, Kochendoerfer C, Buys W, Leung AKL, Zambidis ET. Proteogenomic reprogramming to a functional human blastomere-like stem cell state via a PARP-DUX4 regulatory axis. Cell Rep 2025; 44:115671. [PMID: 40338744 DOI: 10.1016/j.celrep.2025.115671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 02/17/2025] [Accepted: 04/16/2025] [Indexed: 05/10/2025] Open
Abstract
Here, we show that conventional human pluripotent stem cells cultured with non-specific tankyrase-PARP1-inhibited conditions underwent proteogenomic reprogramming to functional blastomere-like tankyrase/PARP inhibitor-regulated naive stem cells (TIRN-SC). TIRN-SCs concurrently expressed hundreds of pioneer factors in hybrid 2C-8C-morula-ICM programs that were augmented by induced expression of DUX4. Injection of TIRN-SCs into 8C-staged murine embryos equipotently differentiated human cells to the extra-embryonic and embryonic compartments of chimeric blastocysts and fetuses. Ectopic expression of murine-E-Cadherin in TIRN-SCs further enhanced interspecific chimeric tissue targeting. TIRN-SC-derived trophoblast stem cells efficiently generated placental chimeras. Proteome-ubiquitinome analyses revealed increased TNKS and reduced PARP1 levels and an ADP-ribosylation-deficient, hyper-ubiquitinated proteome that impacted expression of both tankyrase and PARP1 substrates. ChIP-seq of NANOG-SOX2-OCT4 and PARP1 (NSOP) revealed genome-wide NSOP co-binding at DUX4-accessible enhancers of embryonic lineage factors; suggesting a DUX4-NSOP axis regulated TIRN-SC lineage plasticity. TIRN-SCs may serve as valuable models for studying the proteogenomic regulation of pre-lineage human embryogenesis. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Ludovic Zimmerlin
- Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Ariana Angarita
- Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Tea Soon Park
- Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Rebecca Evans-Moses
- Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Justin Thomas
- Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sirui Yan
- Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Isabel Uribe
- Departments of Biochemistry and Molecular Biology, The Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Isabella Vegas
- Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Clara Kochendoerfer
- Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Willem Buys
- Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Anthony K L Leung
- Departments of Biochemistry and Molecular Biology, The Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Elias T Zambidis
- Institute for Cell Engineering, The Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
14
|
Wang H, Qiu J, Lin Y, Bai X, Wei X. A Cartilaginous Organoid System Derived From Human Expanded Pluripotent Stem Cells (hEPSCs). Bio Protoc 2025; 15:e5304. [PMID: 40364989 PMCID: PMC12067298 DOI: 10.21769/bioprotoc.5304] [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: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
The development of human organotypic models of cartilage provides essential insights into chondrogenesis and chondrocyte hypertrophy while enabling advanced applications in drug discovery, gene editing, and tissue regeneration. Here, we present a robust and efficient protocol for differentiating human expanded pluripotent stem cells (hEPSCs) into hypertrophic chondrocytes through a sclerotome intermediate. The protocol involves initial sclerotome induction, followed by 3D chondrogenic culture and subsequent hypertrophic maturation induced by bone morphogenetic protein-4 (BMP4), thyroid hormone (T3), and β-glycerophosphate. This protocol also allows for sensitive testing of the effects of various compounds on hypertrophic differentiation during the maturation process. Furthermore, we identify an α-adrenergic receptor antagonist, phentolamine, as an inhibitor of hypertrophic differentiation. This organoid system provides a practical platform for exploring cartilage hypertrophy mechanisms and testing therapeutic strategies for cartilage regeneration. Key features • This differentiation protocol generates hypertrophic chondrocytes from hEPSCs through a sclerotome intermediate. • This protocol facilitates sensitive testing of compounds during the hypertrophic maturation stage, enabling the study of molecular mechanisms and therapeutic interventions for cartilage hypertrophy. • This protocol identifies the α-adrenergic receptor antagonist phentolamine as a modulator of hypertrophic differentiation.
Collapse
Affiliation(s)
- Hong Wang
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Jingyang Qiu
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yin Lin
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaochun Bai
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xiaocui Wei
- Department of Stomatology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Wang H, Zhong L, Wang Z, Xiang J, Pei D. Wnt Inhibition Safeguards Porcine Embryonic Stem Cells From the Acquisition of Extraembryonic Endoderm Cell Fates. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416802. [PMID: 40063421 PMCID: PMC12061302 DOI: 10.1002/advs.202416802] [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] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/25/2025] [Indexed: 05/10/2025]
Abstract
Porcine embryonic stem cells (ESCs) are excellent models for exploring embryogenesis, producing genetically enhanced farm animals, and improving breeding. Various chemicals have been applied to generate porcine ESCs from embryos, which differ from mouse and human ESC derivation. Wnt inhibitors XAV939 or IWR1 are required to isolate and maintain porcine ESCs. How Wnt inhibitors specify porcine ESC fate decisions remains poorly understood. Additionally, whether porcine ESCs can be converted to extraembryonic endoderm (XEN) cells without genetic interventions has not been reported. Here, it is reported that Wnt inhibitors (i.e., XAV939 and IWR1) safeguard porcine ESCs from acquiring the XEN lineage. Porcine ESCs rely on Wnt inhibitors to maintain pluripotency. Without them, porcine ESCs exit from pluripotency and convert to XEN cells. An efficient strategy and culture conditions are further developed to directly derive porcine XEN cells from ESCs without gene editing. The resulting XEN cells from ESCs exhibit similar transcriptome and chromatin accessibility features to XEN cells from embryos and contribute to mouse extraembryonic tissues. This study will deepen the understanding of porcine pluripotency, lay the foundation for deriving high-quality porcine ESCs with germline chimerism and transmission, and provide valuable materials to study extraembryonic development and lineage segregation in livestock.
Collapse
Affiliation(s)
- Hanning Wang
- Laboratory of Cell Fate ControlSchool of Life SciencesWestlake UniversityHangzhou310030China
| | - Liang Zhong
- Hebei Provincial Key Laboratory of Basic Medicine for DiabetesThe Shijiazhuang Second HospitalShijiazhuang050051China
| | - Zhuangfei Wang
- Laboratory of Cell Fate ControlSchool of Life SciencesWestlake UniversityHangzhou310030China
| | - Jinzhu Xiang
- Laboratory of Cell Fate ControlSchool of Life SciencesWestlake UniversityHangzhou310030China
| | - Duanqing Pei
- Laboratory of Cell Fate ControlSchool of Life SciencesWestlake UniversityHangzhou310030China
- Westlake Laboratory of Life Sciences and BiomedicineHangzhou310030China
| |
Collapse
|
16
|
Cai L, Lv M, Wei J, Liu C, Li Y, Liao Z, Li T, Zhang H, Xi L, Sui C. Mir-218-5p from Extracellular Vesicles of Endometrium in Patients with Recurrent Implantation Failure Impairs Pre-Implantation Embryo Development. Int J Nanomedicine 2025; 20:5661-5679. [PMID: 40331233 PMCID: PMC12052006 DOI: 10.2147/ijn.s508491] [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: 12/10/2024] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
Background Recurrent implantation failure (RIF) presents a crucial obstacle to in vitro fertilization success. Previous research has shown that small extracellular vesicles (EVs) from endometrial RIF patients hinder embryo development, yet the underlying mechanism and potential solutions remain largely unexplored. In this study, we aimed to investigate the effectiveness of miR-218-5p as a molecular factor in RIF-EVs. Our findings revealed that miR-218-5p disrupted mouse embryo development, and this effect could be reversed by engineered extracellular vesicles (E-EVs) containing anti-miR-218-5p. Methods The percentage of blastocyst development and hatching rates, embryo morphology, and the total cell number were measured. RNA-sequencing was used to analyze transcriptional changes in embryos post miR-218-5p agomir treatment. The abnormal segregation genes of trophectoderm (TE) and inner cell mass (ICM) were visualized via qRT-PCR and immunofluorescence staining. The E-EVs were using the EVs derived from Human Umbilical Cord Mesenchymal Stem Cells (HUMSCs). Characteristics of the EVs were measured using Western blotting, nanoparticle tracking analysis, and transmission electron microscopy. EVs internalization was visualized using BODIPY TR ceramide staining. Results Mouse embryos were arrested at the morula stage and demonstrated reduced blastocyst and hatching rates following miR-218-5p agomir treatment (P < 0.001). Essential transcription factors for TE and ICM, such as Cdx2, Yap1, Sox2, Nanog, Tead4, were reduced at the mRNA level in the miR-218-5p treated morula. This was accompanied by decreased Cdx2 protein levels at the 8-16-cell stage (P < 0.001) and disruption of co-localization of Yap1 and Cdx2. The blastocyte rate was increased by anti-miR-218-5p-encapsulated E-EVs compared with miR-218-5p group (P < 0.001). Conclusion This study offers valuable insights into the potential role of miR-218-5p in RIF and presents. The utilization of engineered vesicles containing anti-miR-218-5p may present a promising avenue for patients facing challenges with RIF.
Collapse
Affiliation(s)
- Lei Cai
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Mingwei Lv
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center, Key Laboratory of the Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jianbo Wei
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Chang Liu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medicine School, Nanjing, 210000, People’s Republic of China
| | - Yuehan Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Zhiqi Liao
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Tianhui Li
- State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - Hanwang Zhang
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Ling Xi
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center, Key Laboratory of the Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Cong Sui
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| |
Collapse
|
17
|
Hosseinabadi SK, Nazari H, Arabi M, Shams Esfandabadi N, Ahmadi E, Afzali A, Ghanaei H. Improving Bovine Embryo Development and Quality Using Bovine Oviductal Epithelial Cell-Derived Conditioned Medium (bOEC-CM). Vet Med Sci 2025; 11:e70179. [PMID: 40159373 PMCID: PMC11955017 DOI: 10.1002/vms3.70179] [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: 05/17/2024] [Revised: 11/19/2024] [Accepted: 12/08/2024] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND The embryo co-culture systems with the monolayer cultured cells are complex, unreproducible, and have a high probability of biological contamination. Therefore, nowadays, using conditioned media is a suitable alternative to these methods. OBJECTIVES This study aimed to investigate the impact of utilizing bovine oviductal epithelial cell-derived conditioned medium (bOEC-CM) on subsequent embryo development and quality. METHODS Bovine embryos produced in vitro were cultured in a specific medium supplemented with either 5% charcoal-stripped FBS (Charcoled Strip Serum [CSS]) or 10% bOEC-CM from either Days 1 or 3 post-fertilization. Various parameters, such as cleavage rate, blastocyst formation, hatching rate and blastocyst quality, were assessed. RESULTS The results indicated that adding 10% CM from Day 1 significantly reduced the cleavage rate compared to using CSS on the same day (p < 0.05). Furthermore, the CSS and CM from both Days 1 and 3 increased blastocyst formation rates (p < 0.05). Notably, the addition of 10% CM on Day 3 significantly improved the hatching rate compared to the other groups (p < 0.05). Both CM and CSS were found to enhance the inner cell mass (ICM), trophectoderm (TE) and total cell numbers in blastocysts when used on both Days 1 and 3 (p < 0.05). Additionally, CM from Day 3 positively influenced the expression levels of development-specific genes in cultured embryos (p < 0.05). CONCLUSION Overall, the findings suggest that using bOEC-CM at a 10% concentration may provide a promising supplement even better than serum and traditional co-culture methods during the last 5 days of embryo culture.
Collapse
Affiliation(s)
| | - Hassan Nazari
- Research Institute of Animal Embryo TechnologyShahrekord UniversityShahrekordIran
| | - Mehran Arabi
- Department of Animal PhysiologyFaculty of Basic SciencesShahrekord UniversityShahrekordIran
| | - Naser Shams Esfandabadi
- Research Institute of Animal Embryo TechnologyShahrekord UniversityShahrekordIran
- Department of Clinical SciencesFaculty of Veterinary MedicineShahrekord UniversityShahrekordIran
| | - Ebrahim Ahmadi
- Research Institute of Animal Embryo TechnologyShahrekord UniversityShahrekordIran
| | - Azita Afzali
- Clinical EmbryologistShahrekord University of Medical SciencesShahrekordIran
| | | |
Collapse
|
18
|
Wen S, Zheng R, Cai C, Jiang W. Chemical-based epigenetic reprogramming to advance pluripotency and totipotency. Nat Chem Biol 2025; 21:635-647. [PMID: 40251434 DOI: 10.1038/s41589-025-01874-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 03/06/2025] [Indexed: 04/20/2025]
Abstract
Reprogramming technology, breaking the inherent limitations of cellular identity and turning somatic cells into pluripotent cells with more developmental potential, holds great promise for cell therapy and regenerative medicine. Compared with traditional methods based on overexpressing transcription factors, chemical reprogramming with small molecules exhibits substantial advantages in safety and convenience, thus being the leading edge. Over the past decade, a notable focus has been reshaping cellular pluripotency and totipotency using pure small-molecule systems. Here, we provide a concise Review comparing the chemical approaches that have emerged to date and discussing the epigenetic regulatory mechanisms involved in chemical reprogramming. This Review highlights the remarkable potential of small-molecule potions to reformulate cell fate through epigenetic reprogramming and newly discovered actions. We aim to offer insights into chemically controlled cell manipulation and key challenges and future application prospects of chemical reprogramming.
Collapse
Affiliation(s)
- Shanshan Wen
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Ran Zheng
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, China
| | - Cheguo Cai
- Shenzhen Beike Biotechnology Co., Ltd, Shenzhen, China.
| | - Wei Jiang
- Department of Biological Repositories, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China.
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China.
| |
Collapse
|
19
|
Shen L, Xu Y, Li B, Long Y. Risk Factors for Postpartum Hemorrhage Following Vaginal Deliveries in China: A Case-Control Study of Second Births After IUD Removal. Int J Womens Health 2025; 17:1203-1214. [PMID: 40331130 PMCID: PMC12053772 DOI: 10.2147/ijwh.s516263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Objective This study aims to evaluate the predictive value of risk factors for postpartum hemorrhage (PPH) following vaginal delivery, with a special focus on the impact of intrauterine device (IUD) usage in the context of recent policy changes allowing more childbirths in China. Methods A total of 6879 women who underwent vaginal deliveries from January 2021 to December 2023 in the Lianyungang regional maternal care system and met the inclusion and exclusion criteria were enrolled. A case-control design was employed, comprising 524 women with PPH (blood loss ≥500 mL) 24h after giving birth and an equal number of controls (blood loss <500 mL) matched on a 1:1 ratio. Univariate and multivariate logistic regression analyses, alongside ROC curve analysis, were conducted to identify risk factors for PPH. Results Univariate analysis revealed significant differences in age (OR = 1.86, 95% CI: 1.35-2.57, P < 0.01), number of miscarriages (OR = 1.97, 95% CI: 1.46-2.65, P < 0.001), gestational week, number of fetuses, weight of the second child (OR = 10.78, 95% CI: 7.88-14.75, P < 0.001), placental adhesion, and uterine atony (OR = 2.40, 95% CI: 1.60-3.61, P < 0.001). No significant differences were observed regarding occupation, educational level, mode of first delivery, IUD use and duration, and presence of gestational diabetes or hypertension (P > 0.05). Multivariate logistic regression identified age, number of miscarriages, weight of the second child, and uterine atony as independent risk factors for PPH (P < 0.05). ROC curve analysis showed that the combined predictive value of these factors was superior, with an AUC of 0.805 for the combined predictive model. Conclusion PPH is influenced by advanced maternal age, multiple miscarriages, high birth weight, and uterine atony. IUD use may not independently impact the likelihood of PPH under the conditions studied.
Collapse
Affiliation(s)
- Liping Shen
- Department of Family Planning, Maternal and Child Health Hospital of Lianyungang City, Lianyungang, Jiangsu Province, 222000, People’s Republic of China
| | - Yang Xu
- Department of Family Planning, Maternal and Child Health Hospital of Lianyungang City, Lianyungang, Jiangsu Province, 222000, People’s Republic of China
| | - Baoxia Li
- Department of Family Planning, Maternal and Child Health Hospital of Lianyungang City, Lianyungang, Jiangsu Province, 222000, People’s Republic of China
| | - Yuyun Long
- Department of Family Planning, Maternal and Child Health Hospital of Lianyungang City, Lianyungang, Jiangsu Province, 222000, People’s Republic of China
| |
Collapse
|
20
|
Li B, Jin N, Lu J, Wang M, Wang J, Chen S. Decreased OGT Attenuates Endometrial Decidualization and Embryo Implantation by Affecting HIF-1α Stability. Mol Reprod Dev 2025; 92:e70025. [PMID: 40342239 DOI: 10.1002/mrd.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/02/2025] [Accepted: 04/16/2025] [Indexed: 05/11/2025]
Abstract
Hypoxia-inducible factor 1-alpha (HIF-1α) is essential for glycolysis regulation. Its expression in the endometrium is significantly reduced in recurrent implantation failure (RIF), indicating that lower levels of HIF-1α may contribute to embryo implantation failure. O-GlcNAcylation is a dynamic posttranslational modification mediated by O-GlcNAc transferase (OGT), known to regulate HIF-1α in cancer cells. However, it remains unclear whether OGT affects glycolytic processes in uterine endometrial stromal cells (ESCs) and its potential role in embryo implantation. This study utilized In Vitro and In Vivo experiments to investigate the role of OGT in decidualization and embryo implantation, along with its underlying mechanisms. Our findings show that OGT expression is significantly reduced in the endometrium of patients with RIF. Additionally, OGT knockdown led to failed embryo implantation in mice. Further analysis revealed that OGT promotes decidualization by stabilizing HIF-1α, which enhances glycolytic activity. Inhibiting OGT resulted in insufficient decidualization among human ESCs. Moreover, our results indicate that OGT partially regulates CCL2 secretion by maintaining HIF-1α levels within human ESCs, which is essential for successful embryo implantation. Based on these findings, we propose that OGT represents a novel and promising therapeutic target for both the diagnosis and treatment of RIF.
Collapse
Affiliation(s)
- Bo Li
- Department of Obstetrics and Gynecology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ni Jin
- Department of Obstetrics and Gynecology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jie Lu
- Department of Obstetrics and Gynecology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ming Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jun Wang
- Department of Obstetrics and Gynecology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Shuqiang Chen
- Department of Obstetrics and Gynecology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
21
|
Wang M, Zhang L, Liu Z, Guo A, Yang G, Yu T. Host-Microbiota Interactions in the Pathogenesis of Porcine Fetal Mummification. Microorganisms 2025; 13:1052. [PMID: 40431225 PMCID: PMC12113762 DOI: 10.3390/microorganisms13051052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
The number of mummies (MUM) in pigs is a major factor affecting sow reproductive performance. Reducing the incidence of MUM can effectively improve sow utilization efficiency. However, the complex mechanisms by which the host genome, gut microbiome, and metabolome interact to influence sow MUM remain unclear. Based on the current research landscape, this study systematically reveals the regulatory mechanisms of the host genome-gut microbiome-metabolome interaction network on sow MUM. By conducting a multi-omics analysis on the intestinal contents of Yorkshire sows during late gestation across different parities, we constructed a dynamic atlas of the gut microbiota and identified 385 core microbial taxa. Through multi-model MWAS and meta-analysis, we screened six key microbial taxa significantly associated with MUM, including Bacteroidales_RF16_group, Prevotellaceae_Ga6A1_group, Comamonas, Paraprevotella, Dorea, and Gallicola. An mGWAS analysis further identified Bacteroidales_RF16_group as regulated by host genetics, as well as candidate genes such as EGF, ENPEP, and CASP6, and important SNP loci such as rs345237235 and rs3475666995. The study found that the abundance of Proteobacteria in the sow's gut increased progressively from the first parity, providing a theoretical basis for pathogen suppression mechanisms. By integrating fecal metabolomics data, we constructed a four-dimensional regulatory network of host gene-gut microbiota-metabolite-host phenotype. This study innovatively combines quantitative genetics with multi-omics approaches, not only providing a theoretical foundation for understanding host-microbiota interaction mechanisms but also offering critical scientific guidance for reducing sow MUM incidence and improving reproductive efficiency.
Collapse
Affiliation(s)
| | | | | | | | - Gongshe Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| | - Taiyong Yu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| |
Collapse
|
22
|
Hua L, Peng Y, Yan L, Yuan P, Qiao J. Moving toward totipotency: the molecular and cellular features of totipotent and naive pluripotent stem cells. Hum Reprod Update 2025:dmaf006. [PMID: 40299455 DOI: 10.1093/humupd/dmaf006] [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: 05/25/2023] [Revised: 01/06/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Dissecting the key molecular mechanism of embryonic development provides novel insights into embryogenesis and potential intervention strategies for clinical practices. However, the ability to study the molecular mechanisms of early embryo development in humans, such as zygotic genome activation and lineage segregation, is meaningfully constrained by methodological limitations and ethical concerns. Totipotent stem cells have an extended developmental potential to differentiate into embryonic and extraembryonic tissues, providing a suitable model for studying early embryo development. Recently, a series of ground-breaking results on stem cells have identified totipotent-like cells or induced pluripotent stem cells into totipotent-like cells. OBJECTIVE AND RATIONALE This review followed the PRISMA guidelines, surveys the current works of literature on totipotent, naive, and formative pluripotent stem cells, introduces the molecular and biological characteristics of those stem cells, and gives advice for future research. SEARCH METHODS The search method employed the terms 'totipotent' OR 'naive pluripotent stem cell' OR 'formative pluripotent stem cell' for unfiltered search on PubMed, Web of Science, and Cochrane Library. Papers included were those with information on totipotent stem cells, naive pluripotent stem cells, or formative pluripotent stem cells until June 2024 and were published in the English language. Articles that have no relevance to stem cells, or totipotent, naive pluripotent, or formative pluripotent cells were excluded. OUTCOMES There were 152 records included in this review. These publications were divided into four groups according to the species of the cells included in the studies: 67 human stem cell studies, 70 mouse stem cell studies, 9 porcine stem cell studies, and 6 cynomolgus stem cell studies. Naive pluripotent stem cell models have been established in other species such as porcine and cynomolgus. Human and mouse totipotent stem cells, e.g. human 8-cell-like cells, human totipotent blastomere-like cells, and mouse 2-cell-like cells, have been successfully established and exhibit high developmental potency for both embryonic and extraembryonic contributions. However, the observed discrepancies between these cells and real embryos in terms of epigenetics and transcription suggest that further research is warranted. Our results systematically reviewed the established methods, molecular characteristics, and developmental potency of different naive, formative pluripotent, and totipotent stem cells. Furthermore, we provide a parallel comparison between animal and human models, and offer recommendations for future applications to advance early embryo research and assisted reproduction technologies. WIDER IMPLICATIONS Totipotent cell models provide a valuable resource to understand the underlying mechanisms of embryo development and forge new paths toward future treatment of infertility and regenerative medicine. However, current in vitro cell models exhibit epigenetic and transcriptional differences from in vivo embryos, and many cell models are unstable across passages, thus imperfectly recapitulating embryonic development. In this regard, standardizing and expanding current research on totipotent stem cell models are essential to enhance our capability to resemble and decipher embryogenesis.
Collapse
Affiliation(s)
- Lingyue Hua
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Yuyang Peng
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Liying Yan
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Peng Yuan
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jie Qiao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Beijing Advanced Innovation Center for Genomics, Beijing, China
| |
Collapse
|
23
|
Ruan D, Chen ACH, Tam TTKK, Huang W, Guo J, Xu S, Ruan H, Fong SW, Liu X, Gao X, Yeung WSB, Lee YL, Liu P. Establishment of human expanded potential stem cell lines via preimplantation embryo cultivation and somatic cell reprogramming. Nat Protoc 2025:10.1038/s41596-025-01168-2. [PMID: 40301626 DOI: 10.1038/s41596-025-01168-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 03/04/2025] [Indexed: 05/01/2025]
Abstract
We previously reported the derivation of expanded potential stem cells (EPSCs) by modulating signaling pathways involved in preimplantation embryogenesis. These cells exhibit expanded developmental potential into embryonic and extraembryonic lineages, and we have shown that human EPSCs (hEPSCs) possess trophoblast differentiation potency for generating human trophoblast stem cells. Here we report protocols for deriving stable hEPSC lines directly from morula or early blastocyst stages of human preimplantation embryos (hEPSC-em) and by reprogramming human dermal fibroblasts (human induced EPSCs) using six exogenous factors, as an extension to our previous protocols on deriving porcine EPSCs from preimplantation embryos and by reprogramming somatic cells. These hEPSC lines proliferate robustly over long-term passaging and are amenable to both simple indels and precision genome editing. We provide guidance for characterizing these newly established hEPSCs, including cell-cycle analysis, pluripotency validation and karyotyping. The hEPSCs form teratomas with embryonic and extraembryonic cell lineages and readily differentiate into human trophoblast stem cells in vitro. At the molecular level, hEPSCs have unique features such as high expression of core histone genes and low H3K27me3 levels resembling eight-cell/morula stage embryos. These properties make hEPSCs a valuable tool not only for studying early human development but also for potential applications in regenerative medicine. The protocols presented in this manuscript can be readily performed by postgraduate students or postdoctoral fellows and completed within around 2 months.
Collapse
Affiliation(s)
- Degong Ruan
- Center for Translational Stem Cell Biology, Science Park, Hong Kong Special Administrative Region, China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Andy Chun Hang Chen
- Center for Translational Stem Cell Biology, Science Park, Hong Kong Special Administrative Region, China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Timothy Theodore Ka Ki Tam
- Stem Cell and Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Wen Huang
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jilong Guo
- Center for Translational Stem Cell Biology, Science Park, Hong Kong Special Administrative Region, China
| | - Shao Xu
- Center for Translational Stem Cell Biology, Science Park, Hong Kong Special Administrative Region, China
| | - Hanzhang Ruan
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Sze Wan Fong
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xueyan Liu
- Center for Translational Stem Cell Biology, Science Park, Hong Kong Special Administrative Region, China
| | - Xuefei Gao
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - William Shu Biu Yeung
- Center for Translational Stem Cell Biology, Science Park, Hong Kong Special Administrative Region, China.
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Yin Lau Lee
- Center for Translational Stem Cell Biology, Science Park, Hong Kong Special Administrative Region, China.
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Pentao Liu
- Center for Translational Stem Cell Biology, Science Park, Hong Kong Special Administrative Region, China.
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
- Stem Cell and Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
24
|
Bone RA, Lowndes MP, Raineri S, R Riveiro A, Lundregan SL, Dall M, Sulek K, Romero JAH, Malzard L, Koigi S, Heckenbach IJ, Solis-Mezarino V, Völker-Albert M, Vasilopoulou CG, Meier F, Trusina A, Mann M, L Nielsen M, Treebak JT, Brickman JM. Altering metabolism programs cell identity via NAD +-dependent deacetylation. EMBO J 2025:10.1038/s44318-025-00417-0. [PMID: 40281356 DOI: 10.1038/s44318-025-00417-0] [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: 12/18/2024] [Revised: 02/03/2025] [Accepted: 03/03/2025] [Indexed: 04/29/2025] Open
Abstract
Cells change their metabolic profiles in response to underlying gene regulatory networks, but how can alterations in metabolism encode specific transcriptional instructions? Here, we show that forcing a metabolic change in embryonic stem cells (ESCs) promotes a developmental identity that better approximates the inner cell mass (ICM) of the early mammalian blastocyst in cultures. This shift in cellular identity depends on the inhibition of glycolysis and stimulation of oxidative phosphorylation (OXPHOS) triggered by the replacement of D-glucose by D-galactose in ESC media. Enhanced OXPHOS in turn activates NAD + -dependent deacetylases of the Sirtuin family, resulting in the deacetylation of histones and key transcription factors to focus enhancer activity while reducing transcriptional noise, which results in a robustly enhanced ESC phenotype. This exploitation of a NAD + /NADH coenzyme coupled to OXPHOS as a means of programming lineage-specific transcription suggests new paradigms for how cells respond to alterations in their environment, and implies cellular rejuvenation exploits enzymatic activities for simultaneous activation of a discrete enhancer set alongside silencing genome-wide transcriptional noise.
Collapse
Affiliation(s)
- Robert A Bone
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Molly P Lowndes
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Silvia Raineri
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alba R Riveiro
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sarah L Lundregan
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Dall
- Novo Nordisk Foundation Center for Basic Metabolic Research, Copenhagen, Denmark
| | - Karolina Sulek
- Novo Nordisk Foundation Center for Basic Metabolic Research, Copenhagen, Denmark
| | - Jose A H Romero
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Luna Malzard
- Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
| | - Sandra Koigi
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Catherine G Vasilopoulou
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Florian Meier
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Ala Trusina
- Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Michael L Nielsen
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Copenhagen, Denmark
| | - Joshua M Brickman
- Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
25
|
Haddadi M, Safari R, Hantoushzadeh S. The Diagnostic Role of miRNAs in Identifying Placenta Accreta: A Systematic Review. Am J Obstet Gynecol MFM 2025:101682. [PMID: 40280487 DOI: 10.1016/j.ajogmf.2025.101682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/22/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025]
Abstract
OBJECTIVE This systematic review evaluates the diagnostic accuracy of circulating microRNAs (miRNAs) as potential biomarkers for detecting placenta accreta spectrum (PAS) disorders, a condition characterized by abnormal placental adherence with significant maternal health risks. DATA SOURCES A comprehensive literature search was conducted in PubMed, Embase, and Scopus databases up to October 30, 2024, using predefined keywords such as "miRNA" and "placenta accreta." STUDY ELIGIBILITY Studies investigating miRNA expression in PAS cases compared to controls, using either blood or placental tissue, were included. Articles were screened independently by two reviewers, with discrepancies resolved by consensus. STUDY APPRAISAL AND SYNTHESIS METHODS The methodological quality of eligible studies was assessed using the Newcastle-Ottawa Scale. Extracted data were synthesized to identify miRNAs with diagnostic potential for PAS disorders. Due to significant variations in the comparisons conducted across studies and the diverse outcome measures reported, a meta-analysis of the included studies was not feasible. RESULTS Out of 82 articles identified, 14 met the inclusion criteria after duplicate removal and screening. The studies reported distinct differential expression patterns of miRNAs in PAS cases. Notably, a combination of miR-26a-5p and miR-17-5p demonstrated 100% sensitivity and 82% specificity for predicting PAS in the first-trimester of pregnancy. CONCLUSIONS PAS disorders are typically diagnosed during the third trimester through imaging techniques like ultrasonography. However, miRNAs exhibit promise as non-invasive, early biomarkers, potentially enabling earlier diagnosis and improved clinical management. These findings support the incorporation of miRNA analysis into diagnostic guidelines for PAS.
Collapse
Affiliation(s)
- Mohammad Haddadi
- Vali-E-Asr Reproductive Health Research Center, Family Health Research Institute, Tehran University Of Medical Sciences, Tehran, Iran
| | - Roxana Safari
- Vali-E-Asr Reproductive Health Research Center, Family Health Research Institute, Tehran University Of Medical Sciences, Tehran, Iran
| | - Sedigheh Hantoushzadeh
- Vali-E-Asr Reproductive Health Research Center, Family Health Research Institute, Tehran University Of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
Ren H, Jia X, Yu L. The building blocks of embryo models: embryonic and extraembryonic stem cells. Cell Discov 2025; 11:40. [PMID: 40258839 PMCID: PMC12012135 DOI: 10.1038/s41421-025-00780-6] [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: 04/24/2024] [Accepted: 01/10/2025] [Indexed: 04/23/2025] Open
Abstract
The process of a single-celled zygote developing into a complex multicellular organism is precisely regulated at spatial and temporal levels in vivo. However, understanding the mechanisms underlying development, particularly in humans, has been constrained by technical and ethical limitations associated with studying natural embryos. Harnessing the intrinsic ability of embryonic stem cells (ESCs) to self-organize when induced and assembled, researchers have established several embryo models as alternative approaches to studying early development in vitro. Recent studies have revealed the critical role of extraembryonic cells in early development; and many groups have created more sophisticated and precise ESC-derived embryo models by incorporating extraembryonic stem cell lines, such as trophoblast stem cells (TSCs), extraembryonic mesoderm cells (EXMCs), extraembryonic endoderm cells (XENs, in rodents), and hypoblast stem cells (in primates). Here, we summarize the characteristics of existing mouse and human embryonic and extraembryonic stem cells and review recent advancements in developing mouse and human embryo models.
Collapse
Affiliation(s)
- Hongan Ren
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaojie Jia
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Leqian Yu
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
27
|
Zhang W, Fu H, Huang Y, Zeng M, Ouyang X, Wang X, Ruan D, Ma L, Hu X, Guo J, Galardi JW, Dougan G, Yeung WSB, Li L, Liu J, Feschotte C, Liu P. METTL3-dependent m6A RNA methylation regulates transposable elements and represses human naïve pluripotency through transposable element-derived enhancers. Nucleic Acids Res 2025; 53:gkaf349. [PMID: 40298111 PMCID: PMC12038396 DOI: 10.1093/nar/gkaf349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 03/06/2025] [Accepted: 04/26/2025] [Indexed: 04/30/2025] Open
Abstract
N 6-methyladenosine (m6A) is the most prevalent messenger RNA modification with diverse regulatory roles in mammalian cells. While its functions are well-documented in mouse embryonic stem cells (mESCs), its role in human pluripotent stem cells (hPSCs) remains to be fully explored. METTL3 is the main enzyme responsible for m6A deposition. Here, using a METTL3 inducible knockout (iKO) system, we uncovered that, unlike in mESCs, METTL3 was indispensable for hPSC maintenance. Importantly, loss of METTL3 caused significant upregulation of pluripotency factors including naïve pluripotency genes and failure to exit pluripotency, thus impairing stem cell differentiation towards both embryonic and extraembryonic cell lineages. Mechanistically, METTL3 iKO in hPSCs promoted expression and enhancer activities of two primate-specific transposable elements (TEs), SVA_D and HERVK/LTR5_Hs. At SVA_D elements, loss of METTL3 leads to reduced H3K9me3 deposition. On the other hand, the activation of LTR5_Hs in the METTL3 iKO cells is accompanied by increased chromatin accessibility and binding pluripotency factors. The activated SVA_D and LTR5_Hs elements can act as enhancers and promote nearby naïve gene expression by directly interacting with their promoters. Together these findings reveal that METTL3-dependent m6A RNA methylation plays critical roles in suppressing TE expression and in regulating the human pluripotency network.
Collapse
Affiliation(s)
- Weiyu Zhang
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, United States
| | - Haifeng Fu
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yunying Huang
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ming Zeng
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
- Key laboratory for reproductive Medicine of Guangdong Province, The Third affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Xiangyu Ouyang
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xiao Wang
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Degong Ruan
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Liyang Ma
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xinning Hu
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jilong Guo
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Justin W Galardi
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, United States
| | - Gordon Dougan
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Medicine, Jeffrey Cheah Biomedical Centre, Cambridge University, Puddicombe Way, Cambridge Biomedical Campus, Cambridge CB2 0AW, United Kingdom
| | - William S B Yeung
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Department of Obstetrics and Gynaecology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, The University of Hong Kong ‐ Shenzhen Hospital, Shenzhen 518000, China
| | - Lei Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
- Key laboratory for reproductive Medicine of Guangdong Province, The Third affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Jianqiao Liu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
- Key laboratory for reproductive Medicine of Guangdong Province, The Third affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Cedric Feschotte
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, United States
| | - Pentao Liu
- Centre for Translational Stem Cell Biology, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
28
|
Kuang Z, Pang C, Wang H, Wei X, Ye X, Gao X, Sun L. Generation of kidney organoids derived from human expanded potential stem cells. Cells Dev 2025:204025. [PMID: 40189048 DOI: 10.1016/j.cdev.2025.204025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 04/02/2025] [Accepted: 04/02/2025] [Indexed: 04/24/2025]
Abstract
The establishment of human expanded potential stem cell (hEPSC) presents a unique cellular platform for translational research in kidney organoids. We generated SIX2 reporter and doxycycline (DOX)-inducible YAP overexpression in hEPSC lines using CRISPR-Cas9. Chemical compounds and DOX were added to the culture medium to induce Hippo-YAP signaling, respectively. The hEPSC line containing the SIX2-mCherry reporter gene accurately reflected SIX2 expression in vitro, enabling the real-time tracking of kidney organoid development. A comparative analysis revealed that inhibiting the Hippo-YAP signaling pathway before nephron progenitor cell (NPC) generation effectively increased the number of NPCs, resulting in a more nephron-like structure. However, prolonged inhibition hindered the further maturation of the kidney organoids, leading to differentiation stagnation. Therefore, activating YAP before NPC generation facilitates their maturation, offering effective induction strategies improving kidney organoid differentiation efficiency.
Collapse
Affiliation(s)
- Zhanpeng Kuang
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Changmiao Pang
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Haiyan Wang
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Department of Pediatrics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xiaocui Wei
- Guangdong provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xianhua Ye
- Guangdong provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xuefei Gao
- Guangdong provincial Key Laboratory of Bone and Joint Degenerative Diseases, Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Liangzhong Sun
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
29
|
Martins F, Ribeiro MHL. Quality and Regulatory Requirements for the Manufacture of Master Cell Banks of Clinical Grade iPSCs: The EU and USA Perspectives. Stem Cell Rev Rep 2025; 21:645-679. [PMID: 39821060 DOI: 10.1007/s12015-024-10838-9] [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] [Accepted: 12/23/2024] [Indexed: 01/19/2025]
Abstract
The discovery of induced pluripotent stem cells (iPSCs) and protocols for their differentiation into various cell types have revolutionized the field of tissue engineering and regenerative medicine. Developing manufacturing guidelines for safe and GMP-compliant final products has become essential. Allogeneic iPSCs-derived cell therapies are now the preferred manufacturing alternative. This option requires the establishment of clinical-grade master cell banks of iPSCs. This study aimed at reviewing the Quality and Regulatory requirements from the two main authorities in the world-Europe (EMA) and the United States (FDA)-regarding the manufacture of clinical grade master cell banks (iPSCs). The minimum requirements for iPSCs to be used in first-in-human clinical trials were also reviewed, as well as current best practices currently followed by iPSC bank manufacturers for final product characterisation. The methodology used for this work was a review of various sources of information ranging from scientific literature, published guidance documents available on the EMA and FDA websites, GMP and ICH guidelines, and applicable compendial monographs. Manufacturers of iPSCs cell banks looking to qualify them for clinical use are turning to the ICH guidelines and trying to adapt their requirements. Specifically with the impact of the field of iPSC cell banks, the following areas should be subject to guidance and harmonisation: i) expression vectors authorized for iPSC generation; ii) minimum identity testing; iii) minimum purity testing (including adventitious agent testing); and iv) stability testing. Current ICH guidelines for biotechnological/biological products should be extended to cover cell banks used for cell therapies.
Collapse
Affiliation(s)
- Fernando Martins
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
- Stemamatters S.A., 4805-017, Guimarães, Portugal
| | - Maria H L Ribeiro
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal.
- Research Institute for Medicines (i-Med.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal.
| |
Collapse
|
30
|
Li SY, DeMayo FJ. Revolutionizing Implantation Studies: Uterine-Specific Models and Advanced Technologies. Biomolecules 2025; 15:450. [PMID: 40149986 PMCID: PMC11940528 DOI: 10.3390/biom15030450] [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: 01/30/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Implantation is a complex and tightly regulated process essential for the establishment of pregnancy. It involves dynamic interactions between a receptive uterus and a competent embryo, orchestrated by ovarian hormones such as estrogen and progesterone. These hormones regulate proliferation, differentiation, and gene expression within the three primary uterine tissue types: myometrium, stroma, and epithelium. Advances in genetic manipulation, particularly the Cre/loxP system, have enabled the in vivo investigation of the role of genes in a uterine compartmental and cell type-specific manner, providing valuable insights into uterine biology during pregnancy and disease. The development of endometrial organoids has further revolutionized implantation research. They mimic the native endometrial structure and function, offering a powerful platform for studying hormonal responses, implantation, and maternal-fetal interactions. Combined with omics technologies, these models have uncovered the molecular mechanisms and signaling pathways that regulate implantation. This review provides a comprehensive overview of uterine-specific genetic tools, endometrial organoids, and omics. We explore how these advancements enhance our understanding of implantation biology, uterine receptivity, and decidualization in reproductive research.
Collapse
Affiliation(s)
| | - Francesco John DeMayo
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC 27709, USA;
| |
Collapse
|
31
|
Karmakar A, Augustine ABHR, Thummer RP. Genes as Genome Stabilizers in Pluripotent Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40095244 DOI: 10.1007/5584_2025_853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Pluripotent stem cells, comprising embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), are characterized by their self-renewal capacity and the ability to differentiate into cells of all three germ layers of an adult animal. Out of the two, iPSCs are generated through the reprogramming of somatic cells by inducing a pluripotency-specific transcriptional program. This process requires a resetting of the somatic cell genome to a pluripotent cell-specific genome, resulting in cellular stress at genomic, epigenetic, and transcriptional levels. Notably, in contrast to the predominant compact and inactive organization of chromatin in somatic cells, the chromatin in ESCs and iPSCs is open. Furthermore, maintaining a pluripotent state needs a plethora of changes in the genetic landscape of the cells. Here, we attempt to elucidate how certain genes safeguard genomic stability in ESCs and iPSCs, aiding in the complex cellular mechanisms that regulate self-renewal, pluripotency, and somatic reprogramming.
Collapse
Affiliation(s)
- Asmita Karmakar
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Allan Blessing Harison Raj Augustine
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India.
| |
Collapse
|
32
|
Guiltinan C, Botigelli RC, Candelaria JI, Smith JM, Arcanjo RB, Denicol AC. Primed bovine embryonic stem cell lines can be derived at diverse stages of blastocyst development with similar efficiency and molecular characteristics. Biol Open 2025; 14:BIO061819. [PMID: 39957479 PMCID: PMC11911636 DOI: 10.1242/bio.061819] [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: 11/15/2024] [Accepted: 02/04/2025] [Indexed: 02/18/2025] Open
Abstract
In this study, we established bovine embryonic stem cell (bESC) lines from early (eBL) and full (BL) blastocysts to determine the efficiency of bESC derivation from an earlier embryonic stage and compare the characteristics of the resulting lines. Using established medium and protocols for derivation of primed bESCs from expanded blastocysts, we derived bESC lines from eBLs and BLs with the same efficiency (4/12 each, 33%). Regardless of original blastocyst stage, bESC lines had a similar phenotype, including differentiation capacity, stable karyotype, and pluripotency marker expression over feeder-free transition and long-term culture. Transcriptome and functional analyses indicated that eBL- and BL-derived lines were in primed pluripotency. We additionally compared RNA-sequencing data from our lines to bovine embryos and stem cells from other recent reports, finding that base medium was the predominant source of variation among cell lines. In conclusion, our results show that indistinguishable bESC lines can be readily derived from eBL and BL, widening the pool of embryos available for bESC establishment. Finally, our investigation points to sources of variation in cell phenotype among recently reported bESC conditions, opening the door to future studies investigating the impact of factors aside from signaling molecules on ESC derivation, maintenance, and performance.
Collapse
Affiliation(s)
- Carly Guiltinan
- Department of Animal Science, University of California Davis, Davis, CA, 95616, USA
| | - Ramon C. Botigelli
- Department of Animal Science, University of California Davis, Davis, CA, 95616, USA
| | | | - Justin M. Smith
- Department of Animal Science, University of California Davis, Davis, CA, 95616, USA
| | - Rachel B. Arcanjo
- Department of Animal Science, University of California Davis, Davis, CA, 95616, USA
| | - Anna C. Denicol
- Department of Animal Science, University of California Davis, Davis, CA, 95616, USA
| |
Collapse
|
33
|
Elmorsy EM, Al Doghaither HA, Al-Ghafari AB, Amer S, Fawzy MS, Toraih EA. Fucoxanthin mitigates mercury-induced mitochondrial toxicity in the human ovarian granulosa cell line. Reprod Toxicol 2025; 132:108855. [PMID: 39947444 DOI: 10.1016/j.reprotox.2025.108855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/16/2025]
Abstract
Mercury (Hg) is known to be a hazardous toxin with a significant negative impact on female reproduction through mechanisms that remain unclear. The carotenoid fucoxanthin (FX) is an antioxidant with several positive effects on human health. This study aimed to examine the potential protective role of FX in reducing the Hg-induced bioenergetic disturbances in a human ovarian granulosa cell line model. (methods briefly) Hg was found to reduce the viability of granulosa cells in a concentration-dependent manner, with an estimated 72-hour EC50 of 10 µM. In contrast, FX (10 and 20 µM) improved cell viability. Hg (1 and 10 µM) significantly reduced cellular ATP levels, mitochondrial membrane potential, oxygen consumption rates, and lactate production. Additionally, Hg impaired the activities and kinetics of mitochondrial complexes I and III and reduced the expression of mitochondrial genes ND1, ND5, cytochrome B, cytochrome C oxidase, and ATP synthase subunits 6 and 8. According to tests on mitochondrial membranes, Hg increased membrane fluidity by reducing saturated fatty acid levels and increasing those of unsaturated fatty acids. Hg also promoted mitochondrial swelling and enhanced the inner mitochondrial membrane permeability to hydrogen and potassium ions. FX (10 µM) was shown to mitigate the negative effects of Hg on the viability of treated granulosa cells, bioenergetics parameters, and mitochondrial membrane integrity in a concentration-dependent manner. Based on these findings, bioenergetic disruption may be a key underlying cause of Hg-induced ovarian dysfunction. Furthermore, FX may have a potential therapeutic role in treating ovarian disorders caused by Hg-induced disruption of granulosa cell bioenergetics.
Collapse
Affiliation(s)
- Ekramy M Elmorsy
- Center for Health Research, Northern Border University, Arar 91431, Saudi Arabia.
| | - Huda A Al Doghaither
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Ayat B Al-Ghafari
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Saad Amer
- Translational Medical Sciences, School of Medicine, Royal Derby Hospital Centre, University of Nottingham, Derby DE22 3DT, UK.
| | - Manal S Fawzy
- Center for Health Research, Northern Border University, Arar 91431, Saudi Arabia.
| | - Eman A Toraih
- Department of Surgery, School of Medicine, Tulane University, New Orleans, LA 70112, USA; Department of Cardiovascular Perfusion, Interprofessional Research, College of Health Professions, Upstate Medical University, New York 13210, USA; Genetics Unit, Department of Histology and Cell Biology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt.
| |
Collapse
|
34
|
de Almeida Roque A, Zablocki da Luz J, Filipak Neto F, Barjhoux I, Rioult D, de Oliveira Ribeiro CA. Low concentrations of complex mixtures of pesticides and metabolites are toxic to common Carp brain cells ( Cyprinus carpio carpio). Drug Chem Toxicol 2025; 48:314-324. [PMID: 39210515 DOI: 10.1080/01480545.2024.2397432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Pesticide use increases annually, and Brazil is the world's largest consumer. However, unlike the European Union (EU), there is no established limit value for pesticide mixtures in drinking water, and therefore the concentration of pesticides can reach 3354 times the EU limit. Thus, determining the risk of exposure to pesticide mixtures and their main metabolites is challenging and requires the use of alternative methods. In the present study, the Common Carp Brain (CCB) cell line was used to evaluate the in vitro toxicity of relevant pesticide mixtures (glyphosate, 2,4-D, atrazine, and mancozeb) and their main metabolites after 72 h of exposure. The tested concentrations were based on the Acceptable Daily Intake (ADI) defined by Brazilian legislation. The results showed that cells exposed to lower concentrations of the pesticide mixtures and the pesticide + metabolite mixtures were affected by a decrease in cell confluence, resazurin metabolism, and wound healing capacity. The IBR index showed that lower concentrations had more severe effects, suggesting the absence of safe concentrations of these pesticide and metabolite mixtures for the CCB cell line within the tested concentration range. These findings raise concerns about the effects of exposure to these substances on animal and human health.
Collapse
Affiliation(s)
- Aliciane de Almeida Roque
- Department of Cell Biology, Laboratory of Cell Toxicology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Jessica Zablocki da Luz
- Department of Cell Biology, Laboratory of Cell Toxicology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Francisco Filipak Neto
- Department of Cell Biology, Laboratory of Cell Toxicology, Federal University of Paraná, Curitiba, PR, Brazil
| | - Iris Barjhoux
- UMR-I 02 INERIS-URCA-ULH SEBIO - Stress Environnementaux et BIOsurveillance des milieux aquatiques, Université de Reims Champagne-Ardenne, Reims, France
| | - Damien Rioult
- UMR-I 02 INERIS-URCA-ULH SEBIO - Stress Environnementaux et BIOsurveillance des milieux aquatiques, Université de Reims Champagne-Ardenne, Reims, France
- URCATech Plateau Technique Mobile de Cytométrie Environnementale URCATech- MOBICYTE, Université de Reims Champagne-Ardenne, Reims, France
| | | |
Collapse
|
35
|
Lee YJ, Song JH, Lee JW, Hong TK, Uhm SJ, Hong K, Do JT. Mitochondrial morphology and energy metabolism in reprogrammed porcine expanded potential stem cells. Anim Biosci 2025; 38:444-453. [PMID: 39483037 PMCID: PMC11917424 DOI: 10.5713/ab.24.0521] [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: 07/22/2024] [Accepted: 08/26/2024] [Indexed: 11/03/2024] Open
Abstract
OBJECTIVE Expanded potential stem cells (EPSCs) are stem cells that can differentiate into embryonic and extraembryonic lineages, including extraembryonic endoderm and trophoblast lineages. Therefore, EPSCs have great potential in advancing regenerative medicine, elucidating disease mechanisms, and exploring early embryonic development. However, the generation and characterization of EPSCs in pigs have not been thoroughly explored. In this study, we successfully generated porcine EPSCs (pEPSCs). METHODS We reprogrammed porcine fetal fibroblasts (PFFs) using an integration-free method with Sendai virus vectors. RESULTS The resulting pEPSCs expressed key pluripotency markers and demonstrated the ability to differentiate between embryonic and extraembryonic lineages. Notably, reprogramming into pEPSCs was associated with a transformation of mitochondrial morphology from the elongated form observed in PFFs to a globular shape, reflecting potential alterations in energy metabolism. We observed significant remodeling of mitochondrial morphology and a subsequent shift towards glycolytic energy dependence during the reprogramming of PFFs into pEPSCs. CONCLUSION Our findings provide valuable insights into the characteristics of EPSCs in pigs and highlight their potential applications in regenerative medicine, disease modeling, and emerging fields such as cell-based meat production.
Collapse
Affiliation(s)
- Yun Ju Lee
- Department of Stem Cell and Regenerative Biotechnology, Konkuk Institute of Technology, Konkuk University, Seoul 05029, Korea
- Biotechnology Research Institute, MGENSolutions Co., Ltd., Seoul 06591, Korea
| | - Jae Hoon Song
- Department of Stem Cell and Regenerative Biotechnology, Konkuk Institute of Technology, Konkuk University, Seoul 05029, Korea
| | - Je Woo Lee
- Department of Stem Cell and Regenerative Biotechnology, Konkuk Institute of Technology, Konkuk University, Seoul 05029, Korea
| | - Tae Kyung Hong
- Department of Stem Cell and Regenerative Biotechnology, Konkuk Institute of Technology, Konkuk University, Seoul 05029, Korea
| | - Sang Jun Uhm
- Department of Animal Science, Sangji University, Wonju 26339, Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, Konkuk Institute of Technology, Konkuk University, Seoul 05029, Korea
| | - Jeong Tae Do
- Department of Stem Cell and Regenerative Biotechnology, Konkuk Institute of Technology, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
36
|
Dimova T, Alexandrova M, Vangelov I, You Y, Mor G. The modeling of human implantation and early placentation: achievements and perspectives. Hum Reprod Update 2025; 31:133-163. [PMID: 39673726 DOI: 10.1093/humupd/dmae033] [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] [Revised: 10/29/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND Successful implantation is a critical step for embryo survival. The major losses in natural and assisted human reproduction appeared to occur during the peri-implantation period. Because of ethical constraints, the fascinating maternal-fetal crosstalk during human implantation is difficult to study and thus, the possibility for clinical intervention is still limited. OBJECTIVE AND RATIONALE This review highlights some features of human implantation as a unique, ineffective and difficult-to-model process and summarizes the pros and cons of the most used in vivo, ex vivo and in vitro models. We point out the variety of cell line-derived models and how these data are corroborated by well-defined primary cells of the same nature. Important aspects related to the handling, standardization, validation, and modus operandi of the advanced 3D in vitro models are widely discussed. Special attention is paid to blastocyst-like models recapitulating the hybrid phenotype and HLA profile of extravillous trophoblasts, which are a unique yet poorly understood population with a major role in the successful implantation and immune mother-embryo recognition. Despite raising new ethical dilemmas, extended embryo cultures and synthetic embryo models are also in the scope of our review. SEARCH METHODS We searched the electronic database PubMed from inception until March 2024 by using a multi-stage search strategy of MeSH terms and keywords. In addition, we conducted a forward and backward reference search of authors mentioned in selected articles. OUTCOMES Primates and rodents are valuable in vivo models for human implantation research. However, the deep interstitial, glandular, and endovascular invasion accompanied by a range of human-specific factors responsible for the survival of the fetus determines the uniqueness of the human implantation and limits the cross-species extrapolation of the data. The ex vivo models are short-term cultures, not relevant to the period of implantation, and difficult to standardize. Moreover, the access to tissues from elective terminations of pregnancy raises ethical and legal concerns. Easy-to-culture cancer cell lines have many limitations such as being prone to spontaneous transformation and lacking decent tissue characteristics. The replacement of the original human explants, primary cells or cancer cell lines with cultures of immortalized cell lines with preserved stem cell characteristics appears to be superior for in vitro modeling of human implantation and early placentation. Remarkable advances in our understanding of the peri-implantation stages have also been made by advanced three dimensional (3D) models i.e. spheroids, organoids, and assembloids, as placental and endometrial surrogates. Much work remains to be done for the optimization and standardization of these integrated and complex models. The inclusion of immune components in these models would be an asset to delineate mechanisms of immune tolerance. Stem cell-based embryo-like models and surplus IVF embryos for research bring intriguing possibilities and are thought to be the trend for the next decade for in vitro modeling of human implantation and early embryogenesis. Along with this research, new ethical dilemmas such as the moral status of the human embryo and the potential exploitation of women consenting to donate their spare embryos have emerged. The careful appraisal and development of national legal and ethical frameworks are crucial for better regulation of studies using human embryos and embryoids to reach the potential benefits for human reproduction. WIDER IMPLICATIONS We believe that our data provide a systematization of the available information on the modeling of human implantation and early placentation and will facilitate further research in this field. A strict classification of the advanced 3D models with their pros, cons, applicability, and availability would help improve the research quality to provide reliable outputs.
Collapse
Affiliation(s)
- Tanya Dimova
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Marina Alexandrova
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Ivaylo Vangelov
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Yuan You
- C.S. Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| | - Gil Mor
- C.S. Mott Center for Human Growth and Development, Wayne State University, Detroit, MI, USA
| |
Collapse
|
37
|
Xu S, Hu D, Ye Y, Mu Y, Xiong Y, Zhang Y. Identification of serum small non-coding RNA as biomarkers for endometrial receptivity. Genomics 2025; 117:111002. [PMID: 39848478 DOI: 10.1016/j.ygeno.2025.111002] [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: 07/16/2024] [Revised: 12/30/2024] [Accepted: 01/19/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Current endometrial receptivity analysis is invasive, preventing embryo transfer during the biopsy cycle. This study aims to screen serum sncRNAs as non-invasive biomarkers for ERA tests. METHODS The study included 12 infertile patients undergoing IVF-ET and ERA, whose serum samples were collected for high-energy sequencing technology to detect sncRNA expression profiles. We overexpressed and knocked down tsRNA-35:73-Asp-GTC-1 in the decidualized Immortalized Human Eutopic Endometrial Stromal Cells (HESC) model cultured in vitro to further investigate the its effect on decidualization. The predicted tsRNA-35:73-Asp-GTC-1 target gene was verified by PCR analysis. RESULTS We screened 286 differentially expressed tsRNAs, 46 miRNAs, and 106 piRNAs. KEGG analysis indicated that differentially expressed tsRNAs were associated with pathways such as 'Calcium signaling pathway,' 'Sphingolipid signaling pathway,' etc. The results of RT-qPCR validation showed that the trends of four significantly differentially expressed tsRNAs in serum and endometrium were consistent with sequencing results. ROC curves demonstrated that these four tsRNAs have good predictive value for endometrial receptivity. Overexpression of tsRNA-35:73-Asp-GTC-1 affected the morphology of decidualized cells, and the decidualization indicators also showed a decreasing trend. While knocking down tsRNA-35:73-Asp-GTC-1 had the opposite effect. The RT-qPCR results showed that tsRNA-35:73-Asp-GTC-1 was associated with the Wnt3 target gene. CONCLUSION Serum sncRNA analysis shows potential for studying the molecular mechanisms of endometrial receptivity. Four serum tsRNAs can serve as novel biomarkers for non-invasive endometrial receptivity detection. TsRNA-35:73-Asp-GTC-1 may further regulate endometrial receptivity by targeting Wnt3.
Collapse
Affiliation(s)
- Shaoyuan Xu
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China; Reproductive Medicine Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China; Hubei Clinical Research Center for Reproductive Medicine, Shiyan, Hubei 442000, China
| | - Dongling Hu
- Reproductive Medicine Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China; Hubei Clinical Research Center for Reproductive Medicine, Shiyan, Hubei 442000, China
| | - Yanqin Ye
- Reproductive Medicine Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China; Hubei Clinical Research Center for Reproductive Medicine, Shiyan, Hubei 442000, China
| | - Yanli Mu
- Reproductive Medicine Center, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China; Hubei Clinical Research Center for Reproductive Medicine, Shiyan, Hubei 442000, China
| | - Yao Xiong
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China; Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan, Hubei 430071, China.
| | - Yuanzhen Zhang
- Reproductive Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, China; Hubei Clinical Research Center for Prenatal Diagnosis and Birth Health, Wuhan, Hubei 430071, China.
| |
Collapse
|
38
|
Díaz M, Quesada-López T, Villarroya F, Casano P, López-Bermejo A, de Zegher F, Ibáñez L. The Proteome of Exosomes at Birth Predicts Insulin Resistance, Adrenarche and Liver Fat in Childhood. Int J Mol Sci 2025; 26:1721. [PMID: 40004184 PMCID: PMC11854951 DOI: 10.3390/ijms26041721] [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: 01/17/2025] [Revised: 02/13/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
It is unknown whether there are differentially expressed proteins (DEPs) in the circulating exosomes of appropriate- vs. small-for-gestational-age (AGA vs. SGA) infants, and if so, whether such DEPs relate to measures of endocrine-metabolic health and body composition in childhood. Proteomic analysis in cord-blood-derived exosomes was performed by label-free quantitative mass spectrometry in AGA (n = 20) and SGA infants (n = 20) and 91 DEPs were identified. Enrichment analysis revealed that they were related to complement and coagulation cascades, lipid metabolism, neural development, PI3K/Akt and RAS/RAF/MAPK signaling pathways, phagocytosis and focal adhesion. Protein-protein interaction (PPI) analysis identified 39 DEPs involved in the pathways enriched by the KEGG and Reactome. Those DEPs were associated with measures of adiposity and insulin resistance and with liver fat at age 7 (all p < 0.01). Multivariate linear regression analysis uncovered that two DEPs (up-regulated in SGA), namely PCYOX1 (related to adipogenesis) and HSP90AA1 (related to lipid metabolism and metabolic-dysfunction-associated steatotic liver disease progression), were independent predictors of the hepatic fat fraction at age 7 (β = 0.634; p = 0.002; R2 = 52% and β = 0.436; p = 0.009; R2 = 24%, respectively). These data suggest that DEPs at birth may predict insulin resistance, adrenarche and/or ectopic adiposity in SGA children at age 7, when an early insulin-sensitizing intervention could be considered.
Collapse
Affiliation(s)
- Marta Díaz
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Tania Quesada-López
- Department of Biomedicine, Institut de Recerca Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Network Biomedical Research Center of Physiopathology of Obesity and Nutrition (CIBEROBN), Health Institute Carlos III, 28029 Madrid, Spain;
| | - Francesc Villarroya
- Network Biomedical Research Center of Physiopathology of Obesity and Nutrition (CIBEROBN), Health Institute Carlos III, 28029 Madrid, Spain;
- Biochemistry and Molecular Biomedicine Department, Institute of Biomedicine, University of Barcelona, 08007 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues, 08950 Barcelona, Spain
| | - Paula Casano
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Abel López-Bermejo
- Pediatric Endocrinology Research Group, Girona Institute for Biomedical Research (IDIBGI), Faculty of Medicine, University of Girona and Dr. Josep Trueta Hospital, 17007 Girona, Spain;
| | - Francis de Zegher
- Leuven Research & Development, University of Leuven, 3000 Leuven, Belgium;
| | - Lourdes Ibáñez
- Endocrinology Department, Institut de Recerca Sant Joan de Déu, University of Barcelona, 08950 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
39
|
Tongu Y, Kasahara T, Akiyama Y, Suzuki T, Ho HJ, Matsumoto Y, Kujirai R, Kikuchi K, Nata K, Kanzaki M, Suzuki K, Watanabe S, Kawabe C, Miyata Y, Itai S, Toyohara T, Suzuki C, Tanaka T, Wada J, Tomioka Y, Abe T. Hypoglycemia and hyperinsulinemia induced by phenolic uremic toxins in CKD and DKD patients. Sci Rep 2025; 15:5762. [PMID: 39962199 PMCID: PMC11833073 DOI: 10.1038/s41598-025-87501-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: 02/17/2024] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
Patients with end-stage renal disease have lower fasting plasma glucose and HbA1c levels, with significantly higher insulin levels. For a long time, it has been believed that this higher insulin level in renal failure is due to decreased insulin clearance caused by reduced renal function. However, here we reported that accumulation of the gut microbiota-derived uremic toxin, phenyl sulfate (PS) in the renal failure, increased insulin secretion from the pancreas by enhanced glucose-stimulated insulin secretion. Other endogenous sulfides compounds which accumulated as in the renal failure also increased glucose-stimulated insulin secretion from β-cell. With RNA-seq analyses and gene knock down, we demonstrated that insulin secretion evoked by PS was mediated by Ddah2. In addition, we also found that PS increased insulin resistance through lncRNA expression and Erk phosphorylation in the adipocytes. To confirm the relationship between PS and glucose metabolism in human, we recruited 2 clinical cohort studies (DKD and CKD) including 462 patients, and found that there was a weak negative correlation between PS and HbA1c. Because these trials did not measure fasting insulin level, we alternatively used the urinary C-peptide/creatinine ratio (UCPCR) as an indicator of insulin resistance. We found that PS may induce insulin resistance in patients with eGFR < 60 mL/min/1.73 m2. These data suggest that the accumulation of uremic toxins modulates glucose metabolism and induced insulin resistance in CKD and DKD patients. Considering HbA1c as a reflection of chronic hyperglycemia and UCPCR as a reflection of chronic hyperinsulinemia, our findings indicate that PS is negatively associated with hyperglycemia independent of CKD, and positively associated with hyperinsulinemia in DKD patients.
Collapse
Affiliation(s)
- Yoshiyasu Tongu
- Tohoku University School of Medicine, Sendai, Japan
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoko Kasahara
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yasutoshi Akiyama
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Takehiro Suzuki
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Medical Science, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan
| | - Hsin-Jung Ho
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Faculty of Health Sciences, Hokkaido University, Sapporo, Japan
| | - Yotaro Matsumoto
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Ryota Kujirai
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Koichi Kikuchi
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Koji Nata
- Department of Medical Biochemistry, School of Pharmacy, Iwate Medical University, Morioka, Japan
| | - Makoto Kanzaki
- Department of Biomedical Engineering, Tohoku University, Sendai, Japan
| | - Kenshin Suzuki
- Tohoku University School of Medicine, Sendai, Japan
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shun Watanabe
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chiharu Kawabe
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yui Miyata
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shun Itai
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takafumi Toyohara
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chitose Suzuki
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuhiro Tanaka
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshihisa Tomioka
- Laboratory of Oncology, Pharmacy Practice and Sciences, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Takaaki Abe
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine, Sendai, Japan.
- Division of Nephrology, Endocrinology, and Vascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
- Division of Medical Science, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan.
| |
Collapse
|
40
|
Correia CD, Calado SM, Matos A, Esteves F, De Sousa-Coelho AL, Campinho MA, Fernandes MT. Advancing Glioblastoma Research with Innovative Brain Organoid-Based Models. Cells 2025; 14:292. [PMID: 39996764 PMCID: PMC11854129 DOI: 10.3390/cells14040292] [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: 01/12/2025] [Revised: 02/06/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
Glioblastoma (GBM) is a relatively rare but highly aggressive form of brain cancer characterized by rapid growth, invasiveness, and resistance to standard therapies. Despite significant progress in understanding its molecular and cellular mechanisms, GBM remains one of the most challenging cancers to treat due to its high heterogeneity and complex tumor microenvironment. To address these obstacles, researchers have employed a range of models, including in vitro cell cultures and in vivo animal models, but these often fail to replicate the complexity of GBM. As a result, there has been a growing focus on refining these models by incorporating human-origin cells, along with advanced genetic techniques and stem cell-based bioengineering approaches. In this context, a variety of GBM models based on brain organoids were developed and confirmed to be clinically relevant and are contributing to the advancement of GBM research at the preclinical level. This review explores the preparation and use of brain organoid-based models to deepen our understanding of GBM biology and to explore novel therapeutic approaches. These innovative models hold significant promise for improving our ability to study this deadly cancer and for advancing the development of more effective treatments.
Collapse
Affiliation(s)
- Cátia D. Correia
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve (UAlg), Campus de Gambelas, 8005-139 Faro, Portugal
| | - Sofia M. Calado
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
- Faculdade de Ciências e Tecnologia (FCT), Universidade dos Açores (UAc), 9500-321 Ponta Delgada, Portugal
| | - Alexandra Matos
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
| | - Filipa Esteves
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve (UAlg), Campus de Gambelas, 8005-139 Faro, Portugal
| | - Ana Luísa De Sousa-Coelho
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
- Escola Superior de Saúde (ESS), Universidade do Algarve (UAlg), Campus de Gambelas, 8005-139 Faro, Portugal
| | - Marco A. Campinho
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
- Faculdade de Medicina e Ciências Biomédicas (FMCB), Universidade do Algarve (UAlg), Campus de Gambelas, 8005-139 Faro, Portugal
| | - Mónica T. Fernandes
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, 8005-139 Faro, Portugal; (C.D.C.); (S.M.C.); (M.A.C.)
- Escola Superior de Saúde (ESS), Universidade do Algarve (UAlg), Campus de Gambelas, 8005-139 Faro, Portugal
| |
Collapse
|
41
|
Soto-Heras S, Volz LJ, Bovin N, Miller DJ. Porcine sperm bind to an oviduct glycan coupled to glass surfaces as a model of sperm interaction with the oviduct. Sci Rep 2025; 15:4680. [PMID: 39920342 PMCID: PMC11806115 DOI: 10.1038/s41598-025-88986-2] [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: 09/27/2024] [Accepted: 02/03/2025] [Indexed: 02/09/2025] Open
Abstract
During transport through the oviduct, sperm interact with epithelial cells by attaching to specific glycans, a mechanism believed to select sperm and prolong their viability. An in vitro model of sperm-oviduct interactions was developed, consisting of a glass surface (either a slide or a coverslip) to which an oviduct glycan (sulfated Lewis X trisaccharide; suLeX) is coupled. The ability of porcine sperm to attach to suLeX-surfaces and detach in response to progesterone and mature cumulus-oocyte complexes (COCs) was validated. The suLeX-coverslip was adapted for in vitro fertilization (IVF), termed glycan-IVF, by allowing porcine sperm to first bind suLeX before transferring mature COCs. The glycan-IVF method produced a percentage of fertilized oocytes comparable to that of conventional IVF (75.1 vs. 72.0%). Finally, the ability of the suLeX-coverslip to maintain sperm fertilizing ability over time was assessed. After 24 h of incubation, fertilization by sperm bound to the suLeX-coverslip was sustained, compared to sperm with unmodified coverslips (12.0 vs. 1.0%, p < 0.05). Furthermore, the percentage of polyspermic zygotes was reduced in the suLeX-coverslip method (17.7 vs. 41.3%, p < 0.05). This study validated an in vitro model for studying sperm-oviduct interactions, with potential applications in assisted reproductive technologies.
Collapse
Affiliation(s)
- Sandra Soto-Heras
- Department of Animal Sciences, Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA
- Epivara, 2109 S. Oak Street, Champaign, IL, 61820, USA
| | - Larissa J Volz
- Department of Animal Sciences, Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA
- , Cryomate, 320 N. Meridian St, Suite 606, Indianapolis, IN, 46204, USA
| | - Nicolai Bovin
- Shemyakin Institute of Bioorganic Chemistry, Moscow, Russia
| | - David J Miller
- Department of Animal Sciences, Institute of Genomic Biology, University of Illinois at Urbana-Champaign, 1207 West Gregory Drive, Urbana, IL, 61801, USA.
| |
Collapse
|
42
|
Zhang H, Yu M, Li L, Chen C, He Q. Obesity-related indices are associated with self-reported infertility in women: findings from the National Health and Nutrition Examination Survey. J Int Med Res 2025; 53:3000605251315019. [PMID: 39932267 PMCID: PMC11815785 DOI: 10.1177/03000605251315019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/06/2025] [Indexed: 02/14/2025] Open
Abstract
OBJECTIVE Obesity can contribute to infertility, but the exact relationship between infertility risk and obesity-related measurements like waist-to-height ratio (WHtR), body roundness index (BRI), conicity index (CoI), and A body shape index (ABSI) in women is uncertain. We investigated the association between these indices and female infertility. METHODS In this cross-sectional study, we used National Health and Nutrition Examination Survey data (2013-2018). We used weighted multivariable logistic regression analysis, receiver operating characteristic (ROC) curves, and subgroup analysis, as well as propensity score matching. RESULTS Among 3373 participants, 344 (10.2%) reported infertility. A significant link between higher infertility risk and increased WHtR, BRI, CoI, ABSI, and body mass index (BMI) was found. Multivariable logistic regression analysis showed WHtR (odds ratio [OR] = 1.27, 95% confidence interval [CI]: 1.14-1.42), BRI (OR = 1.09, 95% CI: 1.05-1.14), CoI (OR = 1.36, 95% CI: 1.18-1.56), ABSI (OR = 1.22, 95% CI: 1.12-1.33), and BMI (OR = 1.03, 95% CI: 1.02-1.05) were significantly associated with female infertility. CoI had the best diagnostic performance (area under the ROC curve 0.628, 95% CI: 0.597-0.658). CONCLUSIONS Obesity-related indices were positively linked to infertility risk among women in the United States. These indices serve as valuable tools for assessing female infertility risk.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Minmin Yu
- Department of Obstetrics and Gynecology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Li Li
- Department of Ultrasound, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chen Chen
- Department of Infectious Disease, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Qinyuan He
- Department of Obstetrics and Gynecology, The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
43
|
Vygonskaya M, Wu Y, Price TJ, Chen Z, Smith MT, Klyne DM, Han FY. The role and treatment potential of the complement pathway in chronic pain. THE JOURNAL OF PAIN 2025; 27:104689. [PMID: 39362355 DOI: 10.1016/j.jpain.2024.104689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/05/2024]
Abstract
The role of the complement system in pain syndromes has garnered attention on the back of preclinical and clinical evidence supporting its potential as a target for new analgesic pharmacotherapies. Of the components that make up the complement system, component 5a (C5a) and component 3a (C3a) are most strongly and consistently associated with pain. Receptors for C5a are widely found in immune resident cells (microglia, astrocytes, sensory neuron-associated macrophages (sNAMs)) in the central nervous system (CNS) as well as hematogenous immune cells (mast cells, macrophages, T-lymphocytes, etc.). When active, as is often observed in chronic pain conditions, these cells produce various inflammatory mediators including pro-inflammatory cytokines. These events can trigger nervous tissue inflammation (neuroinflammation) which coexists with and potentially maintains peripheral and central sensitization. C5a has a likely critical role in initiating this process highlighting its potential as a promising non-opioid target for treating pain. This review summarizes the most up-to-date research on the role of the complement system in pain with emphasis on the C5 pathway in peripheral tissue, dorsal root ganglia (DRG) and the CNS, and explores advances in complement-targeted drug development and sex differences. A perspective on the optimal application of different C5a inhibitors for different types (e.g., neuropathic, post-surgical and chemotherapy-induced pain, osteoarthritis pain) and stages (e.g., acute, subacute, chronic) of pain is also provided to help guide future clinical trials. PERSPECTIVE: This review highlights the role and mechanisms of complement components and their receptors in physiological and pathological pain. The potential of complement-targeted therapeutics for the treatment of chronic pain is also explored with a focus on C5a inhibitors to help guide future clinical trials.
Collapse
Affiliation(s)
- Marina Vygonskaya
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Youzhi Wu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Theodore J Price
- Center for Advanced Pain Studies, Department of Neuroscience, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Zhuo Chen
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Maree T Smith
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - David M Klyne
- NHMRC Centre of Clinical Research Excellence in Spinal Pain, Injury and Health, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Felicity Y Han
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
44
|
Xiao Y, Wang Y, Zhang M, Zhang Y, Ju Z, Wang J, Zhang Y, Yang C, Wang X, Jiang Q, Gao Y, Wei X, Liu W, Gao Y, Hu P, Huang J. Tankyrase inhibitor IWR-1 modulates HIPPO and Transforming Growth Factor β signaling in primed bovine embryonic stem cells cultured on mouse embryonic fibroblasts. Theriogenology 2025; 233:100-111. [PMID: 39613494 DOI: 10.1016/j.theriogenology.2024.11.010] [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: 08/25/2024] [Revised: 11/17/2024] [Accepted: 11/17/2024] [Indexed: 12/01/2024]
Abstract
The use of tankyrase inhibitors is essential for capturing livestock embryonic stem cells (ESC), yet their mechanisms of action remain largely uncharacterized. Previous studies indicate that their roles extend beyond the suppression of canonical WNT signaling. This study investigates the effects of the tankyrase inhibitor IWR-1 on maintaining the pluripotency of bovine embryonic stem cells (bESC) cultured on mitotically inactivated mouse embryonic fibroblasts (MEF). Notably, bESC exhibited significant differentiation after one month of IWR-1 withdrawal, without a clear bias toward any specific germ layer. IWR-1 effectively inhibited TNKS2 activity in bESC, whereas it suppressed TNKS1 protein level in growth-arrested MEF. Early differentiation upon IWR-1 removal induced more substantial transcriptomic changes in MEF than in bESC. Furthermore, cell communication analysis predicted that IWR-1 influenced several paracrine and autocrine signals within the culture system. We also observed that IWR-1 repressed protein abundance of the HIPPO pathway components including TEAD4 and YAP1 in bESC and decreased transcription of HIPPO targeted genes CYR61. Protein analysis in growth-arrested MEF suggested that IWR-1 modulated MEF function by impeding TGF-β1 activation and activin A secretion which mitigated nuclear localization of SMAD2/3 in the bESC. This study underscores the role of tankyrase inhibitors in ESC self-renewal by modulating key signaling pathways and orchestrating cell-cell interactions, which may be meaningful in understanding the delicate signaling control of pluripotency in livestock and improving the culture system.
Collapse
Affiliation(s)
- Yao Xiao
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Technical Innovation Center of Dairy Cattle Breeding Industry of Shandong Province, Jinan, 250100, China
| | - Yujie Wang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; College of Life Sciences, Shandong Normal University, Jinan, 250358, China
| | - Minghao Zhang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Yan Zhang
- Key Laboratory of Efficient Dairy Cattle Propagation and Germplasm Innovation of Ministry of Agriculture and Rural Affairs, Shandong OX Livestock Breeding Co., Ltd, Jinan, 250100, China
| | - Zhihua Ju
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Jinpeng Wang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Yaran Zhang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Chunhong Yang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Xiuge Wang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Qiang Jiang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Yaping Gao
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Xiaochao Wei
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China
| | - Wenhao Liu
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Technical Innovation Center of Dairy Cattle Breeding Industry of Shandong Province, Jinan, 250100, China
| | - Yundong Gao
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Technical Innovation Center of Dairy Cattle Breeding Industry of Shandong Province, Jinan, 250100, China; Key Laboratory of Efficient Dairy Cattle Propagation and Germplasm Innovation of Ministry of Agriculture and Rural Affairs, Shandong OX Livestock Breeding Co., Ltd, Jinan, 250100, China
| | - Peng Hu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China
| | - Jinming Huang
- Key Laboratory of Livestock and Poultry Multi-omics of Ministry of Agriculture and Rural Affairs, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Shandong Key Laboratory of Animal Disease Control and Breeding, Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, 250100, China; Technical Innovation Center of Dairy Cattle Breeding Industry of Shandong Province, Jinan, 250100, China; College of Life Sciences, Shandong Normal University, Jinan, 250358, China.
| |
Collapse
|
45
|
Olenic M, Deelkens C, Heyman E, De Vlieghere E, Zheng X, van Hengel J, De Schauwer C, Devriendt B, De Smet S, Thorrez L. Review: Livestock cell types with myogenic differentiation potential: Considerations for the development of cultured meat. Animal 2025; 19 Suppl 1:101242. [PMID: 39097434 DOI: 10.1016/j.animal.2024.101242] [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: 12/08/2023] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 08/05/2024] Open
Abstract
With the current environmental impact of large-scale animal production and societal concerns about the welfare of farm animals, researchers are questioning whether we can cultivate animal cells for the purpose of food production. This review focuses on a pivotal aspect of the cellular agriculture domain: cells. We summarised information on the various cell types from farm animals currently used for the development of cultured meat, including mesenchymal stromal cells, myoblasts, and pluripotent stem cells. The review delves into the advantages and limitations of each cell type and considers factors like the selection of the appropriate cell source, as well as cell culture conditions that influence cell performance. As current research in cultured meat seeks to create muscle fibers to mimic the texture and nutritional profile of meat, we focused on the myogenic differentiation capacity of the cells. The most commonly used cell type for this purpose are myoblasts or satellite cells, but given their limited proliferation capacity, efforts are underway to formulate myogenic differentiation protocols for mesenchymal stromal cells and pluripotent stem cells. The multipotent character of the latter cell types might enable the creation of other tissues found in meat, such as adipose and connective tissues. This review can help guiding the selection of a cell type or culture conditions in the context of cultured meat development.
Collapse
Affiliation(s)
- M Olenic
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium; Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - C Deelkens
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium; Medical Cell Biology, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - E Heyman
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - E De Vlieghere
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium; Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Belgium
| | - X Zheng
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium
| | - J van Hengel
- Medical Cell Biology, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - C De Schauwer
- Veterinary Stem Cell Research Unit, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - B Devriendt
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - S De Smet
- Laboratory for Animal Nutrition and Animal Product Quality, Department of Animal Sciences and Aquatic Ecology, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - L Thorrez
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, Kortrijk, Belgium.
| |
Collapse
|
46
|
Ma G, Fu X, Zhou L, Babarinde IA, Shi L, Yang W, Chen J, Xiao Z, Qiao Y, Ma L, Ou Y, Li Y, Chang C, Deng B, Zhang R, Sun L, Tong G, Li D, Li Y, Hutchins AP. The nuclear matrix stabilizes primed-specific genes in human pluripotent stem cells. Nat Cell Biol 2025; 27:232-245. [PMID: 39789220 DOI: 10.1038/s41556-024-01595-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 12/11/2024] [Indexed: 01/12/2025]
Abstract
The nuclear matrix, a proteinaceous gel composed of proteins and RNA, is an important nuclear structure that supports chromatin architecture, but its role in human pluripotent stem cells (hPSCs) has not been described. Here we show that by disrupting heterogeneous nuclear ribonucleoprotein U (HNRNPU) or the nuclear matrix protein, Matrin-3, primed hPSCs adopted features of the naive pluripotent state, including morphology and upregulation of naive-specific marker genes. We demonstrate that HNRNPU depletion leads to increased chromatin accessibility, reduced DNA contacts and increased nuclear size. Mechanistically, HNRNPU acts as a transcriptional co-factor that anchors promoters of primed-specific genes to the nuclear matrix with POLII to promote their expression and their RNA stability. Overall, HNRNPU promotes cell-type stability and when reduced promotes conversion to earlier embryonic states.
Collapse
Affiliation(s)
- Gang Ma
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Xiuling Fu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Lulu Zhou
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Isaac A Babarinde
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Liyang Shi
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Wenting Yang
- Department of Reproductive Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiao Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhen Xiao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Yu Qiao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Lisha Ma
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Yuhao Ou
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Yuhao Li
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Chen Chang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Boping Deng
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Ran Zhang
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Li Sun
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Guoqing Tong
- Department of Reproductive Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dongwei Li
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Yiming Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China.
| | - Andrew P Hutchins
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
47
|
Ma S, Xia E, Zhang M, Hu Y, Tian S, Zheng X, Li B, Ma G, Su R, Sun K, Fan Q, Yang F, Guo G, Guo C, Shang Y, Zhou X, Zhou X, Wang J, Han Y. Role of the FOXM1/CMA/ER stress axis in regulating the progression of nonalcoholic steatohepatitis. Clin Transl Med 2025; 15:e70202. [PMID: 39924645 PMCID: PMC11807764 DOI: 10.1002/ctm2.70202] [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: 06/12/2024] [Revised: 01/09/2025] [Accepted: 01/16/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND/AIMS The molecular mechanisms driving nonalcoholic steatohepatitis (NASH) progression are poorly understood. This research examines the involvement of chaperone-mediated autophagy (CMA) in NASH progression. METHODS Hepatic CMA activity was analysed in NASH mice and patients. Lysosome-associated membrane protein 2A (LAMP2A) was knocked down or overexpressed to assess the effects of hepatocyte-specific CMA on NASH progression. Mice received a high-fat diet or a methionine and choline-deficient diet to induce NASH. Palmitic acid was employed to mimic lipotoxicity-induced hepatocyte damage in vitro. The promoter activity of FOXM1 was evaluated via ChIP and dual-luciferase reporter assays. RESULTS Hepatic CMA activity was substantially low in NASH mice and patients. LAMP2A knockdown resulted in hepatocyte-specific CMA deficiency, which promoted fibrosis and hepatic inflammation in NASH mice. Both in vitro and in vivo, CMA deficiency also exacerbated hepatocyte damage and endoplasmic reticulum (ER) stress. Mechanistically, CMA deficiency in hepatocytes increased cholesterol accumulation by blocking the degradation of 3-hydroxy-3-methylglutaryl coenzyme A (HMGCR), a key cholesterol synthesis-related enzyme, and the accumulated cholesterol subsequently induced ER stress and hepatocyte damage. The restoration of hepatocyte-specific CMA activity effectively ameliorated diet-induced NASH and ER stress in vivo and in vitro. FOXM1 directly bound to LAMP2A promoter and negatively regulated its transcription. The upregulation of FOXM1 expression impaired CMA and enhanced ER stress, which in turn increased FOXM1 expression, resulting in a vicious cycle and promoting NASH development. CONCLUSIONS This study highlights the significance of the FOXM1/CMA/ER stress axis in NASH progression and proposes novel therapeutic targets for NASH. KEY POINTS Chaperone-mediated autophagy (CMA) deficiency in hepatocytes promotes hepatic inflammation and fibrosis in mice with nonalcoholic steatohepatitis (NASH) by inducing cholesterol accumulation and endoplasmic reticulum (ER) stress. Upregulated FOXM1 impairs CMA by suppressing the transcription of lysosome-associated membrane protein 2A (LAMP2A), a rate-limiting component of CMA. ER stress increases FOXM1 expression and cholesterol accumulation. FOXM1/CMA/ER stress axis forms a vicious circle and promotes the development of NASH.
Collapse
Affiliation(s)
- Shuoyi Ma
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Erzhuo Xia
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Miao Zhang
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Yinan Hu
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Siyuan Tian
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Xiaohong Zheng
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Bo Li
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Gang Ma
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Rui Su
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Keshuai Sun
- Department of GastroenterologyThe Air Force Hospital From Eastern Theater of PLANanjingChina
| | - Qingling Fan
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Fangfang Yang
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Guanya Guo
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Changcun Guo
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Yulong Shang
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Xinmin Zhou
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Xia Zhou
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| | - Jingbo Wang
- Science and Technology Innovation Research InstituteTangdu Hospital, The Fourth Military Medical UniversityXi'anChina
| | - Ying Han
- State Key Laboratory of Cancer BiologyXijing Hospital of Digestive Diseases, The Fourth Military Medical UniversityXi'anChina
| |
Collapse
|
48
|
Batty L, Park J, Qin L, Riaz M, Lin Y, Xu Z, Gao X, Li X, Lopez C, Zhang W, Hoareau M, Fallon ME, Huang Y, Luo H, Luo J, Ménoret S, Li P, Jiang Z, Smith P, Sachs DH, Tellides G, Ignacio Anegon, Pober JS, Liu P, Qyang Y. Vascular endothelial cells derived from transgene-free pig induced pluripotent stem cells for vascular tissue engineering. Acta Biomater 2025; 193:171-184. [PMID: 39681154 DOI: 10.1016/j.actbio.2024.12.033] [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: 07/08/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 12/18/2024]
Abstract
Induced pluripotent stem cells (iPSCs) hold great promise for the treatment of cardiovascular diseases through cell-based therapies, but these therapies require extensive preclinical testing that is best done in species-in-species experiments. Pigs are a good large animal model for these tests due to the similarity of their cardiovascular system to humans. However, a lack of adequate pig iPSCs (piPSCs) that are analogous to human iPSCs has greatly limited the potential usefulness of this model system. Herein, transgene-free piPSCs with true pluripotency were generated by using reprogramming factors in an optimized pig pluripotency medium. Using an effective differentiation protocol, piPSCs were used to derive endothelial cells (ECs) which displayed EC markers and functionality comparable to native pig ECs. Further, piPSC-ECs demonstrated suitability for vascular tissue engineering, producing a tissue engineered vascular conduit (TEVC) that displayed the upregulation of flow responding markers. In an in vivo functional study, these piPSC-EC-TEVCs maintained the expression of endothelial markers and prevented thrombosis as interposition inferior vena cava grafts in immunodeficient rats. The piPSCs described in this study open up the possibility of unique preclinical species-in-species large animal modeling for the furtherance of modeling of cell-based cardiovascular tissue engineering therapies. STATEMENT OF SIGNIFICANCE: While there has been significant progress in the development of cellularized cardiovascular tissue engineered therapeutics using stem cells, few of them have moved into clinical trials. This is due to the lack of a robust preclinical large animal model to address the high safety and efficacy standards for transplanted therapeutics. In this study, pig stem cells that are analagous to human's were created to address this bottleneck. They demonstrated the ability to differentiate into functional endothelial cells and were able to create a tissue engineered therapeutic that is analogous to a human therapy. With these cells, future experiments testing the safety and efficacy of tissue engineered constructs are possible, bringing these crucial therapeutics closer to the patients that need them.
Collapse
Affiliation(s)
- Luke Batty
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA; Department of Pathology, Yale University, New Haven, CT 06510, USA
| | - Jinkyu Park
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA; Department of Physiology, College of Medicine, Hallym University, Hallymdaehak-gil, Chuncheon-si, Gangwon-Do, 24252, South Korea
| | - Lingfeng Qin
- Department of Surgery, Yale University, New Haven, CT 06520, USA
| | - Muhammad Riaz
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Yuyao Lin
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA; Department of Plastic, Aesthetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhen Xu
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Xuefei Gao
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Xin Li
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Colleen Lopez
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Wei Zhang
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Marie Hoareau
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Meghan E Fallon
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Yan Huang
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Hangqi Luo
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Jiesi Luo
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA
| | - Séverine Ménoret
- Center for Research in Transplantation and Translational Immunology UMR1064, INSERM, Nantes Université, Nantes, France; Nantes Université, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
| | - Peining Li
- Department of Genetics, Yale University, New Haven, CT 06519, USA
| | - Zhenting Jiang
- Department of Earth & Planetary Sciences, Yale University, New Haven, CT 06511, USA
| | - Peter Smith
- Department of Comparative Medicine, Yale University, New Haven, CT 06520, USA
| | - David H Sachs
- Department of Surgery, Columbia Center for Translational Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA; Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - George Tellides
- Department of Surgery, Yale University, New Haven, CT 06520, USA
| | - Ignacio Anegon
- Center for Research in Transplantation and Translational Immunology UMR1064, INSERM, Nantes Université, Nantes, France; Nantes Université, SFR Santé, Inserm UMS 016, CNRS UMS 3556, Nantes, France
| | - Jordan S Pober
- Department of Pathology, Yale University, New Haven, CT 06510, USA; Department of Immunobiology, Yale University, New Haven, CT 06520, USA
| | - Pentao Liu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Stem Cell and Regenerative Medicine Consortium, Pokfulam, Hong Kong, China
| | - Yibing Qyang
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, New Haven, CT 06511, USA; Yale Stem Cell Center, 10 Amistad Street, New Haven, CT 06511, USA; Department of Pathology, Yale University, New Haven, CT 06510, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06519, USA.
| |
Collapse
|
49
|
Stefanović M, Vukomanović P, Kutlesic R, Trenkić M, Dimitrov V, Stefanović A, Cvetanović V. The Effect of Autologous Platelet Rich Plasma on Endometrial Receptivity: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:134. [PMID: 39859116 PMCID: PMC11767130 DOI: 10.3390/medicina61010134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/03/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
Background and Objectives: Autologous platelet-rich plasma (PRP) transfusions are a relatively new treatment method used in different fields of medicine, including the field of reproductive medicine. One of the applications of these concentrated platelet infusions is the treatment of endometrial receptivity, which is a key factor for embryo implantation. There are implications that PRP infusions can lead to increased endometrial thickness, endometrial receptivity, and significantly elevated clinical pregnancy rates. Our objective is to briefly understand what PRP is and to, through a narrative review, summarize the findings from studies focused on evaluating the benefits of PRP infusions to treat thin endometrium with the goal of achieving better endometrial receptivity. Materials and Methods: Reference data was searched using Medline, PubMed, and EMBASE to identify reports from 2015 to 2024. The combination of search words used was "PRP" and "platelet-rich plasma" with "thin endometrium", "endometrial receptivity", "endometrial thickness", and "endometrial implantation". Obtained articles were screened, and suited studies (randomized controlled trials, case reports, case series, pilot studies, and reviews) were included in the present review. Reports not available in the English language were eliminated from the current review. Results: The results from most of the reviewed studies showed a positive effect of autologous PRP infusions on increasing endometrial thickness, enhancing endometrial receptivity, and elevating clinical pregnancy rates. The majority of the evaluated findings revealed endometrial thickness > 7 mm (increased endometrial thickness was observed in each evaluated study) following the PRP treatment. More than 50% of the evaluated studies resulted in enhanced endometrial thickness, increased endometrial receptivity, and an elevated pregnancy rate after the PRP application. Conclusions: Autologous PRP infusions for treating endometrium are a relatively new method that has shown promising results. Its major strengths are availability and proper application, which eliminates possible immunological reactions or disease transmission. The main drawbacks are not enough data on safety (i.e., its effect on endometriosis) and the lack of uniformity in the PRP preparation, which would provide optimal standardized quality and quantity of the PRP product and, thus, optimal treatment results.
Collapse
Affiliation(s)
- Milan Stefanović
- Faculty of Medicine, University of Niš, Blvd. Dr Zoran Đinđić 81, 18000 Niš, Serbia; (P.V.); (M.T.); (V.D.); (A.S.); (V.C.)
- Gynecology and Obstetrics Clinic, Clinical Center Niš, Blvd. Dr Zoran Đinđić 48, 18000 Niš, Serbia
| | - Predrag Vukomanović
- Faculty of Medicine, University of Niš, Blvd. Dr Zoran Đinđić 81, 18000 Niš, Serbia; (P.V.); (M.T.); (V.D.); (A.S.); (V.C.)
- Gynecology and Obstetrics Clinic, Clinical Center Niš, Blvd. Dr Zoran Đinđić 48, 18000 Niš, Serbia
| | - Ranko Kutlesic
- Faculty of Medicine, University of Niš, Blvd. Dr Zoran Đinđić 81, 18000 Niš, Serbia; (P.V.); (M.T.); (V.D.); (A.S.); (V.C.)
- Gynecology and Obstetrics Clinic, Clinical Center Niš, Blvd. Dr Zoran Đinđić 48, 18000 Niš, Serbia
| | - Milan Trenkić
- Faculty of Medicine, University of Niš, Blvd. Dr Zoran Đinđić 81, 18000 Niš, Serbia; (P.V.); (M.T.); (V.D.); (A.S.); (V.C.)
- Gynecology and Obstetrics Clinic, Clinical Center Niš, Blvd. Dr Zoran Đinđić 48, 18000 Niš, Serbia
| | - Vanja Dimitrov
- Faculty of Medicine, University of Niš, Blvd. Dr Zoran Đinđić 81, 18000 Niš, Serbia; (P.V.); (M.T.); (V.D.); (A.S.); (V.C.)
- Gynecology and Obstetrics Clinic, Clinical Center Niš, Blvd. Dr Zoran Đinđić 48, 18000 Niš, Serbia
| | - Aleksa Stefanović
- Faculty of Medicine, University of Niš, Blvd. Dr Zoran Đinđić 81, 18000 Niš, Serbia; (P.V.); (M.T.); (V.D.); (A.S.); (V.C.)
- Gynecology and Obstetrics Clinic, Clinical Center Niš, Blvd. Dr Zoran Đinđić 48, 18000 Niš, Serbia
| | - Vladimir Cvetanović
- Faculty of Medicine, University of Niš, Blvd. Dr Zoran Đinđić 81, 18000 Niš, Serbia; (P.V.); (M.T.); (V.D.); (A.S.); (V.C.)
- Gynecology and Obstetrics Clinic, Clinical Center Niš, Blvd. Dr Zoran Đinđić 48, 18000 Niš, Serbia
| |
Collapse
|
50
|
Davalieva K, Kocarev D, Plaseska-Karanfilska D. Decoding recurrent pregnancy loss: insights from comparative proteomics studies. Biol Reprod 2025; 112:1-17. [PMID: 39288094 DOI: 10.1093/biolre/ioae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/28/2024] [Accepted: 09/16/2024] [Indexed: 09/19/2024] Open
Abstract
Recurrent pregnancy loss represents a common disorder that affects up to 2% of the women aiming at childbirth with long-term consequences on family and society. Factors contributing to it in more than half of the cases are still unknown. Comparative proteomic analysis can provide new insights into the biological pathways underlining the pathogenesis of recurrent pregnancy loss. Until now, chorionic villi, decidua, placenta, endometrium, and maternal blood from women with recurrent pregnancy loss have been analyzed by comparative proteomics studies. In this review, we aimed to provide a critical evaluation of the published comparative studies of recurrent pregnancy loss on human samples, gathered by systematic literature search using PubMed and Google Scholar databases. We provide a detailed overview of the analyzed materials, proteomics platforms, proposed candidate biomarkers and altered pathways and processes linked with recurrent pregnancy loss. The top, most identified and validated biomarker candidates from all studies are discussed, followed by bioinformatics analysis of the available high-throughput data and presentation of common altered processes and pathways in recurrent pregnancy loss. Finally, future directions aimed at developing new and efficient therapeutic strategies are discussed as well.
Collapse
Affiliation(s)
- Katarina Davalieva
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, Krste Misirkov 2, 1000 Skopje, North Macedonia
| | - Damjan Kocarev
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, Krste Misirkov 2, 1000 Skopje, North Macedonia
| | - Dijana Plaseska-Karanfilska
- Research Centre for Genetic Engineering and Biotechnology "Georgi D Efremov", Macedonian Academy of Sciences and Arts, Krste Misirkov 2, 1000 Skopje, North Macedonia
| |
Collapse
|