1
|
Li L, Baek KH. Exploring Potential Biomarkers in Recurrent Pregnancy Loss: A Literature Review of Omics Studies to Molecular Mechanisms. Int J Mol Sci 2025; 26:2263. [PMID: 40076883 PMCID: PMC11900470 DOI: 10.3390/ijms26052263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Recurrent pregnancy loss (RPL) is characterized by the occurrence of three or more consecutive spontaneous pregnancy losses before 20-24 weeks of gestation. Despite significant progress in the investigation of the biological pathways associated with unexplained RPL, the precise molecular mechanisms remain elusive. Recent advances in multi-omics approaches have identified numerous biomarkers that offer potential avenues for understanding the underlying complexities of RPL. The aim of this comprehensive literature review was to investigate the functional roles of these candidate markers and explore the possible key mechanisms that may contribute to RPL. We also aimed to elucidate the functional networks predicted by omics analyses, which hold promise for providing invaluable insights into novel diagnostic and therapeutic strategies for women experiencing RPL. Furthermore, this review expands on clinical implications and possible applications, highlighting those currently moving towards clinical use and ongoing studies developing in this direction.
Collapse
Affiliation(s)
- Lan Li
- The Key Laboratory for Preclinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 063210, China;
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA University, CHA General Hospital, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| |
Collapse
|
2
|
Chen Z, Yin H, Long Y, Zhu H, Xiong R, Duan X, Liu H, Li J. Metabonomics analysis of decidual tissue in patients with recurrent spontaneous abortion. J Reprod Immunol 2025; 167:104398. [PMID: 39608278 DOI: 10.1016/j.jri.2024.104398] [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: 09/22/2024] [Revised: 11/01/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024]
Abstract
OBJECTIVE This study aimed to delineate the metabolic differences and identify enriched pathways in the decidual tissue of patients with recurrent spontaneous abortion (RSA) compared to normal pregnant women. METHODS A cohort of 25 RSA patients and 25 normal pregnant women was recruited for the study. Non-targeted metabolomic analysis of decidual tissue was conducted using high-performance liquid chromatography coupled with mass spectrometry (HPLC-MS). Principal component analysis (PCA) and orthogonal partial least squares discriminant analysis (OPLS-DA) were employed to identify differential metabolites. Pathway enrichment analysis was performed using the Kyoto Encyclopedia of Genes and Genomes (KEGG) database to elucidate associated metabolic pathways. Quantitative polymerase chain reaction (qPCR) was utilized to assess the expression levels of key proteins related to these pathways, including acyl-CoA synthetase long-chain family member 4 (ACSL4), glutathione peroxidase 4 (GPX4), and indoleamine 2,3-dioxygenase (IDO). RESULTS A total of 54 metabolites were identified with significant differences between the decidual tissues of RSA patients and normal controls, corresponding to 29 significantly enriched metabolic pathways (P<0.05). The expression of ACSL4 was markedly upregulated, while the expression of GPX4 and IDO were significantly downregulated in RSA patients (P<0.05). CONCLUSIONS This study elucidates substantial metabolic disruptions in the decidual tissue of RSA patients, identifying 54 differential metabolites and 29 enriched pathways. The altered expression of ACSL4, GPX4, and IDO underscores their potential involvement in the pathogenesis of RSA. These findings provide critical insights into the metabolic mechanisms underlying RSA and suggest promising targets for diagnostic and therapeutic interventions.
Collapse
Affiliation(s)
- Zhihui Chen
- School of Life Sciences, Central South University, Changsha, Hunan, China; Changsha Hospital for Maternal and Child Health Care, Changsha, Hunan, China
| | - Huifang Yin
- School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Youmei Long
- School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Huiling Zhu
- School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Renmei Xiong
- School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Xin Duan
- School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Hongyu Liu
- School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Jiada Li
- School of Life Sciences, Central South University, Changsha, Hunan, China.
| |
Collapse
|
3
|
Yue L, Xu H. MicroRNA-200c promotes trophoblast cell dysfunction via inhibition of PI3K/Akt signaling in unexplained recurrent spontaneous abortion. Reprod Biol 2024; 24:100951. [PMID: 39243437 DOI: 10.1016/j.repbio.2024.100951] [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: 02/25/2024] [Revised: 07/24/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
Dysfunction in trophoblast cells is closely associated with the development of recurrent spontaneous abortion (RSA). Previous reports have indicated that microRNA (miR)-200c was upregulated in the serum of patients who have had abortions. This study aimed to investigate the regulatory effects and mechanisms of miR-200c in trophoblast cells. The human extravillous trophoblast cell line HTR-8/SVneo was either subjected to knockdown or overexpression of miR-200c, and its levels were measured using RT-qPCR. The cell behaviors of HTR-8/SVneo were assessed using CCK-8, Transwell, wound healing assays, and flow cytometry. Western blotting was used to detect the protein levels of Ki67, Bcl-2, Bax, MMP2/9, and PI3K/Akt-related markers. The findings revealed that miR-200c levels were higher in the villous tissues of URSA patients. Depletion of miR-200c impeded HTR-8/SVneo cell apoptosis and enhanced cell migration, invasiveness, and proliferation, while overexpression of miR-200c exhibited the opposite effects. The data suggested that mechanistically, miR-200c inactivated PI3K/Akt signaling in trophoblast cells. Furthermore, rescue experiments demonstrated that blocking PI3K/Akt signaling reversed the effects of miR-200c depletion on HTR-8/SVneo cell behavior. Therefore, miR-200c depletion can potentially improve trophoblast cell function by activating PI3K/Akt signaling.
Collapse
Affiliation(s)
- Lei Yue
- Department of Obstetrics and Gynecology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China.
| | - Hui Xu
- Department of Obstetrics and Gynecology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| |
Collapse
|
4
|
Lai H, Yang Y, Zhang J. Advances in post-translational modifications and recurrent spontaneous abortion. Gene 2024; 927:148700. [PMID: 38880188 DOI: 10.1016/j.gene.2024.148700] [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: 01/31/2024] [Revised: 05/25/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Recurrent spontaneous abortion (RSA) is defined as two or more pregnancy loss, which affects approximately 1-2% of women's fertility. The etiology of RSA has not yet been fully revealed, which poses a great problem for clinical treatment. Post- translational modifications(PTMs) are chemical modifications that play a crucial role in the functional proteome. A considerable number of published studies have shown the relationship between post-translational modifications of various proteins and RSA. The study of PTMs contributes to elucidating the role of modified proteins in the pathogenesis of RSA, as well as the design of more effective diagnostic/prognostic tools and more targeted treatments. Most reviews in the field of RSA have only focused on RNA epigenomics research. The present review reports the latest research developments of PTMs related to RSA, such as glycosylation, phosphorylation, Methylation, Acetylation, Ubiquitination, etc.
Collapse
Affiliation(s)
- Hanhong Lai
- Jinan University, Guangzhou, Guangdong 510632, People's Republic of China
| | - Yi Yang
- Jinan University, Guangzhou, Guangdong 510632, People's Republic of China
| | - Jun Zhang
- Jinan University, Guangzhou, Guangdong 510632, People's Republic of China.
| |
Collapse
|
5
|
Wang D, Zhao XR, Li YF, Wang RL, Li XB, Wang CX, Li YW. Quercetin promotes the proliferation, migration, and invasion of trophoblast cells by regulating the miR-149-3p/AKT1 axis. Kaohsiung J Med Sci 2024; 40:903-915. [PMID: 39162596 DOI: 10.1002/kjm2.12887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/21/2024] Open
Abstract
Recurrent spontaneous abortion (RSA) has a complex pathogenesis with an increasing prevalence and is one of the most intractable clinical challenges in the field of reproductive medicine. Quercetin (QCT) is an effective active ingredient extracted from Semen Cuscutae and Herba Taxilli used in traditional Chinese medicine for tonifyng the kidneys and promoting fetal restoration. Although QCT helps improve adverse pregnancy outcomes, the specific mechanism remains unclear. The trophoblast cell line HTR-8/SVneo cultured in vitro was treated with different concentrations of QCT, and the cell counting kit-8 assay, wound healing assay, transwell assay, and western blotting were used to evaluate the effects and mechanisms of QCT on the proliferation, migration, and invasion of HTR-8/SVneo cells, respectively. To assess the expression levels of miR-149-3p and AKT serine/threonine kinase 1 (AKT1), quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting analysis were performed. A dual-luciferase reporter assay was used to investigate the potential regulatory relationship between miR-149-3p and AKT1. Our results showed that QCT promoted the proliferation, migration, and invasion of trophoblast cells, promoted the expression of MMP2, MMP9, and vimentin, and downregulated the expression of E-cadherin. Mechanistically, QCT downregulated the expression of miR-149-3p and upregulated the expression of AKT1, and miR-149-3p directly targets AKT1, negatively regulating its expression. Overexpression of miR-149-3p and silencing of AKT1 counteracted the promotional effects of QCT on trophoblast proliferation, migration, and invasion. Taken together, QCT regulates the migration and invasion abilities of HTR-8/SVneo cells through the miR-149-3p/AKT1 axis, which may provide a promising therapeutic approach for RSA.
Collapse
Affiliation(s)
- Dan Wang
- The Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou City, Henan Province, China
| | - Xin-Rui Zhao
- Chinese Medicine College, Hong Kong Baptist University, Kowloon City, Hong Kong, China
| | - Yi-Fan Li
- The Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou City, Henan Province, China
| | - Rui-Lin Wang
- The Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou City, Henan Province, China
| | - Xue-Bing Li
- Department of Clinical Laboratory Centre, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou City, Henan Province, China
| | - Chun-Xia Wang
- The Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou City, Henan Province, China
- Department of Clinical Laboratory Centre, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou City, Henan Province, China
| | - Yong-Wei Li
- The Second Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou City, Henan Province, China
- Department of Clinical Laboratory Centre, Henan Province Hospital of Traditional Chinese Medicine, The Second Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou City, Henan Province, China
| |
Collapse
|
6
|
De Falco F, Cutarelli A, Leonardi L, Marcus I, Roperto S. Vertical Intrauterine Bovine and Ovine Papillomavirus Coinfection in Pregnant Cows. Pathogens 2024; 13:453. [PMID: 38921751 PMCID: PMC11206582 DOI: 10.3390/pathogens13060453] [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: 04/10/2024] [Revised: 05/08/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
There is very little information available about transplacental infections by the papillomavirus in ruminants. However, recent evidence has emerged of the first report of vertical infections of bovine papillomavirus (BPV) in fetuses from naturally infected, pregnant cows. This study reports the coinfection of BPV and ovine papillomavirus (OaPV) in bovine fetuses from infected pregnant cows suffering from bladder tumors caused by simultaneous, persistent viral infections. Some molecular mechanisms involving the binary complex composed of Eras and platelet-derived growth factor β receptor (PDGFβR), by which BPVs and OaPVs contribute to reproductive disorders, have been investigated. A droplet digital polymerase chain reaction (ddPCR) was used to detect and quantify the nucleic acids of the BPVs of the Deltapapillomavirus genus (BPV1, BPV2, BPV13, and BPV14) and OaPVs belonging to the Deltapapillomavirus (OaPV1, OaPV2, and OaPV4) and Dyokappapapillomavirus (OaPV3) genera in the placenta and fetal organs (heart, lung, liver, and kidneys) of four bovine fetuses from four pregnant cows with neoplasia of the urinary bladder. A papillomaviral evaluation was also performed on the bladder tumors and peripheral blood of these pregnant cows. In all fetal and maternal samples, the genotype distribution of BPVs and OaPVs were evaluated using both their DNA and RNA. A BPV and OaPV coinfection was seen in bladder tumors, whereas only BPV infection was found in peripheral blood. The genotype distribution of both the BPVs and OaPVs detected in placentas and fetal organs indicated a stronger concordance with the viral genotypes detected in bladder tumors rather than in peripheral blood. This suggests that the viruses found in placentas and fetuses may have originated from infected bladders. Our study highlights the likelihood of vertical infections with BPVs and OaPVs and emphasizes the importance of gaining further insights into the mechanisms and consequences of this exposure. This study warrants further research as adverse pregnancy outcomes are a major source of economic losses in cattle breeding.
Collapse
Affiliation(s)
- Francesca De Falco
- Dipartimento di Medicina Veterinaria e delle Produzioni Animali, Università degli Studi di Napoli Federico II, 80137 Naples, Italy;
- Area Science Park, Campus di Baronissi, Università degli Studi di Salerno, 84081 Baronissi, Italy
| | - Anna Cutarelli
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, 80055 Portici, Italy;
| | - Leonardo Leonardi
- Dipartimento di Medicina Veterinaria, Università degli Studi di Perugia, 06126 Perugia, Italy;
| | - Ioan Marcus
- Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 400000 Cluj-Napoca, Romania;
| | - Sante Roperto
- Dipartimento di Medicina Veterinaria e delle Produzioni Animali, Università degli Studi di Napoli Federico II, 80137 Naples, Italy;
| |
Collapse
|
7
|
Han Y, Wang Y, Zhang C, Li Y, Guo J, Tian C. Metastasis-associated lung adenocarcinoma transcript 1 induces methyl-CpG-binding domain protein 4 in mice with recurrent spontaneous abortion caused by anti-phospholipid antibody positivity. Placenta 2023; 137:38-48. [PMID: 37068447 DOI: 10.1016/j.placenta.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/27/2023] [Accepted: 04/07/2023] [Indexed: 04/19/2023]
Abstract
INTRODUCTION Antiphospholipid syndrome is an autoimmune disease characterized by pregnancy-related morbidity, related to persistent positivity of antiphospholipid antibodies (APL). One of the characteristics of pregnancy-related morbidity in patients with antiphospholipid syndrome is recurrent spontaneous abortion (RSA). This study aimed to examine the mechanism through which metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) regulates methyl-CpG-binding domain protein 4 (MBD4) expression in APL-positive RSA. METHODS Clinical samples were subjected to microarray analysis to filter differentially expressed genes. RSA mice with APL positivity were generated, followed by adenoviral vector injection to artificially upregulate MALAT1. The effects of MALAT1 on the biological behavior of trophoblast cells were assessed. The downstream mechanism of MALAT1 was analyzed using subcellular fractionation and bioinformatics prediction, and the relationship between MALAT1 and CREB binding protein (CREBBP) or MBD4 was investigated in trophoblast cells. RESULTS MALAT1 was downregulated in APL-positive RSA patients. MALAT1 was predominantly localized in the nucleus and recruited CREBBP to mediate the MBD4 transcription. In the APL-positive RSA mice overexpressing MALAT1, the expression of soluble Fms-related tyrosine kinase 1 and anticardiolipin antibody and the embryonic resorption rate were decreased, indicating that MALAT1 reduced the occurrence of RSA in mice. Moreover, MALAT1 enhanced proliferation, migration, and invasion of trophoblast cells through recruiting CREBBP to promote MBD4 expression. Silencing of CREBBP or MBD4 increased embryonic resorption rate in RSA mice overexpressing MALAT1. DISCUSSION MALAT1 suppresses APL-positive RSA by promoting MBD4 transcription through recruitment of CREBBP to the MBD4 promoter region.
Collapse
Affiliation(s)
- Yongmei Han
- College of Integrated Traditional Chinese and Western Medicine, Henan University of Traditional Chinese Medicine, Zhengzhou, 450046, Henan, PR China.
| | - Ying Wang
- Reproductive Center, Nanyang First People's Hospital, Nanyang, 473000, Henan, PR China
| | - Chenyu Zhang
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, Henan, PR China
| | - Yanru Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, Henan, PR China
| | - Jing Guo
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, Henan, PR China
| | - Chao Tian
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, Henan, PR China
| |
Collapse
|
8
|
Fu K, Chen X, Guo W, Zhou Z, Zhang Y, Ji T, Yang P, Tian X, Wang W, Zou Y. Effects of N Acetylcysteine on the Expression of Genes Associated with Reproductive Performance in the Goat Uterus during Early Gestation. Animals (Basel) 2022; 12:2431. [PMID: 36139290 PMCID: PMC9495183 DOI: 10.3390/ani12182431] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/10/2022] [Accepted: 09/11/2022] [Indexed: 11/18/2022] Open
Abstract
N acetylcysteine (NAC) affects antioxidation and reactive oxygen species scavenging in the body and thereby promotes embryonic development and implantation and inhibits inflammation. The mechanism through which NAC regulates reproductive performance in the uteri of goats during early gestation remains unclear. In this study, the treatment group was fed 0.07% NAC for the first 35 days of gestation, whereas the control group received no NAC supplementation. The regulatory genes and key pathways associated with goat reproductive performance under NAC supplementation were identified by RNA-seq. RT-qPCR was used to verify the sequencing results and subsequently construct tissue expression profiles of the relevant genes. RNA-seq identified 19,796 genes coexpressed in the control and treatment groups and 1318 differentially expressed genes (DEGs), including 787 and 531 DEGs enriched in the treatment and control groups, respectively. A GO analysis revealed that the identified genes mapped to pathways such as cell activation, cytokine production, cell mitotic processes, and angiogenesis, and a KEGG enrichment analysis showed that the DEGs were enriched in pathways associated with reproductive regulation, immune regulation, resistance to oxidative stress, and cell adhesion. The RT-qPCR analysis showed that BDNF and CSF-1 were most highly expressed in the uterus, that WIF1 and ESR2 showed low expression in the uterus, and that CTSS, PTX3, and TGFβ-3 were most highly expressed in the oviduct, which indicated that these genes may be directly or indirectly involved in the modulation of reproduction in early-gestation goats. These findings provide fundamental data for the NAC-mediated modulation of the reproductive performance of goats during early gestation.
Collapse
Affiliation(s)
- Kaibin Fu
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Xiang Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Wei Guo
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Zhinan Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Yan Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Taotao Ji
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Peifang Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Xingzhou Tian
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Weiwei Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Yue Zou
- Key Laboratory of Animal Genetics, Breeding and Reproduction in The Plateau Mountainous Region, Ministry of Education, Guizhou University, Guiyang 550025, China
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Guiyang 550025, China
- College of Animal Science, Guizhou University, Guiyang 550025, China
| |
Collapse
|
9
|
Zhang X, Yan J, Dai Z, Long X, Jin J, Yang Q, Lin C, Yang Y, Chen Y, Zhu J. Long non-coding RNA LINC01347 suppresses trophoblast cell migration, invasion and EMT by regulating miR-101-3p/PTEN/AKT axis. Reprod Biol 2022; 22:100670. [PMID: 35810709 DOI: 10.1016/j.repbio.2022.100670] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/11/2022] [Accepted: 06/27/2022] [Indexed: 10/17/2022]
Abstract
Recurrent miscarriage (RM) is one of the common complications of pregnancy, which is closely related to gene mutation. The profiling of non-coding RNAs showed that the expression level of long non-coding RNA LINC01347 (LINC01347) in the serum of patients with recurrent abortion was significantly increased, which could serve as a potential marker for early diagnosis. However, the biological functions of LINC01347 in the miscarriage remain to be elucidated. In this study, LINC01347 expression levels in HTR-8/SVneo cells and placenta samples were measured by RT-qPCR. The migration ability of HTR-8/SVneo cells was detected by wound-healing assay. Western blotting (WB) assay was conducted to measure E-cadherin, Vimentin, N-cadherin, PTEN, phospho-AKT(S473), phospho-AKT(T308) and AKT levels. Dual luciferase reporter assay and RNA pull-down analysis were performed to validate the molecular interactions. The results showed an upregulation of LINC01347 in the placenta samples of RM patients and HTR-8/SVneo cells. LINC01347 overexpression impaired the invasion and migration of trophoblast cells, while LINC01347 silencing promoted cell migration and invasion. LINC01347 level was also negatively correlated with the changes of epithelial-mesenchymal transition (EMT) markers in trophoblasts. We further demonstrated that miR-101-3p/PTEN/AKT axis played an important role in mediating the biological roles of LINC01347 in the invasion and migration of trophoblasts. In conclusion, our results revealed that LINC01347 suppresses the migratory ability and regulates the EMT processes in trophoblasts by regulating miR-101-3p/PTEN/AKT axis, suggesting that targeting LINC01347 may serve as a strategy to ameliorate RM.
Collapse
Affiliation(s)
- Xiahui Zhang
- Department of Obstetrics and Gynecology, The First People's Hospital of Wenling, 317500, China
| | - Jinyu Yan
- Department of Obstetrics and Gynecology, The First People's Hospital of Wenling, 317500, China
| | - Zhenzhen Dai
- Department of Obstetrics and Gynecology, The First People's Hospital of Wenling, 317500, China
| | - Xiaoxi Long
- Department of Obstetrics and Gynecology, The First People's Hospital of Wenling, 317500, China
| | - Jiaxi Jin
- Department of Obstetrics and Gynecology, The First People's Hospital of Wenling, 317500, China
| | - Qian Yang
- Department of Obstetrics and Gynecology, The First People's Hospital of Wenling, 317500, China
| | - Chenxiao Lin
- Department of Obstetrics and Gynecology, The First People's Hospital of Wenling, 317500, China
| | - Youlin Yang
- Department of Obstetrics and Gynecology, The First People's Hospital of Wenling, 317500, China
| | - Yi Chen
- Department of Obstetrics and Gynecology, The First People's Hospital of Wenling, 317500, China.
| | - Jun Zhu
- Department of Obstetrics and Gynecology, The First People's Hospital of Wenling, 317500, China.
| |
Collapse
|