Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
Cancers (Basel) ; 16(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38730709

ABSTRACT

In patients with high-risk prostate cancer (HRPC) after radical prostatectomy (RP), biochemical recurrence (BCR) increases the risk of distant metastasis. Accordingly, additional prognostic biomarkers are required to identify the subpopulation of patients with HRPC who develop clinical recurrence (CR) after BCR. The objective of this study was to identify biomarkers in formalin-fixed paraffin-embedded (FFPE) RP samples that are prognostic for CR in patients with HRPC who experience BCR after RP (post-RP BCR). First, we performed a preliminary RNA sequencing analysis to comprehensively profile RNA expression in FFPE RP samples obtained from patients with HRPC who developed CR after post-RP BCR and found that many snRNAs were very abundant in preserved FFPE samples. Subsequently, we used quantitative polymerase chain reaction (qPCR) to compare the expression levels of highly abundant snRNAs in FFPE RP samples from patients with HRPC with and without CR after post-RP BCR (21 CR patients and 46 non-CR patients who had more than 5 years of follow-up after BCR). The qPCR analysis revealed that the expression levels of snRNA RNU1-1/1-2 and RNU4-1 were significantly higher in patients with CR than in patients without CR. These snRNAs were significantly correlated with clinical recurrence-free survival (RFS) in patients with HRPC who experienced post-RP BCR. Furthermore, snRNA RNU1-1/1-2 could serve as an independent prognostic factor for clinical RFS in post-RP BCR of HRPC cases where known prognostic factors (e.g., Gleason score) cannot distinguish between CR and non-CR patients. Our findings provide new insights into the involvement of snRNAs in prostate cancer progression.

2.
J Reprod Immunol ; 161: 104187, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38199177

ABSTRACT

Extracellular vesicles (EVs), including exosomes, are carriers of extracellular microRNAs (miRNAs). Exomeres, non-vesicular extracellular nanoparticles (NVEPs), are novel extracellular cargo carriers. However, little is known of the characteristics of placental trophoblast-derived exomeres. In this study, we characterized trophoblast-derived exomeres and investigated the cell-cell communication of placenta-specific miRNAs carried by those exomeres using an in vitro model system (BeWo trophoblasts and Jurkat T cells). BeWo exomeres (∼ 40 nm diameter) had pilling-like nanoparticle structures, which were distinct from cup-shaped exosomes (∼ 90-110 nm diameter). BeWo cells secreted more exomeres than exosomes. Exomeres were positive for AGO2 but negative for exosome markers (CD63, CD9, CD81, FLOT1, and TSG101). The levels of placenta-specific miRNAs in exomeres were significantly higher than in exosomes. In a cell-cell communication analysis using a placenta-specific miRNA, BeWo exomeres delivered significantly more miR-517a-3p to recipient Jurkat cells compared with exosomes. Moreover, exomere-miR-517a-3p significantly reduced the expression of PRKG1 in miR-517a-3p-inhibitor (-) Jurkat cells compared with miR-517a-3p-inhibitor (+) cells, suggesting that miR-517a-3p inhibition reversed the exomere-miR-517a-3p-mediated repression of PRKG1 expression in recipient cells. Therefore, BeWo trophoblast exomeres are enriched with bioactive extracellular placenta-specific miRNAs, which were formerly considered to be carried by exosomes. Our findings provide insight into trophoblast-derived NVEPs.


Subject(s)
Exosomes , Extracellular Vesicles , MicroRNAs , Humans , Female , Pregnancy , MicroRNAs/genetics , MicroRNAs/metabolism , Placenta/metabolism , Exosomes/genetics , Exosomes/metabolism , Extracellular Vesicles/metabolism , Trophoblasts/metabolism
3.
J Reprod Immunol ; 162: 104189, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38241848

ABSTRACT

In villous trophoblasts, DROSHA is a key ribonuclease III enzyme that processes pri-microRNAs (pri-miRNAs) into pre-miRNAs at the placenta-specific, chromosome 19 miRNA cluster (C19MC) locus. However, little is known of its other functions. We performed formaldehyde crosslinking, immunoprecipitation, and sequencing (fCLIP-seq) analysis of terminal chorionic villi to identify DROSHA-binding RNAs in villous trophoblasts. In villous trophoblasts, DROSHA predominantly generated placenta-specific C19MC pre-miRNAs, including antiviral C19MC pre-miRNAs. The fCLIP-seq analysis also identified non-miRNA transcripts with hairpin structures potentially capable of binding to DROSHA (e.g., SNORD100 and VTRNA1-1). Moreover, in vivo immunohistochemical analysis revealed DROSHA in the cytoplasm of villous trophoblasts. DROSHA was abundant in the cytoplasm of villous trophoblasts, particularly in the apical region of syncytiotrophoblast, in the full-term placenta. Furthermore, in BeWo trophoblasts infected with Sindbis virus (SINV), DROSHA translocated to the cytoplasm and recognized the genomic RNA of SINV. Therefore, in trophoblasts, DROSHA not only regulates RNA metabolism, including the biogenesis of placenta-specific miRNAs, but also recognizes viral RNAs. After SINV infection, BeWo DROSHA-binding VTRNA1-1 was significantly upregulated, and cellular VTRNA1-1 was significantly downregulated, suggesting that DROSHA soaks up VTRNA1-1 in response to viral infection. These results suggest that the DROSHA-mediated recognition of RNAs defends against viral infection in villous trophoblasts. Our data provide insight into the antiviral functions of DROSHA in villous trophoblasts of the human placenta.


Subject(s)
MicroRNAs , Virus Diseases , Humans , Ribonuclease III/genetics , Ribonuclease III/chemistry , Ribonuclease III/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cytoplasm/metabolism , Trophoblasts/metabolism , Antiviral Agents
4.
Histochem Cell Biol ; 159(4): 363-375, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36484822

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.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Pregnancy , Female , Mice , Animals , Trophoblasts/metabolism , Placenta/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Endothelial Cells/metabolism
5.
Cells ; 11(15)2022 08 05.
Article in English | MEDLINE | ID: mdl-35954272

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.


Subject(s)
MicroRNAs , Pre-Eclampsia , Aspirin/therapeutic use , Biomarkers , Female , Humans , Infant, Newborn , MicroRNAs/metabolism , Pre-Eclampsia/diagnosis , Pre-Eclampsia/genetics , Pregnancy , Pregnancy Trimester, First
6.
Histochem Cell Biol ; 158(1): 65-78, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35486179

ABSTRACT

A few long noncoding RNAs (long ncRNAs, lncRNAs) exhibit trophoblast cell type-specific expression patterns and functional roles in mouse placenta. However, the cell- and stage-specific expression patterns and functions of most placenta-derived lncRNAs remain unclear. In this study, we explored mouse placenta-associated lncRNAs using a combined bioinformatic and experimental approach. We used the FANTOM5 database to survey lncRNA expression in mouse placenta and found that 1600012P17Rik (MGI: 1919275, designated P17Rik), a long intergenic ncRNA, was the most highly expressed lncRNA at gestational day 17. Polymerase chain reaction analysis confirmed that P17Rik was exclusively expressed in placenta and not in any of the adult organs examined in this study. In situ hybridization analysis revealed that it was highly expressed in spongiotrophoblast cells and to a lesser extent in glycogen trophoblast cells, including migratory glycogen trophoblast cells invading the decidua. Moreover, we found that it is a polyadenylated lncRNA localized mainly to the cytoplasm of these trophoblast cells. As these trophoblast cells also expressed the neighboring protein-coding gene, pappalysin 2 (Pappa2), we investigated the effects of P17Rik on Pappa2 expression using Pappa2-expressing MC3T3-E1 cells and found that P17Rik transfection increased the messenger RNA (mRNA) and protein levels of Pappa2. These results indicate that mouse placenta-specific lncRNA P17Rik modulates the expression of the neighboring protein-coding gene Pappa2 in spongiotrophoblast and glycogen trophoblast cells of mouse placenta during late gestation.


Subject(s)
RNA, Long Noncoding , Trophoblasts , Animals , Female , Glycogen/metabolism , In Situ Hybridization , Mice , Pregnancy , Pregnancy-Associated Plasma Protein-A/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/metabolism , Trophoblasts/metabolism
8.
Genes (Basel) ; 12(2)2021 01 27.
Article in English | MEDLINE | ID: mdl-33514011

ABSTRACT

Long noncoding RNAs (lncRNAs) are emerging as critical regulators of gene expression, which play fundamental roles in cancer development. In this study, we found that homeobox A11 antisense RNA (HOXA11-AS), a highly expressed lncRNA in cell lines derived from prostate cancer bone metastases, promoted the cell invasion and proliferation of PC3 prostate cancer cells. Transcription factor homeobox B13 (HOXB13) was identified as an upstream regulator of HOXA11-AS.HOXA11-AS regulated bone metastasis-associated C-C motif chemokine ligand 2 (CCL2)/C-C chemokine receptor type 2 (CCR2) signaling in both PC3 prostate cancer cells and SaOS2 osteoblastic cells. The HOXB13/HOXA11-AS axis also regulated integrin subunits (ITGAV and ITGB1) specific to prostate cancer bone metastasis. HOXB13, in combination with HOXA11-AS, directly regulated the integrin-binding sialoprotein (IBSP) promoter. Furthermore, conditioned medium containing HOXA11-AS secreted from PC3 cells could induce the expression of CCL2 and IBSP in SaOS2 osteoblastic cells. These results suggest that prostate cancer HOXA11-AS and HOXB13 promote metastasis by regulation of CCL2/CCR2 cytokine and integrin signaling in autocrine and paracrine manners.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/secondary , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Long Noncoding/genetics , Cell Line, Tumor , Chemokine CCL2/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Osteoblasts/metabolism , Prostatic Neoplasms/pathology , Transcriptome
9.
Int J Mol Sci ; 22(3)2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33513878

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.


Subject(s)
GATA2 Transcription Factor/antagonists & inhibitors , Matrix Metalloproteinases/metabolism , MicroRNAs/metabolism , Placenta/metabolism , RNA, Long Noncoding/metabolism , Trophoblasts/metabolism , Cell Line , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation/genetics , Cell Proliferation/physiology , Female , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression Regulation, Developmental/genetics , Humans , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinases/genetics , MicroRNAs/genetics , Pregnancy , Pregnancy Trimester, First , RNA, Long Noncoding/genetics , RNA, Small Interfering , RNA-Seq , Trophoblasts/enzymology
10.
J Matern Fetal Neonatal Med ; 34(20): 3377-3385, 2021 Oct.
Article in English | MEDLINE | ID: mdl-31736372

ABSTRACT

INTRODUCTION: Appropriate extravillous trophoblast (EVT) invasion is essential for successful pregnancy. Previously, we showed that EVTs express CD44, which accelerated EVT invasion. However, its regulation mechanism via CD44 remains unknown. Our hypothesis was that WNT signaling enhanced EVT invasion via CD44. To test this hypothesis, we investigated the effects of WNT ligands on CD44 expression and EVT invasion using EVT cell lines and isolated primary EVTs. METHODS: We used EVT cell lines (HTR8/SVneo and HChEpC1b) and isolated primary EVTs, extracted from first-trimester trophoblasts. The cells were supplemented with WNT3A, 5A, and 10B. We examined cell invasion and the expressions of CD44 and matrix metalloproteinase (MMP) 9. Next, to clarify the pathway of WNT10B in EVTs, we knock-downed WNT10B using siRNA and activated or inhibited the WNT canonical pathway using an activator (lithium chloride) or inhibitor (FH535, XAV939) with WNT10B addition. RESULTS: WNT3A, 5A, and 10B accelerated the invasion in the EVT lines and isolated primary EVTs. The expressions of CD44 and MMP9 were also upregulated by WNT ligands. WNT10B knockdown significantly inhibited EVT invasion concomitantly with CD44 expression. The WNT canonical pathway activator upregulated CD44 expression and its inhibitor downregulated it with WNT10B addition. CONCLUSIONS: The present study is the first to show the possibility that WNT3A, WNT5A, and WNT10B exist upstream of CD44 in EVTs. Among them, WNT10B may be a novel accelerator of EVT invasion. WNT signaling mediated by multiple WNT ligands may contribute to EVT invasion.


Subject(s)
Matrix Metalloproteinase 9 , Trophoblasts , Cell Movement , Female , Humans , Hyaluronan Receptors/genetics , Matrix Metalloproteinase 9/metabolism , Pregnancy , Pregnancy Trimester, First , Trophoblasts/metabolism , Wnt Signaling Pathway , Wnt3A Protein
11.
Histochem Cell Biol ; 149(5): 517-527, 2018 May.
Article in English | MEDLINE | ID: mdl-29411102

ABSTRACT

Long non-coding RNAs (lncRNAs; > 200 nucleotides in length) have attracted attention as fine-tuners of gene expression. However, little is known about the cell- and stage-specific expression pattern and function of lncRNAs in spermatogenesis. The purpose of this study was to identify mouse testis-associated lncRNAs using a combination of computational and experimental approaches. We first used the FANTOM5 database to survey lncRNA expression in the mouse testis and performed reverse transcription quantitative polymerase chain reaction (real-time PCR) and in situ hybridization (ISH) analyses. In silico analysis showed that most of the highly expressed lncRNAs in the adult mouse testis were testis-specific lncRNAs and were expressed at and following the initiation of spermatogenesis. We selected the antisense lncRNA 1700108J01Rik and long intergenic non-coding RNA 1700101O22Rik from the most highly expressed lncRNAs in the adult testis for further analysis. Real-time PCR analysis confirmed that 1700108J01Rik and 1700101O22Rik were specifically expressed in the testis. ISH analysis revealed that the two mouse-testis-specific lncRNAs were expressed exclusively in testicular germ cells in meiotic prophase and the round spermatid stage, which coincide with the period of transcriptional reactivation during spermatogenesis. The cytoplasmic distribution of these lncRNAs revealed by ISH suggests their involvement in post-transcriptional gene regulation rather than in epigenetic or transcriptional regulation. Our data provide new insight into testis-associated lncRNAs that will be useful in expression and functional studies of spermatogenesis.


Subject(s)
RNA, Long Noncoding/genetics , Testis/metabolism , Animals , Computational Biology , Gene Expression Profiling , Male , Mice , Mice, Inbred Strains , Real-Time Polymerase Chain Reaction
12.
Audiol Neurootol ; 22(3): 135-145, 2017.
Article in English | MEDLINE | ID: mdl-28968605

ABSTRACT

The molecular mechanisms underlying age-related hearing loss are unknown, and currently, there is no treatment for this condition. Recent studies have shown that microRNAs (miRNAs) and age-related diseases are intimately linked, suggesting that some miRNAs may present attractive therapeutic targets. In this study, we obtained 8 human temporal bones from 8 elderly subjects at brain autopsy in order to investigate the expression profile of miRNAs in the inner ear with miRNA arrays. A mean of 478 different miRNAs were expressed in the samples, of which 348 were commonly expressed in all 8 samples. Of these, levels of 16 miRNAs significantly differed between young elderly and old elderly subjects. miRNAs, which play important roles in inner ear development, were detected in all samples, i.e., in both young and old elderly subjects, whether with or without hearing loss. Our results suggest that these miRNAs play important roles not only in development, but also in the maintenance of inner ear homeostasis.


Subject(s)
Ear, Inner/metabolism , Hearing Loss/genetics , MicroRNAs/genetics , Aged , Aged, 80 and over , Female , Gene Expression Profiling/methods , Hearing Loss/metabolism , Humans , Male , MicroRNAs/metabolism , Middle Aged , Real-Time Polymerase Chain Reaction
14.
Mol Med Rep ; 15(4): 1989-1996, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28259929

ABSTRACT

Mesenteric lymph is vital for immune cell trafficking and intestinal fluid and chyle transport, which aid homeostatic maintenance. There have been few reports investigating the profiles and circulatory dynamics of mesenteric lymph microRNAs (miRNAs). The present study aimed to provide a comprehensive analysis of miRNAs in normal rodent mesenteric lymph. Reverse transcription­quantitative polymerase chain reaction (RT­qPCR)­based array analysis was performed to examine the expression levels of 375 miRNAs in normal rat mesenteric lymph. Using differential centrifugation, the presence of miR­150, a representative lymph miRNA, in exosomes was assessed. Rat small intestine epithelial cell line IEC­6­derived exosomes were prepared from culture supernatants of cells transfected with cel­miR­238­3p, and were used to trace the administered exosomes in vivo and to investigate the in vivo delivery of lymph miRNAs via mesenteric lymphatics into the systemic circulation following injection of cel­miR­238­3p­exosomes. RT­qPCR­based array analysis detected 287 miRNAs in lymph, and 21 miRNAs that were significantly differentially expressed between lymph and plasma. Lymph fractionation analysis demonstrated that some cell­free lymph miR­150 was distributed in the exosome­containing microsomal fraction. Furthermore, in vivo analysis of lymph miRNA delivery revealed that exosomal cel­miR­238­3p was markedly distributed in the lung compared with in the liver, kidney and spleen, thus indicating that the lung is the major organ responsible for clearance of exosomal lymph miRNAs. These findings provide novel insights into the modulation of gene expression by mesenteric lymph miRNAs in the lung.


Subject(s)
Exosomes/genetics , Lymph/metabolism , Mesentery/metabolism , MicroRNAs/analysis , MicroRNAs/genetics , Animals , Cell Line , Gene Expression Profiling , Male , Mesentery/blood supply , MicroRNAs/administration & dosage , RNA Stability , Rats, Sprague-Dawley
15.
Placenta ; 50: 25-31, 2017 02.
Article in English | MEDLINE | ID: mdl-28161058

ABSTRACT

INTRODUCTION: Adequate extravillous trophoblast (EVT) invasion is essential for successful placentation. Although miR-520c-3p plays an important role in CD44-mediated invasion in cancer cells, there is little information on whether miR-520c-3p is involved in the regulatory mechanisms of CD44-mediated EVT invasion. METHODS: We screened first trimester trophoblast cells and trophoblast cell lines for expression of miR-520c-3p using real-time polymerase chain reaction. The cell invasion assay was performed using EVT cell lines, HTR8/SVneo and HChEpC1b, to investigate the capability of suppressing EVT invasion by miR-520c-3p. Laser microdissection analysis was then used to determine whether miR-520c-3p was present in the first trimester decidua. Finally, the possibility of chorionic villous trophoblast (CVT)-EVT communication via exosomal miR-520c-3p was determined using an in vitro model based on BeWo exosomes and the EVT cell lines as recipient cells. RESULTS: The miR-520c-3p level was significantly downregulated in EVT cell lines and EVTs. Cell invasion was significantly inhibited in miR-520c-3p-overexpressing cell lines, involving a significant reduction of CD44. Laser microdissection analysis showed that miR-520c-3p in the periarterial area of the decidua was significantly higher than that in the non-periarterial area. Using an in vitro model system, BeWo exosomal miR-520c-3p was internalized into the EVT cells with subsequently reduced cell invasion via CD44 repression. CONCLUSIONS: EVT invasion is synergistically enhanced by the reciprocal expression of endogenous miR-520c-3p and CD44. The present study supports a novel model involving a placenta-associated miRNA function in cell-cell communication in which CVT exosomal miR-520c-3p regulates cell invasion by targeting CD44 in EVTs.


Subject(s)
Cell Movement/physiology , Hyaluronan Receptors/metabolism , MicroRNAs/metabolism , Trophoblasts/metabolism , Cell Line , Female , Humans , MicroRNAs/genetics , Placenta/metabolism , Pregnancy , Pregnancy Trimester, First , Trophoblasts/cytology
17.
Hepatol Res ; 46(5): 391-406, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26490438

ABSTRACT

Hepatocellular carcinoma (HCC) is the fifth most frequent cancer and the third cause of cancer-related mortality worldwide. The primary risk factor for HCC is liver cirrhosis secondary to persistent infection with hepatitis B virus or hepatitis C virus. Although a number of cellular phenomena and molecular events have been reported to facilitate tumor initiation, progression and metastasis, the exact etiology of HCC has not yet been fully uncovered. miRNA, a class of non-coding RNA, negatively regulate post-transcriptional processes that participate in crucial biological processes, including development, differentiation, apoptosis and proliferation. In the liver, specific miRNA can be negative regulators of gene expression. Recent studies have uncovered the contribution of miRNA to cancer pathogenesis as they can function as oncogenes or tumor suppressor genes. In addition, other studies have demonstrated their potential value in the clinical management of patients with HCC as some miRNA may be used as prognostic or diagnostic markers. In this review, we summarize the current knowledge about the roles of miRNA in the carcinogenesis and progression of HCC.

18.
World J Hepatol ; 7(4): 696-702, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25866606

ABSTRACT

A large number of studies have demonstrated that the synergistic collaboration of a number of microRNAs (miRNAs), their growth factors and their downstream agents is required for the initiation and completion of pathogenesis in the liver. miRNAs are thought to exert a profound effect on almost every aspect of liver biology and pathology. Accumulating evidence indicates that several miRNAs are involved in the hepatitis B virus (HBV) life cycle and infectivity, in addition to HBV-associated liver diseases including fibrosis, cirrhosis and hepatocellular carcinoma (HCC). In turn, HBV can modulate the expression of several cellular miRNAs, thus promoting a favorable environment for its replication and survival. In this review, we focused on the involvement of host cellular miRNAs that are directly and indirectly associated with HBV RNA or HBV associated transcription factors. Exploring different facets of the interactions among miRNA, HBV and HCV infections, and the carcinogenesis and progress of HCC, could facilitate the development of novel and effective treatment approaches for liver disease.

19.
Int J Mol Med ; 35(6): 1511-24, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25824636

ABSTRACT

Although recent studies have demonstrated that microRNAs (miRNAs or miRs) regulate fundamental natural killer (NK) cellular processes, including cytotoxicity and cytokine production, little is known about the miRNA-gene regulatory relationships in maternal peripheral blood NK (pNK) cells during pregnancy. In the present study, to determine the roles of miRNAs within gene regulatory networks of maternal pNK cells, we performed comprehensive miRNA and gene expression profiling of maternal pNK cells using a combination of reverse transcription quantitative PCR (RT-qPCR)-based miRNA array and DNA microarray analyses and analyzed the differential expression levels between first- and third-trimester pNK cells. Furthermore, we constructed regulatory networks for miRNA-mediated gene expression in pNK cells during pregnancy by Ingenuity Pathway Analysis (IPA). PCR-based array analysis revealed that the placenta-derived miRNAs [chromosome 19 miRNA cluster (C19MC) miRNAs] were detected in pNK cells during pregnancy. Twenty-five miRNAs, including six C19MC miRNAs, were significantly upregulated in the third- compared to first-trimester pNK cells. The rapid clearance of C19MC miRNAs also occurred in the pNK cells following delivery. Nine miRNAs, including eight C19MC miRNAs, were significantly downregulated in the post-delivery pNK cells compared to those of the third-trimester. DNA microarray analysis identified 69 NK cell function-related genes that were differentially expressed between the first- and third-trimester pNK cells. On pathway and network analysis, the observed gene expression changes of pNK cells likely contribute to the increase in the cytotoxicity, as well as the cell cycle progression of third- compared to first-trimester pNK cells. Thirteen of the 69 NK cell function-related genes were significantly downregulated between the first- and third-trimester pNK cells. Nine of the 13 downregulated NK-function-associated genes were in silico target candidates of 12 upregulated miRNAs, including C19MC miRNA miR-512-3p. The results of this study suggest that the transfer of placental C19MC miRNAs into maternal pNK cells occurs during pregnancy. The present study provides new insight into maternal NK cell functions.


Subject(s)
Killer Cells, Natural/metabolism , MicroRNAs/blood , Parturition/blood , Placenta/metabolism , Pregnancy Trimester, Third/blood , Pregnancy/blood , Adult , Female , Humans
20.
PLoS One ; 10(3): e0121819, 2015.
Article in English | MEDLINE | ID: mdl-25793409

ABSTRACT

Environmental diseases related to cadmium exposure primarily develop owing to industrial wastewater pollution and/or contaminated food. In regions with high cadmium exposure in Japan, cadmium accumulation occurs primarily in the kidneys of individuals who are exposed to the metal. In contrast, in the itai-itai disease outbreak that occurred in the Jinzu River basin in Toyama Prefecture in Japan, cadmium primarily accumulated in the liver. On the other hand, high concentration of cadmium caused renal tubular disorder and osteomalacia (multiple bone fracture), probably resulting from the renal tubular dysfunction and additional pathology. In this study, we aimed to establish a mouse model of chronic cadmium intake. We administered cadmium-containing drinking water (32 mg/l) to female and male mice ad libitum for 11 weeks. Metal analysis using inductively coupled plasma mass spectrometry revealed that cadmium accumulated in the kidneys (927 x 10 + 185 ng/g in females and 661 x 10 + 101 ng/g in males), liver (397 x 10 + 199 ng/g in females and 238 x 10 + 652 ng/g in males), and thyroid gland (293 + 93.7 ng/g in females and 129 + 72.7 ng/g in males) of mice. Female mice showed higher cadmium accumulation in the kidney, liver, and thyroid gland than males did (p = 0.00345, p = 0.00213, and p = 0.0331, respectively). Shotgun proteome analyses after chronic oral administration of cadmium revealed that protein levels of glutathione S-transferase Mu2, Mu4, and Mu7 decreased in the liver, and those of A1 and A2 decreased in the kidneys in both female and male mice.


Subject(s)
Cadmium/metabolism , Proteome/metabolism , Proteomics/methods , Sex Characteristics , Administration, Oral , Animals , Down-Regulation , Female , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...