1
|
Buczyńska A, Sidorkiewicz I, Niemira M, Krętowski AJ, Węgrzyn P, Kosiński P, Zbucka-Krętowska M. Identification of MicroRNA Profiles in Fetal Spina Bifida: The Role in Pathomechanism and Diagnostic Significance. Int J Mol Sci 2024; 25:2896. [PMID: 38474143 DOI: 10.3390/ijms25052896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/16/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Distinct miRNA expression patterns may reflect anomalies related to fetal congenital malformations such as spinal bifida (SB). The aim of this preliminary study was to determine the maternal miRNA expression profile of women carrying fetuses with SB. Therefore, six women carrying fetuses with SB and twenty women with euploid healthy fetuses were enrolled in this study. Using NanoString technology, we evaluated the expression level of 798 miRNAs in both plasma and amniotic fluid samples. A downregulation of miR-1253, miR-1290, miR-194-5p, miR-302d-3p, miR-3144-3p, miR-4536-5p, miR-548aa + miR-548t-3p, miR-548ar-5p, miR-548n, miR-590-5p, miR-612, miR-627-5p, miR-644a, and miR-122-5p, and an upregulation of miR-320e, let-7b-5p, miR-23a-3p, miR-873-3p, and miR-30d-5p were identified in maternal amniotic fluid samples in SB when compared to the control group. The target genes of these miRNAs play a predominant role in regulating the synthesis of several biological compounds related to signaling pathways such as those regulating the pluripotency of stem cells. Moreover, the maternal plasma expression of miR-320e was increased in pregnancies with SB, and this marker could serve as a valuable non-invasive screening tool. Our results highlight the SB-specific miRNA signature and the differentially expressed miRNAs that may be involved in SB pathogenesis. Our findings emphasize the role of miRNA as a predictive factor that could potentially be useful in prenatal genetic screening for SB.
Collapse
Affiliation(s)
- Angelika Buczyńska
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Iwona Sidorkiewicz
- Clinical Research Support Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Magdalena Niemira
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Adam Jacek Krętowski
- Clinical Research Centre, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Piotr Węgrzyn
- Department of Obstetrics, Perinatology and Gynecology, Medical University of Warsaw, 63A Zwirki i Wigury, 02-091 Warsaw, Poland
| | - Przemysław Kosiński
- Department of Obstetrics, Perinatology and Gynecology, Medical University of Warsaw, 63A Zwirki i Wigury, 02-091 Warsaw, Poland
| | - Monika Zbucka-Krętowska
- Department of Gynecological Endocrinology and Adolescent Gynecology, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| |
Collapse
|
2
|
Cheng J, Ho WK, Wu BT, Liu HP, Lin WY. miRNA profiling as a complementary diagnostic tool for amyotrophic lateral sclerosis. Sci Rep 2023; 13:13805. [PMID: 37612427 PMCID: PMC10447559 DOI: 10.1038/s41598-023-40879-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS), the most prevalent motor neuron disease characterized by its complex genetic structure, lacks a single diagnostic test capable of providing a conclusive diagnosis. In order to demonstrate the potential for genetic diagnosis and shed light on the pathogenic role of miRNAs in ALS, we developed an ALS diagnostic rule by training the model using 80% of a miRNA profiling dataset consisting of 253 ALS samples and 103 control samples. Subsequently, we validated the diagnostic rule using the remaining 20% of unseen samples. The diagnostic rule we developed includes miR-205-5p, miR-206, miR-376a-5p, miR-412-5p, miR-3927-3p, miR-4701-3p, miR-6763-5p, and miR-6801-3p. Remarkably, the rule achieved an 82% true positive rate and a 73% true negative rate when predicting the unseen samples. Furthermore, the identified miRNAs target 21 genes in the PI3K-Akt pathway and 27 genes in the ALS pathway, including notable genes such as BCL2, NEFH, and OPTN. We propose that miRNA profiling may serve as a complementary diagnostic tool to supplement the clinical presentation and aid in the early recognition of ALS.
Collapse
Affiliation(s)
- Jack Cheng
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
- Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Wen-Kuang Ho
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Bor-Tsang Wu
- Department of Senior Citizen Service Management, National Taichung University of Science and Technology, Taichung City, 40343, Taiwan
| | - Hsin-Ping Liu
- Graduate Institute of Acupuncture Science, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan.
| | - Wei-Yong Lin
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan.
- Department of Medical Research, China Medical University Hospital, Taichung, 40447, Taiwan.
| |
Collapse
|
3
|
Guerrero-Santoro J, Morizane M, Oh SY, Mishima T, Goff JP, Bildirici I, Sadovsky E, Ouyang Y, Tyurin VA, Tyurina YY, Kagan VE, Sadovsky Y. The lipase cofactor CGI58 controls placental lipolysis. JCI Insight 2023; 8:168717. [PMID: 37212279 DOI: 10.1172/jci.insight.168717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/12/2023] [Indexed: 05/23/2023] Open
Abstract
In eutherians, the placenta plays a critical role in the uptake, storage, and metabolism of lipids. These processes govern the availability of fatty acids to the developing fetus, where inadequate supply has been associated with substandard fetal growth. Whereas lipid droplets are essential for the storage of neutral lipids in the placenta and many other tissues, the processes that regulate placental lipid droplet lipolysis remain largely unknown. To assess the role of triglyceride lipases and their cofactors in determining placental lipid droplet and lipid accumulation, we assessed the role of patatin like phospholipase domain containing 2 (PNPLA2) and comparative gene identification-58 (CGI58) in lipid droplet dynamics in the human and mouse placenta. While both proteins are expressed in the placenta, the absence of CGI58, not PNPLA2, markedly increased placental lipid and lipid droplet accumulation. These changes were reversed upon restoration of CGI58 levels selectively in the CGI58-deficient mouse placenta. Using co-immunoprecipitation, we found that, in addition to PNPLA2, PNPLA9 interacts with CGI58. PNPLA9 was dispensable for lipolysis in the mouse placenta yet contributed to lipolysis in human placental trophoblasts. Our findings establish a crucial role for CGI58 in placental lipid droplet dynamics and, by extension, in nutrient supply to the developing fetus.
Collapse
Affiliation(s)
- Jennifer Guerrero-Santoro
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mayumi Morizane
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Soo-Young Oh
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Takuya Mishima
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Julie P Goff
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ibrahim Bildirici
- Department of Obstetrics and Gynecology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Elena Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yingshi Ouyang
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Vladimir A Tyurin
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
| | - Yulia Y Tyurina
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Department of Chemistry
- Department of Pharmacology and Chemical Biology
- Department of Radiation Oncology; and
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
4
|
Huang CC, Hsueh YW, Chang CW, Hsu HC, Yang TC, Lin WC, Chang HM. Establishment of the fetal-maternal interface: developmental events in human implantation and placentation. Front Cell Dev Biol 2023; 11:1200330. [PMID: 37266451 PMCID: PMC10230101 DOI: 10.3389/fcell.2023.1200330] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Early pregnancy is a complex and well-orchestrated differentiation process that involves all the cellular elements of the fetal-maternal interface. Aberrant trophoblast-decidual interactions can lead to miscarriage and disorders that occur later in pregnancy, including preeclampsia, intrauterine fetal growth restriction, and preterm labor. A great deal of research on the regulation of implantation and placentation has been performed in a wide range of species. However, there is significant species variation regarding trophoblast differentiation as well as decidual-specific gene expression and regulation. Most of the relevant information has been obtained from studies using mouse models. A comprehensive understanding of the physiology and pathology of human implantation and placentation has only recently been obtained because of emerging advanced technologies. With the derivation of human trophoblast stem cells, 3D-organoid cultures, and single-cell analyses of differentiated cells, cell type-specific transcript profiles and functions were generated, and each exhibited a unique signature. Additionally, through integrative transcriptomic information, researchers can uncover the cellular dysfunction of embryonic and placental cells in peri-implantation embryos and the early pathological placenta. In fact, the clinical utility of fetal-maternal cellular trafficking has been applied for the noninvasive prenatal diagnosis of aneuploidies and the prediction of pregnancy complications. Furthermore, recent studies have proposed a viable path toward the development of therapeutic strategies targeting placenta-enriched molecules for placental dysfunction and diseases.
Collapse
|
5
|
Treeck O, Haerteis S, Ortmann O. Non-Coding RNAs Modulating Estrogen Signaling and Response to Endocrine Therapy in Breast Cancer. Cancers (Basel) 2023; 15:cancers15061632. [PMID: 36980520 PMCID: PMC10046587 DOI: 10.3390/cancers15061632] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
The largest part of human DNA is transcribed into RNA that does not code for proteins. These non-coding RNAs (ncRNAs) are key regulators of protein-coding gene expression and have been shown to play important roles in health, disease and therapy response. Today, endocrine therapy of ERα-positive breast cancer (BC) is a successful treatment approach, but resistance to this therapy is a major clinical problem. Therefore, a deeper understanding of resistance mechanisms is important to overcome this resistance. An increasing amount of evidence demonstrate that ncRNAs affect the response to endocrine therapy. Thus, ncRNAs are considered versatile biomarkers to predict or monitor therapy response. In this review article, we intend to give a summary and update on the effects of microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) on estrogen signaling in BC cells, this pathway being the target of endocrine therapy, and their role in therapy resistance. For this purpose, we reviewed articles on these topics listed in the PubMed database. Finally, we provide an assessment regarding the clinical use of these ncRNA types, particularly their circulating forms, as predictive BC biomarkers and their potential role as therapy targets to overcome endocrine resistance.
Collapse
Affiliation(s)
- Oliver Treeck
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany
- Correspondence:
| | - Silke Haerteis
- Institute for Molecular and Cellular Anatomy, University of Regensburg, 93053 Regensburg, Germany
| | - Olaf Ortmann
- Department of Gynecology and Obstetrics, University Medical Center Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
6
|
Devor EJ, Santillan DA, Warrier A, Scroggins SM, Santillan MK. Placenta-specific protein 1 (PLAC1) expression is significantly down-regulated in preeclampsia via a hypoxia-mediated mechanism. J Matern Fetal Neonatal Med 2022; 35:8419-8425. [PMID: 34565269 PMCID: PMC8959068 DOI: 10.1080/14767058.2021.1977792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/03/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Examine a mechanism of PLAC1 regulation and its potential role in preeclampsia (PE). MATERIALS AND METHODS Placental tissue samples and detailed clinical information were obtained through the University of Iowa Maternal Fetal Tissue Bank (IRB# 200910784) from gestational and maternal age-matched control (n = 17) and PE affected pregnancies (n = 12). PLAC1 and PLAC1 promoter-specific expression was measured using quantitative polymerase chain reaction (qPCR) and differences were assessed via the standard ΔΔCt method. In addition, the role of hypoxia in PLAC1 transcription was investigated through the exposure of HTR8/SVneo human trophoblast cells to the hypoxia mimic dimethyloxaloylglycine (DMOG). RESULTS PLAC1 expression is seen to be 8.9-fold lower in human placentas affected by preeclampsia in comparison with controls (p < .05). Further, this decrease is paralleled by a significantly lower expression of the P2 or proximal PLAC1 promoter (p < .05). Expression of mediator complex subunit 1 (MED1), a known hypoxia-sensitive transcription coactivator and PLAC1 effector, is significantly correlated with PLAC 1 expression (r2 = 0.607, p < .001). These data suggest that PLAC1 expression is significantly down-regulated in preeclampsia at least in part via a MED1 hypoxia-mediated mechanism. CONCLUSIONS We confirm that PLAC1 transcription is suppressed in the placentae of women affected by preeclampsia. We further demonstrate that this suppression is driven through the P2 or proximal PLAC1 promoter. This demonstration led to the identification of the MED1-TRAP cofactor complex as the hypoxia-sensitive driver.
Collapse
Affiliation(s)
- Eric J. Devor
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Donna A. Santillan
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Akshaya Warrier
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Sabrina M. Scroggins
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Mark K. Santillan
- Department of Obstetrics and Gynecology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
7
|
Micro-RNAs in Human Placenta: Tiny Molecules, Immense Power. Molecules 2022; 27:molecules27185943. [PMID: 36144676 PMCID: PMC9501247 DOI: 10.3390/molecules27185943] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/30/2022] [Accepted: 09/09/2022] [Indexed: 12/06/2022] Open
Abstract
Micro-RNAs (miRNAs) are short non-coding single-stranded RNAs that modulate the expression of various target genes after transcription. The expression and distribution of kinds of miRNAs have been characterized in human placenta during different gestational stages. The identified miRNAs are recognized as key mediators in the regulation of placental development and in the maintenance of human pregnancy. Aberrant expression of miRNAs is associated with compromised pregnancies in humans, and dysregulation of those miRNAs contributes to the occurrence and development of related diseases during pregnancy, such as pre-eclampsia (PE), fetal growth restriction (FGR), gestational diabetes mellitus (GDM), recurrent miscarriage, preterm birth (PTB) and small-for-gestational-age (SGA). Thus, having a better understanding of the expression and functions of miRNAs in human placenta during pregnancy and thereby developing novel drugs targeting the miRNAs could be a potentially promising method in the prevention and treatment of relevant diseases in future. Here, we summarize the current knowledge of the expression pattern and function regulation of miRNAs in human placental development and related diseases.
Collapse
|
8
|
Exploring the Expression of Pro-Inflammatory and Hypoxia-Related MicroRNA-20a, MicroRNA-30e, and MicroRNA-93 in Periodontitis and Gingival Mesenchymal Stem Cells under Hypoxia. Int J Mol Sci 2022; 23:ijms231810310. [PMID: 36142220 PMCID: PMC9499533 DOI: 10.3390/ijms231810310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/21/2022] [Accepted: 08/28/2022] [Indexed: 11/22/2022] Open
Abstract
Hypoxia associated with inflammation are common hallmarks observed in several diseases, and it plays a major role in the expression of non-coding RNAs, including microRNAs (miRNAs). In addition, the miRNA target genes for hypoxia-inducible factor-1α (HIF-1α) and nuclear factor of activated T cells-5 (NFAT5) modulate the adaptation to hypoxia. The objective of the present study was to explore hypoxia-related miRNA target genes for HIF-1α and NFAT5, as well as miRNA-20a, miRNA-30e, and miRNA-93 expression in periodontitis versus healthy gingival tissues and gingival mesenchymal stem cells (GMSCs) cultured under hypoxic conditions. Thus, a case-control study was conducted, including healthy and periodontitis subjects. Clinical data and gingival tissue biopsies were collected to analyze the expression of miRNA-20a, miRNA-30e, miRNA-93, HIF-1α, and NFAT5 by qRT-PCR. Subsequently, GMSCs were isolated and cultured under hypoxic conditions (1% O2) to explore the expression of the HIF-1α, NFAT5, and miRNAs. The results showed a significant upregulation of miRNA-20a (p = 0.028), miRNA-30e (p = 0.035), and miRNA-93 (p = 0.026) in periodontitis tissues compared to healthy gingival biopsies. NFAT5 mRNA was downregulated in periodontitis tissues (p = 0.037), but HIF-1α was not affected (p = 0.60). Interestingly, hypoxic GMSCs upregulated the expression of miRNA-20a and HIF-1α, but they downregulated miRNA-93e. In addition, NFAT5 mRNA expression was not affected in hypoxic GMSCs. In conclusion, in periodontitis patients, the expression of miRNA-20a, miRNA-30e, and miRNA-93 increased, but a decreased expression of NFAT5 mRNA was detected. In addition, GMSCs under hypoxic conditions upregulate the HIF-1α and increase miRNA-20a (p = 0.049) expression. This study explores the role of inflammatory and hypoxia-related miRNAs and their target genes in periodontitis and GMSCs. It is crucial to determine the potential therapeutic target of these miRNAs and hypoxia during the periodontal immune–inflammatory response, which should be analyzed in greater depth in future studies.
Collapse
|
9
|
Kochhar P, Vukku M, Rajashekhar R, Mukhopadhyay A. microRNA signatures associated with fetal growth restriction: a systematic review. Eur J Clin Nutr 2022; 76:1088-1102. [PMID: 34741137 DOI: 10.1038/s41430-021-01041-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/17/2021] [Accepted: 10/19/2021] [Indexed: 12/20/2022]
Abstract
Placental-origin microRNA (miRNA) profiles can be useful toward early diagnosis and management of fetal growth restriction (FGR) and associated complications. We conducted a systematic review to identify case-control studies that have examined miRNA signatures associated with human FGR. We systematically searched PubMed and ScienceDirect databases for relevant articles and manually searched reference lists of the relevant articles till May 18th, 2021. Of the 2133 studies identified, 21 were included. FGR-associated upregulation of miR-210 and miR-424 and downregulation of a placenta-specific miRNA cluster miRNA located on C19MC (miR-518b, miR-519d) and miR-221-3p was reported by >1 included studies. Analysis of the target genes of these miRNA as well as pathway analysis pointed to the involvement of angiogenesis and growth signaling pathways, such as the phosphatidylinositol 3-kinase- protein kinase B (PI3K-Akt) pathway. Only 3 out of the 21 included studies reported FGR-associated miRNAs in matched placental and maternal blood samples. We conclude that FGR-associated placental miRNAs could be utilized to inform clinical practice towards early diagnosis of FGR, provided enough evidence from studies on matched placental and maternal blood samples become available.Prospective Register of Systematic Reviews (PROSPERO) registration number: CRD42019136762.
Collapse
Affiliation(s)
- P Kochhar
- Division of Nutrition, St. John's Research Institute, A Recognized Research Centre of University of Mysore, Bangalore, India
| | - M Vukku
- Division of Nutrition, St. John's Research Institute, A Recognized Research Centre of University of Mysore, Bangalore, India
| | - R Rajashekhar
- Division of Nutrition, St. John's Research Institute, A Recognized Research Centre of University of Mysore, Bangalore, India.,Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - A Mukhopadhyay
- Division of Nutrition, St. John's Research Institute, A Recognized Research Centre of University of Mysore, Bangalore, India.
| |
Collapse
|
10
|
Wu S, Liu H, Zhou M, Shang Y, Luo L, Chen J, Yang J. The miR-410-5p /ITGA6 axis participates in the pathogenesis of recurrent abortion by regulating the biological function of trophoblast. J Reprod Immunol 2022; 152:103647. [DOI: 10.1016/j.jri.2022.103647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/30/2022] [Accepted: 05/18/2022] [Indexed: 11/15/2022]
|
11
|
Rutigliano HM, Thomas AJ, Umbaugh JJ, Wilhelm A, Sessions BR, Kaundal R, Duhan N, Hicks BA, Schlafer DH, White KL, Davies CJ. Increased expression of pro-inflammatory cytokines at the fetal-maternal interface in bovine pregnancies produced by cloning. Am J Reprod Immunol 2022; 87:e13520. [PMID: 34974639 PMCID: PMC9285385 DOI: 10.1111/aji.13520] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 12/10/2021] [Accepted: 12/29/2021] [Indexed: 11/26/2022] Open
Abstract
PROBLEM A significant rate of spontaneous abortion is observed in cattle pregnancies produced by somatic cell nuclear transfer (SCNT). Major histocompatibility complex class I (MHC-I) proteins are abnormally expressed on the surface of trophoblast cells from SCNT conceptuses. METHOD OF STUDY MHC-I homozygous compatible (n = 9), homozygous incompatible (n = 8), and heterozygous incompatible (n = 5) pregnancies were established by SCNT. Eight control pregnancies were established by artificial insemination. Uterine and trophoblast samples were collected on day 35 ±1 of pregnancy, the expression of immune-related genes was examined by qPCR, and the expression of trophoblast microRNAs was assessed by sequencing. RESULTS Compared to the control group, trophoblast from MHC-I heterozygous incompatible pregnancies expressed increased levels of CD28, CTLA4, CXCL8, IFNG, IL1A, IL2, IL10, IL12B, TBX21, and TNF, while GNLY expression was downregulated. The MHC-I homozygous incompatible treatment group expressed increased levels of IFNG, IL1A, and IL2 while the MHC-I homozygous compatible group did not differentially express any genes compared to the control group. In the endometrium, relative to the control group, MHC-I heterozygous incompatible pregnancies expressed increased levels of CD28, CTLA4, CXCL8, IFNG, IL10, IL12B, and TNF, while GATA3 expression was downregulated. The MHC-I homozygous incompatible group expressed decreased amounts of CSF2 transcripts compared with the control group but did not have abnormal expression of any other immune-related genes. MHC-I incompatible pregnancies had 40 deregulated miRNAs compared to control pregnancies and 62 deregulated microRNAs compared to MHC-I compatible pregnancies. CONCLUSIONS MHC-I compatibility between the dam and fetus prevented an exacerbated maternal immune response from being mounted against fetal antigens.
Collapse
Affiliation(s)
- Heloisa M Rutigliano
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, USA.,School of Veterinary Medicine, Utah State University, Logan, Utah, USA.,Center for Integrated BioSystems, Utah State University, Logan, Utah, USA
| | - Aaron J Thomas
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, USA.,Center for Integrated BioSystems, Utah State University, Logan, Utah, USA
| | - Janae J Umbaugh
- School of Veterinary Medicine, Utah State University, Logan, Utah, USA
| | - Amanda Wilhelm
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, USA.,Center for Integrated BioSystems, Utah State University, Logan, Utah, USA
| | - Benjamin R Sessions
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, USA.,Center for Integrated BioSystems, Utah State University, Logan, Utah, USA
| | - Rakesh Kaundal
- Center for Integrated BioSystems, Utah State University, Logan, Utah, USA.,Department of Plants, Soils and Climate, Utah State University, Logan, Utah, USA
| | - Naveen Duhan
- Center for Integrated BioSystems, Utah State University, Logan, Utah, USA.,Department of Plants, Soils and Climate, Utah State University, Logan, Utah, USA
| | - Brady A Hicks
- J.R. Simplot Company Cattle Reproduction Facility, Boise, Idaho, USA
| | - Donald H Schlafer
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Kenneth L White
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, USA.,School of Veterinary Medicine, Utah State University, Logan, Utah, USA.,Center for Integrated BioSystems, Utah State University, Logan, Utah, USA
| | - Christopher J Davies
- Department of Animal, Dairy and Veterinary Sciences, Utah State University, Logan, Utah, USA.,School of Veterinary Medicine, Utah State University, Logan, Utah, USA.,Center for Integrated BioSystems, Utah State University, Logan, Utah, USA
| |
Collapse
|
12
|
Assessing hypoxic damage to placental trophoblasts by measuring membrane viscosity of extracellular vesicles. Placenta 2022; 121:14-22. [PMID: 35245720 PMCID: PMC9010367 DOI: 10.1016/j.placenta.2022.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 02/06/2022] [Accepted: 02/22/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION As highly sophisticated intercellular communication vehicles in biological systems, extracellular vesicles (EVs) have been investigated as both promising liquid biopsy-based disease biomarkers and drug delivery carriers. Despite tremendous progress in understanding their biological and physiological functions, mechanical characterization of these nanoscale entities remains challenging due to the limited availability of proper techniques. Especially, whether damage to parental cells can be reflected by the mechanical properties of their EVs remains unknown. METHODS In this study, we characterized membrane viscosities of different types of EVs collected from primary human trophoblasts (PHTs), including apoptotic bodies, microvesicles and small extracellular vesicles, using fluorescence lifetime imaging microscopy (FLIM). The biochemical origin of EV membrane viscosity was examined by analyzing their phospholipid composition, using mass spectrometry. RESULTS We found that different EV types derived from the same cell type exhibit different membrane viscosities. The measured membrane viscosity values are well supported by the lipidomic analysis of the phospholipid compositions. We further demonstrate that the membrane viscosity of microvesicles can faithfully reveal hypoxic injury of the human trophoblasts. More specifically, the membrane of PHT microvesicles released under hypoxic condition is less viscous than its counterpart under standard culture condition, which is supported by the reduction in the phosphatidylethanolamine-to-phosphatidylcholine ratio in PHT microvesicles. DISCUSSION Our study suggests that biophysical properties of released trophoblastic microvesicles can reflect cell health. Characterizing EV's membrane viscosity may pave the way for the development of new EV-based clinical applications.
Collapse
|
13
|
Role of the Mediator Complex and MicroRNAs in Breast Cancer Etiology. Genes (Basel) 2022; 13:genes13020234. [PMID: 35205279 PMCID: PMC8871970 DOI: 10.3390/genes13020234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 12/16/2022] Open
Abstract
Transcriptional coactivators play a key role in RNA polymerase II transcription and gene regulation. One of the most important transcriptional coactivators is the Mediator (MED) complex, which is an evolutionary conserved large multiprotein complex. MED transduces the signal between DNA-bound transcriptional activators (gene-specific transcription factors) to the RNA polymerase II transcription machinery to activate transcription. It is known that MED plays an essential role in ER-mediated gene expression mainly through the MED1 subunit, since estrogen receptor (ER) can interact with MED1 by specific protein–protein interactions; therefore, MED1 plays a fundamental role in ER-positive breast cancer (BC) etiology. Additionally, other MED subunits also play a role in BC etiology. On the other hand, microRNAs (miRNAs) are a family of small non-coding RNAs, which can regulate gene expression at the post-transcriptional level by binding in a sequence-specific fashion at the 3′ UTR of the messenger RNA. The miRNAs are also important factors that influence oncogenic signaling in BC by acting as both tumor suppressors and oncogenes. Moreover, miRNAs are involved in endocrine therapy resistance of BC, specifically to tamoxifen, a drug that is used to target ER signaling. In metazoans, very little is known about the transcriptional regulation of miRNA by the MED complex and less about the transcriptional regulation of miRNAs involved in BC initiation and progression. Recently, it has been shown that MED1 is able to regulate the transcription of the ER-dependent miR-191/425 cluster promoting BC cell proliferation and migration. In this review, we will discuss the role of MED1 transcriptional coactivator in the etiology of BC and in endocrine therapy-resistance of BC and also the contribution of other MED subunits to BC development, progression and metastasis. Lastly, we identified miRNAs that potentially can regulate the expression of MED subunits.
Collapse
|
14
|
Baker BC, Lui S, Lorne I, Heazell AEP, Forbes K, Jones RL. Sexually dimorphic patterns in maternal circulating microRNAs in pregnancies complicated by fetal growth restriction. Biol Sex Differ 2021; 12:61. [PMID: 34789323 PMCID: PMC8597318 DOI: 10.1186/s13293-021-00405-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/27/2021] [Indexed: 12/18/2022] Open
Abstract
Background Current methods fail to accurately predict women at greatest risk of developing fetal growth restriction (FGR) or related adverse outcomes, including stillbirth. Sexual dimorphism in these adverse pregnancy outcomes is well documented as are sex-specific differences in gene and protein expression in the placenta. Circulating maternal serum microRNAs (miRNAs) offer potential as biomarkers that may also be informative of underlying pathology. We hypothesised that FGR would be associated with an altered miRNA profile and would differ depending on fetal sex. Methods miRNA expression profiles were assessed in maternal serum (> 36 weeks’ gestation) from women delivering a severely FGR infant (defined as an individualised birthweight centile (IBC) < 3rd) and matched control participants (AGA; IBC = 20–80th), using miRNA arrays. qPCR was performed using specific miRNA primers in an expanded cohort of patients with IBC < 5th (n = 15 males, n = 16 females/group). Maternal serum human placental lactogen (hPL) was used as a proxy to determine if serum miRNAs were related to placental dysfunction. In silico analyses were performed to predict the potential functions of altered miRNAs. Results Initial analyses revealed 11 miRNAs were altered in maternal serum from FGR pregnancies. In silico analyses revealed all 11 altered miRNAs were located in a network of genes that regulate placental function. Subsequent analysis demonstrated four miRNAs showed sexually dimorphic patterns. miR-28-5p was reduced in FGR pregnancies (p < 0.01) only when there was a female offspring and miR-301a-3p was only reduced in FGR pregnancies with a male fetus (p < 0.05). miR-454-3p was decreased in FGR pregnancies (p < 0.05) regardless of fetal sex but was only positively correlated to hPL when the fetus was female. Conversely, miR-29c-3p was correlated to maternal hPL only when the fetus was male. Target genes for sexually dimorphic miRNAs reveal potential functional roles in the placenta including angiogenesis, placental growth, nutrient transport and apoptosis. Conclusions These studies have identified sexually dimorphic patterns for miRNAs in maternal serum in FGR. These miRNAs may have potential as non-invasive biomarkers for FGR and associated placental dysfunction. Further studies to determine if these miRNAs have potential functional roles in the placenta may provide greater understanding of the pathogenesis of placental dysfunction and the differing susceptibility of male and female fetuses to adverse in utero conditions. Supplementary Information The online version contains supplementary material available at 10.1186/s13293-021-00405-z. Detection and treatment of pregnancies at high risk of fetal growth restriction (FGR) and stillbirth remains a major obstetric challenge; circulating maternal serum microRNAs (miRNAs) offer potential as novel biomarkers. Unbiased analysis of serum miRNAs in women in late pregnancy identified a specific profile of circulating miRNAs in women with a growth-restricted infant. Some altered miRNAs (miR-28-5p, miR-301a-3p) showed sexually dimorphic expression in FGR pregnancies and others a fetal-sex dependent association to a hormonal marker of placental dysfunction (miR-454-3p, miR-29c-3p). miR-301a-3p and miR-28-5p could potentially be used to predict FGR specifically in pregnancies with a male or female baby, respectively, however larger cohort studies are required. Further investigations of these miRNAs and their relationship to placental dysfunction will lead to a better understanding of the pathophysiology of FGR and why there is differing susceptibility of male and female fetuses to FGR and stillbirth.
Collapse
Affiliation(s)
- Bernadette C Baker
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK.
| | - Sylvia Lui
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK.,Division of Inflammation and Repair, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Isabel Lorne
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| | - Alexander E P Heazell
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK.,St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M13 9WL, UK
| | - Karen Forbes
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK.
| | - Rebecca L Jones
- Division of Developmental Biology and Medicine, Maternal and Fetal Health Research Centre, University of Manchester, Manchester, UK
| |
Collapse
|
15
|
Smith MD, Pillman K, Jankovic-Karasoulos T, McAninch D, Wan Q, Bogias KJ, McCullough D, Bianco-Miotto T, Breen J, Roberts CT. Large-scale transcriptome-wide profiling of microRNAs in human placenta and maternal plasma at early to mid gestation. RNA Biol 2021; 18:507-520. [PMID: 34412547 PMCID: PMC8677031 DOI: 10.1080/15476286.2021.1963105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) are increasingly seen as important regulators of placental development and opportunistic biomarker targets. Given the difficulty in obtaining samples from early gestation and subsequent paucity of the same, investigation of the role of miRNAs in early gestation human placenta has been limited. To address this, we generated miRNA profiles using 96 placentas from presumed normal pregnancies, across early gestation, in combination with matched profiles from maternal plasma. Placenta samples range from 6 to 23 weeks' gestation, a time period that includes placenta from the early, relatively low but physiological (6-10 weeks' gestation) oxygen environment, and later, physiologically normal oxygen environment (11-23 weeks' gestation).We identified 637 miRNAs with expression in 86 samples (after removing poor quality samples), showing a clear gestational age gradient from 6 to 23 weeks' gestation. We identified 374 differentially expressed (DE) miRNAs between placentas from 6-10 weeks' versus 11-23 weeks' gestation. We see a clear gestational age group bias in miRNA clusters C19MC, C14MC, miR-17 ~ 92 and paralogs, regions that also include many DE miRNAs. Proportional change in expression of placenta-specific miRNA clusters was reflected in maternal plasma.The presumed introduction of oxygenated maternal blood into the placenta (between ~10 and 12 weeks' gestation) changes the miRNA profile of the chorionic villus, particularly in placenta-specific miRNA clusters. Data presented here comprise a clinically important reference set for studying early placenta development and may underpin the generation of minimally invasive methods for monitoring placental health.
Collapse
Affiliation(s)
- Melanie D Smith
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| | - Katherine Pillman
- Centre for Cancer Biology, University of South Australia/SA Pathology, Adelaide, SA, Australia
| | - Tanja Jankovic-Karasoulos
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| | - Dale McAninch
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Qianhui Wan
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| | - K Justinian Bogias
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Dylan McCullough
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| | - Tina Bianco-Miotto
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.,School of Agriculture Food and Wine, Waite Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - James Breen
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,South Australian Genomics Centre, South Australian Health & Medical Research Institute, Adelaide, SA, Australia
| | - Claire T Roberts
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| |
Collapse
|
16
|
Gonzalez TL, Eisman LE, Joshi NV, Flowers AE, Wu D, Wang Y, Santiskulvong C, Tang J, Buttle RA, Sauro E, Clark EL, DiPentino R, Jefferies CA, Chan JL, Lin Y, Zhu Y, Afshar Y, Tseng HR, Taylor K, Williams J, Pisarska MD. High-throughput miRNA sequencing of the human placenta: expression throughout gestation. Epigenomics 2021; 13:995-1012. [PMID: 34030457 PMCID: PMC8244582 DOI: 10.2217/epi-2021-0055] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Aim: To understand miRNA changes across gestation in healthy human placentae. This is essential before miRNAs can be used as biomarkers or prognostic indicators during pregnancy. Materials & methods: Using next-generation sequencing, we characterize the normative human placenta miRNome in first (n = 113) and third trimester (n = 47). Results & conclusion: There are 801 miRNAs expressed in both first and third trimester, including 182 with similar expression across gestation (p ≥ 0.05, fold change ≤2) and 180 significantly different (false discovery rate <0.05, fold change >2). Of placenta-specific miRNA clusters, chromosome 14 miRNA cluster decreases across gestation and chromosome 19 miRNA cluster is overall highly expressed. Chromosome 13 clusters are upregulated in first trimester. This work provides a rich atlas of healthy pregnancies to direct functional studies investigating the epigenetic differences in first and third trimester placentae.
Collapse
Affiliation(s)
- Tania L Gonzalez
- Department of Obstetrics & Gynecology, Center for Reproductive Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Laura E Eisman
- Department of Obstetrics & Gynecology, Center for Reproductive Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nikhil V Joshi
- Department of Obstetrics & Gynecology, Center for Reproductive Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amy E Flowers
- Department of Obstetrics & Gynecology, Center for Reproductive Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Di Wu
- Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yizhou Wang
- Genomics Core, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Chintda Santiskulvong
- CS Cancer Applied Genomics Shared Resource, CS Cancer, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jie Tang
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Rae A Buttle
- Department of Obstetrics & Gynecology, Center for Reproductive Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Erica Sauro
- Department of Obstetrics & Gynecology, Center for Reproductive Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ekaterina L Clark
- Department of Obstetrics & Gynecology, Center for Reproductive Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Rosemarie DiPentino
- Department of Obstetrics & Gynecology, Center for Reproductive Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Caroline A Jefferies
- Division of Rheumatology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jessica L Chan
- Department of Obstetrics & Gynecology, Center for Reproductive Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yayu Lin
- Department of Obstetrics & Gynecology, Center for Reproductive Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yazhen Zhu
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular & Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Yalda Afshar
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hsian-Rong Tseng
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular & Medical Pharmacology, University of California, Los Angeles, CA 90095, USA
| | - Kent Taylor
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- The Institute for Translational Genomics & Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - John Williams
- Department of Obstetrics & Gynecology, Center for Reproductive Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Margareta D Pisarska
- Department of Obstetrics & Gynecology, Center for Reproductive Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
17
|
Zhao L, Xiong M, Liu Y. Baicalin enhances the proliferation and invasion of trophoblasts and suppresses vascular endothelial damage by modulating long non-coding RNA NEAT1/miRNA-205-5p in hypertensive disorder complicating pregnancy. J Obstet Gynaecol Res 2021; 47:3060-3070. [PMID: 34101306 DOI: 10.1111/jog.14789] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/08/2021] [Accepted: 03/22/2021] [Indexed: 12/31/2022]
Abstract
AIM Trophoblastic and vascular endothelial injuries were closely associated with the pathogenesis of hypertensive disorder complicating pregnancy (HDCP). The present study was designed to determine the functional role of baicalin in the proliferation and invasion of trophoblasts and vascular endothelial injury. METHODS Ang II was adopted to stimulate HTR-8/SVneo and human umbilical vein endothelial cells (HUVECs). Cell viability was examined by CCK-8 assay. Flow cytometry and TUNEL staining determined cell apoptosis. Invasive ability of HTR-8/SVneo cells was measured by transwell assay. In vitro angiogenesis of HUVECs was assessed by Tube formation assay. In addition, the production of reactive oxygen species (ROS) was determined by DCFH-DA staining. Furthermore, long non-coding RNA (lncRNA) NEAT1 and miRNA-205-5p levels were detected using real-time quantitative polymerase chain reaction and the binding relationship between lncRNA NEAT1 and miRNA-205-5p was verified by dual-luciferase reporter assay. Moreover, interactions among lncRNA NEAT1, miRNA-205-5p, and MMP9 or vascular endothelial growth factor (VEGF) were confirmed by RNA immunoprecipitation assay. RESULTS Baicalin visibly improved cell viability, reduced the apoptosis of Ang II-stimulated HTR-8/SVneo and HUVEC cells, and repressed overproduction of ROS. Additionally, baicalin promoted the invasion of Ang II-stimulated HTR-8/SVneo cells and induced a stronger in vitro angiogenesis of Ang II-stimulated HUVECs. What's more, baicalin upregulated lncRNA NEAT1 expression and downregulated miR-205-5p expression. LncRNA NEAT1 sponged miR-205-5p and inhibited the combination of miR-205-5p and MMP9 or VEGF. CONCLUSIONS Baicalin can facilitate the proliferation and invasion of trophoblasts and alleviate vascular endothelial damage by upregulating lncRNA NEAT1 to impede the interaction between miR-205-5p and MMP9 or VEGF.
Collapse
Affiliation(s)
- Lidong Zhao
- Department of Emergency Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai JiaoTong University, Shanghai, China
| | - Miao Xiong
- Department of Obstetrics, Shanghai Sixth People's Hospital Affiliated to Shanghai JiaoTong University, Shanghai, China
| | - Yang Liu
- Department of Obstetrics, Shanghai Sixth People's Hospital Affiliated to Shanghai JiaoTong University, Shanghai, China
| |
Collapse
|
18
|
Chu T, Mouillet JF, Cao Z, Barak O, Ouyang Y, Sadovsky Y. RNA Network Interactions During Differentiation of Human Trophoblasts. Front Cell Dev Biol 2021; 9:677981. [PMID: 34150771 PMCID: PMC8209545 DOI: 10.3389/fcell.2021.677981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
In the human placenta, two trophoblast cell layers separate the maternal blood from the villous basement membrane and fetal capillary endothelial cells. The inner layer, which is complete early in pregnancy and later becomes discontinuous, comprises the proliferative mononuclear cytotrophoblasts, which fuse together and differentiate to form the outer layer of multinucleated syncytiotrophoblasts. Because the syncytiotrophoblasts are responsible for key maternal-fetal exchange functions, tight regulation of this differentiation process is critical for the proper development and the functional role of the placenta. The molecular mechanisms regulating the fusion and differentiation of trophoblasts during human pregnancy remain poorly understood. To decipher the interactions of non-coding RNAs (ncRNAs) in this process, we exposed cultured primary human trophoblasts to standard in vitro differentiation conditions or to conditions known to hinder this differentiation process, namely exposure to hypoxia (O2 < 1%) or to the addition of dimethyl sulfoxide (DMSO, 1.5%) to the culture medium. Using next generation sequencing technology, we analyzed the differential expression of trophoblastic lncRNAs, miRNAs, and mRNAs that are concordantly modulated by both hypoxia and DMSO. Additionally, we developed a model to construct a lncRNA-miRNA-mRNA co-expression network and inferred the functions of lncRNAs and miRNAs via indirect gene ontology analysis. This study improves our knowledge of the interactions between ncRNAs and mRNAs during trophoblast differentiation and identifies key biological processes that may be impaired in common gestational diseases, such as fetal growth restriction or preeclampsia.
Collapse
Affiliation(s)
- Tianjiao Chu
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jean-Francois Mouillet
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Zhishen Cao
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Oren Barak
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yingshi Ouyang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Yoel Sadovsky
- Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
19
|
Chen A, Yu R, Jiang S, Xia Y, Chen Y. Recent Advances of MicroRNAs, Long Non-coding RNAs, and Circular RNAs in Preeclampsia. Front Physiol 2021; 12:659638. [PMID: 33995125 PMCID: PMC8121253 DOI: 10.3389/fphys.2021.659638] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/12/2021] [Indexed: 12/26/2022] Open
Abstract
Preeclampsia is a clinical syndrome characterized by multiple-organ dysfunction, such as maternal hypertension and proteinuria, after 20 weeks of gestation. It is a common cause of fetal growth restriction, fetal malformation, and maternal death. At present, termination of pregnancy is the only way to prevent the development of the disease. Non-coding RNAs, including microRNAs, long non-coding RNAs, and circular RNAs, are involved in important pathological and physiological functions in life cycle activities including ontogeny, reproduction, apoptosis, and cell reprogramming, and are closely associated with human diseases. Accumulating evidence suggests that non-coding RNAs are involved in the pathogenesis of preeclampsia through regulation of various physiological functions. In this review, we discuss the current evidence of the pathogenesis of preeclampsia, introduce the types and biological functions of non-coding RNA, and summarize the roles of non-coding RNA in the pathophysiological development of preeclampsia from the perspectives of oxidative stress, hypoxia, angiogenesis, decidualization, trophoblast invasion and proliferation, immune regulation, and inflammation. Finally, we briefly discuss the potential clinical application and future prospects of non-coding RNA as a biomarker for the diagnosis of preeclampsia.
Collapse
Affiliation(s)
- Ailing Chen
- Translational Medicine Laboratory, Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Renqiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Shiwen Jiang
- Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China
| | - Ying Chen
- Translational Medicine Laboratory, Research Institute for Reproductive Health and Genetic Diseases, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
20
|
Kajiwara K, Beharier O, Chng CP, Goff JP, Ouyang Y, St Croix CM, Huang C, Kagan VE, Hsia KJ, Sadovsky Y. Ferroptosis induces membrane blebbing in placental trophoblasts. J Cell Sci 2021; 135:jcs.255737. [PMID: 33414166 DOI: 10.1242/jcs.255737] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
Ferroptosis is a regulated, non-apoptotic form of cell death, characterized by hydroxy-peroxidation of discrete phospholipid hydroperoxides, particularly hydroperoxyl (Hp) forms of arachidonoyl- and adrenoyl-phosphatidylethanolamine, with a downstream cascade of oxidative damage to membrane lipids, proteins and DNA, culminating in cell death. We recently showed that human trophoblasts are particularly sensitive to ferroptosis caused by depletion or inhibition of glutathione peroxidase 4 (GPX4) or the lipase PLA2G6. Here, we show that trophoblastic ferroptosis is accompanied by a dramatic change in the trophoblast plasma membrane, with macro-blebbing and vesiculation. Immunofluorescence revealed that ferroptotic cell-derived blebs stained positive for F-actin, but negative for cytoplasmic organelle markers. Transfer of conditioned medium that contained detached macrovesicles or co-culture of wild-type target cells with blebbing cells did not stimulate ferroptosis in target cells. Molecular modeling showed that the presence of Hp-phosphatidylethanolamine in the cell membrane promoted its cell ability to be stretched. Together, our data establish that membrane macro-blebbing is characteristic of trophoblast ferroptosis and can serve as a useful marker of this process. Whether or not these blebs are physiologically functional remains to be established.
Collapse
Affiliation(s)
- Kazuhiro Kajiwara
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Obstetrics and Gynecology, Jikei University School of Medicine, Tokyo, Japan105-8461
| | - Ofer Beharier
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Choon-Peng Chng
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Republic of Singapore
| | - Julie P Goff
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yingshi Ouyang
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Changjin Huang
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Republic of Singapore.,School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Republic of Singapore
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - K Jimmy Hsia
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Republic of Singapore.,School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore 637459, Republic of Singapore
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
21
|
Abel T, Moodley J, Naicker T. The Involvement of MicroRNAs in SARS-CoV-2 Infection Comorbid with HIV-Associated Preeclampsia. Curr Hypertens Rep 2021; 23:20. [PMID: 33847825 PMCID: PMC8042355 DOI: 10.1007/s11906-021-01138-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2021] [Indexed: 02/07/2023]
Abstract
Purpose of Review This review investigated the potential role of microRNAs (miRNAs) in the synergy of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, preeclampsia (PE), and human immunodeficiency virus (HIV) infection. Maternal health is a great concern when treating pregnant women fighting this triad of diseases, which is highly prevalent in South Africa. MicroRNAs are involved in fine-tuning of physiological processes. Disruptions to the balance of this minute protein can lead to various physiological changes that are sometimes pathological. Recent Findings MicroRNAs have recently been implicated in PE and have been linked to the anti-angiogenic imbalance evident in PE. Recent in silico studies have identified potential host miRNAs with anti-viral properties against SARS-CoV-2 infection. Studies have demonstrated dysregulated expression of several miRNAs in HIV-1 infection along with the ability of HIV-1 to downregulate anti-viral host microRNAs. Summary This review has highlighted the significant gap in literature on the potential of miRNAs in women with HIV-associated PE in synergy with the novel SARS-CoV-2 infection. In addition, this review has provided evidence of the critical role that the epigenetic regulatory mechanism of miRNA plays in viral infections and PE, thereby providing a foundation for further research investigating the potential of therapeutic miRNA development with fewer side-effects for pregnant women.
Collapse
Affiliation(s)
- Tashlen Abel
- Optics and Imaging Centre, Doris Duke Medical Research Institution, College of Health Sciences, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa.
| | - Jagidesa Moodley
- Women's Health and HIV Research Group, Department of Obstetrics & Gynaecology, School of Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institution, College of Health Sciences, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| |
Collapse
|
22
|
Colson A, Sonveaux P, Debiève F, Sferruzzi-Perri AN. Adaptations of the human placenta to hypoxia: opportunities for interventions in fetal growth restriction. Hum Reprod Update 2020; 27:531-569. [PMID: 33377492 DOI: 10.1093/humupd/dmaa053] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/15/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The placenta is the functional interface between the mother and the fetus during pregnancy, and a critical determinant of fetal growth and life-long health. In the first trimester, it develops under a low-oxygen environment, which is essential for the conceptus who has little defense against reactive oxygen species produced during oxidative metabolism. However, failure of invasive trophoblasts to sufficiently remodel uterine arteries toward dilated vessels by the end of the first trimester can lead to reduced/intermittent blood flow, persistent hypoxia and oxidative stress in the placenta with consequences for fetal growth. Fetal growth restriction (FGR) is observed in ∼10% of pregnancies and is frequently seen in association with other pregnancy complications, such as preeclampsia (PE). FGR is one of the main challenges for obstetricians and pediatricians, as smaller fetuses have greater perinatal risks of morbidity and mortality and postnatal risks of neurodevelopmental and cardio-metabolic disorders. OBJECTIVE AND RATIONALE The aim of this review was to examine the importance of placental responses to changing oxygen environments during abnormal pregnancy in terms of cellular, molecular and functional changes in order to highlight new therapeutic pathways, and to pinpoint approaches aimed at enhancing oxygen supply and/or mitigating oxidative stress in the placenta as a mean of optimizing fetal growth. SEARCH METHODS An extensive online search of peer-reviewed articles using PubMed was performed with combinations of search terms including pregnancy, placenta, trophoblast, oxygen, hypoxia, high altitude, FGR and PE (last updated in May 2020). OUTCOMES Trophoblast differentiation and placental establishment are governed by oxygen availability/hypoxia in early pregnancy. The placental response to late gestational hypoxia includes changes in syncytialization, mitochondrial functions, endoplasmic reticulum stress, hormone production, nutrient handling and angiogenic factor secretion. The nature of these changes depends on the extent of hypoxia, with some responses appearing adaptive and others appearing detrimental to the placental support of fetal growth. Emerging approaches that aim to increase placental oxygen supply and/or reduce the impacts of excessive oxidative stress are promising for their potential to prevent/treat FGR. WIDER IMPLICATIONS There are many risks and challenges of intervening during pregnancy that must be considered. The establishment of human trophoblast stem cell lines and organoids will allow further mechanistic studies of the effects of hypoxia and may lead to advanced screening of drugs for use in pregnancies complicated by placental insufficiency/hypoxia. Since no treatments are currently available, a better understanding of placental adaptations to hypoxia would help to develop therapies or repurpose drugs to optimize placental function and fetal growth, with life-long benefits to human health.
Collapse
Affiliation(s)
- Arthur Colson
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium
| | - Frédéric Debiève
- Pole of Obstetrics, Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, Brussels, Belgium.,Department of Obstetrics, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, UK
| |
Collapse
|
23
|
Krawczynski K, Ouyang Y, Mouillet JF, Chu T, Coyne CB, Sadovsky Y. Unc-13 homolog D mediates an antiviral effect of the chromosome 19 microRNA cluster miR-517a. J Cell Sci 2020; 134:jcs246769. [PMID: 33093239 PMCID: PMC7687871 DOI: 10.1242/jcs.246769] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/07/2020] [Indexed: 11/20/2022] Open
Abstract
The function of microRNAs (miRNAs) can be cell autonomous or communicated to other cell types and has been implicated in diverse biological processes. We previously demonstrated that miR-517a-3p (miR-517a), a highly expressed member of the chromosome 19 miRNA cluster (C19MC) that is transcribed almost exclusively in human trophoblasts, attenuates viral replication via induction of autophagy in non-trophoblastic recipient cells. However, the molecular mechanisms underlying these effects remain unknown. Here, we identified unc-13 homolog D (UNC13D) as a direct, autophagy-related gene target of miR-517a, leading to repression of UNC13D. In line with the antiviral activity of miR-517a, silencing UNC13D suppressed replication of vesicular stomatitis virus (VSV), whereas overexpression of UNC13D increased VSV levels, suggesting a role for UNC13D silencing in the antiviral activity of miR-517a. We also found that miR-517a activated NF-κB signaling in HEK-293XL cells expressing TLR8, but the effect was not specific to C19MC miRNA. Taken together, our results define mechanistic pathways that link C19MC miRNA with inhibition of viral replication.
Collapse
Affiliation(s)
- Kamil Krawczynski
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Department of Obstetrics and Gynecology and Reproductive Science, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yingshi Ouyang
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Department of Obstetrics and Gynecology and Reproductive Science, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jean-Francois Mouillet
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Department of Obstetrics and Gynecology and Reproductive Science, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tianjiao Chu
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Department of Obstetrics and Gynecology and Reproductive Science, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Carolyn B Coyne
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Center for Microbial Pathogenesis, Children's Hospital of Pittsburgh of the University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15224, USA
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Pittsburgh, PA 15213, USA
- Department of Obstetrics and Gynecology and Reproductive Science, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
24
|
Unveiling the ups and downs of miR-205 in physiology and cancer: transcriptional and post-transcriptional mechanisms. Cell Death Dis 2020; 11:980. [PMID: 33191398 PMCID: PMC7667162 DOI: 10.1038/s41419-020-03192-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022]
Abstract
miR-205 plays important roles in the physiology of epithelia by regulating a variety of pathways that govern differentiation and morphogenesis. Its aberrant expression is frequently found in human cancers, where it was reported to act either as tumor-suppressor or oncogene depending on the specific tumor context and target genes. miR-205 expression and function in different cell types or processes are the result of the complex balance among transcription, processing and stability of the microRNA. In this review, we summarize the principal mechanisms that regulate miR-205 expression at the transcriptional and post-transcriptional level, with particular focus on the transcriptional relationship with its host gene. Elucidating the mechanisms and factors regulating miR-205 expression in different biological contexts represents a fundamental step for a better understanding of the contribution of such pivotal microRNA to epithelial cell function in physiology and disease, and for the development of modulation strategies for future application in cancer therapy.
Collapse
|
25
|
Abstract
The recently identified ferroptotic cell death is characterized by excessive accumulation of hydroperoxy-arachidonoyl (C20:4)- or adrenoyl (C22:4)- phosphatidylethanolamine (Hp-PE). The selenium-dependent glutathione peroxidase 4 (GPX4) inhibits ferroptosis, converting unstable ferroptotic lipid hydroperoxides to nontoxic lipid alcohols in a tissue-specific manner. While placental oxidative stress and lipotoxicity are hallmarks of placental dysfunction, the possible role of ferroptosis in placental dysfunction is largely unknown. We found that spontaneous preterm birth is associated with ferroptosis and that inhibition of GPX4 causes ferroptotic injury in primary human trophoblasts and during mouse pregnancy. Importantly, we uncovered a role for the phospholipase PLA2G6 (PNPLA9, iPLA2beta), known to metabolize Hp-PE to lyso-PE and oxidized fatty acid, in mitigating ferroptosis induced by GPX4 inhibition in vitro or by hypoxia/reoxygenation injury in vivo. Together, we identified ferroptosis signaling in the human and mouse placenta, established a role for PLA2G6 in attenuating trophoblastic ferroptosis, and provided mechanistic insights into the ill-defined placental lipotoxicity that may inspire PLA2G6-targeted therapeutic strategies.
Collapse
|
26
|
Liu E, Zhou Y, Li J, Zhang D. MicroRNA‑491‑5p inhibits trophoblast cell migration and invasion through targeting matrix metalloproteinase‑9 in preeclampsia. Mol Med Rep 2020; 22:5033-5040. [PMID: 33174053 PMCID: PMC7646938 DOI: 10.3892/mmr.2020.11604] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/04/2020] [Indexed: 01/03/2023] Open
Abstract
Insufficient invasion of trophoblasts is correlated with the development of preeclampsia (PE). MicroRNA (miR)-491-5p has been reported to be implicated in human cancer cell invasion; however, whether miR-491-5p is involved in the development of PE remains largely unclear. The aim of the present study was to investigate the role of miR-491-5p in trophoblastic invasion in vitro and to determine its underlying mechanism of action. The expression levels of miR-491-5p were validated using reverse transcription-quantitative PCR. The effects of miR-491-5p on trophoblast cell invasion were evaluated in vitro. Then, the association between miR-491-5p and its downstream target was investigated in both cell lines and clinical specimens. miR-491-5p expression levels were observed to be significantly increased in the placental tissues from patients with PE. The invasive capacity of HTR-8/SVneo trophoblast cells was suppressed following the upregulation of miR-491-5p and increased following the inhibition of miR-491-5p. Matrix metalloproteinase-9 (MMP-9), a well-known regulator of trophoblast cell invasion, was discovered to be a direct target of miR-491-5p in HTR-8/SVneo trophoblast cells. Moreover, miR-491-5p expression levels were found to be inversely correlated with MMP-9 expression levels in placental tissues from patients with PE. The overexpression of MMP-9 partly attenuated the inhibitory effects of miR-491-5p on HTR-8/SVneo trophoblast cells invasion. Collectively, these findings suggested that the aberrant expression of miR-491-5p may contribute to PE through suppressing trophoblast invasion, thus highlighting the novel roles of miR-491-5p in the molecular pathogenesis of PE. The present study also showed that the miR-491-5p/MMP-9 axis may be an effective biomarker or a viable drug target for therapeutic intervention in PE.
Collapse
Affiliation(s)
- Enling Liu
- Department of Obstetrics and Gynecology, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Yuxiu Zhou
- Department of Immunity, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Jun Li
- Department of Obstetrics and Gynecology, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| | - Donghong Zhang
- Department of Obstetrics and Gynecology, Tangshan Gongren Hospital, Hebei Medical University, Tangshan, Hebei 063000, P.R. China
| |
Collapse
|
27
|
Lee J, Hong HK, Peng SB, Kim TW, Lee WY, Yun SH, Kim HC, Liu J, Ebert PJ, Aggarwal A, Jung S, Cho YB. Identifying metastasis-initiating miRNA-target regulations of colorectal cancer from expressional changes in primary tumors. Sci Rep 2020; 10:14919. [PMID: 32913235 PMCID: PMC7484763 DOI: 10.1038/s41598-020-71868-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 07/21/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is prevalent with high mortality, with liver metastasis contributing as a major factor that worsens the survival of patients. The roles of miRNAs in CRC have been elucidated, subsequent to recent studies that suggest the involvement of miRNAs in cancer biology. In this study, we compare the miRNA and gene expression profiles of primary tumors between two groups of patients (with and without liver metastasis) to identify the metastasis-initiating microRNA-target gene regulations. Analysis from 33 patients with metastasis and 14 patients without metastasis revealed that 17 miRNAs and their 198 predicted target genes are differentially expressed, where the target genes showed association with cancer progression and metastasis with statistical significance. In order to evaluate the clinical implications of the findings, we classified CRC patients of independent data into two groups based on the identified miRNA-target regulations, where one group was closer to primary tumors with metastasis than the other group. The comparison of survival showed statistically significant difference, thereby implying the roles of the identified miRNA-target regulations in cancer progression and metastasis. The identification of metastasis-initiating miRNA-target regulations in this study will lead to better understanding of the roles of miRNAs in CRC progression.
Collapse
Affiliation(s)
- Jongmin Lee
- Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Hye Kyung Hong
- Research Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | | | - Tae Won Kim
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Woo Yong Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.,Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Seong Hyun Yun
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hee Cheol Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | | | - Sungwon Jung
- Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon, Republic of Korea. .,Department of Genome Medicine and Science, Gachon University College of Medicine, Incheon, Republic of Korea.
| | - Yong Beom Cho
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea. .,Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
28
|
Li H, Pinilla-Macua I, Ouyang Y, Sadovsky E, Kajiwara K, Sorkin A, Sadovsky Y. Internalization of trophoblastic small extracellular vesicles and detection of their miRNA cargo in P-bodies. J Extracell Vesicles 2020; 9:1812261. [PMID: 32944196 PMCID: PMC7480505 DOI: 10.1080/20013078.2020.1812261] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pregnancy is a unique situation, in which placenta-derived small extracellular vesicles (sEVs) may communicate with maternal and foetal tissues. While relevant to homoeostatic and pathological functions, the mechanisms underlying sEV entry and cargo handling in target cells remain largely unknown. Using fluorescently or luminescently labelled sEVs, derived from primary human placental trophoblasts or from a placental cell line, we interrogated the endocytic pathways used by these sEVs to enter relevant target cells, including the neighbouring primary placental fibroblasts and human uterine microvascular endothelial cells. We found that trophoblastic sEVs can enter target cells, where they retain biological activity. Importantly, using a broad series of pharmacological inhibitors and siRNA-dependent silencing approaches, we showed that trophoblastic sEVs enter target cells using macropinocytosis and clathrin-mediated endocytosis pathways, but not caveolin-dependent endocytosis. Tracking their intracellular course, we localized the sEVs to early endosomes, late endosomes, and lysosomes. Finally, we used coimmunoprecipitation to demonstrate the association of the sEV microRNA (miRNA) with the P-body proteins AGO2 and GW182. Together, our data systematically detail endocytic pathways used by placental sEVs to enter relevant fibroblastic and endothelial target cells, and provide support for “endocytic escape” of sEV miRNA to P-bodies, a key site for cytoplasmic RNA regulation.
Collapse
Affiliation(s)
- Hui Li
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Reproductive Department of Xiangya Hospital, Central South University, Changsha, Hunan, China.,The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Itziar Pinilla-Macua
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yingshi Ouyang
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Elena Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kazuhiro Kajiwara
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alexander Sorkin
- Department of Cell Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
29
|
Liu LL, Qiao S, Wang ML, Wu HK, Su YX, Wang KM, Liu XW. MiR224-5p Inhibitor Restrains Neuronal Apoptosis by Targeting NR4A1 in the Oxygen-Glucose Deprivation (OGD) Model. Front Neurosci 2020; 14:613. [PMID: 32670010 PMCID: PMC7330102 DOI: 10.3389/fnins.2020.00613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/18/2020] [Indexed: 01/28/2023] Open
Abstract
This study was designed to investigate the molecular mechanism of stroke and to explore the effect of miR-224-5p in hypoxic cortical neurons. Firstly, we established a middle cerebral artery occlusion (MCAO) model with Sprague–Dawley rats. Triphenyltetrazolium chloride (TTC) staining showed the brain infarction of an MCAO rat. Longa scores of rats were significantly increased in 12th, 24th, and 48th hours after MCAO. Then, we found that miR-224-5p was increased after MCAO in rats by qRT-PCR. In order to investigate the effect of miR-224-5p in hypoxic neurons, we established an oxygen-glucose deprivation (OGD) model with cortical neurons. MiR-224-5p was also upregulated in neurons after OGD by qRT-PCR. After transfection of the miR-224-5p inhibitor, the number of neurons in the anti-miR-224-5p group significantly increased (P < 0.01) in comparison to the anti-NC group. Furthermore, Tuj1+ (neuronal marker) staining and TUNEL assay (to detect apoptotic cells) were performed in neurons. The survival of neurons in the anti-miR-224-5p group was significantly improved (P < 0.01), while the apoptosis of neurons in the anti-miR-224-5p group was significantly decreased (P < 0.01), when compared with that of the anti-NC group. In addition, we predicted that potential target genes of miR-224-5p were nuclear receptor subfamily 4 group A member 1 (NR4A1), interleukin 1 receptor antagonist (IL1RN), and ring finger protein 38 (RNF38) with bioinformatics databases, such as TargetScan, miRDB, miRmap, and miRanda. The result of qRT-PCR confirmed that NR4A1 was significantly decreased after hypoxic injury (P < 0.01). Meanwhile, luciferase reporter’s assay indicated that NR4A1 was the direct target of miR-224-5p. Compared with the anti-miR-224-5p + siNC group, the number of cortical neurons and the length of the neuron axon in the anti-miR-224-5p + si-NR4A1 group were significantly decreased (P < 0.01), and the number of neuronal apoptosis in the anti-miR-224-5p + si-NR4A1 group was increased (P < 0.01). In conclusion, miR-224-5p played a crucial role in hypoxic neuron injury through NR4A1, which might be an important regulatory mechanism in OGD injury of neurons.
Collapse
Affiliation(s)
- Ling-Ling Liu
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Neurology, Liaocheng People's Hospital, Liaocheng, China
| | - Shan Qiao
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated With Shandong First Medical University, Jinan, China
| | - Mei-Ling Wang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Huai-Kuan Wu
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yong-Xin Su
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ke-Mo Wang
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xue-Wu Liu
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
30
|
Tian J, Liu Y, Hu M, Zheng Y, Xu P, Zhang L, Liao J, Wu Y, Wen L, Tong C, Yan J, Qi H, Saffery R, Baker PN, Kilby MD. Upregulated LncZBTB39 in pre-eclampsia and its effects on trophoblast invasion and migration via antagonizing the inhibition of miR-210 on THSD7A expression. Eur J Obstet Gynecol Reprod Biol 2020; 248:164-171. [PMID: 32222649 DOI: 10.1016/j.ejogrb.2020.03.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 03/10/2020] [Accepted: 03/16/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Pre-eclampsia (PE) is a major cause of maternal morbidity and mortality, but its etiology remains to be elucidated. Accumulating evidence suggests that placental long noncoding RNAs (lncRNAs) might contribute to the pathogenesis of pre-eclampsia. STUDY DESIGN In the present study, the expression levels of lncRNAs in human placenta were first determined by microarray analysis and then validated by secondary RT-qPCR and FISH. LncZBTB39 expression manipulation in HTR8/SVneo trophoblast cells was achieved by shRNA and plasmid transfection. Then, the invasion and migration of lncZBTB39-deficient and lncZBTB39-overexpressing trophoblast cells were evaluated by transwell assays and wound-healing assays, respectively. MMP2 activity was measured by gelatin zymography. The downstream target genes of lncZBTB39 were then identified by a transcriptomic microarray, followed by RT-qPCR validation. RESULTS We found that lncZBTB39 was upregulated in PE-complicated human placentas, and overexpression of lncZBTB39 inhibited invasion and migration, as well as MMP2 activity in HTR8/SVneo cells, while downregulation of lncZBTB39 enhanced invasion, migration and MMP2 activity. In addition, THSD7A expression was elevated by lncZBTB39 overexpression but reduced in lncZBTB39-deficient cells; moreover, lncZBTB39 antagonized the inhibitory effects of miR-210 on THSD7A expression. CONCLUSION PE-complicated placentas are associated with upregulated lncZBTB39, which negatively regulates trophoblast invasion and migration, most likely by preserving the expression of THSD7A mRNA through sponging miR-210. The results of this study not only provide novel evidence that lncRNAs regulate trophoblastic activities but also suggest that lncZBTB39 may be a potential interventional target for PE.
Collapse
Affiliation(s)
- Jing Tian
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing 400016, China; Department of Obstetrics and Gynecology, University-Town Hospital of Chongqing Medical University, Chongqing 401331, China
| | - Yamin Liu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing 400016, China; Department of Obstetrics, Chongqing Health Center for Women and Children, Chongqing 401147, China
| | - Mingyu Hu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing 400016, China
| | - Yangxi Zheng
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing 400016, China
| | - Ping Xu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing 400016, China
| | - Lan Zhang
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing 400016, China
| | - Jiujiang Liao
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing 400016, China
| | - Yue Wu
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing 400016, China
| | - Li Wen
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing 400016, China
| | - Chao Tong
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing 400016, China.
| | - Jianying Yan
- Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350001, China.
| | - Hongbo Qi
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; International Collaborative Laboratory of Reproduction and Development, Ministry of Education, Chongqing 400016, China; State Key Laboratory of Maternal and Fetal Medicine of Chongqing Municipality, Chongqing 400016, China.
| | - Richard Saffery
- Department of Pediatrics, University of Melbourne, Parkville, VIC 3052, Australia
| | - Philip N Baker
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; College of Life Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Mark D Kilby
- Fetal Medicine Centre, Birmingham Women's & Children's Foundation Trust, Birmingham, B15 2TG, UK; Institute of Metabolism & Systems Research, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
31
|
Preeclampsia: The Interplay Between Oxygen-Sensitive miRNAs and Erythropoietin. J Clin Med 2020; 9:jcm9020574. [PMID: 32093169 PMCID: PMC7073952 DOI: 10.3390/jcm9020574] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 12/21/2022] Open
Abstract
Changes in the oxygen partial pressure caused by a violation of uteroplacental perfusion are considered a powerful inducer of a cascade of reactions leading to the clinical manifestation of preeclampsia (PE). At the same time, the induction of oxygen-dependent molecule expression, in particular, miRNA and erythropoietin, is modulated. Therefore, the focus of our study was aimed at estimating the miRNA expression profile of placental tissue and blood plasma in pregnant women with preeclampsia using deep sequencing and quantitative RT-PCR, as well as determining the concentration of erythropoietin. The expression of miR-27b-3p, miR-92b-3p, miR-125b-5p, miR-181a-5p, and miR-186-5p, as regulated by hypoxia/reoxygenation, was significantly increased in blood plasma during early-onset preeclampsia. The possibility of detecting early PE according to the logistic regression model (miR-92b-3p, miR-125b-5p, and miR-181a-5p (AUC = 0.91)) was evaluated. Furthermore, the erythropoietin level, which is regulated by miR-125b-5p, was significantly increased. According to PANTHER14.1, the participation of these miRNAs in the regulation of pathways, such as the hypoxia’s response via HIF activation, oxidative stress response, angiogenesis, and the VEGF signaling pathway, were determined.
Collapse
|
32
|
Femia MR, Evans RM, Zhang J, Sun X, Lebegue CJ, Roggero VR, Allison LA. Mediator subunit MED1 modulates intranuclear dynamics of the thyroid hormone receptor. J Cell Biochem 2019; 121:2909-2926. [PMID: 31692077 DOI: 10.1002/jcb.29532] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 10/10/2019] [Indexed: 12/26/2022]
Abstract
The thyroid hormone receptors (TRs) mediate thyroid hormone (T3 )-dependent gene expression. The nuclear import and export signals that direct TR shuttling are well characterized, but little is known about factors modulating nuclear retention. We used fluorescence-based nucleocytoplasmic scoring and fluorescence recovery after photobleaching in transfected cells to investigate whether Mediator subunits MED1 and MED13 play a role in nuclear retention of TR. When MED1 was overexpressed, there was a striking shift towards a greater nuclear localization of TRβ1 and the oncoprotein v-ErbA, subtypes with cytosolic populations at steady-state, and TRβ1 intranuclear mobility was reduced. For TRα1, there was no observable change in its predominantly nuclear distribution pattern or mobility. Consistent with a role for MED1 in nuclear retention, the cytosolic TRα1 and TRβ1 population were significantly greater in MED1-/- cells, compared with MED1+/+ cells. Exposure to T3 and epidermal growth factor, which induces MED1 phosphorylation, also altered TR intranuclear dynamics. Overexpression of miR-208a, which downregulates MED13, led to a more cytosolic distribution of nuclear-localized TRα1; however, overexpression of MED13 had no effect on TRβ1 localization. The known binding site of MED1 overlaps with a transactivation domain and nuclear export signal in helix 12 of TR's ligand-binding domain (LBD). Coimmunoprecipitation assays demonstrated that TR's LBD interacts directly with exportins 5 and 7, suggesting that binding of exportins and MED1 to TR may be mutually exclusive. Collectively, our data provide evidence that MED1 promotes nuclear retention of TR, and highlight the dual functionality of helix 12 in TR transactivation and nuclear export.
Collapse
Affiliation(s)
- Matthew R Femia
- Department of Biology, William and Mary, Williamsburg, Viginia
| | | | - Jibo Zhang
- Department of Biology, William and Mary, Williamsburg, Viginia
| | - Xiaopeng Sun
- Department of Biology, William and Mary, Williamsburg, Viginia
| | | | | | | |
Collapse
|
33
|
Hemmatzadeh M, Shomali N, Yousefzadeh Y, Mohammadi H, Ghasemzadeh A, Yousefi M. MicroRNAs: Small molecules with a large impact on pre-eclampsia. J Cell Physiol 2019; 235:3235-3248. [PMID: 31595979 DOI: 10.1002/jcp.29286] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 09/03/2019] [Indexed: 12/22/2022]
Abstract
As critical mediators in biological processes, microRNAs (miRNAs) which are small and endogenous noncoding RNAs have been associated with disease progression, cell proliferation, and development. Pre-eclampsia (PE), a pregnancy-related disorder with no early markers or symptoms is recognized as the main reason for fetal and maternal mortality and morbidity in the initial steps or even during pregnancy, worldwide. Clinical symptoms usually appear in the third trimester of the pregnancy. Although numerous research have unraveled several aspects of placenta development abnormalities associated with abnormal trophoblastic invasion and angiogenesis modification, many questions about the PE pathogenesis remains unanswered. A large number of studies have shown the important role of miRNAs as potential biomarkers in the PE prognosis and diagnosis. Here, the latest investigations about the PE and placental miRNAs expression, as well as, the crucial role of miRNA molecules including miR-210 and miR-155 which are deregulated in patients with PE, will be argued.
Collapse
Affiliation(s)
- Maryam Hemmatzadeh
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Navid Shomali
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Yousefzadeh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.,Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Aliyeh Ghasemzadeh
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
34
|
The untapped potential of placenta-enriched molecules for diagnostic and therapeutic development. Placenta 2019; 84:28-31. [PMID: 30745114 DOI: 10.1016/j.placenta.2019.02.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/19/2019] [Accepted: 02/02/2019] [Indexed: 12/27/2022]
Abstract
Pregnancy complications such as fetal growth restriction and preeclampsia are diseases with limited biomarkers for prediction, and a complete lack of therapeutic options. We define placenta-enriched molecules as those that are highly expressed in the placenta relative to all other human tissues. Many exist including mRNAs, miRNAs and proteins. It is now well established that placenta-enriched mRNAs are found within the maternal circulation and are cleared rapidly after birth. Similarly, distinct clusters of miRNAs that are placenta-enriched have been identified and are measurable within the circulation. However, perhaps the most established potential diagnostics thus far are circulating placental proteins such as placental growth factor (PlGF), pregnancy associated pregnancy protein-A (PAPP-A) and soluble FMS-like tyrosine kinase 1 (sFlt-1). There has also been much interest in targeting placenta-enriched molecules as a means to treat diseases of pregnancy. We have shown promising results in targeting placenta-enriched epidermal growth factor receptor (EGFR) to treat ectopic pregnancy. Others have focused on using placenta-enriched molecules as a means of homing therapeutic-filled nanoparticles to the placenta, or to directly target sFlt-1 to improve disease outcomes. Importantly, many placenta-enriched molecules remain largely unstudied. We propose that a better understanding of their biology, and potential contribution to the pathogenesis of diseases, may yield more predictive diagnostic and therapeutic targets.
Collapse
|
35
|
Skalis G, Katsi V, Miliou A, Georgiopoulos G, Papazachou O, Vamvakou G, Nihoyannopoulos P, Tousoulis D, Makris T. MicroRNAs in Preeclampsia. Microrna 2019; 8:28-35. [PMID: 30101723 DOI: 10.2174/2211536607666180813123303] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 05/22/2018] [Accepted: 07/27/2018] [Indexed: 11/22/2022]
Abstract
Preeclampsia (PE) continues to represent a worldwide problem and challenge for both clinicians and laboratory-based doctors. Despite many efforts, the knowledge acquired regarding its pathogenesis and pathophysiology does not allow us to treat it efficiently. It is not possible to arrest its progressive nature, and the available therapies are limited to symptomatic treatment. Furthermore, both the diagnosis and prognosis are frequently uncertain, whilst the ability to predict its occurrence is very limited. MicroRNAs are small non-coding RNAs discovered two decades ago, and present great interest given their ability to regulate almost every aspect of the cell function. A lot of evidence regarding the role of miRNAs in pre-eclampsia has been accumulated in the last 10 years. Differentially expressed miRNAs are characteristic of both mild and severe PE. In many cases they target signaling pathway-related genes that result in altered processes which are directly involved in PE. Immune system, angiogenesis and trophoblast proliferation and invasion, all fundamental aspects of placentation, are controlled in various degrees by miRNAs which are up- or downregulated. Finally, miRNAs represent a potential therapeutic target and a diagnostic tool.
Collapse
Affiliation(s)
- Georgios Skalis
- Department of Cardiology, Helena Venizelou Hospital, Athens, Greece
| | - Vasiliki Katsi
- Cardiology Department, Hippokration Hospital, National Health System, Athens, Greece
| | - Antigoni Miliou
- 1st Department of Cardiology, Hippokration Hospital, National & Kapodistrian University of Athens, Athens, Greece
| | | | | | - Georgia Vamvakou
- Department of Cardiology, Helena Venizelou Hospital, Athens, Greece
| | - Petros Nihoyannopoulos
- 1st Department of Cardiology, Hippokration Hospital, National & Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Tousoulis
- 1st Department of Cardiology, Hippokration Hospital, National & Kapodistrian University of Athens, Athens, Greece
| | - Thomas Makris
- Department of Cardiology, Helena Venizelou Hospital, Athens, Greece
| |
Collapse
|
36
|
Bildirici I, Schaiff WT, Chen B, Morizane M, Oh SY, O’Brien M, Sonnenberg-Hirche C, Chu T, Barak Y, Nelson DM, Sadovsky Y. PLIN2 Is Essential for Trophoblastic Lipid Droplet Accumulation and Cell Survival During Hypoxia. Endocrinology 2018; 159:3937-3949. [PMID: 30351430 PMCID: PMC6240902 DOI: 10.1210/en.2018-00752] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
Trophoblast hypoxia and injury, key components of placental dysfunction, are associated with fetal growth restriction and other complications of pregnancy. Accumulation of lipid droplets has been found in hypoxic nonplacental cells. Unique to pregnancy, lipid accumulation in the placenta might perturb lipid transport to the fetus. We tested the hypothesis that hypoxia leads to accumulation of lipid droplets in human trophoblasts and that trophoblastic PLIN proteins play a key role in this process. We found that hypoxia promotes the accumulation of lipid droplets in primary human trophoblasts. A similar accretion of lipid droplets was found in placental villi in vivo from pregnancies complicated by fetal growth restriction. In both situations, these changes were associated with an increased level of cellular triglycerides. Exposure of trophoblasts to hypoxia led to reduced fatty acid efflux and oxidation with no change in fatty acid uptake or synthesis. We further found that hypoxia markedly stimulated PLIN2 mRNA synthesis and protein expression, which colocalized to lipid droplets. Knockdown of PLIN2, but not PLIN3, enhanced trophoblast apoptotic death, and overexpression of PLIN2 promoted cell viability. Collectively, our data indicate that hypoxia enhances trophoblastic lipid retention in the form of lipid droplets and that PLIN2 plays a key role in this process and in trophoblast defense against apoptotic death. These findings also imply that this protective mechanism may lead to diminished trafficking of lipids to the developing fetus.
Collapse
Affiliation(s)
- Ibrahim Bildirici
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - W Timothy Schaiff
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Baosheng Chen
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Mayumi Morizane
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Soo-Young Oh
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew O’Brien
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Tianjiao Chu
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yaacov Barak
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - D Michael Nelson
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
- Correspondence: Yoel Sadovsky, MD, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, Pennsylvania 15213. E-mail:
| |
Collapse
|
37
|
Gunel T, Kamali N, Hosseini MK, Gumusoglu E, Benian A, Aydinli K. Regulatory effect of miR-195 in the placental dysfunction of preeclampsia. J Matern Fetal Neonatal Med 2018; 33:901-908. [PMID: 30078346 DOI: 10.1080/14767058.2018.1508439] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose: Preeclampsia (PE) is a pregnancy specific disease soon after 20 weeks of gestation where major symptoms are hypertension and proteinuria. The underlying pathology is believed to be abnormal placentation. Epigenetic and genetic factors have significant roles in abnormal placental development. MicroRNA's (miRNAs), being one of the most important epigenetic regulators, take part in abnormal placentation. Hsa-miR-195 is a molecule associated with abnormal placental growth mechanisms such as impaired cellular proliferation, inadequate trophoblastic invasion causing defective spiral artery remodeling, and apoptosis. We aimed to evaluate miRNA functions, namely miR-195 expression profile, in order to divulge PE pathogenesis.Methods: In this study, we extracted circulating miRNAs from maternal plasma and placenta from 20 PE patients and 20 normotensive pregnant women. miR-195 was quantified using quantitative real time reverse transcriptase PCR (qRT-PCR). The target genes of miR-195 were predicted by Diana Tools-mirPath, TargetScan, and miRDB databases.Results: We found that miR-195 levels were downregulated (3.83-fold decrease, p < .05) in preeclamptic placenta samples, however miR-195 were undetected in preeclamptic and normotensive plasma samples. The steep down-regulation of miR-195 points to its importance of PE pathogenesis.Conclusion: miR-195 is suggested to regulate PE via its target genes manipulating biological processes such as placental proliferation, apoptosis, and angiogenesis. We propose that detection of decreased miR-195 levels in preeclamptic placentas could be used to enlighten the pathophysiology of PE.
Collapse
Affiliation(s)
- Tuba Gunel
- Faculty of Science, Department of Molecular Biology and Genetics, Istanbul University, Istanbul, Turkey
| | - Nilufer Kamali
- Faculty of Science, Department of Molecular Biology and Genetics, Istanbul University, Istanbul, Turkey
| | - Mohammad K Hosseini
- Faculty of Science, Department of Molecular Biology and Genetics, Istanbul University, Istanbul, Turkey
| | - Ece Gumusoglu
- Faculty of Science, Department of Molecular Biology and Genetics, Istanbul University, Istanbul, Turkey
| | - Ali Benian
- Cerrahpasa Medical Faculty, Department of Obstetrics and Gynecology, Istanbul University, Istanbul, Turkey
| | | |
Collapse
|
38
|
Nagpal N, Sharma S, Maji S, Durante G, Ferracin M, Thakur JK, Kulshreshtha R. Essential role of MED1 in the transcriptional regulation of ER-dependent oncogenic miRNAs in breast cancer. Sci Rep 2018; 8:11805. [PMID: 30087366 PMCID: PMC6081450 DOI: 10.1038/s41598-018-29546-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 07/12/2018] [Indexed: 01/24/2023] Open
Abstract
Mediator complex has been extensively shown to regulate the levels of several protein-coding genes; however, its role in the regulation of miRNAs in humans remains unstudied so far. Here we show that MED1, a Mediator subunit in the Middle module of Mediator complex, is overexpressed in breast cancer and is a negative prognostic factor. The levels of several miRNAs (miR-100-5p, -191-5p, -193b-3p, -205-5p, -326, -422a and -425-5p) were found to be regulated by MED1. MED1 induces miR-191/425 cluster in an estrogen receptor-alpha (ER-α) dependent manner. Occupancy of MED1 on estrogen response elements (EREs) upstream of miR-191/425 cluster is estrogen and ER-α-dependent and ER-α-induced expression of these miRNAs is MED1-dependent. MED1 mediates induction of cell proliferation and migration and the genes associated with it (JUN, FOS, EGFR, VEGF, MMP1, and ERBB4) in breast cancer, which is abrogated when used together with miR-191-inhibition. Additionally, we show that MED1 also regulates the levels of direct miR-191 target genes such as SATB1, CDK6 and BDNF. Overall, the results show that MED1/ER-α/miR-191 axis promotes breast cancer cell proliferation and migration and may serve as a novel target for therapy.
Collapse
Affiliation(s)
- Neha Nagpal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India.,Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shivani Sharma
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Sourobh Maji
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Giorgio Durante
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126, Bologna, Italy
| | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126, Bologna, Italy
| | - Jitendra K Thakur
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India.
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India.
| |
Collapse
|
39
|
Wang Y, Liu HZ, Liu Y, Wang HJ, Pang WW, Zhang JJ. Downregulated MALAT1 relates to recurrent pregnancy loss via sponging miRNAs. Kaohsiung J Med Sci 2018; 34:503-510. [PMID: 30173780 DOI: 10.1016/j.kjms.2018.04.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 03/14/2018] [Accepted: 04/24/2018] [Indexed: 01/26/2023] Open
Abstract
Recurrent pregnancy loss (RPL) is three or more times of consecutive spontaneous loss of pregnancy. The underlying cause is complicated and the etiology of over 50% of RPL patients is unclear. Metastasis associated lung adenocarcinoma transcript-1 (MALAT-1), a multiple lncRNA functions as key regulators of diverse cellular processes. In this study, we found a reduced MALAT1 level in the villus samples of 36 RPL patients. Predicted by bioinformatics tool and confirmed by dual luciferase assay, we identified that MALAT1 directly interacts with miRNAs. Subsequent functional study in HTR-8/SVneo and HUVEC cells indicated that MALAT1 modulates the cell proliferation, apoptosis, migration and invasion via directly interact with miR-383, miR-15, miR-205 and miR-375. By modulating the VEGFA expression, MALAT1 controls the capillary formation of HUVEC cells. In conclusion, MALAT1 as a functional lncRNA controls cell proliferation, apoptosis, migration, invasion and modulates blood vessel formation. Down regulated MALAT1 induced disordered cross-talk between embryo and mother is one of the factor contributes to the pathogenesis of RPL.
Collapse
Affiliation(s)
- Yan Wang
- Department of Obstetrics, Affiliated Hospital of Weifang Medical University, China
| | - Hui-Ze Liu
- Department of Obstetrics, Affiliated Hospital of Weifang Medical University, China
| | - Yang Liu
- Department of Obstetrics, Affiliated Hospital of Weifang Medical University, China
| | - Hui-Juan Wang
- Department of Obstetrics, Affiliated Hospital of Weifang Medical University, China
| | - Wen-Wen Pang
- Department of Obstetrics, Affiliated Hospital of Weifang Medical University, China
| | - Jian-Jun Zhang
- Department of Obstetrics, Affiliated Hospital of Weifang Medical University, China.
| |
Collapse
|
40
|
Vidal DO, Ramão A, Pinheiro DG, Muys BR, Lorenzi JCC, de Pádua Alves C, Zanette DL, de Molfetta GA, Duarte G, Silva WA. Highly expressed placental miRNAs control key biological processes in human cancer cell lines. Oncotarget 2018; 9:23554-23563. [PMID: 29805755 PMCID: PMC5955126 DOI: 10.18632/oncotarget.25264] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 03/24/2018] [Indexed: 12/31/2022] Open
Abstract
Despite being a healthy tissue, the constituent cells of the placenta, share similar characteristics with tumor cells, such as increased cell growth, migration, and invasion. However, while these processes are stochastic and uncontrolled in cancer cells, in placenta they are precisely controlled. Since miRNAs have been reported to regulate genes that control the molecular mechanisms necessary for the development of both human placenta and cancer, we addressed for miRNAs highly expressed in the placenta that could be involved in tumorigenesis. Here, we assessed the miRNA profile in placenta samples using microarray analysis. The results showed that miR-451 and miR-720, highly expressed placental miRNAs, presented very low or undetectable expression in cancer cell lines compared to the normal placenta and healthy tissues. Additionally, transfection of miR-451 or miR-720 mimics in choriocarcinoma cell line (JEG3) and colorectal adenocarcinoma cell line (HT-29) resulted in impaired cell proliferation, decreased cell migration and invasion and reduced ability of colony formation. These findings provide evidence that placenta may work as an alternative model to identify novel miRNAs involved in pathways controlling tumorigenesis.
Collapse
Affiliation(s)
- Daniel Onofre Vidal
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, SP, Brazil
| | - Anelisa Ramão
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Center for Cell-Based Therapy (CEPID/FAPESP), National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Riberão Preto, SP, Brazil
| | - Daniel Guariz Pinheiro
- Center for Cell-Based Therapy (CEPID/FAPESP), National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Riberão Preto, SP, Brazil
| | - Bruna Rodrigues Muys
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Center for Cell-Based Therapy (CEPID/FAPESP), National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Riberão Preto, SP, Brazil
| | - Julio Cesar Cetrulo Lorenzi
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Center for Cell-Based Therapy (CEPID/FAPESP), National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Riberão Preto, SP, Brazil
| | - Cleidson de Pádua Alves
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Center for Cell-Based Therapy (CEPID/FAPESP), National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Riberão Preto, SP, Brazil
| | - Dalila Luciola Zanette
- Center for Cell-Based Therapy (CEPID/FAPESP), National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Riberão Preto, SP, Brazil
| | - Greice Andreotti de Molfetta
- Center for Cell-Based Therapy (CEPID/FAPESP), National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Riberão Preto, SP, Brazil
| | - Geraldo Duarte
- Department of Gynecology and Obstetrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Wilson Araújo Silva
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Center for Cell-Based Therapy (CEPID/FAPESP), National Institute of Science and Technology in Stem Cell and Cell Therapy (INCTC/CNPq), Riberão Preto, SP, Brazil.,Center for Medical Genomics (HCFMRP/USP), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil.,Center for Integrative Systems Biology (CISBi-NAP/USP), Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
41
|
Wang N, Feng Y, Xu J, Zou J, Chen M, He Y, Liu H, Xue M, Gu Y. miR-362-3p regulates cell proliferation, migration and invasion of trophoblastic cells under hypoxia through targeting Pax3. Biomed Pharmacother 2018; 99:462-468. [PMID: 29665647 DOI: 10.1016/j.biopha.2018.01.089] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/08/2018] [Accepted: 01/16/2018] [Indexed: 12/21/2022] Open
Abstract
Preeclampsia (PE), a common obstetrical disorder, is one of the leading causes of pregnancy associated death. PE is closely linked with impaired migration and invasion ability of trophoblastic cells. miR-362-3p recently received our particular attention due not only to its aberrant expression in the placentas of patients with PE, but also to its important roles in regulating migration and invasion of various cells. This study was thus conducted to investigate the roles of miR-362-3p in PE and the related mechanism. The expression of miR-362-3p and Pax3 was examined in placentas of patients with PE and in normal placentas. HTR8/SVneo cells were cultured under hypoxia and transfected with miR-362-3p mimics, miR-362-3p inhibitors or Pax3 over-expression vectors. Results showed up-regulation of miR-362-3p but down-regulation of Pax3 in placentas of preeclamptic pregnancies. Luciferase report assay confirmed that Pax3 is a direct target of miR-362-3p. Although Pax3 was predicted to be targeted by miR-30a-3p and miR-181a-5p as well, their expression either had no difference between placentas of PE patients and normal placentas or showed less increment in placentas of PE patients than miR-362-3p. Exposure to hypoxia inhibited cell viability, migration and invasion of HTR8/SVneo cells. Increasing miR-362-3p by the mimics conferred improved effects on the inhibition. However, deletion of miR-362-3p or overexpression of Pax3 abolished the inhibiton. These results suggest that miR-362-3p/Pax3 axis regulates cell viability, migration and invasion of HTR8/SVneo cells under hypoxia. The present study adds to the further understanding of the pathogenesis of PE.
Collapse
Affiliation(s)
- Nan Wang
- Department of Obstetrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Yaling Feng
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, 214002, PR China.
| | - Jianjuan Xu
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, 214002, PR China
| | - Jinfang Zou
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, 214002, PR China
| | - Minghua Chen
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, 214002, PR China
| | - Yue He
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, 214002, PR China
| | - Huan Liu
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, 214002, PR China
| | - Min Xue
- Department of Obstetrics, The Third Xiangya Hospital of Central South University, Changsha, Hunan, 410013, PR China
| | - Yanfang Gu
- Department of Obstetrics and Gynecology, Wuxi Matemal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, 214002, PR China.
| |
Collapse
|
42
|
Bayer A, Lennemann NJ, Ouyang Y, Sadovsky E, Sheridan MA, Roberts RM, Coyne CB, Sadovsky Y. Chromosome 19 microRNAs exert antiviral activity independent from type III interferon signaling. Placenta 2017; 61:33-38. [PMID: 29277269 DOI: 10.1016/j.placenta.2017.11.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/18/2017] [Accepted: 11/08/2017] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Cultured primary human trophoblasts (PHT), derived from term placentas, are relatively resistant to infection by diverse viruses. The resistance can be conferred to non-trophoblastic cells by pre-exposing them to medium that was conditioned by PHT cells. This antiviral effect is mediated, at least in part, by microRNAs (miRNA) expressed from the chromosome 19 microRNA cluster (C19MC). Recently we showed that PHT cells and cells pre-exposed to PHT medium are also resistant to infection by Zika virus (ZIKV), an effect mediated by the constitutive release of the type III interferons (IFN) IFN lambda-1 and IFN lambda-2 in trophoblastic medium. We hypothesized that trophoblastic C19MC miRNA are active against ZIKV, and assessed the interaction of this pathway with IFN lambda-1 - mediated resistance. METHODS Term PHT cells were cultured using standard techniques. An osteosarcoma cell line (U2OS) was used as non-trophoblastic cells, which were infected with either ZIKV or vesicular stomatitis virus (VSV). Trophoblastic extracellular vesicles (EVs) were produced by gradient ultracentrifugation. RT-qPCR was used to determine viral infection, cellular or medium miRNA levels and the expression of interferon-stimulated genes. RESULTS We showed that C19MC miRNA attenuate infection of U2OS cells by ZIKV, and that C19MC miRNA or exosomes that contain C19MC miRNA did not influence the type III IFN pathway. Similarly, cell exposure to recombinant IFN lambda-1 had no effect on miRNA expression, and these pathways did not exhibit synergistic interaction. DISCUSSION PHT cells exert antiviral activity by at least two independent mechanisms, mediated by C19MC miRNA and by type III IFNs.
Collapse
Affiliation(s)
- Avraham Bayer
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Obstetrics and Gynecology, and Reproductive Science, University of Pittsburgh, PA 15213, USA
| | | | - Yingshi Ouyang
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Obstetrics and Gynecology, and Reproductive Science, University of Pittsburgh, PA 15213, USA
| | - Elena Sadovsky
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Megan A Sheridan
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA; Department of Biochemistry and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - R Michael Roberts
- Division of Animal Sciences and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA; Department of Biochemistry and Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, USA
| | - Carolyn B Coyne
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Obstetrics and Gynecology, and Reproductive Science, University of Pittsburgh, PA 15213, USA; Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15219, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Yoel Sadovsky
- Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Obstetrics and Gynecology, and Reproductive Science, University of Pittsburgh, PA 15213, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
43
|
Abstract
Epidemiological evidence links an individual's susceptibility to chronic disease in adult life to events during their intrauterine phase of development. Biologically this should not be unexpected, for organ systems are at their most plastic when progenitor cells are proliferating and differentiating. Influences operating at this time can permanently affect their structure and functional capacity, and the activity of enzyme systems and endocrine axes. It is now appreciated that such effects lay the foundations for a diverse array of diseases that become manifest many years later, often in response to secondary environmental stressors. Fetal development is underpinned by the placenta, the organ that forms the interface between the fetus and its mother. All nutrients and oxygen reaching the fetus must pass through this organ. The placenta also has major endocrine functions, orchestrating maternal adaptations to pregnancy and mobilizing resources for fetal use. In addition, it acts as a selective barrier, creating a protective milieu by minimizing exposure of the fetus to maternal hormones, such as glucocorticoids, xenobiotics, pathogens, and parasites. The placenta shows a remarkable capacity to adapt to adverse environmental cues and lessen their impact on the fetus. However, if placental function is impaired, or its capacity to adapt is exceeded, then fetal development may be compromised. Here, we explore the complex relationships between the placental phenotype and developmental programming of chronic disease in the offspring. Ensuring optimal placentation offers a new approach to the prevention of disorders such as cardiovascular disease, diabetes, and obesity, which are reaching epidemic proportions.
Collapse
Affiliation(s)
- Graham J Burton
- Centre for Trophoblast Research and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; and Department of Medicine, Knight Cardiovascular Institute, and Moore Institute for Nutrition and Wellness, Oregon Health and Science University, Portland, Oregon
| | - Abigail L Fowden
- Centre for Trophoblast Research and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; and Department of Medicine, Knight Cardiovascular Institute, and Moore Institute for Nutrition and Wellness, Oregon Health and Science University, Portland, Oregon
| | - Kent L Thornburg
- Centre for Trophoblast Research and Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom; and Department of Medicine, Knight Cardiovascular Institute, and Moore Institute for Nutrition and Wellness, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
44
|
Jiang M, Zhong T, Zhang W, Xiao Z, Hu G, Zhou H, Kuang H. Reduced expression of miR-205-5p promotes apoptosis and inhibits proliferation and invasion in lung cancer A549 cells by upregulation of ZEB2 and downregulation of erbB3. Mol Med Rep 2017; 15:3231-3238. [DOI: 10.3892/mmr.2017.6398] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 02/09/2017] [Indexed: 11/06/2022] Open
|
45
|
Schanzenbach CI, Kirchner B, Ulbrich SE, Pfaffl MW. Can milk cell or skim milk miRNAs be used as biomarkers for early pregnancy detection in cattle? PLoS One 2017; 12:e0172220. [PMID: 28234939 PMCID: PMC5325256 DOI: 10.1371/journal.pone.0172220] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 02/01/2017] [Indexed: 01/07/2023] Open
Abstract
The most critical phase of pregnancy is the first three weeks following insemination. During this period about 50% of high yielding lactating cows suffer embryonic loss prior to implantation, which poses a high economic burden on dairy farmers. Early diagnosis of pregnancy in cattle is therefore essential for monitoring breeding outcome and efficient production intervals. Regulated microRNAs (miRNAs) that reach easily accessible body fluids via a ‘liquid biopsy’ could be a new class of pregnancy predicting biomarkers. As milk is obtained regularly twice daily and non-invasively from the animal, it represents an ideal sample material. Our aim was to establish a pregnancy test system based on the discovery of small RNA biomarkers derived from the bovine milk cellular fraction and skim milk of cows. Milk samples were taken on days 4, 12 and 18 of cyclic cows and after artificial insemination, respectively, of the same animals (n = 6). miRNAs were analysed using small RNA sequencing (small RNA Seq). The miRNA profiles of milk cells and skim milk displayed similar profiles despite the presence of immune cell related miRNAs in milk cells. Trends in regulation of miRNAs between the oestrous cycle and pregnancy were found in miR-cluster 25~106b and its paralog cluster 17~92, miR-125 family, miR-200 family, miR-29 family, miR-15a, miR-21, miR-26b, miR-100, miR-140, 193a-5p, miR-221, miR-223, miR-320a, miR-652, miR-2898 and let-7i. A separation of cyclic and pregnant animals was achieved in a principal component analysis. Bta-miRs-29b, -221, -125b and -200b were successfully technically validated using quantitative real-time PCR, however biological validation failed. Therefore we cannot recommend the diagnostic use of these miRNAs in milk as biomarkers for detection of bovine pregnancy for now.
Collapse
Affiliation(s)
- Corina I. Schanzenbach
- Animal Physiology and Immunology, Department of Animal Sciences, Wissenschaftszentrum Weihenstephan, Technische Universität München, Freising, Germany
- Animal Physiology, Institute of Agricultural Science, Department of Environmental Systems Science, ETH Zürich, Zurich, Switzerland
- * E-mail:
| | - Benedikt Kirchner
- Animal Physiology and Immunology, Department of Animal Sciences, Wissenschaftszentrum Weihenstephan, Technische Universität München, Freising, Germany
| | - Susanne E. Ulbrich
- Animal Physiology, Institute of Agricultural Science, Department of Environmental Systems Science, ETH Zürich, Zurich, Switzerland
| | - Michael W. Pfaffl
- Animal Physiology and Immunology, Department of Animal Sciences, Wissenschaftszentrum Weihenstephan, Technische Universität München, Freising, Germany
| |
Collapse
|
46
|
Gunel T, Hosseini MK, Gumusoglu E, Kisakesen HI, Benian A, Aydinli K. Expression profiling of maternal plasma and placenta microRNAs in preeclamptic pregnancies by microarray technology. Placenta 2017; 52:77-85. [PMID: 28454701 DOI: 10.1016/j.placenta.2017.02.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/15/2017] [Accepted: 02/21/2017] [Indexed: 10/20/2022]
Abstract
Preeclampsia (PE) is one of the leading causes of maternal and fetal morbidity and mortality, occurring usually in the second half of pregnancy and affecting approximately 5-8% of pregnancies in the world. miRNAs play critical role in the regulation of placental development processes. We aimed to determine specific novel miRNAs for early diagnosis of preeclampsia which is one of the most dangerous pregnancy diseases. In this study 72 samples, maternal age 22 ≤ and ≤36, have been analyzed; maternal plasma and placental miRNAs were isolated from 18 severe preeclampsia (sPE) patients and 18 controls, respectively. Profiling of human miRNAs (1368 probe) was performed in samples with Agilent v16 microarrays for detection of the differences in miRNA expression between two groups. The results were validated by using TaqMan RT-qPCR method. The analysis indicated that 406 of these miRNAs in all placentas and 42 of these miRNAs in all maternal plasma were expressed. The relative expression analysis has shown that 12 miRNAs (p < 0.05 and >2-fold) in maternal plasma were differentially expressed in PE and control group. However, five miRNAs were validated by qRT-PCR. Once validated miRNAs have been searched in databases for their target genes and function, it has been shown that there are some preeclampsia related pathways as a target such as angiogenesis, cardiovascular, hypertension, placental abruption and preeclampsia disorders. Differentially expressed and validated plasma miRNAs might be used as notable biomarkers for non-invasive early diagnosis of preeclampsia and treatment of disease.
Collapse
Affiliation(s)
- Tuba Gunel
- Istanbul University, Faculty of Science, Department of Molecular Biology and Genetics, Istanbul, Turkey.
| | - Mohammad Kazem Hosseini
- Istanbul University, Faculty of Science, Department of Molecular Biology and Genetics, Istanbul, Turkey
| | - Ece Gumusoglu
- Istanbul University, Faculty of Science, Department of Molecular Biology and Genetics, Istanbul, Turkey
| | - Halil Ibrahim Kisakesen
- Istanbul Technical University, Faculty of Science, Department of Molecular Biology and Genetics, Istanbul, Turkey
| | - Ali Benian
- Istanbul University, Department of Obstetrics and Gynecology, Cerrahpasa Medical Faculty, Istanbul, Turkey
| | | |
Collapse
|
47
|
First trimester screening of circulating C19MC microRNAs and the evaluation of their potential to predict the onset of preeclampsia and IUGR. PLoS One 2017; 12:e0171756. [PMID: 28182660 PMCID: PMC5300267 DOI: 10.1371/journal.pone.0171756] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/25/2017] [Indexed: 12/17/2022] Open
Abstract
Objectives A nested case control study of a longitudinal cohort comparing pregnant women enrolled at 10 to 13 gestational weeks was carried out to evaluate risk assessment for preeclampsia and IUGR based on circulating placental specific C19MC microRNAs in early pregnancy. Methods The expression of placental specific C19MC microRNAs (miR-516b-5p, miR-517-5p, miR-518b, miR-520a-5p, miR-520h, and miR-525-5p) was determined in plasma samples from pregnancies that subsequently developed preeclampsia (n = 21), IUGR (n = 18), and 58 normal pregnancies using real-time PCR and comparative Ct method relative to synthetic Caenorhabditis elegans microRNA (cel-miR-39). Results Circulating C19MC microRNAs were up-regulated (miR-517-5p, p = 0.005; miR-518b, p = 0.013; miR-520h, p = 0.021) or showed a trend toward up-regulation in patients destined to develop preeclampsia (miR-520a-5p, p = 0.067; miR-525-5p, p = 0.073). MiR-517-5p had the best predictive performance for preeclampsia with a sensitivity of 42.9%, a specificity of 86.2%, a PPV of 52.9% and a NPV of 80.6%. The combination of all examined circulating C19MC microRNAs had no advantage over using only the miR-517-5p biomarker to predict the occurrence of preeclampsia (a sensitivity of 20.6%, a specificity of 90.8%, a PPV of 44.8%, and a NPV of 76.0%). Conclusions Up-regulation of miR-517-5p, miR-518b and miR-520h was associated with a risk of later development of preeclampsia. First trimester screening of extracellular miR-517-5p identified a proportion of women with subsequent preeclampsia. No circulating C19MC microRNA biomarkers were identified that could predict later occurrence of IUGR.
Collapse
|
48
|
He C, Wang L, Zhang J, Xu H. Hypoxia-inducible microRNA-224 promotes the cell growth, migration and invasion by directly targeting RASSF8 in gastric cancer. Mol Cancer 2017; 16:35. [PMID: 28173803 PMCID: PMC5297251 DOI: 10.1186/s12943-017-0603-1] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 01/20/2017] [Indexed: 11/10/2022] Open
Abstract
Background Hypoxia plays an important role in the development of various cancers. MicroRNAs (miRNAs) act as post-transcriptional regulators of gene expression and modulate the tumorigenesis, including gastric cancer. However, the roles and molecular mechanism of miR-224 in gastric cancer under hypoxia remain poorly understood. Method Real-time PCR and Northern blot assay were used to examine the effects of hypoxia and HIF-1α on miR-224 expression. Luciferase and ChIP assays were performed to determine whether miR-224 was a transcriptional target of HIF-1α. Then MTT, colony formation, in vitro scratch and invasion assays were used to detect the effects of miR-224 on cell growth, migration and invasion under hypoxia, as well as the in vivo animal study. Luciferase assay and Western blot were performed to validate the targets of miR-224. Functional studies were performed to determine the roles of RASSF8 as that of miR-224 under hypoxia. The effects of RASSF8 knockdown on the transcriptional activity and translocation of NF-κB were investigated using Luciferase assay and Western blot, respectively. Finally, the expression levels of miR-224 and RASSF8 were detected using real-time PCR in gastric cancer tissues as well as lymph node metastasis tissues. Results We demonstrated that miR-224 was upregulated by hypoxia and HIF-1α. HIF-1α affected miR-224 expression at the transcriptional level. MiR-224 inhibition suppressed cell growth, migration and invasion induced by hypoxia, while miR-224 overexpression resulted in opposite effects. MiR-224 inhibition also suppressed tumor growth in vivo. We then validated that RASSF8 was a direct target of miR-224. RASSF8 overexpression inhibited cell growth and invasion, while RASSF8 knockdown ameliorated the inhibitory effects of miR-224 inhibition on cell growth and invasion. Furthermore, we found that RASSF8 knockdown enhanced the transcriptional activity of NF-κB and p65 translocation, while RASSF8 overexpression resulted in opposite effects. Inhibition of NF-κB activity by PDTC attenuated the effects of RASSF8 knockdown on cell proliferation and invasion. Finally, miR-224 was upregulated in both gastric cancer tissues and lymph node metastasis positive tissues, while RASSF8 expression was opposite to that of miR-224. Conclusion These results indicate that hypoxia-inducible miR-224 promotes gastric cancer cell growth, migration and invasion by downregulating RASSF8 and acts as an oncogene, implying that inhibition of miR-224 may have potential as a therapeutic target for patients with hypoxic gastric tumors.
Collapse
Affiliation(s)
- Chuan He
- Department of Gastroenterology, First Hospital of Jilin University, No.71 Xinmin Street, Changchun, Jilin, 130021, People's Republic of China
| | - Libo Wang
- Department of Gastroenterology, First Hospital of Jilin University, No.71 Xinmin Street, Changchun, Jilin, 130021, People's Republic of China
| | - Jiantao Zhang
- Department of Colorectal and anal surgery, First Hospital of Jilin University, Changchun, Jilin, 130021, People's Republic of China
| | - Hong Xu
- Department of Gastroenterology, First Hospital of Jilin University, No.71 Xinmin Street, Changchun, Jilin, 130021, People's Republic of China.
| |
Collapse
|
49
|
Ouyang Y, Bayer A, Chu T, Tyurin VA, Kagan VE, Morelli AE, Coyne CB, Sadovsky Y. Isolation of human trophoblastic extracellular vesicles and characterization of their cargo and antiviral activity. Placenta 2016; 47:86-95. [PMID: 27780544 PMCID: PMC5123854 DOI: 10.1016/j.placenta.2016.09.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/08/2016] [Accepted: 09/13/2016] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Primary human trophoblasts release a repertoire of extracellular vesicles (EVs). Among them are nano-sized exosomes, which we found to suppress the replication of a wide range of diverse viruses. These exosomes contain trophoblastic microRNAs (miRNAs) that are expressed from the chromosome 19 miRNA cluster and exhibit antiviral properties. Here, we report our investigation of the cargo of placental EVs, focusing on the composition and the antiviral properties of exosomes, microvesicles, and apoptotic blebs. METHODS We isolated EVs using ultracentrifugation and defined their purity using immunoblotting, electron microscopy, and nanoparticle tracking. We used liquid chromatography-electrospray ionization-mass spectrometry, protein mass spectrometry, and miRNA TaqMan card PCR to examine the phospholipids, proteins, and miRNA cargo of trophoblastic EVs and an in vitro viral infection assay to assess the antiviral properties of EVs. RESULTS We found that all three EV types contain a comparable repertoire of miRNA. Interestingly, trophoblastic exosomes harbor a protein and phospholipid profile that is distinct from that of microvesicles or apoptotic blebs. Functionally, trophoblastic exosomes exhibit the highest antiviral activity among the EVs. Consistently, plasma exosomes derived from pregnant women recapitulate the antiviral effect of trophoblastic exosomes derived from in vitro cultures of primary human trophoblasts. DISCUSSION When compared to other trophoblastic EVs, exosomes exhibit a unique repertoire of proteins and phospholipids, but not miRNAs, and a potent viral activity. Our work suggests that human trophoblastic EVs may play a key role in maternal-placental-fetal communication.
Collapse
Affiliation(s)
- Yingshi Ouyang
- Magee-Womens Research Institute, Department of OBGYN and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Avraham Bayer
- Magee-Womens Research Institute, Department of OBGYN and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tianjiao Chu
- Magee-Womens Research Institute, Department of OBGYN and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Vladimir A Tyurin
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Adrian E Morelli
- T.E. Starzl Institute and Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Carolyn B Coyne
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of OBGYN and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
50
|
Jain CV, Jessmon P, Kilburn BA, Jodar M, Sendler E, Krawetz SA, Armant DR. Regulation of HBEGF by Micro-RNA for Survival of Developing Human Trophoblast Cells. PLoS One 2016; 11:e0163913. [PMID: 27701455 PMCID: PMC5049799 DOI: 10.1371/journal.pone.0163913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/17/2016] [Indexed: 11/30/2022] Open
Abstract
Introduction The growth factor HBEGF is upregulated post-transcriptionally in the low O2 environment of the human placenta during the first 10 weeks of pregnancy. We have examined the possible roles of HBEGF turnover and micro-RNA (miRNA) in its regulation by O2 in human first trimester trophoblast. Methods HTR-8/SVneo trophoblast cells were cultured at 2% or 20% O2. The cells were transfected with a dual luciferase reporter construct (psiCHECK-2) containing no insert (control), the HBEGF 3’ untranslated region (3’UTR), or sub-regions of the 3’UTR, as well as with siRNA for DGCR8. RNA was extracted from trophoblast cells cultured at 2% O2 for 0–4 h for next-generation sequencing. HBEGF was quantified by ELISA. HBEGF, DGCR8, and β–actin were examined by western blotting. Results Protein turnover studies, using 10 μg/ml cyclohexamide, 1 μg/ml lactocystin, or 100 μg/ml MG132, demonstrated faster HBEGF degradation at 20% O2 than 2% O2, mediated by the proteasome. However, proteasome inhibition failed to initiate HBEGF accumulation at 20% O2. Reporter assays, comparing to empty vector, demonstrated that the intact HBEGF 3’ UTR inhibited expression (0.26), while fragments containing only its flanking regions increased reporter activity (3.15; 3.43). No differential expression of miRNAs was found in trophoblast cells cultured at 2% and 20% O2. Nevertheless, HBEGF upregulation at 2% O2 was blocked when the miRNA-processing protein DGCR8 was silenced, suggesting a role for miRNA. Conclusion Our findings suggest involvement of flanking regions of the 3’UTR in activating HBEGF protein synthesis in response to 2% O2, possibly through a miRNA-mediated mechanism.
Collapse
Affiliation(s)
- Chandni V. Jain
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Philip Jessmon
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Brian A. Kilburn
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Meritxell Jodar
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Edward Sendler
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - Stephen A. Krawetz
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States of America
| | - D. Randall Armant
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, Detroit, Michigan, United States of America
- Program in Reproductive and Adult Endocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, DHHS, Bethesda, MD, United States of America
- * E-mail:
| |
Collapse
|