1
|
Sinha T, Sadhukhan S, Panda AC. Computational Prediction of Gene Regulation by lncRNAs. Methods Mol Biol 2025; 2883:343-362. [PMID: 39702716 DOI: 10.1007/978-1-0716-4290-0_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
High-throughput sequencing technologies and innovative bioinformatics tools discovered that most of the genome is transcribed into RNA. However, only a fraction of the RNAs in cell translates into proteins, while the majority of them are categorized as noncoding RNAs (ncRNAs). The ncRNAs with more than 200 nt without protein-coding ability are termed long noncoding RNAs (lncRNAs). Hundreds of studies established that lncRNAs are a crucial RNA family regulating gene expression. Regulatory RNAs, including lncRNAs, modulate gene expression by interacting with RNA, DNA, and proteins. Several databases and computational tools have been developed to explore the functions of lncRNAs in cellular physiology. This chapter discusses the tools available for lncRNA functional analysis and provides a detailed workflow for the computational analysis of lncRNAs.
Collapse
Affiliation(s)
- Tanvi Sinha
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, India
| | - Susovan Sadhukhan
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, India
| | - Amaresh C Panda
- Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, India.
| |
Collapse
|
2
|
Senousy MA, Shaker OG, Elmaasrawy AH, Ashour AM, Alsufyani SE, Arab HH, Ayeldeen G. Serum lncRNAs TUG1, H19, and NEAT1 and their target miR-29b/SLC3A1 axis as possible biomarkers of preeclampsia: Potential clinical insights. Noncoding RNA Res 2024; 9:995-1008. [PMID: 39026605 PMCID: PMC11254728 DOI: 10.1016/j.ncrna.2024.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/26/2024] [Accepted: 06/07/2024] [Indexed: 07/20/2024] Open
Abstract
To date, the epigenetic signature of preeclampsia (PE) is not completely deciphered. Oxidative stress-responsive long non-coding RNAs (lncRNAs) are deregulated in preeclamptic placenta; however, their circulating profiles and diagnostic abilities are still unexplored. We investigated serum redox-sensitive lncRNAs TUG1, H19, and NEAT1, and their target miR-29b/cystine/neutral/dibasic amino acids transporter solute carrier family 3, member 1 (SLC3A1) as potential non-invasive biomarkers of PE risk, onset, and severity. We recruited 82 patients with PE and 78 healthy pregnant women. We classified PE patients into early-onset (EOPE) and late-onset (LOPE) subgroups at a cut-off 34 gestational weeks and into severe and mild PE subgroups by blood pressure and proteinuria criteria. Bioinformatics analysis was employed to select lncRNAs/microRNA/target gene interactions. Serum H19, NEAT1, and SLC3A1 mRNA expression were reduced, meanwhile miR-29b levels were elevated, whereas there was no significant difference in TUG1 levels between PE patients and healthy pregnancies. Serum H19 levels were lower, whereas miR-29b levels were higher in EOPE versus LOPE. Serum miR-29b and H19 levels were higher in severe versus mild PE. ROC analysis identified serum H19, NEAT1, miR-29b, and SLC3A1 as potential diagnostic markers, with H19 (AUC = 0.818, 95%CI = 0.744-0.894) and miR-29b (AUC = 0.82, 95%CI = 0.755-0.885) were superior discriminators. Only H19 and miR-29b discriminated EOPE and severe PE cases. In multivariate logistic analysis, miR-29b and H19 were associated with EOPE, using maternal age and gestational age as covariates, while miR-29b was associated with severe PE, using maternal age as covariate. Studied markers were correlated with clinical and ultrasound data in the overall PE group. Serum H19 and TUG1 were negatively correlated with albuminuria in EOPE and LOPE, respectively. NEAT1 and SLC3A1 were correlated with ultrasound data in EOPE. Likewise, TUG1, miR-29b, and SLC3A1 showed significant correlations with ultrasound data in LOPE. Conclusively, this study configures SLC3A1 expression as a novel potential serum biomarker of PE and advocates serum H19 and miR-29b as biomarkers of EOPE and miR-29b as a biomarker of PE severity.
Collapse
Affiliation(s)
- Mahmoud A. Senousy
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Olfat G. Shaker
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ahmed H.Z. Elmaasrawy
- Department of Obstetrics and Gynecology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Ahmed M. Ashour
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al Qura University, P.O. Box 13578, Makkah, 21955, Saudi Arabia
| | - Shuruq E. Alsufyani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Hany H. Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Ghada Ayeldeen
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
3
|
Cannarella R, Rando OJ, Condorelli RA, Chamayou S, Romano S, Guglielmino A, Yin Q, Hans TG, Mancuso F, Arato I, Bellucci C, Luca G, Lundy SD, La Vignera S, Calogero AE. Sperm-carried IGF2: towards the discovery of a spark contributing to embryo growth and development. Mol Hum Reprod 2024; 30:gaae034. [PMID: 39312692 PMCID: PMC11975288 DOI: 10.1093/molehr/gaae034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 09/06/2024] [Indexed: 09/25/2024] Open
Abstract
Spermatozoa have been shown to carry key RNAs which, according to animal evidence, seem to play a role in early embryo development. In this context, a potential key growth regulator is insulin-like growth factor 2 (IGF2), a highly conserved paternally expressed imprinted gene involved in cell growth and proliferation which, recent observations indicate, is expressed in human spermatozoa. We herein hypothesized that sperm IGF2 gene expression and transmission at fertilization is required to support early embryo development. To test this hypothesis, we analyzed sperm IGF2 mRNA levels in the same semen aliquot used for homologous assisted reproductive technique (ART) in infertile couples and correlated these levels with embryo morphokinetics. To find a mechanistic explanation for the observed results, the transcriptomes of blastocysts obtained after injection of Igf2 mRNA in mouse parthenotes were analyzed. Sperm IGF2 mRNA negatively correlated with time of 2-cell stage (t2), t3, t4, t5, and time of expanded blastocyst (tEB), independently of maternal age, body mass index, anti-Müllerian hormone levels, and oocyte quality. An IGF2 mRNA index >4.9 predicted the ability of the embryos to reach the blastocyst stage on Day 5, with a sensitivity of 100% and a specificity of 71.6% (AUC 0.845; P < 0.001). In the animal study, transcriptome analysis demonstrated that 65 and 36 genes were, respectively, up- and down-regulated in the experimental group compared to the control group. These genes belong to pathways that regulate early embryo development, thus supporting the findings found in humans. This study has the potential to challenge the longstanding tenet that spermatozoa are simply vehicles carrying paternal DNA. Instead, it suggests that IGF2 mRNA in healthy spermatozoa provides critical support for early embryo development. Pre-ART sperm-carried IGF2 mRNA levels may be used as a marker to predict the chances of obtaining blastocysts to be transferred for infertile couples undergoing ART.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Oliver J Rando
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | - Simona Romano
- Centro HERA—Unità di Medicina della Riproduzione, Catania, Italy
| | | | - Qiangzong Yin
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Tobias Gustafsson Hans
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Iva Arato
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Catia Bellucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giovanni Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Scott D Lundy
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
4
|
Gallagher LT, Bardill J, Sucharov CC, Wright CJ, Karimpour-Fard A, Zarate M, Breckenfelder C, Liechty KW, Derderian SC. Dysregulation of miRNA-mRNA expression in fetal growth restriction in a caloric restricted mouse model. Sci Rep 2024; 14:5579. [PMID: 38448721 PMCID: PMC10918062 DOI: 10.1038/s41598-024-56155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/01/2024] [Indexed: 03/08/2024] Open
Abstract
Fetal growth restriction (FGR) is associated with aberrant placentation and accounts for a significant proportion of perinatal deaths. microRNAs have been shown to be dysregulated in FGR. The purpose of this study was to determine microRNA-regulated molecular pathways altered using a caloric restricted mouse model of FGR. Pregnant mice were subjected to a 50% caloric restricted diet beginning at E9. At E18.5, RNA sequencing of placental tissue was performed to identify differences in gene expression between caloric restricted and control placentas. Significant differences in gene expression between caloric restricted and control placentas were observed in 228 of the 1546 (14.7%) microRNAs. Functional analysis of microRNA-mRNA interactions demonstrated enrichment of several biological pathways with oxidative stress, apoptosis, and autophagy pathways upregulated and angiogenesis and signal transduction pathways downregulated. Ingenuity pathway analysis also suggested that ID1 signaling, a pathway integral for trophoblast differentiation, is also dysregulated in caloric restricted placentas. Thus, a maternal caloric restriction mouse model of FGR results in aberrant microRNA-regulated molecular pathways associated with angiogenesis, oxidative stress, signal transduction, apoptosis, and cell differentiation. As several of these pathways are dysregulated in human FGR, our findings suggest that this model may provide an excellent means to study placental microRNA derangements seen in FGR.
Collapse
Affiliation(s)
- Lauren T Gallagher
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - James Bardill
- Department of Surgery, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - Carmen C Sucharov
- Division of Cardiology, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Anis Karimpour-Fard
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Miguel Zarate
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Courtney Breckenfelder
- Laboratory for Fetal and Regenerative Biology, Department of Surgery, University of Colorado Denver School of Medicine, Aurora, CO, 80045, USA
| | - Kenneth W Liechty
- Division of Pediatric Surgery, University of Arizona College of Medicine, Tucson, AZ, 85721, USA
| | - S Christopher Derderian
- Colorado Fetal Care Center, Children's Hospital Colorado, University of Colorado, 13123 E 16th Ave, Aurora, CO, 80045, USA.
- Division of Pediatric Surgery, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, 80045, USA.
| |
Collapse
|
5
|
Huang Y, Li S, Tan Y, Xu C, Huang X, Yin Z. Identification and functional analysis of ovarian lncRNAs during different egg laying periods in Taihe Black-Bone Chickens. Front Physiol 2024; 15:1358682. [PMID: 38426211 PMCID: PMC10902129 DOI: 10.3389/fphys.2024.1358682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
Introduction: Long non-coding RNA (lncRNA) refers to a category of non-coding RNA molecules exceeding 200 nucleotides in length, which exerts a regulatory role in the context of ovarian development. There is a paucity of research examining the involvement of lncRNA in the regulation of ovary development in Taihe Black-Bone Chickens. In order to further investigate the egg laying regulation mechanisms of Taihe Black-Bone Chickens at different periods, transcriptome analysis was conducted on the ovarian tissues at different laying periods. Methods: This study randomly selected ovarian tissues from 12 chickens for RNA-seq. Four chickens were selected for each period, including the early laying period (102 days, Pre), the peak laying period (203 days, Peak), and the late laying period (394 days, Late). Based on our previous study of mRNA expression profiles in the same ovarian tissue, we identified three differentially expressed lncRNAs (DE lncRNAs) at different periods and searched for their cis- and trans-target genes to draw an lncRNA-mRNA network. Results and discussion: In three groups of ovarian tissues, we identified 136 DE lncRNAs, with 8 showing specific expression during the early laying period, 10 showing specific expression during the peak laying period, and 4 showing specific expression during the late laying period. The lncRNA-mRNA network revealed 16 pairs of lncRNA-target genes associated with 7 DE lncRNAs, and these 14 target genes were involved in the regulation of reproductive traits. Furthermore, these reproductive-related target genes were primarily associated with signaling pathways related to follicle and ovary development in Taihe Black-Bone Chickens, including cytokine-cytokine receptor interaction, TGF-beta signaling pathway, tyrosine metabolism, ECM-receptor interaction, focal adhesion, neuroactive ligand-receptor interaction, and cell adhesion molecules (CAMs). This study offers valuable insights for a comprehensive understanding of the influence of lncRNAs on poultry reproductive traits.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhaozheng Yin
- College of Animal Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
6
|
da Cunha Agostini L, Almeida TC, da Silva GN. ANRIL, H19 and TUG1: a review about critical long non-coding RNAs in cardiovascular diseases. Mol Biol Rep 2023; 51:31. [PMID: 38155319 DOI: 10.1007/s11033-023-09007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/30/2023] [Indexed: 12/30/2023]
Abstract
Cardiovascular diseases are the leading cause of death worldwide. They are non-transmissible diseases that affect the cardiovascular system and have different etiologies such as smoking, lipid disorders, diabetes, stress, sedentary lifestyle and genetic factors. To date, lncRNAs have been associated with increased susceptibility to the development of cardiovascular diseases such as hypertension, acute myocardial infarction, stroke, angina and heart failure. In this way, lncRNAs are becoming a very promising point for the prevention and diagnosis of cardiovascular diseases. Therefore, this review highlights the most important and recent discoveries about the mechanisms of action of the lncRNAs ANRIL, H19 and TUG1 and their clinical relevance in these pathologies. This may contribute to early detection of cardiovascular diseases in order to prevent the pathological phenotype from becoming established.
Collapse
Affiliation(s)
- Lívia da Cunha Agostini
- Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, Morro do Cruzeiro, s/nº, Ouro Prêto, Minas Gerais, CEP 35402-163, Brazil
| | - Tamires Cunha Almeida
- Escola Superior Instituto Butantan (ESIB), Laboratório de Dor e Sinalização, Instituto Butantan, São Paulo, São Paulo, Brazil
| | - Glenda Nicioli da Silva
- Programa de Pós-Graduação em Ciências Farmacêuticas (CiPharma), Escola de Farmácia, Universidade Federal de Ouro Preto, Morro do Cruzeiro, s/nº, Ouro Prêto, Minas Gerais, CEP 35402-163, Brazil.
- Departamento de Análises Clínicas (DEACL), Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Prêto, Brazil.
| |
Collapse
|
7
|
Ning H, Tao H. Small RNA sequencing of exosomal microRNAs reveals differential expression of microRNAs in preeclampsia. Medicine (Baltimore) 2023; 102:e35597. [PMID: 37861520 PMCID: PMC10589583 DOI: 10.1097/md.0000000000035597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 09/20/2023] [Indexed: 10/21/2023] Open
Abstract
Preeclampsia (PE) is one of the most common hypertensive disorders of pregnancy. It is a dangerous condition with a high mortality rate in mothers and fetuses and is associated with a lack of early diagnosis and effective treatment. While the etiology of the disease is complex and obscure, it is now clear that the placenta is central to disease progression. Exosomal microRNAs (miRNAs) are possible mediators that regulate placenta-related physiological and pathological processes. Placental mesenchymal stem cells have considerable potential to help us understand the pathogenesis and treatment of pregnancy-related diseases. Here, we investigate the exosomal miRNA profiles of human placenta-derived mesenchymal stem cells between healthy pregnant women and those with PE. We performed small RNA sequencing to obtain miRNA profiles, and conducted enrichment analysis of the miRNA target genes to identify differentially expressed miRNAs associated with PE. Overall, we detected 1795 miRNAs; among them, 206 were differentially expressed in women with PE, including 35 upregulated and 171 downregulated miRNAs, when compared with healthy pregnant women. Moreover, we identified possible functions and pathways associated with PE, including angiogenesis, cell proliferation, migration and invasion, and the coagulation-fibrinolysis balance. Eventually, we proposed hsa-miR-675-5p, hsa-miR-3614-5p, and hsa-miR-615-5p as potential regulators of the pathogenesis of PE, and constructed a miRNA-target gene network. Our study identifies possible candidate biomarkers for the diagnosis of PE, and introduces a new direction for further understanding the pathogenesis of PE.
Collapse
Affiliation(s)
- Hui Ning
- Department of Obstetrics, Qingdao Municipal Hospital, Qingdao, China
| | - Hong Tao
- Department of Obstetrics, Qingdao Municipal Hospital, Qingdao, China
| |
Collapse
|
8
|
Mora-Palazuelos C, Villegas-Mercado CE, Avendaño-Félix M, Lizárraga-Verdugo E, Romero-Quintana JG, López-Gutiérrez J, Beltrán-Ontiveros S, Bermúdez M. The Role of ncRNAs in the Immune Dysregulation of Preeclampsia. Int J Mol Sci 2023; 24:15215. [PMID: 37894897 PMCID: PMC10607488 DOI: 10.3390/ijms242015215] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/04/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
The main complications causing practically 75% of all maternal deaths are severe bleeding, infections, and high blood pressure during pregnancy (preeclampsia (PE) and eclampsia). The usefulness of ncRNAs as clinical biomarkers has been explored in an extensive range of human diseases including pregnancy-related diseases such as PE. Immunological dysregulation show that the Th1/17:Th2/Treg ratio is "central and causal" to PE. However, there is evidence of the involvement of placenta-expressed miRNAs and lncRNAs in the immunological regulation of crucial processes of placenta development and function during pregnancy. Abnormal expression of these molecules is related to immune physiopathological processes that occur in PE. Therefore, this work aims to describe the importance of miRNAs and lncRNAs in immune dysregulation in PE. Interestingly, multiple ncRNAS are involved in the immune dysregulation of PE participating in type 1 immune response regulation, immune microenvironment regulation in placenta promoting inflammatory factors, trophoblast cell invasion in women with Early-Onset PE (EOPE), placental development, and angiogenesis, promotion of population of M1 and M2, proliferation, invasion, and migration of placental trophoblast cells, and promotion of invasion and autophagy through vias such as PI3K/AKT/mTOR, VEGF/VEGFR1, and TLR9/STAT3.
Collapse
Affiliation(s)
- Carlos Mora-Palazuelos
- Health Sciences Research and Teaching Center, Autonomous University of Sinaloa, Culiacan 80010, Sinaloa, Mexico; (C.M.-P.); (E.L.-V.); (S.B.-O.)
| | | | - Mariana Avendaño-Félix
- Faculty of Dentistry, Autonomous University of Sinaloa, Culiacan 80010, Sinaloa, Mexico;
| | - Erik Lizárraga-Verdugo
- Health Sciences Research and Teaching Center, Autonomous University of Sinaloa, Culiacan 80010, Sinaloa, Mexico; (C.M.-P.); (E.L.-V.); (S.B.-O.)
| | | | - Jorge López-Gutiérrez
- Faculty of Biology, Autonomous University of Sinaloa, Culiacan 80010, Sinaloa, Mexico;
| | - Saúl Beltrán-Ontiveros
- Health Sciences Research and Teaching Center, Autonomous University of Sinaloa, Culiacan 80010, Sinaloa, Mexico; (C.M.-P.); (E.L.-V.); (S.B.-O.)
| | - Mercedes Bermúdez
- Faculty of Dentistry, Autonomous University of Chihuahua, Chihuahua 31110, Chihuahua, Mexico;
| |
Collapse
|
9
|
Banikazemi Z, Heidar Z, Rezaee A, Taghavi SP, Zadeh Modarres S, Asemi Z, Goleij P, Jahed F, Mazaheri E, Taghizadeh M. Long non-coding RNAs and female infertility: What do we know? Pathol Res Pract 2023; 250:154814. [PMID: 37757620 DOI: 10.1016/j.prp.2023.154814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/24/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023]
Abstract
Ten percent of people who are of reproductive age experience infertility. Sometimes the most effective therapies, including technology for assisted reproduction, may lead to unsuccessful implantation. Because of the anticipated epigenetic alterations of in vitro as well as in vitro fertilization growth of embryos, these fertility techniques have also been linked to unfavorable pregnancy outcomes linked to infertility. In this regard, a variety of non-coding RNAs such as long noncoding RNAs (lncRNAs) act as epigenetic regulators in the various physiological and pathophysiological events such as infertility. LncRNAs have been made up of cytoplasmic and nuclear nucleotides; RNA polymerase II transcribes these, which are lengthier than 200 nt. LncRNAs perform critical roles in a number of biological procedures like nuclear transport, X chromosome inactivation, apoptosis, stem cell pluripotency, as well as genomic imprinting. A significant amount of lncRNAs were linked into a variety of biological procedures as high throughput sequencing technology advances, including the development of the testes, preserving spermatogonial stem cells' capacity for differentiation along with self-renewal, and controlling spermatocyte meiosis. All of them point to possible utility of lncRNAs to be biomarkers and treatment aims for female infertility. Herein, we summarize various lncRNAs that are involved in female infertility.
Collapse
Affiliation(s)
- Zarrin Banikazemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Zahra Heidar
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Pouya Taghavi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran; School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Shahrzad Zadeh Modarres
- Clinical Research Development Center, Mahdiyeh Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Pouya Goleij
- Department of Genetics, Faculty of Biology, Sana Institute of Higher Education, Sari, Iran
| | - Fatemeh Jahed
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Elaheh Mazaheri
- Preventative Gynecology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
10
|
Wang M, Zheng L, Ma S, Lin R, Li J, Yang S. Biogenesis and function of exosome lncRNAs and their role in female pathological pregnancy. Front Endocrinol (Lausanne) 2023; 14:1191721. [PMID: 37745705 PMCID: PMC10515720 DOI: 10.3389/fendo.2023.1191721] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Preeclampsia, gestational diabetes mellitus, and recurrent spontaneous abortion are common maternal pregnancy complications that seriously endanger women's lives and health, and their occurrence is increasing year after year with a rejuvenation trend. In contrast to biomarkers found freely in tissues or body fluids, exosomes exist in a relatively independent environment and provide a higher level of stability. As backbone molecules, guidance molecules, and signaling molecules in the nucleus, lncRNAs can regulate gene expression. In the cytoplasm, lncRNAs can influence gene expression levels by modifying mRNA stability, acting as competitive endogenous RNAs to bind miRNAs, and so on. Exosomal lncRNAs can exist indefinitely and are important in intercellular communication and signal transduction. Changes in maternal serum exosome lncRNA expression can accurately and timely reflect the progression and regression of pregnancy-related diseases. The purpose of this paper is to provide a reference for clinical research on the pathogenesis, diagnosis, and treatment methods of pregnancy-related diseases by reviewing the role of exosome lncRNAs in female pathological pregnancy and related molecular mechanisms.
Collapse
Affiliation(s)
- Min Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Lianwen Zheng
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Shuai Ma
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Ruixin Lin
- Department of Hepato-Biliary-Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jiahui Li
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| | - Shuli Yang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Liao J, Chen B, Zhu Z, Du C, Gao S, Zhao G, Zhao P, Wang Y, Wang A, Schwartz Z, Song L, Hong J, Wagstaff W, Haydon RC, Luu HH, Fan J, Reid RR, He TC, Shi L, Hu N, Huang W. Long noncoding RNA (lncRNA) H19: An essential developmental regulator with expanding roles in cancer, stem cell differentiation, and metabolic diseases. Genes Dis 2023; 10:1351-1366. [PMID: 37397543 PMCID: PMC10311118 DOI: 10.1016/j.gendis.2023.02.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/07/2023] [Accepted: 02/08/2023] [Indexed: 07/04/2023] Open
Abstract
Recent advances in deep sequencing technologies have revealed that, while less than 2% of the human genome is transcribed into mRNA for protein synthesis, over 80% of the genome is transcribed, leading to the production of large amounts of noncoding RNAs (ncRNAs). It has been shown that ncRNAs, especially long non-coding RNAs (lncRNAs), may play crucial regulatory roles in gene expression. As one of the first isolated and reported lncRNAs, H19 has gained much attention due to its essential roles in regulating many physiological and/or pathological processes including embryogenesis, development, tumorigenesis, osteogenesis, and metabolism. Mechanistically, H19 mediates diverse regulatory functions by serving as competing endogenous RNAs (CeRNAs), Igf2/H19 imprinted tandem gene, modular scaffold, cooperating with H19 antisense, and acting directly with other mRNAs or lncRNAs. Here, we summarized the current understanding of H19 in embryogenesis and development, cancer development and progression, mesenchymal stem cell lineage-specific differentiation, and metabolic diseases. We discussed the potential regulatory mechanisms underlying H19's functions in those processes although more in-depth studies are warranted to delineate the exact molecular, cellular, epigenetic, and genomic regulatory mechanisms underlying the physiological and pathological roles of H19. Ultimately, these lines of investigation may lead to the development of novel therapeutics for human diseases by exploiting H19 functions.
Collapse
Affiliation(s)
- Junyi Liao
- Departments of Orthopedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Orthopedic Research Center, Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Bowen Chen
- Departments of Orthopedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Orthopedic Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Zhenglin Zhu
- Departments of Orthopedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Orthopedic Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Chengcheng Du
- Departments of Orthopedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Orthopedic Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Shengqiang Gao
- Departments of Orthopedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Orthopedic Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Guozhi Zhao
- Departments of Orthopedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Piao Zhao
- Departments of Orthopedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Orthopedic Research Center, Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yonghui Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Clinical Laboratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai 200000, China
| | - Annie Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zander Schwartz
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- School of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Lily Song
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jeffrey Hong
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- The Medical Scientist Training Program, The University of Chicago Pritzker School of Medicine, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jiaming Fan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, Department of Clinical Biochemistry, The School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Suture Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Suture Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lewis Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Ning Hu
- Departments of Orthopedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Orthopedic Research Center, Chongqing Medical University, Chongqing 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Wei Huang
- Departments of Orthopedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Orthopedic Research Center, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
12
|
Hosseini SA, Haddadi MH, Fathizadeh H, Nemati F, Aznaveh HM, Taraj F, Aghabozorgizadeh A, Gandomkar G, Bazazzadeh E. Long non-coding RNAs and gastric cancer: An update of potential biomarkers and therapeutic applications. Biomed Pharmacother 2023; 163:114407. [PMID: 37100014 DOI: 10.1016/j.biopha.2023.114407] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 04/28/2023] Open
Abstract
The frequent metastasis of gastric cancer (GC) complicates the cure and therefore the development of effective diagnostic and therapeutic approaches is urgently necessary. In recent years, lncRNA has emerged as a drug target in the treatment of GC, particularly in the areas of cancer immunity, cancer metabolism, and cancer metastasis. This has led to the demonstration of the importance of these RNAs as prognostic, diagnostic and therapeutic agents. In this review, we provide an overview of the biological activities of lncRNAs in GC development and update the latest pathological activities, prognostic and diagnostic strategies, and therapeutic options for GC-related lncRNAs.
Collapse
Affiliation(s)
- Sayedeh Azimeh Hosseini
- Department of Medical Biotechnology, School of Advanced Technology, Shahrekord University of Medical Sciences, Shahrekord, Iran; Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran; USERN office, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Hadis Fathizadeh
- Student Research Committee, Sirjan School of Medical Sciences, Sirjan, Iran; Department of Laboratory sciences, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Foroogh Nemati
- Department of Microbiology, Kashan University of Medical Sciences, Kashan, Iran
| | - Hooman Mahmoudi Aznaveh
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| | - Farima Taraj
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - AmirArsalan Aghabozorgizadeh
- Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Golmaryam Gandomkar
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Elaheh Bazazzadeh
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box: 14115-154, Tehran, Iran
| |
Collapse
|
13
|
Sun Y, Zhang C, Luo L, Lin H, Liu C, Zhang W. Paternal genetic intergenerational and transgenerational effects of cadmium exposure on hormone synthesis disorders in progeny ovarian granulosa cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 322:121175. [PMID: 36731734 DOI: 10.1016/j.envpol.2023.121175] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/27/2023] [Accepted: 01/28/2023] [Indexed: 06/18/2023]
Abstract
To investigate the paternal genetic effects of cadmium (Cd) exposure on hormone synthesis disorders in the ovarian granulosa cells (GCs) of offspring. Here, male Sprague‒Dawley (SD) rats were gavaged with CdCl2 (0, 0.5, 2, 8 mg/kg) from postnatal day (PND) 28-56, followed by mating with newly purchased healthy adult females to produce F1, and F1 adult males (PND 56) were mated with newly purchased healthy adult females to produce F2. The serum levels of estradiol (E2) and progesterone (Pg) decreased in F1 but essentially returned to normal in F2. The levels of StAR, CYP11A1, CYP17A1, CYP19A1, and SF-1 showed different alterations in F1 and F2 ovarian GCs. The expression patterns of miRNAs and imprinted genes related to hormone synthesis in GCs of F1 and F2 differed, but methylation of hormone synthesis-related genes was not significantly altered (except for individual loci in F1). In addition, there were significant changes in the expression of imprinted genes and miRNAs in F0 and F1 sperm. We conclude that paternal Cd exposure causes intergenerational genetic effects (hormone synthesis disorders) and transgenerational effects (reparative changes in hormone synthesis function) in ovarian GCs. These genetic effects were related to the downregulation of StAR in F1 and the upregulation of CYP17A1, CYP19A1, StAR and SF-1 in F2. Important changes in miRNAs and imprinted genes were also observed, but not all alterations originated from paternal inheritance.
Collapse
Affiliation(s)
- Yi Sun
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Chenyun Zhang
- School of Health Management, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Lingfeng Luo
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Hao Lin
- Fuzhou Center for Disease Control and Prevention, Fuzhou, 350005, Fujian, China
| | - Chenchen Liu
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian, China
| | - Wenchang Zhang
- Department of Preventive Medicine, Fujian Provincial Key Laboratory of Environmental Factors and Cancer, Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
14
|
Hou B, Mao M, Dong S, Deng M, Sun B, Guo Y, Li Y, Liu D, Liu G. Transcriptome analysis reveals mRNAs and long non-coding RNAs associated with fecundity in the hypothalamus of high-and low-fecundity goat. Front Vet Sci 2023; 10:1145594. [PMID: 37056233 PMCID: PMC10086355 DOI: 10.3389/fvets.2023.1145594] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/14/2023] [Indexed: 03/30/2023] Open
Abstract
As an important organ that coordinates the neuroendocrine system, the hypothalamus synthesizes and secretes reproductive hormones that act on the goat organism, thereby precisely regulating follicular development and reproductive processes in goats. However, it is still elusive to explore the mechanism of hypothalamic effects on goat fertility alone. Therefore, RNA-seq was used to analyze the gene expression in hypothalamic tissues of goats in high fertility group (HFG: litter size per litter ≥2) and low fertility group (LFG: litter size per litter = 1), and identified the differential lncRNAs and mRNAs and their associated pathways related to their fertility. The results showed that a total of 23 lncRNAs and 57 mRNAs were differentially expressed in the hypothalamic tissue of high and low fertility goats. GO terms and KEGG functional annotation suggest that DE lncRNAs and DE mRNAs were significantly enriched in hormone-related pathways regulating ovarian development, hormone synthesis and secretion, regulation of reproductive processes, Estrogen signaling pathway, Oxytocin signaling pathway and GnRH signaling pathway. And we constructed a co-expression network of lncRNAs and target genes, and identified reproduction-related genes such as NMUR2, FEZF1, and WT1. The sequencing results of the hypothalamic transcriptome have broadened our understanding of lncRNA and mRNA in goat hypothalamic tissue and provided some new insights into the molecular mechanisms of follicle development and regulation of its fertility in goats.
Collapse
|
15
|
Possible transfer of lncRNA H19-derived miRNA miR-675-3p to adjacent H19-non-expressing trophoblast cells in near-term mouse placenta. Histochem Cell Biol 2022; 159:363-375. [PMID: 36484822 DOI: 10.1007/s00418-022-02169-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2022] [Indexed: 12/13/2022]
Abstract
LncRNA H19 serves as a regulatory RNA in mouse placental development. However, there is little information available on the in situ expression of H19 in the late-gestation mouse placenta. In this study, we performed quantitative polymerase chain reaction (qPCR) and in situ hybridization (ISH) analyses of lncRNA H19 and its exon 1-derived miRNA miR-675-3p to identify cell types expressing these non-coding RNAs in the mouse placenta during mid-to-late gestation. By qPCR analysis, we confirmed that H19 was highly expressed during mid-to-late gestation (E10.5-E18.5) and that H19-derived miRNA miR-675-3p was remarkably upregulated in the E18.5 placenta. ISH analysis revealed trophoblast cell type-specific expression of lncRNA H19 and miR-675-3p during later stages of gestation. In the junctional zone and decidua of late-gestation placenta, H19 was expressed in trophoblast giant cells and glycogen trophoblast cells; however, H19 was absent in spongiotrophoblast cells. In the labyrinth and chorionic plate, H19 was present in sinusoidal mononuclear trophoblast giant cells, fetal vascular endothelial cells, and basal chorionic trophoblast cells, but not in syncytiotrophoblasts. As expected, these lncRNA H19-expressing cells exhibited miR-675-3p in the E18.5 placenta. Intriguingly, miR-675-3p was also present in H19-negative spongiotrophoblast cells and syncytiotrophoblasts, implying the possible transfer of miR-675-3p from H19-exprssing cells to adjacent H19-non-expressing trophoblast cells. These findings suggest that the mouse placenta expresses lncRNA H19 in a trophoblast cell type-specific fashion during later stages of gestation.
Collapse
|
16
|
Kannampuzha S, Ravichandran M, Mukherjee AG, Wanjari UR, Renu K, Vellingiri B, Iyer M, Dey A, George A, Gopalakrishnan AV. The mechanism of action of non-coding RNAs in placental disorders. Biomed Pharmacother 2022; 156:113964. [DOI: 10.1016/j.biopha.2022.113964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
|
17
|
Rong W, Shukun W, Xiaoqing W, Wenxin H, Mengyuan D, Chenyang M, Zhang H. Regulatory roles of non-coding RNAs and m6A modification in trophoblast functions and the occurrence of its related adverse pregnancy outcomes. Crit Rev Toxicol 2022; 52:681-713. [PMID: 36794364 DOI: 10.1080/10408444.2022.2144711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Adverse pregnancy outcomes, such as preeclampsia, gestational diabetes mellitus, fetal growth restriction, and recurrent miscarriage, occur frequently in pregnant women and might further induce morbidity and mortality for both mother and fetus. Increasing studies have shown that dysfunctions of human trophoblast are related to these adverse pregnancy outcomes. Recent studies also showed that environmental toxicants could induce trophoblast dysfunctions. Moreover, non-coding RNAs (ncRNAs) have been reported to play important regulatory roles in various cellular processes. However, the roles of ncRNAs in the regulation of trophoblast dysfunctions and the occurrence of adverse pregnancy outcomes still need to be further investigated, especially with exposure to environmental toxicants. In this review, we analyzed the regulatory mechanisms of ncRNAs and m6A methylation modification in the dysfunctions of trophoblast cells and the occurrence of adverse pregnancy outcomes and also summarized the harmful effects of environmental toxicants. In addition to DNA replication, mRNA transcription, and protein translation, ncRNAs and m6A modification might be considered as the fourth and fifth elements that regulate the genetic central dogma, respectively. Environmental toxicants might also affect these processes. In this review, we expect to provide a deeper scientific understanding of the occurrence of adverse pregnancy outcomes and to discover potential biomarkers for the diagnosis and treatment of these outcomes.
Collapse
Affiliation(s)
- Wang Rong
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Wan Shukun
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Wang Xiaoqing
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huang Wenxin
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Dai Mengyuan
- Department of Toxicology, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Mi Chenyang
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Huidong Zhang
- Key Laboratory of Environment and Female Reproductive Health, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
18
|
Non-Coding RNAs and Prediction of Preeclampsia in the First Trimester of Pregnancy. Cells 2022; 11:cells11152428. [PMID: 35954272 PMCID: PMC9368389 DOI: 10.3390/cells11152428] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 12/13/2022] Open
Abstract
Preeclampsia (PE) is a major cause of maternal and perinatal morbidity and mortality. The only fundamental treatment for PE is the termination of pregnancy. Therefore, not only severe maternal complications but also perinatal complications due to immaturity of the infant associated with early delivery are serious issues. The treatment and prevention of preterm onset preeclampsia (POPE) are challenging. In 2017, the ASPRE trial showed that a low oral dose of aspirin administered to POPE high-risk women in early pregnancy reduced POPE by 62%. A prediction algorithm at 11–13 weeks of gestation identifies POPE with 75% sensitivity when the false positive rate is set at 10%. New biomarkers to increase the accuracy of the prediction model for POPE high-risk women in early pregnancy are needed. In this review, we focused on non-coding RNAs (ncRNAs) as potential biomarkers for the prediction of POPE. Highly expressed ncRNAs in the placenta in early pregnancy may play crucial roles in placentation. Furthermore, placenta-specific ncRNAs have been detected in maternal blood. In this review, we summarized ncRNAs that were highly expressed in the primary human placenta in early pregnancy. We also presented highly expressed ncRNAs in the placenta that were associated with or predictive of the development of PE in an expression analysis of maternal blood during the first trimester of pregnancy. These previous studies showed that the chromosome 19 microRNA (miRNA) -derived miRNAs (e.g., miR-517-5p, miR-518b, and miR-520h), the hypoxia-inducible miRNA (miR-210), and long non-coding RNA H19, were not only highly expressed in the early placenta but were also significantly up-regulated in the blood at early gestation in pregnant women who later developed PE. These maternal circulating ncRNAs in early pregnancy are expected to be possible biomarkers for POPE.
Collapse
|
19
|
Schroeder M, Badini G, Sferruzzi-Perri AN, Albrecht C. The Consequences of Assisted Reproduction Technologies on the Offspring Health Throughout Life: A Placental Contribution. Front Cell Dev Biol 2022; 10:906240. [PMID: 35747691 PMCID: PMC9210138 DOI: 10.3389/fcell.2022.906240] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/05/2022] [Indexed: 11/13/2022] Open
Abstract
The use of assisted reproductive technologies (ART) worldwide has led to the conception and birth of over eight million babies since being implemented in 1978. ART use is currently on the rise, given growing infertility and the increase in conception age among men and women in industrialized countries. Though obstetric and perinatal outcomes have improved over the years, pregnancies achieved by ART still bear increased risks for the mother and the unborn child. Moreover, given that the first generation of ART offspring is now only reaching their forties, the long-term effects of ART are currently unknown. This is important, as there is a wealth of data showing that life-long health can be predetermined by poor conditions during intrauterine development, including irregularities in the structure and functioning of the placenta. In the current review, we aim to summarize the latest available findings examining the effects of ART on the cardiometabolic, cognitive/neurodevelopmental, and behavioral outcomes in the perinatal period, childhood and adolescence/adulthood; and to examine placental intrinsic factors that may contribute to the developmental outcomes of ART offspring. Altogether, the latest knowledge about life outcomes beyond adolescence for those conceived by ART appears to suggest a better long-term outcome than previously predicted. There are also changes in placenta structure and functional capacity with ART. However, more work in this area is critically required, since the potential consequences of ART may still emerge as the offspring gets older. In addition, knowledge of the placenta may help to foresee and mitigate any adverse outcomes in the offspring.
Collapse
Affiliation(s)
- Mariana Schroeder
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Gina Badini
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Amanda N. Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Christiane Albrecht
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| |
Collapse
|
20
|
Zhang Y, Wang S. The possible role of long non-coding RNAs in recurrent miscarriage. Mol Biol Rep 2022; 49:9687-9697. [PMID: 35397764 PMCID: PMC9515028 DOI: 10.1007/s11033-022-07427-9] [Citation(s) in RCA: 1] [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: 12/30/2021] [Accepted: 03/24/2022] [Indexed: 12/16/2022]
Abstract
Recurrent miscarriage (RM) is a complicated disease in reproductive medicine that impacts many families. Currently, the etiology of RM is thought to include chromosome abnormalities, reproductive tract malformations, autoimmune dysfunction, infection, and environmental factors. However, the underlying mechanisms of RM remain unknown. At present, research on long non-coding RNAs (lncRNAs) is rapidly emerging and becoming a hot research topic in epigenetic studies. Recent studies revealed that lncRNAs are strongly linked to RM and play a crucial role in epigenetic, cell cycle, cell differentiation regulation, and other life activities. This article mainly reviews the difference in lncRNA expression in patients with RM and regulation of susceptibility, endometrial receptivity, and the maternal-fetal interface. Meanwhile, the correlation between lncRNAs and RM is expounded, which provides new insights for the early diagnosis and treatment of RM.
Collapse
Affiliation(s)
- Yanan Zhang
- Shandong Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to, Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, China
| | - Shan Wang
- Shandong Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to, Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, China.
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, 324 Jingwu Road, Jinan, 250021, China.
| |
Collapse
|
21
|
Sun Y, Li T, Qian X. Biological Role of Nodal Modulator: A Comprehensive Review of the Last Two Decades. DNA Cell Biol 2022; 41:336-341. [PMID: 35133875 DOI: 10.1089/dna.2021.0944] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Nodal modulator (NOMO) is a type I transmembrane protein that is conserved in various human tissues. Humans have three highly similar NOMO proteins, namely NOMO1, NOMO2, and NOMO3. These three proteins are closely related and may have similar functions. NOMO has been identified as a part of a protein complex that mediates a wide range of biological processes such as tumor formation, bone and cartilage formation, embryo formation, facial asymmetry, and development of congenital heart disease. To date, a few studies have focused on the role of NOMO; however, the mechanism underlying its effects remains unknown. To improve our understanding regarding NOMO, we reviewed the role of NOMO in different diseases and investigated the mechanism underlying its effects.
Collapse
Affiliation(s)
- Yuhui Sun
- Department of Pediatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Tao Li
- Department of Pediatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, P.R. China
| | - Xin Qian
- Department of Pulmonary and Critical Care Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, P.R. China
| |
Collapse
|
22
|
Cannarella R, Mancuso F, Arato I, Lilli C, Bellucci C, Gargaro M, Curto R, Aglietti MC, La Vignera S, Condorelli RA, Luca G, Calogero AE. Sperm-carried IGF2 downregulated the expression of mitogens produced by Sertoli cells: A paracrine mechanism for regulating spermatogenesis? Front Endocrinol (Lausanne) 2022; 13:1010796. [PMID: 36523595 PMCID: PMC9744929 DOI: 10.3389/fendo.2022.1010796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 11/07/2022] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Insulin-like growth factor 2 (IGF2) mRNA has been found in human and mouse spermatozoa. It is currently unknown whether the IGF2 protein is expressed in human spermatozoa and, if so, its possible role in the cross-talk between germ and Sertoli cells (SCs) during spermatogenesis. METHODS To accomplish this, we analyzed sperm samples from four consecutive Caucasian men. Furthermore, to understand its role during the spermatogenetic process, porcine SCs were incubated with increasing concentrations (0.33, 3.33, and 10 ng/mL) of recombinant human IGF2 (rhIGF2) for 48 hours. Subsequently, the experiments were repeated by pre-incubating SCs with the non-competitive insulin-like growth factor 1 receptor (IGF1R) inhibitor NVP-AEW541. The following outcomes were evaluated: 1) Gene expression of the glial cell-line derived neurotrophic factor (GDNF), fibroblast growth factor 2 (FGF2), and stem cell factor (SCF) mitogens; 2) gene and protein expression of follicle-stimulating hormone receptor (FSHR), anti-Müllerian hormone (AMH), and inhibin B; 3) SC proliferation. RESULTS We found that the IGF2 protein was present in each of the sperm samples. IGF2 appeared as a cytoplasmic protein localized in the equatorial and post-acrosomal segment and with a varying degree of expression in each cell. In SCs, IGF2 significantly downregulated GDNF gene expression in a concentration-dependent manner. FGF2 and SCF were downregulated only by the highest concentration of IGF2. Similarly, IGF2 downregulated the FSHR gene and FSHR, AMH, and inhibin B protein expression. Finally, IGF2 significantly suppressed the SC proliferation rate. All these findings were reversed by pre-incubation with NVP-AEW541, suggesting an effect mediated by the interaction of IGF2 with the IGFR. CONCLUSION In conclusion, sperm IGF2 seems to downregulate the expression of mitogens, which are known to be physiologically released by the SCs to promote gonocyte proliferation and spermatogonial fate adoption. These findings suggest the presence of paracrine regulatory mechanisms acting on the seminiferous epithelium during spermatogenesis, by which germ cells can influence the amount of mitogens released by the SCs, their sensitivity to FSH, and their rate of proliferation.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- Glickman Urological & Kidney Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
- *Correspondence: Rossella Cannarella,
| | - Francesca Mancuso
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Iva Arato
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cinzia Lilli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Catia Bellucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marco Gargaro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Roberto Curto
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Maria C. Aglietti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A. Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Giovani Luca
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Aldo E. Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
23
|
Sun J, Wang R, Chao T, Wang C. Long Noncoding RNAs Involved in Cardiomyocyte Apoptosis Triggered by Different Stressors. J Cardiovasc Transl Res 2021; 15:588-603. [PMID: 34855148 DOI: 10.1007/s12265-021-10186-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/04/2021] [Indexed: 12/26/2022]
Abstract
Cardiomyocytes are essential to maintain the normal cardiac function. Ischemia, hypoxia, and drug stimulation can induce pathological apoptosis of cardiomyocytes which eventually leads to heart failure, arrhythmia, and other cardiovascular diseases. Understanding the molecular mechanisms that regulate cardiomyocyte apoptosis is of great significance for the prevention and treatment of cardiovascular diseases. In recent years, more and more evidences reveal that long noncoding RNAs (lncRNAs) play important regulatory roles in myocardial cell apoptosis. They can modulate the expression of apoptosis-related genes at post-transcriptional level by altering the translation efficacy of target mRNAs or functioning as a precursor for miRNAs or competing for miRNA-mediated inhibition. Moreover, reversing the abnormal expression of lncRNAs can attenuate and even reverse the pathological apoptosis of cardiomyocytes. Therefore, apoptosis-related lncRNAs may become a potential new field for studying cardiomyocyte apoptosis and provide new ideas for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Jinghui Sun
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ru Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Tiantian Chao
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenglong Wang
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
24
|
Munjas J, Sopić M, Stefanović A, Košir R, Ninić A, Joksić I, Antonić T, Spasojević-Kalimanovska V, Prosenc Zmrzljak U. Non-Coding RNAs in Preeclampsia-Molecular Mechanisms and Diagnostic Potential. Int J Mol Sci 2021; 22:10652. [PMID: 34638993 PMCID: PMC8508896 DOI: 10.3390/ijms221910652] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/25/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Preeclampsia (PE) is a leading cause of maternal and neonatal morbidity and mortality worldwide. Defects in trophoblast invasion, differentiation of extravillous trophoblasts and spiral artery remodeling are key factors in PE development. Currently there are no predictive biomarkers clinically available for PE. Recent technological advancements empowered transcriptome exploration and led to the discovery of numerous non-coding RNA species of which microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are the most investigated. They are implicated in the regulation of numerous cellular functions, and as such are being extensively explored as potential biomarkers for various diseases. Altered expression of numerous lncRNAs and miRNAs in placenta has been related to pathophysiological processes that occur in preeclampsia. In the following text we offer summary of the latest knowledge of the molecular mechanism by which lnRNAs and miRNAs (focusing on the chromosome 19 miRNA cluster (C19MC)) contribute to pathophysiology of PE development and their potential utility as biomarkers of PE, with special focus on sample selection and techniques for the quantification of lncRNAs and miRNAs in maternal circulation.
Collapse
Affiliation(s)
- Jelena Munjas
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Street Vojvode Stepe 450, 11000 Belgrade, Serbia; (J.M.); (M.S.); (A.S.); (A.N.); (T.A.); (V.S.-K.)
| | - Miron Sopić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Street Vojvode Stepe 450, 11000 Belgrade, Serbia; (J.M.); (M.S.); (A.S.); (A.N.); (T.A.); (V.S.-K.)
| | - Aleksandra Stefanović
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Street Vojvode Stepe 450, 11000 Belgrade, Serbia; (J.M.); (M.S.); (A.S.); (A.N.); (T.A.); (V.S.-K.)
| | - Rok Košir
- BIA Separations CRO, Labena Ltd., Street Verovškova 64, 1000 Ljubljana, Slovenia;
| | - Ana Ninić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Street Vojvode Stepe 450, 11000 Belgrade, Serbia; (J.M.); (M.S.); (A.S.); (A.N.); (T.A.); (V.S.-K.)
| | - Ivana Joksić
- Genetic Laboratory Department, Obstetrics and Gynaecology Clinic “Narodni Front”, Street Kraljice Natalije 62, 11000 Belgrade, Serbia;
| | - Tamara Antonić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Street Vojvode Stepe 450, 11000 Belgrade, Serbia; (J.M.); (M.S.); (A.S.); (A.N.); (T.A.); (V.S.-K.)
| | - Vesna Spasojević-Kalimanovska
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Street Vojvode Stepe 450, 11000 Belgrade, Serbia; (J.M.); (M.S.); (A.S.); (A.N.); (T.A.); (V.S.-K.)
| | | |
Collapse
|
25
|
Barberet J, Romain G, Binquet C, Guilleman M, Bruno C, Ginod P, Chapusot C, Choux C, Fauque P. Do frozen embryo transfers modify the epigenetic control of imprinted genes and transposable elements in newborns compared with fresh embryo transfers and natural conceptions? Fertil Steril 2021; 116:1468-1480. [PMID: 34538459 DOI: 10.1016/j.fertnstert.2021.08.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/02/2021] [Accepted: 08/06/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE To determine whether the epigenetic control of imprinted genes (IGs) and transposable elements (TEs) differs at birth between fresh or frozen embryo transfers and natural conceptions. DESIGN Prospective study. SETTING University hospital. PATIENT(S) A total of 202 singleton births were divided into three groups: 84 natural pregnancies (controls), 66 in vitro fertilization/intracytoplasmic sperm injection with fresh embryo transfers, and 52 vitro fertilization/intracytoplasmic sperm injection with frozen embryo transfers. INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Pyrosequencing was used to assess the DNA methylation profiles of three IGs (H19/IGF2:IG-DMR [two sequences], KCNQ1OT1:TSS-DMR, and SNURF:TSS-DMR) and two TEs (LINE-1 and HERV-FRD) in cord blood and placenta. The quantitative reverse transcriptase polymerase chain reaction was used to study the transcription of three IGs (H19, KCNQ1, and SNRPN) and two TEs (LINE-1 and ORF2). RESULT(S) After adjustment, the placental DNA methylation levels of H19/IGF2 were lower in the fresh embryo transfer group than in the control (H19/IGF2-seq1) and frozen embryo transfer (H19/IGF2-seq2) groups. The DNA methylation rate for LINE-1 was lower in placentas from the fresh embryo transfer group than in placentas from the control and frozen embryo transfer groups and for HERV-FRD compared with controls. In cord blood, DNA methylation levels were not significantly associated with the mode of conception. The relative expression of LINE-1 and ORF2 was decreased in both cord blood and placental tissues from fresh embryo transfer conceptions compared with natural conceptions and frozen embryo transfer conceptions. CONCLUSION(S) Compared with natural conceptions and frozen embryo transfers, fresh embryo transfers were associated with methylation and/or transcription changes in some TEs and IGs, mostly in placental samples, which could indicate altered placental epigenetic regulation resulting from ovarian stimulation protocols.
Collapse
Affiliation(s)
- Julie Barberet
- Université Bourgogne Franche-Comté-Equipe Génétique des Anomalies du Développement (GAD), INSERM UMR1231, Dijon, France; Centre Hospitalier Universitaire Dijon-Bourgogne, Laboratoire de Biologie de la Reproduction-CECOS, Dijon, France
| | - Gaelle Romain
- Centre Hospitalier Universitaire Dijon-Bourgogne, Centre d'Investigation Clinique, Module Epidémiologie Clinique/Essais Cliniques (CIC-EC), Dijon, France; INSERM, CIC1432, Module Epidémiologie Clinique, Dijon, France
| | - Christine Binquet
- Centre Hospitalier Universitaire Dijon-Bourgogne, Centre d'Investigation Clinique, Module Epidémiologie Clinique/Essais Cliniques (CIC-EC), Dijon, France; INSERM, CIC1432, Module Epidémiologie Clinique, Dijon, France
| | - Magali Guilleman
- Université Bourgogne Franche-Comté-Equipe Génétique des Anomalies du Développement (GAD), INSERM UMR1231, Dijon, France; Centre Hospitalier Universitaire Dijon-Bourgogne, Laboratoire de Biologie de la Reproduction-CECOS, Dijon, France
| | - Céline Bruno
- Université Bourgogne Franche-Comté-Equipe Génétique des Anomalies du Développement (GAD), INSERM UMR1231, Dijon, France; Centre Hospitalier Universitaire Dijon-Bourgogne, Laboratoire de Biologie de la Reproduction-CECOS, Dijon, France
| | - Perrine Ginod
- Université Bourgogne Franche-Comté-Equipe Génétique des Anomalies du Développement (GAD), INSERM UMR1231, Dijon, France; Centre Hospitalier Universitaire Dijon-Bourgogne, Service de Gynécologie-Obstétrique, Dijon, France
| | - Caroline Chapusot
- Centre Hospitalier Universitaire Dijon-Bourgogne, Plateforme de Génétique des Cancers de Bourgogne, Dijon, France
| | - Cécile Choux
- Université Bourgogne Franche-Comté-Equipe Génétique des Anomalies du Développement (GAD), INSERM UMR1231, Dijon, France; Centre Hospitalier Universitaire Dijon-Bourgogne, Service de Gynécologie-Obstétrique, Dijon, France
| | - Patricia Fauque
- Université Bourgogne Franche-Comté-Equipe Génétique des Anomalies du Développement (GAD), INSERM UMR1231, Dijon, France; Centre Hospitalier Universitaire Dijon-Bourgogne, Laboratoire de Biologie de la Reproduction-CECOS, Dijon, France.
| |
Collapse
|
26
|
Abstract
Cardiac hypertrophy (CH) is generally considered adaptive responses that may occur after myocardial infarction, pressure overload, volume overload, inflammatory heart muscle disease, or idiopathic dilated cardiomyopathy, whereas long-term stimulation eventually leads to heart failure (HF). However, the current molecular mechanisms involved in CH are unclear. Recently, increasing evidences reveal that long non-coding RNAs (lncRNAs) play vital roles in CH. Different lncRNAs can promote or inhibit the pathological process of CH by different mechanisms, while the regulation of lncRNAs expression can improve CH. Thus, CH-related lncRNAs may become a novel field of research on CH.
Collapse
Affiliation(s)
- Jinghui Sun
- Cardiovascular Disease Research Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Haidian District, Beijing, 100091, China
| | - Chenglong Wang
- Cardiovascular Disease Research Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Haidian District, Beijing, 100091, China.
| |
Collapse
|
27
|
Abstract
Preeclampsia (PE) is an idiopathic disease that occurs during pregnancy. It comprises multiple organ and system damage, and can seriously threaten the safety of the mother and infant throughout the perinatal period. As the pathogenesis of PE is unclear, there are few specific remedies. Currently, the only way to eliminate the clinical symptoms is to terminate the pregnancy. Although noncoding RNA (ncRNA) was once thought to be the "junk" of gene transcription, it is now known to be widely involved in pathological and physiological processes, including pregnancy-related disorders. Moreover, there is growing evidence that the unbalanced expression of specific ncRNA is involved in the pathogenesis of PE. In the present review, we summarize the expression patterns of ncRNAs, i.e., microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), and the functional mechanisms by which they affect the development of PE, and examine the clinical significance of ncRNAs as biomarkers for the diagnosis of PE. We also discuss the contributions made by genetic polymorphisms and epigenetic ncRNA regulation to PE. In the present review, we wish to explore and reinforce the clinical value of ncRNAs as noninvasive biomarkers of PE.
Collapse
Affiliation(s)
- Ningxia Sun
- Department of Medical Genetic, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
- Department of Gynecology and obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Shiting Qin
- Department of Medical Genetic, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Lu Zhang
- Department of Medical Genetic, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China.
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Shiguo Liu
- Department of Medical Genetic, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, China.
- Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
28
|
The Role of Long Non-Coding RNAs in Trophoblast Regulation in Preeclampsia and Intrauterine Growth Restriction. Genes (Basel) 2021; 12:genes12070970. [PMID: 34201957 PMCID: PMC8305149 DOI: 10.3390/genes12070970] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/06/2021] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
Preeclampsia (PE) and Intrauterine Growth Restriction (IUGR) are two pregnancy-specific placental disorders with high maternal, fetal, and neonatal morbidity and mortality rates worldwide. The identification biomarkers involved in the dysregulation of PE and IUGR are fundamental for developing new strategies for early detection and management of these pregnancy pathologies. Several studies have demonstrated the importance of long non-coding RNAs (lncRNAs) as essential regulators of many biological processes in cells and tissues, and the placenta is not an exception. In this review, we summarize the importance of lncRNAs in the regulation of trophoblasts during the development of PE and IUGR, and other placental disorders.
Collapse
|
29
|
Adu-Gyamfi EA, Ding YB, Wang YX. Regulation of placentation by the transforming growth factor beta superfamily†. Biol Reprod 2021; 102:18-26. [PMID: 31566220 DOI: 10.1093/biolre/ioz186] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/18/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022] Open
Abstract
During pregnancy, there is increased expression of some cytokines at the fetal-maternal interface; and the clarification of their roles in trophoblast-endometrium interactions is crucial to understanding the mechanism of placentation. This review addresses the up-to-date reported mechanisms by which the members of the transforming growth factor beta superfamily regulate trophoblast proliferation, differentiation, and invasion of the decidua, which are the main phases of placentation. The available information shows that these cytokines regulate placentation in somehow a synergistic and an antagonistic manner; and that dysregulation of their levels can lead to aberrant placentation. Nevertheless, prospective studies are needed to reconcile some conflicting reports; and identify some unknown mediators involved in the actions of these cytokines before their detailed mechanistic regulation of human placentation could be fully characterized. The TGF beta superfamily are expressed in the placenta, and regulate the process of placentation through the activation of several signaling pathways.
Collapse
Affiliation(s)
- Enoch Appiah Adu-Gyamfi
- Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yu-Bin Ding
- Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| | - Ying-Xiong Wang
- Department of Reproductive Sciences, School of Public Health, Chongqing Medical University, Chongqing, People's Republic of China.,Joint International Research Laboratory of Reproduction & Development, Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
30
|
Cannarella R, Crafa A, Condorelli RA, Mongioì LM, La Vignera S, Calogero AE. Relevance of sperm imprinted gene methylation on assisted reproductive technique outcomes and pregnancy loss: a systematic review. Syst Biol Reprod Med 2021; 67:251-259. [PMID: 34080930 DOI: 10.1080/19396368.2021.1909667] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Recent evidence suggests that gamete-imprinted genes play a role in embryo and placenta development and growth. This systematic review aimed to evaluate whether altered methylation of sperm-imprinted genes associates with sperm DNA fragmentation (SDF), pregnancy loss rate and assisted reproductive technique (ART) outcome. To accomplish this, Pubmed, MEDLINE, Cochrane, Academic One Files, Google Scholar, and Scopus databases were used for search strategy from each database inception until December 2020. Specific keywords were used. Studies satisfying the PECOS (Population, Exposure, Comparison/Comparator, Outcomes, Study design) model were retrieved. Ten studies could be included in the qualitative analysis. A significant association was reported between increased SDF rate and aberrant methylation of H19/IGF2 and KCNQ1 genes by two studies. A significantly lower H19 methylation was found in patients with idiopathic recurrent pregnancy loss (RPL) and in infertile patients compared to fertile men. Methylation of GLT2, PEG1/MEST, and ZAC/PLACL1 were similar in patients with RPL and controls. The ART outcome was similar in patients with aberrant and normal methylation of H19, SNRPN, KCNQ1OT1, PEG1/MEST, LIT1, PEG3, NESPAS, and GLT2. By contrast, a study showed an association between altered GLT2 methylation and more inferior ART results. If further confirmed by well-sized studies, these data might be helpful to identify possible epigenetic predictors of ART outcome. Particularly, aberrant methylation of H19/IGF2 and KCNQ1 genes might represent interesting targets that deserve further investigation.
Collapse
Affiliation(s)
- Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Rosita A Condorelli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Laura M Mongioì
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
31
|
He X, Tao L, Zhong Y, Di R, Xia Q, Wang X, Guo X, Gan S, Zhang X, Zhang J, Liu Q, Chu M. Photoperiod induced the pituitary differential regulation of lncRNAs and mRNAs related to reproduction in sheep. PeerJ 2021; 9:e10953. [PMID: 33976954 PMCID: PMC8067910 DOI: 10.7717/peerj.10953] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 01/27/2021] [Indexed: 12/12/2022] Open
Abstract
The pituitary is a vital endocrine organ that regulates animal seasonal reproduction by controlling the synthesis and secretion of the hormone. The change of photoperiod is the key factor affecting the function of the pituitary in animals, but the mechanism is unclear. Here, we studied the transcriptomic variation in pars distalis (PD) of the pituitary between short photoperiod (SP) and long photoperiod (LP) using RNA sequencing based on the OVX+E2 sheep. 346 differentially expressed (DE) lncRNAs and 186 DE-mRNA were found in the PD. Moreover, function annotation analysis indicated that the reproductive hormones and photoperiod response-related pathways including aldosterone synthesis and secretion, insulin secretion, thyroid hormone synthesis, and circadian entrainment were enriched. The interaction analysis of mRNA-lncRNA suggested that MSTRG.240648, MSTRG.85500, MSTRG.32448, and MSTRG.304959 targeted CREB3L1 and DUSP6, which may be involved in the photoperiodic regulation of the PD. These findings provide resources for further study on the seasonal reproductive in ewes.
Collapse
Affiliation(s)
- Xiaoyun He
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lin Tao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yingjie Zhong
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ran Di
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qing Xia
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiangyu Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaofei Guo
- Tianjin Institute of Animal Sciences, Tianjin, China
| | - Shangquan Gan
- Xinjiang Academy of Agricultural and Reclamation Sciences, Xinjiang, China
| | | | - Jinlong Zhang
- Tianjin Institute of Animal Sciences, Tianjin, China
| | - Qiuyue Liu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mingxing Chu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
32
|
Xu P, Ma Y, Wu H, Wang YL. Placenta-Derived MicroRNAs in the Pathophysiology of Human Pregnancy. Front Cell Dev Biol 2021; 9:646326. [PMID: 33777951 PMCID: PMC7991791 DOI: 10.3389/fcell.2021.646326] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
In placental mammals, reproductive success, and maternal-fetal health substantially depend on a well-being placenta, the interface between the fetus and the mother. Disorders in placental cells are tightly associated with adverse pregnancy outcomes including preeclampsia (PE), fetal growth restriction, etc. MicroRNAs (miRNAs) represent small non-coding RNAs that regulate post-transcriptional gene expression and are integral to a wide range of healthy or diseased cellular proceedings. Numerous miRNAs have been detected in human placenta and increasing evidence is revealing their important roles in regulating placental cell behaviors. Recent studies indicate that placenta-derived miRNAs can be released to the maternal circulation via encapsulating into the exosomes, and they potentially target various maternal cells to provide a hormone-like means of intercellular communication between the mother and the fetus. These placental exosome miRNAs are attracting more and more attention due to their differential expression in pregnant complications, which may provide novel biomarkers for prediction of the diseases. In this review, we briefly summarize the current knowledge and the perspectives of the placenta-derived miRNAs, especially the exosomal transfer of placental miRNAs and their pathophysiological relevance to PE. The possible exosomal-miRNA-targeted strategies for diagnosis, prognosis or therapy of PE are highlighted.
Collapse
Affiliation(s)
- Peng Xu
- School of Life Science, Shanxi University, Taiyuan, China
| | - Yeling Ma
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Hongyu Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Yan-Ling Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
33
|
Li C, He X, Zhang Z, Ren C, Chu M. Pineal gland transcriptomic profiling reveals the differential regulation of lncRNA and mRNA related to prolificacy in STH sheep with two FecB genotypes. BMC Genom Data 2021; 22:9. [PMID: 33602139 PMCID: PMC7893892 DOI: 10.1186/s12863-020-00957-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/16/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Long noncoding RNA (lncRNA) has been identified as important regulator in hypothalamic-pituitary-ovarian axis associated with sheep prolificacy. However, little is known of their expression pattern and potential roles in the pineal gland of sheep. Herein, RNA-Seq was used to detect transcriptome expression pattern in pineal gland between follicular phase (FP) and luteal phase (LP) in FecBBB (MM) and FecB++ (ww) STH sheep, respectively, and differentially expressed (DE) lncRNAs and mRNAs associated with reproduction were identified. RESULTS Overall, 135 DE lncRNAs and 1360 DE mRNAs in pineal gland between MM and ww sheep were screened. Wherein, 39 DE lncRNAs and 764 DE mRNAs were identified (FP vs LP) in MM sheep, 96 DE lncRNAs and 596 DE mRNAs were identified (FP vs LP) in ww sheep. Moreover, GO and KEGG enrichment analysis indicated that the targets of DE lncRNAs and DE mRNAs were annotated to multiple biological processes such as phototransduction, circadian rhythm, melanogenesis, GSH metabolism and steroid biosynthesis, which directly or indirectly participate in hormone activities to affect sheep reproductive performance. Additionally, co-expression of lncRNAs-mRNAs and the network construction were performed based on correlation analysis, DE lncRNAs can modulate target genes involved in related pathways to affect sheep fecundity. Specifically, XLOC_466330, XLOC_532771, XLOC_028449 targeting RRM2B and GSTK1, XLOC_391199 targeting STMN1, XLOC_503926 targeting RAG2, XLOC_187711 targeting DLG4 were included. CONCLUSION All of these differential lncRNAs and mRNAs expression profiles in pineal gland provide a novel resource for elucidating regulatory mechanism underlying STH sheep prolificacy.
Collapse
Affiliation(s)
- Chunyan Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.,College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Xiaoyun He
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Zijun Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Chunhuan Ren
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, 230036, China
| | - Mingxing Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Ministry of Agriculture and Rural Affairs, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China.
| |
Collapse
|
34
|
Nuh AM, You Y, Ma M. Information on dysregulation of microRNA in placenta linked to preeclampsia. Bioinformation 2021; 17:240-248. [PMID: 34393443 PMCID: PMC8340720 DOI: 10.6026/97320630017240] [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: 12/17/2020] [Revised: 01/30/2021] [Accepted: 01/31/2021] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs are single-stranded, non-coding RNA molecules, regulate gene expression at the post-transcriptional level. They are expressed in the human body and have a significant impact on the different processes of pathological illness. A developing placenta undergoes a series of stages after successful fertilization, such as cell division, migration, adhesion, apoptosis, and angiogenesis. MicroRNAs dysregulation in placenta has been linked to pregnancy-related complications such as preeclampsia. Therefore, it is of interest to document known information (list of microRNA) on this issue in the development of biological tools for diagnosis, treatment and prevention of the disease.
Collapse
Affiliation(s)
- Abdifatah Mohamed Nuh
- Department of Obstetrics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, 225000, China
- Yangzhou University Medical College, Yangzhou, Jiangsu Province, 225000, China
| | - Yan You
- Department of Obstetrics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, 225000, China
- Yangzhou University Medical College, Yangzhou, Jiangsu Province, 225000, China
| | - Min Ma
- Department of Obstetrics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu Province, 225000, China
- Yangzhou University Medical College, Yangzhou, Jiangsu Province, 225000, China
| |
Collapse
|
35
|
Ogoyama M, Ohkuchi A, Takahashi H, Zhao D, Matsubara S, Takizawa T. LncRNA H19-Derived miR-675-5p Accelerates the Invasion of Extravillous Trophoblast Cells by Inhibiting GATA2 and Subsequently Activating Matrix Metalloproteinases. Int J Mol Sci 2021; 22:ijms22031237. [PMID: 33513878 PMCID: PMC7866107 DOI: 10.3390/ijms22031237] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/18/2021] [Accepted: 01/21/2021] [Indexed: 12/13/2022] Open
Abstract
The invasion of extravillous trophoblast (EVT) cells into the maternal decidua, which plays a crucial role in the establishment of a successful pregnancy, is highly orchestrated by a complex array of regulatory mechanisms. Non-coding RNAs (ncRNAs) that fine-tune gene expression at epigenetic, transcriptional, and post-transcriptional levels are involved in the regulatory mechanisms of EVT cell invasion. However, little is known about the characteristic features of EVT-associated ncRNAs. To elucidate the gene expression profiles of both coding and non-coding transcripts (i.e., mRNAs, long non-coding RNAs (lncRNAs), and microRNAs (miRNAs)) expressed in EVT cells, we performed RNA sequencing analysis of EVT cells isolated from first-trimester placentae. RNA sequencing analysis demonstrated that the lncRNA H19 and its derived miRNA miR-675-5p were enriched in EVT cells. Although miR-675-5p acts as a placental/trophoblast growth suppressor, there is little information on the involvement of miR-675-5p in trophoblast cell invasion. Next, we evaluated a possible role of miR-675-5p in EVT cell invasion using the EVT cell lines HTR-8/SVneo and HChEpC1b; overexpression of miR-675-5p significantly promoted the invasion of both EVT cell lines. The transcription factor gene GATA2 was shown to be a target of miR-675-5p; moreover, small interfering RNA-mediated GATA2 knockdown significantly promoted cell invasion. Furthermore, we identified MMP13 and MMP14 as downstream effectors of miR-675-5p/GATA2-dependent EVT cell invasion. These findings suggest that miR-675-5p-mediated GATA2 inhibition accelerates EVT cell invasion by upregulating matrix metalloproteinases.
Collapse
Affiliation(s)
- Manabu Ogoyama
- Department of Obstetrics and Gynecology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi 329-0498, Japan; (M.O.); (A.O.); (H.T.); (S.M.)
- Department of Molecular Medicine and Anatomy, Nippon Medical School, 1-1-5 Sendagi, Tokyo 113-8602, Japan;
| | - Akihide Ohkuchi
- Department of Obstetrics and Gynecology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi 329-0498, Japan; (M.O.); (A.O.); (H.T.); (S.M.)
| | - Hironori Takahashi
- Department of Obstetrics and Gynecology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi 329-0498, Japan; (M.O.); (A.O.); (H.T.); (S.M.)
| | - Dongwei Zhao
- Department of Molecular Medicine and Anatomy, Nippon Medical School, 1-1-5 Sendagi, Tokyo 113-8602, Japan;
| | - Shigeki Matsubara
- Department of Obstetrics and Gynecology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi 329-0498, Japan; (M.O.); (A.O.); (H.T.); (S.M.)
| | - Toshihiro Takizawa
- Department of Molecular Medicine and Anatomy, Nippon Medical School, 1-1-5 Sendagi, Tokyo 113-8602, Japan;
- Correspondence: ; Tel.: +81-3-3822-2131
| |
Collapse
|
36
|
Massaro C, Safadeh E, Sgueglia G, Stunnenberg HG, Altucci L, Dell’Aversana C. MicroRNA-Assisted Hormone Cell Signaling in Colorectal Cancer Resistance. Cells 2020; 10:cells10010039. [PMID: 33396628 PMCID: PMC7823834 DOI: 10.3390/cells10010039] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 12/26/2020] [Accepted: 12/28/2020] [Indexed: 12/17/2022] Open
Abstract
Despite substantial progress in cancer therapy, colorectal cancer (CRC) is still the third leading cause of cancer death worldwide, mainly due to the acquisition of resistance and disease recurrence in patients. Growing evidence indicates that deregulation of hormone signaling pathways and their cross-talk with other signaling cascades inside CRC cells may have an impact on therapy resistance. MicroRNAs (miRNAs) are small conserved non-coding RNAs thatfunction as negative regulators in many gene expression processes. Key studies have identified miRNA alterations in cancer progression and drug resistance. In this review, we provide a comprehensive overview and assessment of miRNAs role in hormone signaling pathways in CRC drug resistance and their potential as future targets for overcoming resistance to treatment.
Collapse
Affiliation(s)
- Crescenzo Massaro
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio, 7, 80138 Naples, Italy; (C.M.); (E.S.); (G.S.)
| | - Elham Safadeh
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio, 7, 80138 Naples, Italy; (C.M.); (E.S.); (G.S.)
| | - Giulia Sgueglia
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio, 7, 80138 Naples, Italy; (C.M.); (E.S.); (G.S.)
| | | | - Lucia Altucci
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio, 7, 80138 Naples, Italy; (C.M.); (E.S.); (G.S.)
- Correspondence: (L.A.); (C.D.); Tel.: +39-081-566-7564 (L.A.); +39-081-566-7566 (C.D.)
| | - Carmela Dell’Aversana
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via De Crecchio, 7, 80138 Naples, Italy; (C.M.); (E.S.); (G.S.)
- Institute of Experimental Endocrinology and Oncology “Gaetano Salvatore” (IEOS)-National Research Council (CNR), Via Sergio Pansini 5, 80131 Naples, Italy
- Correspondence: (L.A.); (C.D.); Tel.: +39-081-566-7564 (L.A.); +39-081-566-7566 (C.D.)
| |
Collapse
|
37
|
Xu P, Li Z, Wang Y, Yu X, Shao X, Li YX, Peng C, Zhao Y, Wang YL. miR-18a Contributes to Preeclampsia by Downregulating Smad2 (Full Length) and Reducing TGF-β Signaling. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:542-556. [PMID: 33230456 PMCID: PMC7566009 DOI: 10.1016/j.omtn.2020.09.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 09/17/2020] [Indexed: 01/07/2023]
Abstract
The study investigated the regulation of Smad2 by miR-18a and its role in preeclampsia (PE). Bioinformatics analysis showed that both Smad2 and Smad3 were the predicted targets for miR-18a. Mass spectrum analysis showed that two mature Smad2 isoforms existed in human placenta: full length, Smad2(FL), and that lacking exon3, Smad2(Δexon3). The protein level of Smad2(FL), but not Smad2(Δexon3) or Smad3, was significantly increased in severe PE (sPE) placenta, which was inversely correlated with the level of miR-18a. Elevated Smad2(FL) phosphorylation level appeared in sPE placenta, and Smad2 was colocalized with miR-18a in various subtypes of trophoblasts in human placenta. Smad2(FL) was validated as the direct target of miR-18a in HTR8/SVneo cells. miR-18a enhanced trophoblast cell invasion, which was blocked by the overexpression of Smad2(FL). Furthermore, overexpression of miR-18a repressed Smad2 activation and the inhibition of trophoblast cell invasion by transforming growth factor-β (TGF-β). In conclusion, our results suggest that miR-18a inhibits the expression of Smad2(FL), but not Smad2(Δexon3) or Smad3, which can reduce TGF-β signaling, leading to the enhancement of trophoblast cell invasion. A lack of miR-18a, which results in the upregulation of Smad2(FL), contributes to the development of PE.
Collapse
Affiliation(s)
- Peng Xu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- School of Life Science, Shanxi University, Taiyuan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhilang Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yongqing Wang
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Xin Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xuan Shao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu-xia Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chun Peng
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Yangyu Zhao
- Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
| | - Yan-Ling Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
38
|
Xu P, Li Z, Wang Y, Yu X, Shao X, Li YX, Peng C, Zhao Y, Wang YL. miR-18a Contributes to Preeclampsia by Downregulating Smad2 (Full Length) and Reducing TGF-β Signaling. MOLECULAR THERAPY - NUCLEIC ACIDS 2020; 22:542-556. [DOI: pmid: 33230456 doi: 10.1016/j.omtn.2020.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
|
39
|
Xu X, Zhang Y, Li J, Mao B. Urothelial cancer associated 1 (UCA1) regulates trophoblast viability, proliferation, and migration via modulating the UCA1/miR-455/RUNX2 signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2020; 52:1120-1130. [PMID: 33085763 DOI: 10.1093/abbs/gmaa096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 07/24/2020] [Accepted: 10/15/2020] [Indexed: 12/25/2022] Open
Abstract
Spontaneous abortion (SA) is the spontaneous loss of a pregnancy before 20 gestational weeks. The causes of SA are still largely unknown. Long noncoding RNA (lncRNA) urothelial cancer associated 1 (UCA1) plays an important role in cellular progress. However, there is no report focusing on the role of UCA1 in SA. Here, we revealed that, compared with that in clinical samples from elective induced abortion, UCA1 expression was decreased in samples from SA patients as shown by qPCR method. The results demonstrated that UCA1 might be involved in the progress of SA. Then, we found that knockdown of UCA1 reduced cell viability and inhibited cell proliferation and migration of HTR-8/SVneo trophoblast cells as shown by CCK8, EdU, and Transwell methods. Furthermore, we demonstrated that UCA1 could act as a molecular sponge for miR-455 in HTR-8/SVneo cells as shown by luciferase reporter system method. In addition, miR-455 inhibited cell viability, cell proliferation and migration via regulating RUNX2 in HTR-8/SVneo cells. Ultimately, we illustrated that UCA1 plays its role via absorbing miR-455, thus promoting RUNX2 expression in HTR-8/SVneo cells. Collectively, this study first revealed the role and mechanism of UCA1 in the growth and migration of HTR-8/SVneo cells, indicating its potential as a diagnostic biomarker and therapeutic target for SA.
Collapse
Affiliation(s)
- Xiaoying Xu
- Perinatal Medicine center, Gansu Provincial Maternal and Child Health Care Hospital, Lanzhou 730050, China
| | - Yufang Zhang
- Perinatal Medicine center, Gansu Provincial Maternal and Child Health Care Hospital, Lanzhou 730050, China
| | - Jing Li
- Perinatal Medicine center, Gansu Provincial Maternal and Child Health Care Hospital, Lanzhou 730050, China
| | - Baohong Mao
- Perinatal Medicine center, Gansu Provincial Maternal and Child Health Care Hospital, Lanzhou 730050, China
| |
Collapse
|
40
|
Xing W, Li T, Wang Y, Qiang Y, Ai C, Tang H. MiR-33a-5p targets NOMO1 to modulate human cardiomyocyte progenitor cells proliferation and differentiation and apoptosis. J Recept Signal Transduct Res 2020; 41:476-487. [PMID: 33054489 DOI: 10.1080/10799893.2020.1825492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE MicroRNA (miRNA) is known to be involved in the pathological process of congenital heart disease (CHD), and nodal modulator1 (NOMO1) is a critical determinant of heart formation. The present study aims to discover the effect of miR-33a-5p and NOMO1 on CHD. METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was used to detect expressions of miR-33a-5p mimic or inhibitor and overexpressed NOMO1 plasmid orNOMO1 knockdown. Human cardiomyocyte progenitor cells (hCMPCs) proliferation was measured by cell counting kit-8 (CCK-8) at 24, 48 and 72 h. Flow cytometry was applied to investigate hCMPCs cell cycle progression and apoptosis. Expressions of cell apoptotic proteins Bax, Cleaved(C) caspase-3 and Bcl-2, and expressions of cardiomyocyte differentiation markers GATA4, troponin T (cTnT) and myocyte enhancer factor2C (MEF2C) in hCMPCs were identified by qRT-PCR and western blot. Target genes and potential binding sites of NOMO1 and miR-33a-5p were predicted with Targetscan 7.2, and was confirmed through dual-luciferase reporter assay. RESULTS Up-regulation of miR-33a-5p inhibited hCMPCs proliferation, cell cycle G0/S transition but promoted hCMPCs apoptosis, which was partially mitigated by overexpressed NOMO1. NOMO1 was the target gene of miR-33a-5p. Expressions of Bax and C caspase-3 were enhanced but expressions of Bcl-2, GATA4, cTnT and MEF2C were reduced by up-regulation of miR-33a-5p, which was partially mitigated by overexpressed NOMO1. CONCLUSION Up-regulation of miR-33a-5p inhibited hCMPCs proliferation, cell cycle G0/S transition and differentiation into cardiomyocytes but promoted apoptosis via targeting NOMO1.
Collapse
Affiliation(s)
- Wang Xing
- Cardiovascular Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou City, China
| | - Tiangang Li
- Department of Ultrasonography, Gansu Provincial Maternity and Child-care Hospital, Lanzhou City, China
| | - Yixuan Wang
- Department of Ultrasonography, Gansu Provincial Maternity and Child-care Hospital, Lanzhou City, China
| | - Yi Qiang
- Cardiovascular Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou City, China
| | - Chencheng Ai
- Cardiovascular Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou City, China
| | - Hanbo Tang
- Cardiovascular Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou City, China
| |
Collapse
|
41
|
Ghafouri-Fard S, Shoorei H, Taheri M. Role of Non-coding RNAs in the Pathogenesis of Endometriosis. Front Oncol 2020; 10:1370. [PMID: 32850438 PMCID: PMC7417625 DOI: 10.3389/fonc.2020.01370] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 06/29/2020] [Indexed: 12/23/2022] Open
Abstract
Endometriosis is a disorder characterized by the presence of endometrial glands and stroma like lesions outside of the uterus. Although several hypothesis have tried to explain the underlying cause of endometriosis, yet the main cause remained obscure. Recent studies have shown contribution of non-coding RNAs in the pathogenesis of endometriosis. Two classes of these transcripts namely long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) have mostly attracted attention of researchers. Several studies have reported aberrant expression of these transcripts in affected tissues from patients as well as animal models. Modulation of important signaling pathways such as PI3K/AKT, P38-MAPK, ERK1/2-MAPK and Wnt-β catenin by miRNAs and lncRNAs have potentiated these molecules as biomarkers or therapeutic agents in endometriosis. Single nucleotide polymorphisms with miR-126, miR-143 and miR-146b have been associated with risk of endometriosis. Moreover, miRNAs and lncRNAs control inflammatory responses, cell proliferation, angiogenesis and tissue remodeling, thus understanding the role of these transcripts in endometriosis is a possible way to develop novel diagnostic tests and therapeutic targets for this disorder.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Alipoor B, Parvar SN, Sabati Z, Ghaedi H, Ghasemi H. An updated review of the H19 lncRNA in human cancer: molecular mechanism and diagnostic and therapeutic importance. Mol Biol Rep 2020; 47:6357-6374. [PMID: 32743775 DOI: 10.1007/s11033-020-05695-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/26/2020] [Indexed: 12/24/2022]
Abstract
Accumulating evidence has reported that H19 long non-coding RNA (lncRNA) expression level is deregulated in human cancer. It has been also demonstrated that de-regulated levels of H19 could affect cancer biology by various mechanisms including microRNA (miRNA) production (like miR-675), miRNA sponging and epigenetic modifications. Furthermore, lncRNA could act as a potential diagnosis and prognosis biomarkers and also a candidate therapeutic approach for different human cancers. In this narrative review, we shed light on the molecular mechanism of H19 in cancer development and pathogenesis. Moreover, we discussed the expression pattern and diagnostic and therapeutic importance of H19 as a potential biomarker in a range of human malignancies from breast to osteosarcoma cancer.
Collapse
Affiliation(s)
- Behnam Alipoor
- Department of Laboratory Sciences, Faculty of Paramedicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Seyedeh Nasrin Parvar
- Department of Biochemistry, Faculty of Medicine, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Zolfaghar Sabati
- Student Research Committee, Abadan Faculty of Medical Sciences, Abadan, Iran
| | - Hamid Ghaedi
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Ghasemi
- Department of Clinical Biochemistry, Abadan Faculty of Medical Sciences, Abadan, Iran.
| |
Collapse
|
43
|
Association of lncRNA SH3PXD2A-AS1 with preeclampsia and its function in invasion and migration of placental trophoblast cells. Cell Death Dis 2020; 11:583. [PMID: 32719429 PMCID: PMC7385659 DOI: 10.1038/s41419-020-02796-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that the pathogenesis of preeclampsia involves poor placentation caused by insufficient trophoblast invasion and impaired uterine spiral artery remodeling, yet the underlying molecular mechanism remains unclear. We carried out transcriptome profiling on placentae from preeclamptic patients and normal subjects, and identified about four hundred long non-coding RNAs differentially expressed in placentae of patients with early-onset severe preeclampsia. Here, we report our identification of lncRNA SH3PXD2A-AS1 as a potential causal factor for this disease and its downstream pathways involved in placentation. We found that expression level of SH3PXD2A-AS1 in the placentae is positively correlated with clinical severity of the patients. We demonstrated that SH3PXD2A-AS1 inhibited invasion and migration through recruiting CCCTC-binding factor (CTCF) to the promoters of SH3PXD2A and CCR7 to inhibit their transcription. Therefore, we conclude that the upregulation of lncRNA SH3PXD2A-AS1 may contribute to the pathogenesis of preeclampsia through prohibiting trophoblast invasion during placentation.
Collapse
|
44
|
Human amnion-derived mesenchymal stem cells promote osteogenic differentiation of human bone marrow mesenchymal stem cells via H19/miR-675/APC axis. Aging (Albany NY) 2020; 12:10527-10543. [PMID: 32434960 PMCID: PMC7346082 DOI: 10.18632/aging.103277] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/17/2020] [Indexed: 12/29/2022]
Abstract
Bone volume inadequacy is an emerging clinical problem impairing the feasibility and longevity of dental implants. Human bone marrow mesenchymal stem cells (HBMSCs) have been widely used in bone remodeling and regeneration. This study examined the effect of long noncoding RNAs (lncRNAs)-H19 on the human amnion-derived mesenchymal stem cells (HAMSCs)-droved osteogenesis in HBMSCs. HAMSCs and HBMSCs were isolated from abandoned amniotic membrane samples and bone marrow. The coculture system was conducted using transwells, and H19 level was measured by quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR). The mechanism was further verified. We here discovered that osteogenesis of HBMSCs was induced by HAMSCs, while H19 level in HAMSCs was increased during coculturing. H19 had no significant effect on the proliferative behaviors of HBMSCs, while its overexpression of H19 in HAMSCs led to the upregulated osteogenesis of HBMSCs in vivo and in vitro; whereas its knockdown reversed these effects. Mechanistically, H19 promoted miR-675 expression and contributed to the competitively bounding of miR-675 and Adenomatous polyposis coli (APC), thus significantly activating the Wnt/β-catenin pathway. The results suggested that HAMSCs promote osteogenic differentiation of HBMSCs via H19/miR-675/APC pathway, and supply a potential target for the therapeutic treatment of bone-destructive diseases.
Collapse
|
45
|
Frazier S, McBride MW, Mulvana H, Graham D. From animal models to patients: the role of placental microRNAs, miR-210, miR-126, and miR-148a/152 in preeclampsia. Clin Sci (Lond) 2020; 134:1001-1025. [PMID: 32337535 PMCID: PMC7239341 DOI: 10.1042/cs20200023] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/23/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022]
Abstract
Placental microRNAs (miRNAs) regulate the placental transcriptome and play a pathological role in preeclampsia (PE), a hypertensive disorder of pregnancy. Three PE rodent model studies explored the role of placental miRNAs, miR-210, miR-126, and miR-148/152 respectively, by examining expression of the miRNAs, their inducers, and potential gene targets. This review evaluates the role of miR-210, miR-126, and miR-148/152 in PE by comparing findings from the three rodent model studies with in vitro studies, other animal models, and preeclamptic patients to provide comprehensive insight into genetic components and pathological processes in the placenta contributing to PE. The majority of studies demonstrate miR-210 is upregulated in PE in part driven by HIF-1α and NF-κBp50, stimulated by hypoxia and/or immune-mediated processes. Elevated miR-210 may contribute to PE via inhibiting anti-inflammatory Th2-cytokines. Studies report an up- and downregulation of miR-126, arguably reflecting differences in expression between cell types and its multifunctional capacity. MiR-126 may play a pro-angiogenic role by mediating the PI3K-Akt pathway. Most studies report miR-148/152 family members are upregulated in PE. Evidence suggests they may inhibit DNA methylation of genes involved in metabolic and inflammatory pathways. Given the genetic heterogeneity of PE, it is unlikely that a single placental miRNA is a suitable therapeutic target for all patients. Investigating miRNAs in PE subtypes in patients and animal models may represent a more appropriate approach going forward. Developing methods for targeting placental miRNAs and specific placental cell types remains crucial for research seeking to target placental miRNAs as a novel treatment for PE.
Collapse
Affiliation(s)
- Sonya Frazier
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Martin W. McBride
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| | - Helen Mulvana
- Biomedical Engineering, University of Strathclyde, Glasgow, U.K
| | - Delyth Graham
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, U.K
| |
Collapse
|
46
|
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: 1.2] [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
|
47
|
|
48
|
Hussey MR, Burt A, Deyssenroth MA, Jackson BP, Hao K, Peng S, Chen J, Marsit CJ, Everson TM. Placental lncRNA expression associated with placental cadmium concentrations and birth weight. ENVIRONMENTAL EPIGENETICS 2020; 6:dvaa003. [PMID: 32411397 PMCID: PMC7211362 DOI: 10.1093/eep/dvaa003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/01/2020] [Accepted: 02/03/2020] [Indexed: 05/22/2023]
Abstract
Heavy metal exposures, such as cadmium, can have negative effects on infant birth weight (BW)-among other developmental outcomes-with placental dysfunction potentially playing a role in these effects. In this study, we examined how differential placental expression of long non-coding RNAs (lncRNAs) may be associated with cadmium levels in placenta and whether differences in the expression of those lncRNAs were associated with fetal growth. In the Rhode Island Child Health Study, we used data from Illumina HiSeq whole transcriptome RNA sequencing (n = 199) to examine association between lncRNA expression and measures of infant BW as well as placental cadmium concentrations controlled for appropriate covariates. Of the 1191 lncRNAs sequenced, 46 demonstrated associations (q < 0.05) with BW in models controlling for infant sex, maternal age, BMI, maternal education, and smoking during pregnancy. Furthermore, four of these transcripts were associated with placental cadmium concentrations, with MIR22HG and ERVH48-1 demonstrating increases in expression associated with increasing cadmium exposure and elevated odds of small for gestational age birth, while AC114763.2 and LINC02595 demonstrated reduced expression associated with cadmium, but elevated odds of large for gestational age birth with increasing expression. We identified relationships between lncRNA expression with both placental cadmium concentrations and BW. This study provides evidence that disrupted placental expression of lncRNAs may be a part of cadmium's mechanisms of reproductive toxicity.
Collapse
Affiliation(s)
- Michael R Hussey
- Department of Environmental Health, Rollins School of Public Health at Emory University, Atlanta, GA, USA
| | - Amber Burt
- Department of Environmental Health, Rollins School of Public Health at Emory University, Atlanta, GA, USA
| | - Maya A Deyssenroth
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian P Jackson
- Department of Earth Sciences, Dartmouth College, Hanover, NH, USA
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shouneng Peng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Chen
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carmen J Marsit
- Department of Environmental Health, Rollins School of Public Health at Emory University, Atlanta, GA, USA
| | - Todd M Everson
- Department of Environmental Health, Rollins School of Public Health at Emory University, Atlanta, GA, USA
| |
Collapse
|
49
|
|
50
|
Harati-Sadegh M, Kohan L, Teimoori B, Mehrabani M, Salimi S. The effects of placental long noncoding RNA H19 polymorphisms and promoter methylation on H19 expression in association with preeclampsia susceptibility. IUBMB Life 2019; 72:413-425. [PMID: 31769935 DOI: 10.1002/iub.2199] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/28/2019] [Indexed: 12/19/2022]
Abstract
The effect of DNA methylation on gene expression triggered it as a susceptibility factor in various diseases including preeclampsia (PE). The pathogenesis of PE is closely associated with the methylation status and genetic variants of relevant genes. Therefore, the aim of the study was to investigate the possible impacts of the placental DNA methylation and rs3741219, rs217727, and rs2107425 polymorphisms of the H19 gene on the PE susceptibility as well as the its mRNA expression. Moreover, eight haplotypes of three loci in the H19 gene were analyzed. In this case-control study, the placentas of 107 preeclamptic and 113 non-preeclamptic women were collected after delivery. The methylation status was assessed by methylation-specific polymerase chain reaction (PCR). The H19 polymorphisms were genotyped using polymerase chain reaction-restriction fragment length polymorphism or amplification refractory mutation system-polymerase chain reaction methods. The quantitative real time PCR was used for mRNA expression assay. The placental H19 rs3741219 and rs2107425 polymorphisms were not associated with PE. However, H19 rs217727CT and TT genotypes might be associated with a 9.2- and 17.7-fold increased risk of PE, respectively. The Trs3741219 Crs217727 Crs2107425 and Trs3741219 Crs217727 Trs2107425 haplotypes were significantly lower, whereas the Trs3741219 Trs217727 Crs2107425 and Crs3741219 Trs217727 Crs2107425 haplotypes were significantly higher in PE women. Promoter but not upstream region hypermethylation of H19 gene could be led to decreased risk of PE (MM vs. UM + UU). No significant difference was observed in the placental mRNA expression between two groups. The H19 expression was significantly higher in women with unmethylated (UU), compared to methylated promoter (MM). The H19 expression was 17- and 15-fold higher in H19-rs2107425 CC and CT genotypes in PE women. In conclusion, the H19 rs2107425 polymorphism was associated with a higher risk of PE and increased H19 mRNA expression. The promoter hypermethylation of H19 gene was associated with a lower risk of PE and decreased H19 mRNA expression.
Collapse
Affiliation(s)
- Mahdiyeh Harati-Sadegh
- Genetic of Non-Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Leila Kohan
- Department of Biology, Arsanjan Branch, Islamic Azad University, Arsanjan, Iran
| | - Batool Teimoori
- Department of Obstetrics and Gynecology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mehrnaz Mehrabani
- Physiology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeedeh Salimi
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.,Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|