1
|
An S, Hou S, Xu F, Yan H, Zhang W, Xiang J, Chen H, Zhang H, Dong L, Sun X, Huo R, Chen Y, Wang X, Yang Y. WDR36 Regulates Trophectoderm Differentiation During Human Preimplantation Embryonic Development Through Glycolytic Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2412222. [PMID: 39656902 DOI: 10.1002/advs.202412222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/12/2024] [Indexed: 12/17/2024]
Abstract
Mammalian pre-implantation development is a complex process involving sophisticated regulatory dynamics. WD repeat domain 36 (WDR36) is known to play a critical role in mouse early embryonic development, but its regulatory function in human embryogenesis is still elusive due to limited access to human embryos. The human pluripotent stem cell-derived blastocyst-like structure, termed a blastoid, offers an alternative means to study human development in a dish. In this study, after verifying that WDR36 inhibition disrupted polarization in mouse early embryos, it is further demonstrated that WDR36 interference can block human blastoid formation, dominantly hindering the trophectoderm lineage commitment. Both transcriptomics and targeted metabolomics analyses revealed that WDR36 interference downregulated glucose metabolism. WDR36 can interact with glycolytic metabolic protein lactate dehydrogenase A (LDHA), thereby positively regulating glycolysis during the late stage of human blastoid formation. Taken together, the study has established a mechanistic connection between WDR36, glucose metabolism, and cell fate determination during early embryonic lineage commitment, which may provide potential insights into novel therapeutic targets for early adverse pregnancy interventions.
Collapse
Affiliation(s)
- Shiyu An
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Shuyue Hou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Feifei Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Huanyu Yan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Wenyi Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jinfeng Xiang
- Fourth Clinical Medicine College, Nanjing Medical University, Nanjing, 210004, China
- Department of Obstetrics, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Institute, Nanjing, 210004, China
| | - Haoran Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Hanwen Zhang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Lingling Dong
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Xiaobin Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Ran Huo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yun Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xi Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Prenatal Diagnosis of the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yang Yang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Innovation Center of Suzhou Nanjing Medical University, Suzhou, Jiangsu, 215000, China
| |
Collapse
|
2
|
Fang H, Lin D, Zhang Z, Chen H, Zheng Z, Jiang D, Wang W. Association of coexposure to perfluoroalkyl and polyfluoroalkyl compounds and heavy metals with pregnancy loss and reproductive lifespan: The mediating role of cholesterol. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117160. [PMID: 39388969 DOI: 10.1016/j.ecoenv.2024.117160] [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: 09/06/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
Previous studies have demonstrated the toxic effects of per- and polyfluoroalkyl substances (PFASs) and heavy metals on the reproductive system. However, the interactions and combined effects of these substances remain unexplored. This study utilizes data from the National Health and Nutrition Examination Survey to investigate the associations between coexposure to four types of PFASs, lead (Pb), mercury (Hg) and self-reported pregnancy loss and reproductive lifespan in females. Genes associated with these substances and abortion were identified via the Comparative Toxicogenomics Database. The results revealed that Ln-PFOA (IRR=1.88, 95 % CI=1.42-2.50, Ln--: log transformed), Ln-PFOS (IRR=1.58, 95 % CI=1.12-2.22), Ln-PFHxS (IRR=1.99, 95 % CI=1.57-2.52), and Ln-Hg (IRR=1.92, 95 % CI=1.41-2.43) were positively associated with the risk of pregnancy loss. Ln-PFOA (β=1.27, 95 % CI=0.28-2.27), Ln-PFOS (β=1.01, 95 % CI=0.39-1.63), Ln-PFHxS (β=0.71, 95 % CI=0.12-1.63), Ln-PFNA (β=1.15, 95 % CI=0.23-2.08), Ln-Pb (β=3.87, 95 % CI=2.58-5.15), and Ln-Hg (β=1.01, 95 % CI=0.39-1.64) exposures were positively associated with reproductive lifespan. The mixed and overall effects of coexposure to PFASs and heavy metals were positively correlated with the risk of pregnancy loss and reproductive lifespan. Cholesterol partially mediated the association with the risk of pregnancy loss, whereas delay in menopause fully mediated the association with reproductive lifespan. Significant additive interactions were observed between PFOA and Pb and between PFOS, PFHxS, PFNA and Hg at high levels of coexposure. Thirty-nine overlapping genes associated with abortion were identified for these substances, and further analyses revealed that these genes significantly interact and may contribute to abortion through oxidative stress.
Collapse
Affiliation(s)
- Hua Fang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Dai Lin
- Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Department of Nutrition and Food Safety, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Ziqi Zhang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Haoting Chen
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Zixin Zheng
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Dongdong Jiang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenxiang Wang
- Department of Health Inspection and Quarantine, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China; Fujian Province Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, Fujian, China.
| |
Collapse
|
3
|
Zhao Y, Liang Y, Cai L, Cai L, Huang B, Han P, Zhang X, Zhang H, Chen Z, Yin X, Duan P, Shou H, Zhu X, Wang Z, Wan Q, Huang J, Qian J. Comprehensive Proteomic Analysis Reveals Distinct Features and a Diagnostic Biomarker Panel for Early Pregnancy Loss in Histological Subtypes. Mol Cell Proteomics 2024; 23:100848. [PMID: 39321873 PMCID: PMC11541848 DOI: 10.1016/j.mcpro.2024.100848] [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: 04/10/2024] [Revised: 07/21/2024] [Accepted: 08/16/2024] [Indexed: 09/27/2024] Open
Abstract
Early pregnancy loss (EPL) is a common event in human reproduction and is classified into histological subtypes such as hydropic abortion (HA) and hydatidiform moles, including complete hydatidiform moles (CHMs) and partial hydatidiform moles (PHMs). However, accurate diagnosis and improved patient management remain challenging due to high rates of misdiagnosis and diverse prognostic risks. Therefore, diagnostic biomarkers for EPL are urgently needed. Our study aimed to identify biomarkers for EPL through comprehensive proteomic analysis. Ten CHMs, six PHMs, ten HAs, and 10 normal control products of conception were used to obtain a proteomic portrait. Parallel reaction monitoring-targeted proteomic and regression analyses were used to verify and select the diagnostic signatures. Finally, 14 proteins were selected and a panel of diagnostic classifiers (DLK1, SPTB/COL21A1, and SAR1A) was built to represent the CHM, PHM, and normal control groups (area under the receiver operating characteristic curve = 0.900, 0.804/0.885, and 0.991, respectively). This high diagnostic power was further validated in another independent cohort (n = 148) by immunohistochemistry (n = 120) and Western blot analyses (n = 28). The protein SPTB was selected for further biological behavior experiments in vitro. Our data suggest that SPTB maintains trophoblast cell proliferation, angiogenesis, cell motility, and the cytoskeleton network. This study provides a comprehensive proteomic portrait and identifies potential diagnostic biomarkers. These findings enhance our understanding of EPL pathogenesis and offer novel targets for diagnosis and therapeutic interventions.
Collapse
Affiliation(s)
- Yating Zhao
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Yingjiqiong Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Zhejiang Province, PR China; Biomedical Big Data Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China
| | - Luya Cai
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Limeng Cai
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Bo Huang
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Peilin Han
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Xiaofei Zhang
- Department of Pathology, The Zhejiang University School of Medicine Women's Hospital, Hangzhou, China
| | - Huifang Zhang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhen Chen
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangang Yin
- Department of Diagnosis, Ningbo Clinical Pathology Diagnosis Center, Ningbo, China
| | - Ping Duan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huafeng Shou
- Department of Gynecology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang Province, China
| | - Xiaoxu Zhu
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Zhe Wang
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Qihong Wan
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China
| | - Jinyan Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Zhejiang University School of Medicine First Affiliated Hospital, Zhejiang Province, PR China; Biomedical Big Data Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, PR China; Zhejiang University Cancer Center, Zhejiang University, Zhejiang Province, PR China.
| | - Jianhua Qian
- Department of Gynecology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou City, Zhejiang Province, PR China.
| |
Collapse
|
4
|
Li Y, Hou J, Yang L, Zhang T, Jiang Y, Du Z, Ma H, Li G, Zhu J, Chen P. ITRAQ Based Proteomics Reveals the Potential Mechanism of Placental Injury Induced by Prenatal Stress. Int J Mol Sci 2024; 25:9978. [PMID: 39337469 PMCID: PMC11432224 DOI: 10.3390/ijms25189978] [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: 07/19/2024] [Revised: 09/14/2024] [Accepted: 09/14/2024] [Indexed: 09/30/2024] Open
Abstract
Maternal stress experienced during prenatal development is recognized as a significant risk factor for neurodevelopmental and neuropsychiatric disorders across the offspring's lifespan. The placental barrier serves a crucial function in safeguarding the fetus from detrimental exposures during gestation. However, previous investigations have not yet comprehensively elucidated the extensive connections between prenatal stress and the expression of placental proteins. In this study, we used iTRAQ-based quantitative proteomics to elucidate the placental adaptive mechanisms of pregnant rats in response to fear-induced stress. Our results showed that during pregnancy, exposure to fear-induced stress led to a pathological hypercoagulable state in the mother's body. Placental circulation was also disrupted, significantly reducing placental efficiency and blood oxygen saturation in newborn rats. Proteomic analyses showed that most of the DEPs were annotated to the PI3K-Akt and ECM-receptor interaction signaling pathway. In addition, the expressions of CDC37, HSP90β, AKT, p-AKT and p-mTOR were down-regulated significantly in the placenta. Our results demonstrated that prenatal fear-induced stress led to inhibition of the cellular signal transduction of placental PI3K/AKT/mTOR, which affected biological processes such as rRNA processing, translation, protein folding, protein stability, and oxygen transport in the placenta. These abnormalities in biological functions could potentially damage the barrier function of the placenta and thereby result in abnormal development in the offspring.
Collapse
Affiliation(s)
- Yujie Li
- Department of Biochemistry, School of Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China;
| | - Junlin Hou
- Department of Integrated Traditional Chinese and Western Medicine, School of Traditional Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; (J.H.); (T.Z.); (Y.J.); (Z.D.); (H.M.); (G.L.); (J.Z.)
| | - Liping Yang
- Department of Integrated Traditional Chinese and Western Medicine, School of Traditional Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; (J.H.); (T.Z.); (Y.J.); (Z.D.); (H.M.); (G.L.); (J.Z.)
| | - Tong Zhang
- Department of Integrated Traditional Chinese and Western Medicine, School of Traditional Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; (J.H.); (T.Z.); (Y.J.); (Z.D.); (H.M.); (G.L.); (J.Z.)
| | - Yu Jiang
- Department of Integrated Traditional Chinese and Western Medicine, School of Traditional Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; (J.H.); (T.Z.); (Y.J.); (Z.D.); (H.M.); (G.L.); (J.Z.)
| | - Zhixing Du
- Department of Integrated Traditional Chinese and Western Medicine, School of Traditional Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; (J.H.); (T.Z.); (Y.J.); (Z.D.); (H.M.); (G.L.); (J.Z.)
| | - Huizi Ma
- Department of Integrated Traditional Chinese and Western Medicine, School of Traditional Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; (J.H.); (T.Z.); (Y.J.); (Z.D.); (H.M.); (G.L.); (J.Z.)
| | - Gai Li
- Department of Integrated Traditional Chinese and Western Medicine, School of Traditional Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; (J.H.); (T.Z.); (Y.J.); (Z.D.); (H.M.); (G.L.); (J.Z.)
| | - Jianghui Zhu
- Department of Integrated Traditional Chinese and Western Medicine, School of Traditional Medicine, Henan University of Chinese Medicine, Zhengzhou 450046, China; (J.H.); (T.Z.); (Y.J.); (Z.D.); (H.M.); (G.L.); (J.Z.)
| | - Ping Chen
- Gynaecology and Obstetrics, College of First Clinical Medical, Henan University of Chinese Medicine, Zhengzhou 450003, China;
| |
Collapse
|
5
|
Wu D, Wang S, Wang F, Zhang Q, Zhang Z, Li X. Lactate dehydrogenase A (LDHA)-mediated lactate generation promotes pulmonary vascular remodeling in pulmonary hypertension. J Transl Med 2024; 22:738. [PMID: 39103838 PMCID: PMC11302077 DOI: 10.1186/s12967-024-05543-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/26/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND High levels of lactate are positively associated with prognosis and mortality in pulmonary hypertension (PH). Lactate dehydrogenase A (LDHA) is a key enzyme for the production of lactate. This study is undertaken to investigate the role and molecular mechanisms of lactate and LDHA in PH. METHODS Lactate levels were measured by a lactate assay kit. LDHA expression and localization were detected by western blot and Immunofluorescence. Proliferation and migration were determined by CCK8, western blot, EdU assay and scratch-wound assay. The right heart catheterization and right heart ultrasound were measured to evaluate cardiopulmonary function. RESULTS In vitro, we found that lactate promoted proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) in an LDHA-dependent manner. In vivo, we found that LDHA knockdown reduced lactate overaccumulation in the lungs of mice exposed to hypoxia. Furthermore, LDHA knockdown ameliorated hypoxia-induced vascular remodeling and right ventricular dysfunction. In addition, the activation of Akt signaling by hypoxia was suppressed by LDHA knockdown both in vivo and in vitro. The overexpression of Akt reversed the inhibitory effect of LDHA knockdown on proliferation in PASMCs under hypoxia. Finally, LDHA inhibitor attenuated vascular remodeling and right ventricular dysfunction in Sugen/hypoxia mouse PH model, Monocrotaline (MCT)-induced rat PH model and chronic hypoxia-induced mouse PH model. CONCLUSIONS Thus, LDHA-mediated lactate production promotes pulmonary vascular remodeling in PH by activating Akt signaling pathway, suggesting the potential role of LDHA in regulating the metabolic reprogramming and vascular remodeling in PH.
Collapse
Affiliation(s)
- Daiqian Wu
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu Cardiovascular Disease Research Institute, Chengdu, 610014, PR China
| | - Shuo Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, 400042, PR China
| | - Fengxian Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing, 400042, PR China
| | - Qing Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, PR China
| | - Zhen Zhang
- Department of Cardiology, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu Cardiovascular Disease Research Institute, Chengdu, 610014, PR China.
| | - Xingbing Li
- Department of Cardiology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, 400021, PR China.
| |
Collapse
|
6
|
Moses RM, Stenhouse C, Halloran KM, Sah N, Newton MG, Hoskins EC, Washburn SE, Johnson GA, Wu G, Bazer FW. Metabolic pathways of glucose and fructose: II Spatiotemporal expression of genes involved in synthesis and transport of lactate in ovine conceptuses†. Biol Reprod 2024; 111:159-173. [PMID: 38531778 DOI: 10.1093/biolre/ioae047] [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: 12/06/2023] [Revised: 02/14/2024] [Accepted: 03/22/2024] [Indexed: 03/28/2024] Open
Abstract
Lactate, an abundant molecule in fetal fluids and blood of mammalian species, is often overlooked as a metabolic waste product generated during pregnancy. Most of the glucose and fructose consumed by ovine conceptuses is converted to lactate, but proteins involved in lactate metabolism and transport have not been investigated. This study characterized total lactate produced by ovine conceptuses throughout gestation, as well as expression of mRNAs and proteins involved in lactate metabolism. Lactate increased in abundance in the uterine lumen during the preimplantation period and was more abundant than pyruvate. The abundance of lactate in allantoic and amniotic fluids increased with advancing days of gestation and most abundant on Day 125 of pregnancy (P < 0.05). Lactate dehydrogenase subunits A (converts pyruvate to lactate) and B (converts lactate to pyruvate) were expressed by conceptuses throughout gestation. Lactate is transported via monocarboxylic acid transporters SLC16A1 and SLC16A3, both of which were expressed by the conceptus throughout gestation. Additionally, the interplacentomal chorioallantois from Day 126 expressed SLC16A1 and SLC16A3 and transported lactate across the tissue. Hydrocarboxylic acid receptor 1 (HCAR1), a receptor for lactate, was localized to the uterine luminal and superficial glandular epithelia of pregnant ewes throughout gestation and conceptus trophectoderm during the peri-implantation period of gestation. These results provide novel insights into the spatiotemporal profiles of enzymes, transporters, and receptor for lactate by ovine conceptuses throughout pregnancy.
Collapse
Affiliation(s)
- Robyn M Moses
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Claire Stenhouse
- Department of Animal Science, Pennsylvania State University, University Park, PA, USA
| | - Katherine M Halloran
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor MI, USA
| | - Nirvay Sah
- Department of Pathology, University of California-San Diego, San Diego, CA, USA
| | - Makenzie G Newton
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Emily C Hoskins
- College of Biomedical and Diagnostic Sciences, University of Tennessee, Knoxville, TN, USA
| | - Shannon E Washburn
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX, USA
| | - Gregory A Johnson
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| |
Collapse
|
7
|
Hou T, Zhang P, Tian H, Luo Y, Li J, Zhang K, Li Y. Semaphorin 4A Maintains Trophoblastic Function via Activating the STAT3 Pathway. Biomolecules 2024; 14:826. [PMID: 39062540 PMCID: PMC11274653 DOI: 10.3390/biom14070826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
The migration, proliferation, and apoptosis of trophoblastic cells play a crucial role in ensuring the effective preservation of pregnancy at the maternal-fetal interface. Any deviations in the structure and function of these cells might potentially result in the development of numerous pregnancy-related disorders, including missed abortion (MA). This study involved the examination of semaphorin 4A (SEMA4A) expression in missed abortion (n = 18) and normal early pregnancy (n = 18) villus. The findings of this study indicate a statistically significant decrease in the expression of SEMA4A in the villi of individuals diagnosed with missed abortion, as compared to the control group. The results of our vitro study showed that SEMA4A promoted the migration and proliferation of trophoblast cells and inhibited their apoptosis. Subsequent studies have shown that SEMA4A may be involved in regulating p-STAT3/STAT3, MMP9, bcl-2, and BAX levels. In summary, the findings of this study indicate a correlation between the decreased level of SEMA4A in chorionic villi and missed abortion. These results offer novel theoretical insights into the proper implantation and development of SEMA4A embryos at the maternal-fetal interface.
Collapse
Affiliation(s)
- Taotao Hou
- Graduate School of Hebei North University, Zhangjiakou 075000, China
- Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang 050051, China
- Department of Gynecology, Tianjin Beichen Hospital, Tianjin 300400, China
| | - Pingping Zhang
- Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang 050051, China
| | - Haishen Tian
- Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang 050051, China
| | - Yan Luo
- Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang 050051, China
| | - Juan Li
- Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang 050051, China
| | - Kuo Zhang
- Graduate School of University of Science and Technology Beijing, Beijing 100083, China
| | - Yali Li
- Department of Reproductive Genetics, Hebei General Hospital, Shijiazhuang 050051, China
| |
Collapse
|
8
|
Khan NLA, Muhandiram S, Dissanayake K, Godakumara K, Midekessa G, Andronowska A, Heath PR, Kodithuwakku S, Hart AR, Fazeli A. Effect of 3D and 2D cell culture systems on trophoblast extracellular vesicle physico-chemical characteristics and potency. Front Cell Dev Biol 2024; 12:1382552. [PMID: 38835509 PMCID: PMC11148233 DOI: 10.3389/fcell.2024.1382552] [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: 02/05/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
The growing understanding of the role of extracellular vesicles (EVs) in embryo-maternal communication has sparked considerable interest in their therapeutic potential within assisted reproductive technology, particularly in enhancing implantation success. However, the major obstacle remains the large-scale production of EVs, and there is still a gap in understanding how different culture systems affect the characteristics of the EVs. In the current study, trophoblast analogue human chorionic carcinoma cell line was cultivated in both conventional monolayer culture (2D) and as spheroids in suspension culture (3D) and how the cell growth environment affects the physical, biochemical and cellular signalling properties of EVs produced by them was studied. Interestingly, the 3D system was more active in secreting EVs compared to the 2D system, while no significant differences were observed in terms of morphology, size, and classical EV protein marker expression between EVs derived from the two culture systems. There were substantial differences in the proteomic cargo profile and cellular signalling potency of EVs derived from the two culture systems. Notably, 2D EVs were more potent in inducing a cellular response in endometrial epithelial cells (EECs) compared to 3D EVs. Therefore, it is essential to recognize that the biological activity of EVs depends not only on the cell of origin but also on the cellular microenvironment of the parent cell. In conclusion, caution is warranted when selecting an EV production platform, especially for assessing the functional and therapeutic potential of EVs through in vitro studies.
Collapse
Affiliation(s)
- Norhayati Liaqat Ali Khan
- Division of Clinical Medicine, School of Medicine and Population Health, The Medical School, University of Sheffield, Sheffield, United Kingdom
- Centre of Preclinical Science Studies, Faculty of Dentistry, University Teknologi MARA (UiTM), Sg. Buloh, Selangor, Malaysia
| | - Subhashini Muhandiram
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
| | - Keerthie Dissanayake
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Kasun Godakumara
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Getnet Midekessa
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Aneta Andronowska
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Olsztyn, Poland
| | - Paul R Heath
- Division of Clinical Medicine, School of Medicine and Population Health, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Suranga Kodithuwakku
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Animal Science, Faculty of Agriculture, University of Peradeniya, Peradeniya, Sri Lanka
| | - Amber Rose Hart
- Division of Clinical Medicine, School of Medicine and Population Health, The Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Alireza Fazeli
- Division of Clinical Medicine, School of Medicine and Population Health, The Medical School, University of Sheffield, Sheffield, United Kingdom
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, Tartu, Estonia
- Department of Pathophysiology, Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| |
Collapse
|
9
|
Zhang L, Gu X. 4-hydroxysesamin protects rat with right ventricular failure due to pulmonary hypertension by inhibiting JNK/p38 MAPK signaling. Aging (Albany NY) 2024; 16:8142-8154. [PMID: 38728253 PMCID: PMC11131979 DOI: 10.18632/aging.205808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 04/08/2024] [Indexed: 05/12/2024]
Abstract
The specific mechanism of 4-hydroxysesamin (4-HS), a modification of Sesamin, on right ventricular failure due to pulmonary hypertension (PH) is ominous. By creating a rat model of PH in vivo and a model of pulmonary artery smooth muscle cell (PASMC) hypoxia and inflammation in vitro, the current work aimed to investigate in depth the molecular mechanism of the protective effect of 4-HS. In an in vitro model of hypoxia PASMC, changes in cell proliferation and inflammatory factors were detected after treatment with 4-HS, followed by changes in the JNK/p38 MAPK signaling pathway as detected by Western blot signaling pathway. The findings demonstrated that 4-HS was able to minimize PASMC cell death, block the JNK/p38 MAPK signaling pathway, and resist the promoting effect of hypoxia on PASMC cell proliferation. Following that, we found that 4-HS could both mitigate the right ventricular damage brought on by MCT and had a protective impact on rats Monocrotaline (MCT)-induced PH in in vivo investigations. The key finding of this study is that 4-HS may protect against PH by inhibiting the JNK/p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Lingnan Zhang
- Department of Cardiovascular, Affiliated Hospital of Hebei University, Baoding 071000, Hebei, China
- Department of Cardiovascular Sciences, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| | - Xinshun Gu
- Department of Cardiovascular, The Second Hospital of Hebei Medicine University, Shijiazhuang 050000, Hebei, China
- Department of Cardiovascular Sciences, Hebei Medical University, Shijiazhuang 050017, Hebei, China
| |
Collapse
|
10
|
Liu Z, Wang C, Tang Y, Zhang X, Pei J, Liu H, Yu Y, Gu W. ENO1 promotes trophoblast invasion regulated by E2F8 in recurrent miscarriage. FASEB J 2024; 38:e23631. [PMID: 38661062 DOI: 10.1096/fj.202302032rr] [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: 10/07/2023] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024]
Abstract
Recurrent miscarriage (RM) is related to the dysfunction of extravillous trophoblast cells (EVTs), but the comprehensive mechanisms remain largely unexplored. We analyzed single-cell RNA sequencing (scRNA-seq), bulk RNA sequencing and microarray datasets obtained from Gene Expression Omnibus (GEO) database to explore the hub genes in the mechanisms of RM. We identified 1724 differentially expressed genes (DEGs) in EVTs from the RM, and they were all expressed along the trajectory of EVTs. These DEGs were associated with hypoxia and glucose metabolism. Single-cell Regulatory Network Inference and Clustering (SCENIC) analysis revealed that E2F transcription factor (E2F) 8 (E2F8) was a key transcription factor for these DEGs. And the expression of ENO1 can be positively regulated by E2F8 via RNA sequencing analysis. Subsequently, we performed immunofluorescence assay (IF), plasmid transfection, western blotting, chromatin immunoprecipitation (ChIP), real-time quantitative polymerase chain reaction (qRT-PCR), and transwell assays for validation experiments. We found that the expression of alpha-Enolase 1 (ENO1) was lower in the placentas of RM. Importantly, E2F8 can transcriptionally regulate the expression of ENO1 to promote the invasion of trophoblast cells by inhibiting secreted frizzled-related protein 1/4 (SFRP1/4) to activate Wnt signaling pathway. Our results suggest that ENO1 can promote trophoblast invasion via an E2F8-dependent manner, highlighting a potential novel target for the physiological mechanisms of RM.
Collapse
Affiliation(s)
- Zhenzhen Liu
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chengjie Wang
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yao Tang
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xiaoyue Zhang
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jiangnan Pei
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Haiyan Liu
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yi Yu
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Weirong Gu
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
11
|
Yi C, Song H, Liang H, Ran Y, Tang J, Chen E, Li F, Fu L, Wang Y, Chen F, Wang Y, Ding Y, Xie Y. TBX3 reciprocally controls key trophoblast lineage decisions in villi during human placenta development in the first trimester. Int J Biol Macromol 2024; 263:130220. [PMID: 38368983 DOI: 10.1016/j.ijbiomac.2024.130220] [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: 10/29/2023] [Revised: 01/30/2024] [Accepted: 02/03/2024] [Indexed: 02/20/2024]
Abstract
Human trophoblastic lineage development is intertwined with placental development and pregnancy outcomes, but the regulatory mechanisms underpinning this process remain inadequately understood. In this study, based on single-nuclei RNA sequencing (snRNA-seq) analysis of the human early maternal-fetal interface, we compared the gene expression pattern of trophoblast at different developmental stages. Our findings reveal a predominant upregulation of TBX3 during the transition from villous cytotrophoblast (VCT) to syncytiotrophoblast (SCT), but downregulation of TBX3 as VCT progresses into extravillous trophoblast cells (EVT). Immunofluorescence analysis verified the primary expression of TBX3 in SCT, partial expression in MKi67-positive VCT, and absence in HLA-G-positive EVT, consistent with our snRNA-seq results. Using immortalized trophoblastic cell lines (BeWo and HTR8/SVneo) and human primary trophoblast stem cells (hTSCs), we observed that TBX3 knockdown impedes SCT formation through RAS-MAPK signaling, while TBX3 overexpression disrupts the cytoskeleton structure of EVT and hinders EVT differentiation by suppressing FAK signaling. In conclusion, our study suggests that the spatiotemporal expression of TBX3 plays a critical role in regulating trophoblastic lineage development via distinct signaling pathways. This underscores TBX3 as a key determinant during hemochorial placental development.
Collapse
Affiliation(s)
- Cen Yi
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Honglan Song
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Hongxiu Liang
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Yujie Ran
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Jing Tang
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Enxiang Chen
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Fangfang Li
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Lijuan Fu
- Department of Gynecology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China 400021; Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, School of Traditional Chinese Medicine, Chongqing Medical University, Chongqing 400016, China; Department of Basic Medical Sciences, Changsha Medical University, Hunan 410219, China
| | - Yaqi Wang
- Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Fengming Chen
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, 410129, China
| | - Yingxiong Wang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Yubin Ding
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Youlong Xie
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China; Joint International Research Laboratory of Reproduction and Development of the Ministry of Education of China, School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
12
|
Lv C, Li D, Zhang Z, Han Y, Li Y, Song H, Cheng Q, Yang S, Lu Y, Zhao F. Association between urinary polycyclic aromatic hydrocarbons and unexplained recurrent spontaneous abortion from a case-control study. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116093. [PMID: 38364758 DOI: 10.1016/j.ecoenv.2024.116093] [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: 11/22/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/18/2024]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) have been reported to be associated with adverse pregnancy outcomes. However, there is limited knowledge regarding the effects of single or mixed PAHs exposure on unexplained recurrent spontaneous abortion (URSA). This study aimed to investigate the association between monohydroxylated polycyclic aromatic hydrocarbons (OH-PAHs) and URSA in a case-control study. The results showed that 1-NAP, 2-NAP, 9-FLU, and 1-PYR were detected in 100% of the subjects among measured all sixteen OH-PAHs. Compared with those in the lowest quartiles, participants in the highest quartiles of 3-BAA were associated with a higher risk of URSA (OR (95%CI) = 3.56(1.28-9.85)). With each one-unit increase of ln-transformed 3-BAA, the odds of URSA increased by 41% (OR (95%CI) = 1.41(1.05-1.89)). Other OH-PAHs showed negative or non-significant associations with URSA. Weighted quantile sum (WQS) regression, Bayesian kernel machine regression (BKMR), and quantile-based g-computation (qgcomp) analyses consistently identified 3-BAA as the major contributor to the mixture effect of OH-PAHs on URSA. Our findings suggest that exposure to 3-BAA may be a potential risk factor for URSA. However, further prospective studies are needed to validate our findings in the future.
Collapse
Affiliation(s)
- Chunxian Lv
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Dandan Li
- Fengtai District Center for Disease Control and Prevention, Beijing 100071, China
| | - Zheng Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yingying Han
- Fengtai District Center for Disease Control and Prevention, Beijing 100071, China
| | - Yawei Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Haocan Song
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Qianxi Cheng
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China
| | - Siyu Yang
- Institute of Public Health, Henan Provincial Center for Disease Control and Prevention, Zhengzhou, Henan 450016, China
| | - Yifu Lu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China; Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| | - Feng Zhao
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing 100021, China.
| |
Collapse
|
13
|
Wu S, Han L, Zhou M, Li X, Luo L, Wang Z, Yan S, Li F, Chen J, Yang J. LncRNA AOC4P recruits TRAF6 to regulate EZH2 ubiquitination and participates in trophoblast glycolysis and M2 macrophage polarization which is associated with recurrent spontaneous abortion. Int Immunopharmacol 2023; 125:111201. [PMID: 37951195 DOI: 10.1016/j.intimp.2023.111201] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/30/2023] [Accepted: 11/06/2023] [Indexed: 11/13/2023]
Abstract
During embryo implantation, trophoblast cells rely on large amounts of energy produced by glycolysis for their rapid growth and invasion. The disorder of trophoblast metabolism may lead to recurrent spontaneous abortion (RSA). Lactate, which is produced by the glycolysis of trophoblast cells during early pregnancy, can promote the polarization of M2 macrophages and maintain an anti-inflammatory environment at the maternal-fetal interface. Our study found that amine oxidase copper-containing 4 pseudogene (AOC4P) was abnormally increased in villi from RSA patients. It inhibited the glycolysis of trophoblast cells and thus hindered the polarization of M2 macrophages. Further studies showed that AOC4P combines with tumor necrosis factor receptor-associated factor 6 (TRAF6) to upregulate TRAF6 expression. TRAF6 acted as an E3 ubiquitin ligase to promote ubiquitination and degradation of zeste homolog 2 (EZH2). These results provided new insights into the important role played by AOC4P at the maternal-fetal interface.
Collapse
Affiliation(s)
- Shujuan Wu
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei 430060, China
| | - Lu Han
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei 430060, China
| | - Mengqi Zhou
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei 430060, China
| | - Xing Li
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Lingbo Luo
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei 430060, China
| | - Zehao Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei 430060, China
| | - Sisi Yan
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei 430060, China
| | - Faminzi Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei 430060, China
| | - Jiao Chen
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei 430060, China.
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei 430060, China.
| |
Collapse
|
14
|
Gou R, Zhang X. Glycolysis: A fork in the path of normal and pathological pregnancy. FASEB J 2023; 37:e23263. [PMID: 37889786 DOI: 10.1096/fj.202301230r] [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: 06/19/2023] [Revised: 09/17/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023]
Abstract
Glucose metabolism is vital to the survival of living organisms. Since the discovery of the Warburg effect in the 1920s, glycolysis has become a major research area in the field of metabolism. Glycolysis has been extensively studied in the field of cancer and is considered as a promising therapeutic target. However, research on the role of glycolysis in pregnancy is limited. Recent evidence suggests that blastocysts, trophoblasts, decidua, and tumors all acquire metabolic energy at specific stages in a highly similar manner. Glycolysis, carefully controlled throughout pregnancy, maintains a dynamic and coordinated state, so as to maintain the homeostasis of the maternal-fetal interface and ensure normal gestation. In the present review, we investigate metabolic remodeling and the selective propensity of the embryo and placenta for glycolysis. We then address dysregulated glycolysis that occurs in the cellular interactive network at the maternal-fetal interface in miscarriage, preeclampsia, fetal growth restriction, and gestational diabetes mellitus. We provide new insights into the field of maternal-fetal medicine from a metabolic perspective, thus revealing the mystery of human pregnancy.
Collapse
Affiliation(s)
- Rui Gou
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, P.R. China
| | - Xiaohong Zhang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, P.R. China
| |
Collapse
|
15
|
Chen X, Song QL, Wang JY, Ji R, Li ZH, Cao ML, Mu XF, Guo DY, Zhang Y, Yang J. Profilin1 regulates trophoblast invasion and macrophage differentiation. THE AMERICAN JOURNAL OF PATHOLOGY 2023:S0002-9440(23)00165-7. [PMID: 37164274 DOI: 10.1016/j.ajpath.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/08/2023] [Accepted: 04/18/2023] [Indexed: 05/12/2023]
Abstract
Unexplained recurrent spontaneous abortion (URSA) has been associated with the dysfunction of trophoblasts and decidual macrophages. Current evidence suggests that profilin1 (PFN1) plays an important role in many biological processes. However, little is currently known on whether PFN1 is related to URSA. The location of PFN1 was detected by immunohistochemistry. The level of PFN1 were detected by qRT-PCR, western blot and immunohistochemistry. The proliferation of trophoblasts was detected by CCK8 and EdU assays. Apoptosis of trophoblasts was detected by TUNEL assays. The migration and invasion ability of trophoblasts were assessed by the wound-healing test and transwell test. Macrophages were cultured in trophoblast conditioned medium and the polarization of macrophages was detected. PFN1 expression was observed in in cytotrophoblasts, syncytiotrophoblasts, and extravillous trophoblasts and decreased in the villous tissue of URSA patients. The migration and invasion ability and cell viability of trophoblastic cell lines that underwent PFN1 knockdown significantly decreased, and apoptosis increased. Opposite findings were observed following the overexpression of PFN1 in trophoblastic cells. In addition, PFN1 could regulate trophoblast function through PI3K/AKT signal transduction rather than MAPK signaling pathways. In addition, this study also found that knockdown of PFN1 in trophoblast promotes TNF-α secretion to induce macrophage polarization to M1 phenotype, mediated by the NF- κ B signaling pathway.
Collapse
Affiliation(s)
- Xin Chen
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Qian Lin Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jia Yu Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Rui Ji
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Ze Hong Li
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China
| | - Ming Liang Cao
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xue Feng Mu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Duan Ying Guo
- Longgang District People's Hospital of Shenzhen, Shenzhen, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, Hubei, China.
| |
Collapse
|