1
|
Bhardwaj C, Rohilla M, Chopra S, Kaur A, Panigrahi I, Srivastava P. EV-microRNA signatures in pregnant women with idiopathic recurrent pregnancy loss: deciphering microRNAome pathway networks at feto-maternal interface. Front Immunol 2025; 16:1578738. [PMID: 40421018 PMCID: PMC12105548 DOI: 10.3389/fimmu.2025.1578738] [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: 02/18/2025] [Accepted: 04/21/2025] [Indexed: 05/28/2025] Open
Abstract
Background Despite extensive research in the past decade, the exact pathogenesis of recurrent pregnancy loss (RPL) remains unknown. At the time of pregnancy, human placenta releases microRNAs (miRNAs) enclosed in extracellular vesicles (EVs), which enter into maternal circulation and play an important role at feto-maternal interface to sustain a successful pregnancy. Aberrant expression of these miRNAs often results in adverse pregnancy complications. Therefore, studying the expression of these EV-miRNAs in maternal circulation could provide insights into the pathogenesis of RPL. Methods The present study included idiopathic currently pregnant (<22 weeks of gestation) RPL women (n=10) and gestational-age-matched healthy pregnant women as control (n=5). EVs were isolated from plasma samples and characterized for their morphology and cell-surface marker. Total RNA was isolated and subjected to miRNA sequencing on Illumina NovaSeq 6000 platform. Differentially expressed (DE) miRNAs were identified using DESeq package. Target prediction and pathway analysis were done using TargetScan, miRDB, miRTarBase, and DIANA-miRPath v3.0 online tool. Protein-protein interaction was done using STRING, and hub genes were identified using Cytoscape software. Results miRNA sequencing revealed 66 (44 known and 22 novel) significantly DE miRNAs between RPL and healthy pregnant women. Among these, 37 were downregulated and 29 were upregulated, log2|FC| ≥ 1. Network-based analysis showed highest degree for nine miRNAs (hsa-miR-155-5p, hsa-miR-26a-5p, hsa-miR-204-5p, hsa-miR-140-5p, hsa-miR-139-5p, hsa-let-7e-5p, hsa-miR-149-5p, hsa-miR-374a-5p, and hsa-miR-190a-5p). Gene Ontology (GO) and KEGG pathway analysis of target genes showed significant involvement of Hippo, FoxO, TGF-β, and p53 signaling pathways, which play a crucial role in RPL. Top 10 identified hub genes (NFKB1, IL6, JUN, FOS, CXCL8, PTGS2, TGFB1, MMP9, STAT1, and CD4) were significantly enriched in immunological pathways-Th1/Th2/Th17 differentiation, NF-κB pathway, TNF-α signaling, IL-17 signaling pathway, and vascular endothelial growth factor (VEGF) pathway. Conclusion These results suggest that circulating EV-miRNAs in maternal blood could provide clinical insights into the pathogenesis of RPL and dysregulated immunological and molecular pathways at feto-maternal interface.
Collapse
Affiliation(s)
- Chitra Bhardwaj
- Genetic Metabolic Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Minakshi Rohilla
- Department of Obstetrics & Gynaecology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Seema Chopra
- Department of Obstetrics & Gynaecology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Anupriya Kaur
- Genetic Metabolic Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Inusha Panigrahi
- Genetic Metabolic Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Priyanka Srivastava
- Genetic Metabolic Unit, Department of Pediatrics, Advanced Pediatrics Centre, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| |
Collapse
|
2
|
Abdulmonem WA, Ahsan M, Mallick AK, Mohamed AH, Waggiallah HA, Shafie A, Alzahrani HS, Ashour AA, Rab SO, Mirdad MT, Ali HTO. The Role of Exosomal miRNAs in Female Infertility: Therapeutic Potential and Mechanisms of Action. Stem Cell Rev Rep 2025:10.1007/s12015-025-10869-w. [PMID: 40126819 DOI: 10.1007/s12015-025-10869-w] [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] [Accepted: 03/17/2025] [Indexed: 03/26/2025]
Abstract
Reproductive disorders, including preeclampsia (PE), endometriosis, premature ovarian failure (POF), and polycystic ovary syndrome (PCOS), present substantial challenges to women's reproductive health. Exosomes (EXOs) are cell-derived vesicles containing molecules that influence target cells' gene expression and cellular behavior. Among their cargo, microRNAs (miRNAs)-short, non-coding RNAs typically 19-25 nucleotides in length-play a crucial role in post-transcriptional gene regulation and have been extensively studied for their therapeutic potential. miRNAs are considered therapeutic targets because they regulate key cellular pathways such as proliferation, apoptosis, angiogenesis, and tissue repair. This review examines the role of exosomal miRNAs from sources such as mesenchymal stem cells (MSCs), plasma, and amniotic fluid in female reproductive disorders, including PE, POF, PCOS, and endometriosis. We discuss their biological origins, mechanisms of miRNA sorting and packaging, and their therapeutic applications in modulating disease progression. By categorizing miRNAs according to their beneficial or detrimental effects in specific conditions, we aim to simplify the understanding of their roles in female infertility.
Collapse
Affiliation(s)
- Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Marya Ahsan
- Department of Pharmacology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, 13317, Saudi Arabia
| | - Ayaz Khurram Mallick
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Asma'a H Mohamed
- Department of Optometry Techniques, Technical College Al-Mussaib, Al-Furat Al-Awsat Technical University, Najaf, Iraq.
| | - Hisham Ali Waggiallah
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, 11942, Saudi Arabia
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O.Box 11099, Taif, 21944, Saudi Arabia
| | - Hassan Swed Alzahrani
- Counseling Healthy Marriage, Jeddah Regional Laboratory, Jeddah First Cluster , Jeddah, Saudi Arabia
| | - Amal Adnan Ashour
- Department of Oral & Maxillofacial Surgery and Diagnostic Sciences, Faculty of Dentistry, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed Tarek Mirdad
- Medical Intern MBBS, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Hatim T O Ali
- Obstetrics and Gynecology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
3
|
Shan Z, Zhou L, Ma Y, Huang Y. Circ_0090100 induces AHNAK expression to inhibit trophoblast cell proliferation and invasion and accelerate cell apoptosis by segregating miR-139-5p in preeclampsia. Hum Cell 2025; 38:67. [PMID: 40063163 DOI: 10.1007/s13577-025-01185-z] [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: 07/26/2024] [Accepted: 02/06/2025] [Indexed: 05/09/2025]
Abstract
The etiology of preeclampsia (PE) is complex and is known to involve the expression of circular RNAs (circRNAs). Among these, the function of circ_0090100 in PE is yet to be fully understood. This study was conducted to examine the expression profile of circ_0090100 in placental tissues from PE patients and to assess its influence on trophoblast cell functions. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to measure the RNA expression levels of circ_0090100, microRNA-139-5p (miR-139-5p), and AHNAK nucleoprotein (AHNAK). Cell viability, proliferation, apoptosis, and invasion were assessed using the CCK-8 assay, EdU incorporation, flow cytometry, and transwell migration assays, respectively. Western blot analysis was performed to detect the protein expression of N-cadherin, Vimentin, and AHNAK. Dualluciferase reporter assay and RNA immunoprecipitation (RIP) assay were conducted to elucidate the relationships between circ_0090100, miR-139-5p, and AHNAK. We found that the expression of circ_0090100 and AHNAK was upregulated, while miR-139-5p. expression was downregulated in PE placental tissues compared to controls. Transfection of a plasmid overexpressing circ_0090100 into JEG-3 and HTR-8/SVneo trophoblast cell lines resulted in reduced cell proliferation and invasion, but increased apoptosis. Mechanistically, circ_0090100 functioned as a miR-139-5p sponge, and miR-139-5p targeted AHNAK. Furthermore, upregulating miR-139-5p reversed the effects of circ_0090100 in JEG-3 and HTR-8/SVneo cells. AHNAK was found to be involved in the regulation of miR-139-5p effects on trophoblast cells. Additionally, circ_0090100 induced the production of AHNAK through miR-139-5p. Therefore, circ_0090100 activated AHNAK to suppress trophoblast cell proliferation and invasion, and promote cell apoptosis, via miR-139-5p.
Collapse
Affiliation(s)
- Zhenli Shan
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Jing'an District, No. 580 Changle Road, Shanghai, 200092, China
| | - Linfang Zhou
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Jing'an District, No. 580 Changle Road, Shanghai, 200092, China
| | - Yan Ma
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Jing'an District, No. 580 Changle Road, Shanghai, 200092, China
| | - Yiying Huang
- Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Jing'an District, No. 580 Changle Road, Shanghai, 200092, China.
| |
Collapse
|
4
|
Grossini E, Surico D, Venkatesan S, Ola Pour MM, Aquino CI, Remorgida V. Extracellular Vesicles and Pregnancy-Related Hypertensive Disorders: A Descriptive Review on the Possible Implications "From Bench to Bedside". BIOLOGY 2025; 14:240. [PMID: 40136497 PMCID: PMC11939443 DOI: 10.3390/biology14030240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/08/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025]
Abstract
Pregnancy involves extracellular vesicles (EVs) through mechanisms that are poorly understood to date. Furthermore, it is not surprising that EVs may also be involved in the pathophysiology of pre-eclampsia (PE) and gestational hypertension, two clinical conditions with high morbidity and mortality, given their capacity to mediate intracellular communications and regulate inflammation and angiogenesis. We searched major online scientific search engines (PubMed, Google Scholar, Scopus, WES, Embase, etc.) using the terms "Preeclampsia", "Pregnancy", "Hypertension", "Pregnancy-related hypertension", "Extracellular vesicles", "Biomarkers", "Gestation" AND "Obstetrics". Finding potential early biomarkers of risk or illness progression would be essential for the optimum care of expectant mothers with the aforementioned conditions. Nevertheless, none of the various screening assays that have been discovered recently have shown high predictive values. The analysis of EVs in the peripheral blood starting from the first trimester of pregnancy may hold great promise for the possible correlation with gestational hypertension problems and represent a marker of the early stages of the disease. EVs use may be a novel therapeutic approach for the management of various illnesses, as well. In order to define EVs' function in the physiopathology of pregnancy-associated hypertension and PE, as well as their potential as early biomarkers and therapeutic tools, we have compiled the most recent data in this review.
Collapse
Affiliation(s)
- Elena Grossini
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (S.V.); (M.M.O.P.)
| | - Daniela Surico
- Gynecology and Obstetrics Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.S.); (C.I.A.); (V.R.)
| | - Sakthipriyan Venkatesan
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (S.V.); (M.M.O.P.)
| | - Mohammad Mostafa Ola Pour
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (S.V.); (M.M.O.P.)
| | - Carmen Imma Aquino
- Gynecology and Obstetrics Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.S.); (C.I.A.); (V.R.)
| | - Valentino Remorgida
- Gynecology and Obstetrics Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.S.); (C.I.A.); (V.R.)
| |
Collapse
|
5
|
Selvakumar SC, Preethi KA, Ross K, Sekar D. The emerging role of microRNA-based therapeutics in the treatment of preeclampsia. Placenta 2024; 158:38-47. [PMID: 39361986 DOI: 10.1016/j.placenta.2024.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/09/2024] [Accepted: 09/28/2024] [Indexed: 10/05/2024]
Abstract
Preeclampsia (PE) is a pregnancy complication that is often diagnosed due to elevated blood pressure and proteinuria. Though current research focuses on the identification of novel biomarkers and therapeutic targets, still, there is a lack of clinical validation for the use of biomarkers and therapeutic targets for early diagnosis and treatment of PE. Several molecules are being studied for their potential role in PE. Among them, microRNAs are studied vastly for their role in the diagnosis, prognosis, and treatment of PE. But only a few studies are focused on the therapeutic efficacy of miRNAs in PE. Thus, the relevant articles were identified and discussed in this review. These studies provide evidence that miRNAs are indeed important molecules in PE that have the role of both therapeutic targets and therapeutic molecules. However, the studies are limited to in vivo an in vitro models, hence further studies are required to validate the complete potential of miRNA therapeutics. Long non-coding RNA (lncRNA) sponges, miRNA mimics, miRNA inhibitors, exosome-associated miRNAs, and several other molecules have been studied as miRNA-based therapeutics in PE. Thus, miRNAs are postulated to be potential therapeutic targets and miRNA-based therapeutics might pave the way for novel therapeutic approaches for PE.
Collapse
Affiliation(s)
- Sushmaa Chandralekha Selvakumar
- RNA Biology Lab, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences (SIMATS), Saveetha University, Chennai 600077, India
| | - K Auxzilia Preethi
- RNA Biology Lab, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences (SIMATS), Saveetha University, Chennai 600077, India
| | - Kehinde Ross
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, United Kingdom
| | - Durairaj Sekar
- RNA Biology Lab, Saveetha Dental College & Hospitals, Saveetha Institute of Medical & Technical Sciences (SIMATS), Saveetha University, Chennai 600077, India.
| |
Collapse
|
6
|
Zhang K, Zhang H, Wang B, Gao S, Sun C, Jia C, Cui J. NR2F1 overexpression alleviates trophoblast cell dysfunction by inhibiting GDF15/MAPK axis in preeclampsia. Hum Cell 2024; 37:1405-1420. [PMID: 39007956 DOI: 10.1007/s13577-024-01095-6] [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: 02/06/2024] [Accepted: 06/15/2024] [Indexed: 07/16/2024]
Abstract
Abnormal functions of trophoblast cells are associated with the pathogenesis of preeclampsia (PE). Nuclear receptor subfamily 2 group F member 1 (NR2F1) acts as a transcriptionally regulator in many diseases, but its role in PE remains unknown. Hypoxia/reoxygenation (H/R)-stimulated HTR-8/SVneo cells were used to mimic PE injury in vitro. NR2F1 overexpression alleviated trophoblast apoptosis, as evidenced by the decreased number of TUNEL-positive cells and the downregulation of caspase 3 and caspase 9 expression in cells. NR2F1 overexpression increased the invasion and migration ability of HTR-8/SVneo cells, accompanied by increased protein levels of matrix metalloproteinase (MMP)-2 and MMP-9. mRNA-seq was applied to explore the underlying mechanism of NR2F1, identifying growth differentiation factor 15 (GDF15) as the possible downstream effector. Dual-luciferase reporter, ChIP-qPCR, and DNA pull-down assays confirmed that NR2F1 bound to the promoter of GDF15 and transcriptionally inhibited its expression. GDF15 overexpression increased apoptosis and decreased the ability of invasion and migration in HTR-8/SVneo cells expressing NR2F1. MAPK pathway was involved in the regulation of PE. Administration of p38 inhibitor, ERK inhibitor, and JNK inhibitor reversed the effect of simultaneous overexpression NR2F1 and GDF15 on trophoblast apoptosis, invasion, and migration. Our findings demonstrated that NR2F1 overexpression inhibited trophoblast apoptosis and promoted trophoblast invasion and migration. NR2F1 might negatively regulate GDF15 expression by binding to its promoter region, which further inhibited MAPK signaling pathway in PE. Our study highlights that NR2F1 might sever as a potential target in PE.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Hailing Zhang
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Bing Wang
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Shanshan Gao
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Caiping Sun
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Cong Jia
- Department of Obstetrics, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Jinquan Cui
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Zhengzhou University, No. 2, Jingba Road, Zhengzhou, Henan, People's Republic of China.
| |
Collapse
|
7
|
Shi H, Yang Z, Cui J, Tao H, Ma R, Zhao Y. Mesenchymal stem cell-derived exosomes: a promising alternative in the therapy of preeclampsia. Stem Cell Res Ther 2024; 15:30. [PMID: 38317195 PMCID: PMC10845755 DOI: 10.1186/s13287-024-03652-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 01/29/2024] [Indexed: 02/07/2024] Open
Abstract
Preeclampsia (PE) is a common morbid complication during pregnancy, affecting 2%-8% of pregnancies globally and posing serous risks to the health of both mother and fetus. Currently, the only effective treatment for PE is timely termination of pregnancy, which comes with increased perinatal risks. However, there is no effective way to delay pathological progress and improve maternal and fetal outcomes. In light of this, it is of great significance to seek effective therapeutic strategies for PE. Exosomes which are nanoparticles carrying bioactive substances such as proteins, lipids, and nucleic acids, have emerged as a novel vehicle for intercellular communication. Mesenchymal stem cell-derived exosomes (MSC-Exos) participate in various important physiological processes, including immune regulation, cell proliferation and migration, and angiogenesis, and have shown promising potential in tissue repair and disease treatment. Recently, MSC-Exos therapy has gained popularity in the treatment of ischaemic diseases, immune dysfunction, inflammatory diseases, and other fields due to their minimal immunogenicity, characteristics similar to donor cells, ease of storage, and low risk of tumor formation. This review elaborates on the potential therapeutic mechanism of MSC-Exos in treating preeclampsia, considering the main pathogenic factors of the condition, including placental vascular dysplasia, immunological disorders, and oxidative stress, based on the biological function of MSC-Exos. Additionally, we discuss in depth the advantages and challenges of MSC-Exos as a novel acellular therapeutic agent in preeclampsia treatment.
Collapse
Affiliation(s)
- Haoran Shi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Zejun Yang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Jianjian Cui
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Hui Tao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Ruilin Ma
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Yin Zhao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China.
- Shenzhen Huazhong University of Science and Technology Research Institute, Shen Zhen, 518000, China.
| |
Collapse
|
8
|
Sun J, Zhang W. Huc-MSC-derived exosomal miR-144 alleviates inflammation in LPS-induced preeclampsia-like pregnant rats via the FosB/Flt-1 pathway. Heliyon 2024; 10:e24575. [PMID: 38304844 PMCID: PMC10830578 DOI: 10.1016/j.heliyon.2024.e24575] [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: 04/27/2023] [Revised: 12/21/2023] [Accepted: 01/10/2024] [Indexed: 02/03/2024] Open
Abstract
Background Preeclampsia (PE) is a common and severe hypertensive disorder in pregnancy. Mesenchymal stem cell-derived exosomes (Exos-MSC) have been reported to mitigate the progression of inflammatory diseases. The study aimed to explore the effects of human umbilical cord-derived Exos-MSC (huc-Exos-MSC) on PE-like models. Methods Lipopolysaccharide (LPS) was used to construct in vitro and in vivo PE-like models. Exosomes were treated with LPS-induced PE-like cells and rats. Results PE-like inflammatory models of pregnant rats and cells were successfully constructed in vivo and in vitro. miR-144 was screened by bioinformatics analysis. Exosomes were successfully extracted. Silencing FosB, overexpressing miR-144 or treating with exosomes extracted from huc-MSC overexpressing miR-144 in (Exos-MSCmiR-144) reversed the LPS-induced decline in HTR-8/SVneo cell viability and migration. In addition, the above groups decreased LPS-induced increases in interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), phosphorylated nuclear factor-kappaB (p-NF-κB)/NF-κB, soluble FMS-like tyrosine kinase 1 (sFlt-1), and Flt-1 levels. Simultaneously, transfection of miR-144 mimics and overexpressing FosB reversed those changes in the miR-144 mimics group. miR-144 might alleviate LPS-induced HTR-8/SVneo cell inflammation by targeting FosB. Injection of Exos-MSCmiR-144 in PE-like pregnant rats reversed LPS-induced increases in FosB expression, systolic and diastolic blood pressure (SBP and DBP), as well as mean arterial pressure (MAP), heart rate, urine albumin/creatine ratio, inflammatory factors, p-NF-κB/NF-κB, and sFlt-1 levels. Furthermore, compared with the model group, the proportion of live births was significantly higher in the model + Exos-MSCmiR-144 group, while the apoptosis rate of fetal rat brain tissue was significantly lower. Conclusions We found that huc-Exos-MSC-derived miR-144 alleviated gestational hypertension and inflammation in PE-like pregnant rats by regulating the FosB/Flt-1 pathway. In addition, huc-Exos-MSC-derived miR-144 could partially reverse the LPS-induced adverse pregnancy outcome and brain injury in fetal rats, laying the foundation for developing new treatments for PE.
Collapse
Affiliation(s)
- Jingchi Sun
- Department of Medical Administration, The Third People's Hospital of Chengdu, Chengdu, 610014, China
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Weishe Zhang
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Engineering Research Center of Early Life Development and Disease Prevention, Changsha, 410008, China
| |
Collapse
|
9
|
Margiana R. Mesenchymal stem cell-derived exosomes in preeclampsia: A next-generation therapeutic tool. Cell Biochem Funct 2024; 42:e3908. [PMID: 38269498 DOI: 10.1002/cbf.3908] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/29/2023] [Accepted: 12/10/2023] [Indexed: 01/26/2024]
Abstract
Preeclampsia (PE) is a major gestational disorder that causes both long- and short-term damage to both the mother and the fetus. Endometrium decidualization and the formation of the placenta are orchestrated by mesenchymal stem cells (MSCs). MSCs obtained from patients with PE exhibit an elevated rate of aging and apoptosis, which impairs the interplay between MSCs and endothelium, trophoblast, and immune cells in the placenta, accelerating the onset of PE. Preclinical and clinical evidence imply that the MSC-based therapy approach for PE is prospective. Importantly, as a novel cell-free approach, MSC-derived exosomes can improve symptoms and maternal-fetal survival in PE models by raising cell metabolism, encouraging angiogenesis balance, and regulating immune responses. Even following allogeneic administration, the likelihood of immune rejection is very limited as a result of the small quantity of exosome membrane-bound proteins. Furthermore, because exosomes do not expand, developing tumors is not probable. As a result, MSC-derived exosomes show superiority over MSCs in terms of safety. For the first time, we outline the properties of MSC-exosomes and highlight their functions and potential as a new paradigm for PE therapy in this review.
Collapse
Affiliation(s)
- Ria Margiana
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| |
Collapse
|
10
|
Rizano A, Margiana R, Supardi S, Narulita P. Exploring the future potential of mesenchymal stem/stromal cells and their derivatives to support assisted reproductive technology for female infertility applications. Hum Cell 2023; 36:1604-1619. [PMID: 37407748 DOI: 10.1007/s13577-023-00941-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/16/2023] [Indexed: 07/07/2023]
Abstract
Women's infertility impacts the quality of life of both patients and couples and has multifaceted dimensions that increase the number of challenges associated with female infertility and how to face them. Female reproductive disorders, such as premature ovarian failure (POF), endometriosis, Asherman syndrome (AS), polycystic ovary syndrome (PCOS), and preeclampsia, can stimulate infertility. In the last decade, translational medicine has advanced, and scientists are focusing on infertility therapy with innovative attitudes. Recent investigations have suggested that stem cell treatments could be safe and effective. Stem cell therapy has established a novel method for treating women's infertility as part of a regeneration approach. The chief properties and potential of mesenchymal stem/stromal cells (MSCs) in the future of women's infertility should be considered by researchers. Due to their high abundance, great ability to self-renew, and high differentiation capacity, as well as less ethical concerns, MSC-based therapy has been found to be an effective alternative strategy to the previous methods for treating female infertility, such as intrauterine insemination, in vitro fertilization, medicines, and surgical procedures. These types of stem cells exert their beneficial role by releasing active mediators, promoting cell homing, and contributing to immune modulation. Here we first provide an overview of MSCs and their crucial roles in both biological and immunological processes. The next large chapter covers current preclinical and clinical studies on the application of MSCs to treat various female reproductive disorders. Finally, we deliberate on the extant challenges that hinder the application of MSCs in female infertility and suggest plausible measures to alleviate these impediments.
Collapse
Affiliation(s)
- Andrew Rizano
- Department of Medical Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Ria Margiana
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia.
- Department of Anatomy, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
- Master's Programme Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia.
- Indonesia General Academic Hospital, Depok, Indonesia.
- Ciptomangunkusumo General Academic Hospital, Jakarta, Indonesia.
| | - Supardi Supardi
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Pety Narulita
- Andrology Program, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
- Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| |
Collapse
|
11
|
Chang X, He Q, Wei M, Jia L, Wei Y, Bian Y, Duan T, Wang K. Human umbilical cord mesenchymal stem cell derived exosomes (HUCMSC-exos) recovery soluble fms-like tyrosine kinase-1 (sFlt-1)-induced endothelial dysfunction in preeclampsia. Eur J Med Res 2023; 28:277. [PMID: 37559150 PMCID: PMC10413730 DOI: 10.1186/s40001-023-01182-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 06/21/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Preeclampsia is a unique multisystem disorder that affects 5-8% of pregnancies. A high level of soluble fms-like tyrosine kinase-1 (sFlt-1) is a hallmark of preeclampsia that causes endothelial dysfunction. Exosomes derived from mesenchymal stem cells (MSCs) have been indicated to improve endothelial performances by transporting signals to target cells. We hypothesized that exosomes derived from MSCs have potential effects against preeclampsia. METHODS We collected human umbilical cord MSC-derived exosomes (HUCMSC-exos) by ultracentrifugation. The size and morphology of the exosomes were examined using a transmission electron microscope and nanoparticle tracking analysis. Pregnant mice were injected with murine sFlt-1 adenovirus to build the preeclampsia-like mouse model and then treated with HUCMSC-exos. Human umbilical vein endothelial cells (HUVECs) were infected with lentiviruses expressing tet-on-sFlt-1 to obtain cells overexpressing sFlt-1. Cell proliferation and migration assays were used to measure the endothelial functions. The exosomes enriched proteins underlying mechanisms were explored by proteomic analysis. RESULTS In the current study, we successfully collected the cup-shaped HUCMSC-exos with diameters of 30-150 nm. In the sFlt-1-induced preeclampsia mouse model, HUCMSC-exos exhibited beneficial effects on adverse birth events by decreasing blood pressure and improving fetal birth weight. In addition, preeclamptic dams that were injected with HUCMSC-exos had rebuilt dense placental vascular networks. Furthermore, we observed that HUCMSC-exos partially rescued sFlt-1-induced HUVECs dysfunction in vitro. Proteomics analysis of HUCMSC-exos displayed functional enrichment in biological processes related to vesicle-mediated transport, cell communication, cell migration, and angiogenesis. CONCLUSION We propose that exosomes derived from HUCMSCs contain abundant Versican and play beneficial roles in the birth outcomes of sFlt-1-induced preeclamptic mice by promoting angiogenesis.
Collapse
Affiliation(s)
- Xinwen Chang
- Center of Reproductive Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Qizhi He
- Department of Pathology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Mengtian Wei
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Linyan Jia
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Yingying Wei
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Yiding Bian
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Tao Duan
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China.
| | - Kai Wang
- Clinical and Translational Research Center, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China.
| |
Collapse
|
12
|
Wawrzkiewicz-Jałowiecka A, Lalik A, Lukasiak A, Richter-Laskowska M, Trybek P, Ejfler M, Opałka M, Wardejn S, Delfino DV. Potassium Channels, Glucose Metabolism and Glycosylation in Cancer Cells. Int J Mol Sci 2023; 24:ijms24097942. [PMID: 37175655 PMCID: PMC10178682 DOI: 10.3390/ijms24097942] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Potassium channels emerge as one of the crucial groups of proteins that shape the biology of cancer cells. Their involvement in processes like cell growth, migration, or electric signaling, seems obvious. However, the relationship between the function of K+ channels, glucose metabolism, and cancer glycome appears much more intriguing. Among the typical hallmarks of cancer, one can mention the switch to aerobic glycolysis as the most favorable mechanism for glucose metabolism and glycome alterations. This review outlines the interconnections between the expression and activity of potassium channels, carbohydrate metabolism, and altered glycosylation in cancer cells, which have not been broadly discussed in the literature hitherto. Moreover, we propose the potential mediators for the described relations (e.g., enzymes, microRNAs) and the novel promising directions (e.g., glycans-orinented drugs) for further research.
Collapse
Affiliation(s)
- Agata Wawrzkiewicz-Jałowiecka
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Anna Lalik
- Department of Systems Biology and Engineering, Silesian University of Technology, 44-100 Gliwice, Poland
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Agnieszka Lukasiak
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| | - Monika Richter-Laskowska
- The Centre for Biomedical Engineering, Łukasiewicz Research Network-Krakow Institute of Technology, 30-418 Krakow, Poland
| | - Paulina Trybek
- Institute of Physics, University of Silesia in Katowice, 41-500 Chorzów, Poland
| | - Maciej Ejfler
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Maciej Opałka
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Sonia Wardejn
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Domenico V Delfino
- Section of Pharmacology, Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy
| |
Collapse
|
13
|
Wan W, Miao Y, Niu Y, Zhu K, Ma Y, Pan M, Ma B, Wei Q. Human umbilical cord mesenchymal stem cells conditioned medium exerts anti-tumor effects on KGN cells in a cell density-dependent manner through activation of the Hippo pathway. Stem Cell Res Ther 2023; 14:46. [PMID: 36941685 PMCID: PMC10029233 DOI: 10.1186/s13287-023-03273-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 03/08/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND The conditioned medium from human umbilical cord mesenchymal stem cells (UCMSCs-CM) provides a new cell-free therapy for tumors due to its unique secretome. However, there are many contradictory reports about the effect of UCMSCs-CM on tumor cells. The loss of contact inhibition is a common characteristic of tumor cells. A relationship between the effect of UCMSCs-CM on tumor cells and contact inhibition in tumor cells is rarely concerned. Whether the effect of UCMSCs-CM on tumor cells is affected by cell density? Here, we explored the effect of UCMSCs-CM on granulosa tumor cell line (KGN) cells at low or high density. METHODS Growth curve and CCK8 assay were used to assess cell proliferation and viability. Scratch wound and matrigel invasion assay were implicated to detect cell motility of KGN cells. UCMSCs-CM effects on cell cycle, apoptosis and pathway-related proteins were investigated by flow cytometry, TUNEL assay, western blot and immunofluorescence analysis respectively. RESULTS In growth curve analysis, before KGN cells proliferated into confluence, UCMSCs-CM had no effect on cell proliferation. However, once the cells proliferate to contact each other, UCMSCs-CM significantly inhibited proliferation. Meanwhile, when KGN cells were implanted at high density, UCMSCs-CM could induce cell cycle arrest at G1 phase, inhibit cell migration, invasion and promote apoptosis. While it had no similar effect on KGN cells implanted at low density. In mechanism, the UCMSCs-CM treatment activated the Hippo pathway when KGN cells were implanted at high density. Consistently, the MST1/2 inhibitor, XMU-MP-1, inhibited the activation of the Hippo pathway induced by UCMSCs-CM treatment and accordingly declined the anti-tumor effect of UCMSCs-CM on KGN cells. CONCLUSIONS The effect of UCMSCs-CM on tumor cells is affected by cell density. UCMSCs-CM exerted anti-tumor effect on KGN cells by activating Hippo pathway to restore contact inhibition. Our results suggest that UCMSCs-CM is a promising therapeutic candidate for GCT treatment.
Collapse
Affiliation(s)
- Wenjing Wan
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A and F University, Yangling, 712100, Shaanxi, China
- College of Veterinary Medicine, Northwest A and F University, Yangling, 712100, Shaanxi, China
| | - Yuyang Miao
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A and F University, Yangling, 712100, Shaanxi, China
- College of Veterinary Medicine, Northwest A and F University, Yangling, 712100, Shaanxi, China
| | - Yuwei Niu
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A and F University, Yangling, 712100, Shaanxi, China
- College of Veterinary Medicine, Northwest A and F University, Yangling, 712100, Shaanxi, China
| | - Kunyuan Zhu
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A and F University, Yangling, 712100, Shaanxi, China
- College of Veterinary Medicine, Northwest A and F University, Yangling, 712100, Shaanxi, China
| | - Yingwan Ma
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A and F University, Yangling, 712100, Shaanxi, China
- College of Veterinary Medicine, Northwest A and F University, Yangling, 712100, Shaanxi, China
| | - Menghao Pan
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A and F University, Yangling, 712100, Shaanxi, China
- College of Veterinary Medicine, Northwest A and F University, Yangling, 712100, Shaanxi, China
| | - Baohua Ma
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A and F University, Yangling, 712100, Shaanxi, China.
- College of Veterinary Medicine, Northwest A and F University, Yangling, 712100, Shaanxi, China.
| | - Qiang Wei
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A and F University, Yangling, 712100, Shaanxi, China.
- College of Veterinary Medicine, Northwest A and F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
14
|
Fatmous M, Rai A, Poh QH, Salamonsen LA, Greening DW. Endometrial small extracellular vesicles regulate human trophectodermal cell invasion by reprogramming the phosphoproteome landscape. Front Cell Dev Biol 2022; 10:1078096. [PMID: 36619864 PMCID: PMC9813391 DOI: 10.3389/fcell.2022.1078096] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
A series of cyclical events within the uterus are crucial for pregnancy establishment. These include endometrial regeneration following menses, under the influence of estrogen (proliferative phase), then endometrial differentiation driven by estrogen/progesterone (secretory phase), to provide a microenvironment enabling attachment of embryo (as a hatched blastocyst) to the endometrial epithelium. This is followed by invasion of trophectodermal cells (the outer layer of the blastocyst) into the endometrium tissue to facilitate intrauterine development. Small extracellular vesicles (sEVs) released by endometrial epithelial cells during the secretory phase have been shown to facilitate trophoblast invasion; however, the molecular mechanisms that underline this process remain poorly understood. Here, we show that density gradient purified sEVs (1.06-1.11 g/ml, Alix+ and TSG101+, ∼180 nm) from human endometrial epithelial cells (hormonally primed with estrogen and progesterone vs. estrogen alone) are readily internalized by a human trophectodermal stem cell line and promote their invasion into Matrigel matrix. Mass spectrometry-based proteome analysis revealed that sEVs reprogrammed trophectoderm cell proteome and their cell surface proteome (surfaceome) to support this invasive phenotype through upregulation of pro-invasive regulators associated with focal adhesions (NRP1, PTPRK, ROCK2, TEK), embryo implantation (FBLN1, NIBAN2, BSG), and kinase receptors (EPHB4/B2, ERBB2, STRAP). Kinase substrate prediction highlighted a central role of MAPK3 as an upstream kinase regulating target cell proteome reprogramming. Phosphoproteome analysis pinpointed upregulation of MAPK3 T204/T202 phosphosites in hTSCs following sEV delivery, and that their pharmacological inhibition significantly abrogated invasion. This study provides novel molecular insights into endometrial sEVs orchestrating trophoblast invasion, highlighting the microenvironmental regulation of hTSCs during embryo implantation.
Collapse
Affiliation(s)
- Monique Fatmous
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University (LTU), Melbourne, VIC, Australia
| | - Alin Rai
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Central Clinical School, Monash University, Melbourne, VIC, Australia,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia,Baker Department of Cardiovascular Research, Translation and Implementation, LTU, Melbourne, VIC, Australia
| | - Qi Hui Poh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Baker Department of Cardiovascular Research, Translation and Implementation, LTU, Melbourne, VIC, Australia,Department of Biochemistry and Chemistry, LTU, Melbourne, VIC, Australia
| | - Lois A. Salamonsen
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia,Department of Molecular and Translational Medicine, Monash University, Clayton, VIC, Australia
| | - David W. Greening
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Central Clinical School, Monash University, Melbourne, VIC, Australia,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia,Baker Department of Cardiovascular Research, Translation and Implementation, LTU, Melbourne, VIC, Australia,Department of Biochemistry and Chemistry, LTU, Melbourne, VIC, Australia,*Correspondence: David W. Greening,
| |
Collapse
|
15
|
Hayder H, Shan Y, Chen Y, O’Brien JA, Peng C. Role of microRNAs in trophoblast invasion and spiral artery remodeling: Implications for preeclampsia. Front Cell Dev Biol 2022; 10:995462. [PMID: 36263015 PMCID: PMC9575991 DOI: 10.3389/fcell.2022.995462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
It is now well-established that microRNAs (miRNAs) are important regulators of gene expression. The role of miRNAs in placental development and trophoblast function is constantly expanding. Trophoblast invasion and their ability to remodel uterine spiral arteries are essential for proper placental development and successful pregnancy outcome. Many miRNAs are reported to be dysregulated in pregnancy complications, especially preeclampsia and they exert various regulatory effects on trophoblasts. In this review, we provide a brief overview of miRNA biogenesis and their mechanism of action, as well as of trophoblasts differentiation, invasion and spiral artery remodeling. We then discuss the role of miRNAs in trophoblasts invasion and spiral artery remodeling, focusing on miRNAs that have been thoroughly investigated, especially using multiple model systems. We also discuss the potential role of miRNAs in the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Heyam Hayder
- Department of Biology, York University, Toronto, ON, Canada
| | - Yanan Shan
- Department of Biology, York University, Toronto, ON, Canada
| | - Yan Chen
- Department of Biology, York University, Toronto, ON, Canada
| | | | - Chun Peng
- Department of Biology, York University, Toronto, ON, Canada
- Centre for Research on Biomolecular Interactions, York University, Toronto, ON, Canada
- *Correspondence: Chun Peng,
| |
Collapse
|
16
|
Zhang L, Liu M. Circ_0077109 sponges miR-139-5p and upregulates HOXD10 in trophoblast cells as potential mechanism for preeclampsia progression. Am J Reprod Immunol 2022; 88:e13609. [PMID: 35964231 DOI: 10.1111/aji.13609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND One of the important reasons for the development of preeclampsia (PE) is the abnormal function of trophoblast cells. Many circular RNAs (circRNAs) have been confirmed to participate in the regulation of trophoblast cell function to mediate PE progression. However, whether circ_0077109 is involved in PE progression through regulating trophoblast cell function remains unclear. METHODS Quantitative real-time PCR was utilized for measuring the expression of circ_0077109, microRNA (miR)-139-5p and homeobox D10 (HOXD10). Trophoblast cell proliferation, apoptosis, invasion, and angiogenesis was assessed cell counting kit 8 assay, EdU assay, flow cytometry, transwell assay and tube formation assay. In addition, western blot analysis was used to determine protein expression. The interaction between miR-139-5p and circ_0077109 or HOXD10 was verified by dual-luciferase reporter assay and RIP assay. RESULTS Our results pointed out that circ_0077109 was a circRNA with upregulated expression in PE patients. Overexpression of circ_0077109 suppressed trophoblast cell proliferation, invasion, and angiogenesis, while increased apoptosis. MiR-139-5p was found to be sponged by circ_0077109, and its mimic reversed the suppressive effect of circ_0077109 on trophoblast cell function. HOXD10 was a target of miR-139-5p, and its overexpression inhibited trophoblast cell proliferation, invasion, and angiogenesis. MiR-139-5p inhibitor could repress trophoblast cell function, while this effect could be reversed by HOXD10 knockdown. CONCLUSION In summary, we confirmed that circ_0077109 inhibited trophoblast cell function through the regulation of miR-139-5p/HOXD10 axis, which might be a potential target for PE treatment. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Liping Zhang
- Department of Obstetrics, Huai'an Maternal and Child health Care Center, Huai'an, 223002, China
| | - Mengxu Liu
- Department of Obstetrics, Huai'an Maternal and Child health Care Center, Huai'an, 223002, China
| |
Collapse
|
17
|
Jin S, Wu C, Chen M, Sun D, Zhang H. The pathological and therapeutic roles of mesenchymal stem cells in preeclampsia. Front Med (Lausanne) 2022; 9:923334. [PMID: 35966876 PMCID: PMC9370554 DOI: 10.3389/fmed.2022.923334] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/13/2022] [Indexed: 11/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have made progress in the treatment of ischemic and inflammatory diseases. Preeclampsia (PE) is characterized by placenta ischemic and inflammatory injury. Our paper summarized the new role of MSCs in PE pathology and its potency in PE therapy and analyzed its current limitations. Intravenously administered MSCs dominantly distributed in perinatal tissues. There may be additional advantages to using MSCs-based therapies for reproductive disorders. It will provide new ideas for future research in this field.
Collapse
Affiliation(s)
- Sanshan Jin
- Hubei University of Chinese Medicine, Wuhan, China
- Department of Traditional Chinese Medicine, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Canrong Wu
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Ming Chen
- Department of Rehabilitation Physiotherapy, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Dongyan Sun
- Department of Gynecology, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
| | - Hua Zhang
- Hubei University of Chinese Medicine, Wuhan, China
- Department of Traditional Chinese Medicine, Maternal and Child Health Hospital of Hubei Province, Wuhan, China
- *Correspondence: Hua Zhang,
| |
Collapse
|
18
|
Zhang Y, Zhong Y, Zou L, Liu X. Significance of Placental Mesenchymal Stem Cell in Placenta Development and Implications for Preeclampsia. Front Pharmacol 2022; 13:896531. [PMID: 35721156 PMCID: PMC9198303 DOI: 10.3389/fphar.2022.896531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/16/2022] [Indexed: 12/29/2022] Open
Abstract
The well-developed placentation is fundamental for the reproductive pregnancy while the defective placental development is the pathogenetic basis of preeclampsia (PE), a dangerous complication of pregnancy comprising the leading causes of maternal and perinatal morbidity and mortality. Placenta-derived mesenchymal stem cells (PMSCs) are a group of multipotent stem cells that own a potent capacity of differentiating into constitutive cells of vessel walls. Additionally, with the paracrine secretion of various factors, PMSCs inextricably link and interact with other component cells in the placenta, collectively improving the placental vasculature, uterine spiral artery remolding, and uteroplacental interface immunoregulation. Recent studies have further indicated that preeclamptic PMSCs, closely implicated in the abnormal crosstalk between other ambient cells, disturb the homeostasis and development in the placenta. Nevertheless, PMSCs transplantation or PMSCs exosome therapies tend to improve the placental vascular network and trophoblastic functions in the PE model, suggesting PMSCs may be a novel and putative therapeutic strategy for PE. Herein, we provide an overview of the multifaceted contributions of PMSCs in early placental development. Thereinto, the intensive interactions between PMSCs and other component cells in the placenta were particularly highlighted and further extended to the implications in the pathogenesis and therapeutic strategies of PE.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanqi Zhong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxia Liu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Zhang Y, Wang J, Qu Y, Chen Y. 6-Shogaol Suppresses the Progression of Liver Cancer via the Inactivation of Wnt/[Formula: see text]-Catenin Signaling by Regulating TLR4. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 49:2033-2048. [PMID: 34961414 DOI: 10.1142/s0192415x21500968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Liver cancer is a gastrointestinal malignant tumor with high lethality. The prognosis of liver cancer remains poor. Compounds derived from natural products have been confirmed to alleviate the progression of various diseases, including cancers. Additionally, 6-Shogaol has been reported to induce apoptosis in liver cancer cells. However, the mechanism by which 6-shogaol regulates apoptosis in liver cancer cells remains unclear. To investigate the function of 6-shogaol in liver cancer, RT-qPCR and western blotting were used to detect the expression of TLR4 and FOXO3a in liver cancer cells, respectively. The OD value of liver cancer cells was measured using the MTT assay. Flow cytometry was used to measure cell apoptosis. 6-Shogaol inhibited the growth of liver cancer cells. TLR4 and Wnt/[Formula: see text]-catenin were upregulated in liver cancer cells, and FOXO3a was inactivated, but 6-Shogaol reversed the expression of TLR4, Wnt/[Formula: see text]-catenin and FOXO3a in liver cancer cells. Additionally, TLR4 overexpression partially reversed the inhibitory effect of 6-shogaol on the progression of liver cancer cells via Wnt/[Formula: see text]-catenin signaling. Furthermore, the 6-shogaol-induced increase in FOXO3a expression in liver cancer was notably suppressed by TLR4 or Wnt/[Formula: see text]-catenin upregulation. Thus, 6-Shogaol suppresses the progression of liver cancer by mediating Wnt/[Formula: see text]-catenin signaling and is a potential agent for the treatment of liver cancer.
Collapse
Affiliation(s)
- Yi Zhang
- Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan 430065, Hubei Province, P. R. China
| | - Jingjing Wang
- Pharmaceutical Department, Wuhan Third Hospital (Tongren Hospital of Wuhan University, Wuhan 430060, Hubei Province, P. R. China
| | - Yong Qu
- Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan 430065, Hubei Province, P. R. China
| | - Yunzhong Chen
- Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan 430065, Hubei Province, P. R. China
| |
Collapse
|
20
|
Jiang C, Xu M, Zhu J, Yang D, Xue B. CircTHBS1 facilitates the progression of interstitial cystitis depending on the regulation of miR-139-5p/MFN2 axis. Drug Dev Res 2021; 83:351-361. [PMID: 34368980 DOI: 10.1002/ddr.21864] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/16/2021] [Accepted: 06/28/2021] [Indexed: 11/10/2022]
Abstract
Circular RNA (circRNA) have been found to play an important role in the progression of many diseases, including interstitial cystitis (IC). However, the role of circTHBS1 in IC progression is still unclear. Exploring the role and potential molecular mechanism of circTHBS1 in the development of IC. The enzyme-linked immunosorbent assay was used to assess the levels of inflammatory cytokines. The expression levels of circTHBS1, microRNA (miR)-139-5p, and mitofusin 2 (MFN2) were evaluated using quantitative real-time PCR. Cell proliferation and migration were determined using MTT assay, Edu staining, and transwell assay. The protein levels of epithelial-mesenchymal transition (EMT) markers and MFN2 were examined using western blot analysis. The relationship between miR-139-5p and circTHBS1 or MFN2 was confirmed using the dual-luciferase reporter assay and RIP assay. CircTHBS1 was highly repressed in IC tissues and cells, and its expression was positively correlated with the inflammatory response of IC patients. CircTHBS1 could promote the proliferation, migration, EMT process, and inflammation of IC cells, while its knockdown had an opposite effect. CircTHBS1 could serve as a sponge of miR-139-5p, and miR-139-5p could participate in the regulation of circTHBS1 on IC cell progression. In addition, miR-139-5p could target MFN2, and it could inhibit the progression of IC cells by targeting MFN2. Furthermore, circTHBS1 sponged miR-139-5p to positively regulate MFN2. CircTHBS1 promoted IC cell proliferation, migration, EMT process, and inflammation by regulating the miR-139-5p/MFN2 axis indicating that circTHBS1 might be a potential target for IC treatment.
Collapse
Affiliation(s)
- Chen Jiang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ming Xu
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jin Zhu
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Dongrong Yang
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Boxin Xue
- Department of Urology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
21
|
Abstract
Preeclampsia (PE) is an idiopathic disease that occurs during pregnancy. It comprises multiple organ and system damage, and can seriously threaten the safety of the mother and infant throughout the perinatal period. As the pathogenesis of PE is unclear, there are few specific remedies. Currently, the only way to eliminate the clinical symptoms is to terminate the pregnancy. Although noncoding RNA (ncRNA) was once thought to be the "junk" of gene transcription, it is now known to be widely involved in pathological and physiological processes, including pregnancy-related disorders. Moreover, there is growing evidence that the unbalanced expression of specific ncRNA is involved in the pathogenesis of PE. In the present review, we summarize the expression patterns of ncRNAs, i.e., microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), and the functional mechanisms by which they affect the development of PE, and examine the clinical significance of ncRNAs as biomarkers for the diagnosis of PE. We also discuss the contributions made by genetic polymorphisms and epigenetic ncRNA regulation to PE. In the present review, we wish to explore and reinforce the clinical value of ncRNAs as noninvasive biomarkers of PE.
Collapse
Affiliation(s)
- Ningxia Sun
- Department of Medical Genetic, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
- Department of Gynecology and obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Shiting Qin
- Department of Medical Genetic, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Lu Zhang
- Department of Medical Genetic, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China.
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Shiguo Liu
- Department of Medical Genetic, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China.
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
22
|
Exosomal miR-486-5p derived from human placental microvascular endothelial cells regulates proliferation and invasion of trophoblasts via targeting IGF1. Hum Cell 2021; 34:1310-1323. [PMID: 33977502 PMCID: PMC8338855 DOI: 10.1007/s13577-021-00543-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/27/2021] [Indexed: 01/12/2023]
Abstract
Preeclampsia (PE) is a serious complication of pregnancy. Exosomes are known to be upregulated in PE. In this study, we sought to investigate the effect of miR-486-5p from human placental microvascular endothelial cells, on the function of trophoblast cells. To investigate the function of human placental microvascular endothelial cell (HPVEC)-derived exosomes on trophoblast cells, HPVECs were treated with hypoxia/reoxygenation (H/R). The separation efficiency of exosomes was determined by transmission electron microscopy, nanosight and Western blot. Cell Counting Kit-8, EdU staining, wound-healing, and transwell assay were performed to detect the effect of exosomally transferred miR-486-5p inhibitor on proliferation, migration and invasion of trophoblast cells. MiRDB and dual-luciferase report assay were used to find the target of miR-486-5p. Our data revealed that miR-486-5p was significantly upregulated in H/R-treated HPVEC-Exo, and miR-486-5p was enriched in HPVEC-Exo. miR-486-5p inhibitor carried by HPVEC-Exo significantly inhibited the proliferation, migration and invasion of trophoblast cells. Insulin-like growth factor 1 (IGF1) was found to be the target of miR-486-5p, and IGF1 overexpression notably reversed the effect of miR-486-5p inhibitor from HPVEC-Exo on trophoblast cell function. In summary, H/R-treated HPVEC-derived exosomally expressing miR-486-5p inhibitor significantly inhibited the proliferation, migration and invasion of trophoblast cells via downregulation of IGF1. The findings from the present study may be useful in the development of treatments for PE.
Collapse
|
23
|
Extracellular Vesicles from Mesenchymal Stromal Cells for the Treatment of Inflammation-Related Conditions. Int J Mol Sci 2021; 22:ijms22063023. [PMID: 33809632 PMCID: PMC8002312 DOI: 10.3390/ijms22063023] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/06/2021] [Accepted: 03/12/2021] [Indexed: 12/14/2022] Open
Abstract
Over the past two decades, mesenchymal stromal cells (MSCs) have demonstrated great potential in the treatment of inflammation-related conditions. Numerous early stage clinical trials have suggested that this treatment strategy has potential to lead to significant improvements in clinical outcomes. While promising, there remain substantial regulatory hurdles, safety concerns, and logistical issues that need to be addressed before cell-based treatments can have widespread clinical impact. These drawbacks, along with research aimed at elucidating the mechanisms by which MSCs exert their therapeutic effects, have inspired the development of extracellular vesicles (EVs) as anti-inflammatory therapeutic agents. The use of MSC-derived EVs for treating inflammation-related conditions has shown therapeutic potential in both in vitro and small animal studies. This review will explore the current research landscape pertaining to the use of MSC-derived EVs as anti-inflammatory and pro-regenerative agents in a range of inflammation-related conditions: osteoarthritis, rheumatoid arthritis, Alzheimer's disease, cardiovascular disease, and preeclampsia. Along with this, the mechanisms by which MSC-derived EVs exert their beneficial effects on the damaged or degenerative tissues will be reviewed, giving insight into their therapeutic potential. Challenges and future perspectives on the use of MSC-derived EVs for the treatment of inflammation-related conditions will be discussed.
Collapse
|