1
|
Chuang TD, Ton N, Rysling S, Khorram O. The in vivo effects of knockdown of long non-coding RNA XIST on fibroid growth and gene expression. FASEB J 2024; 38:e70140. [PMID: 39475327 DOI: 10.1096/fj.202401982r] [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: 08/23/2024] [Revised: 09/21/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024]
Abstract
The role of long non-coding RNAs in fibroid pathogenesis remains largely unexplored. In a previous study, we found elevated XIST (X-inactive specific transcript) levels in fibroids, which sponged miR-29c and miR-200c, leading to the overexpression of their target genes. This study aimed to assess the therapeutic potential of XIST downregulation in fibroid treatment. Ovariectomized SCID (severe combined immunodeficiency) mice were implanted with fibroid tumors transduced with XIST siRNA or a control via lentivirus. After 1 month, animals were sacrificed and the xenografts were removed for further analysis. XIST knockdown reduced tumor weight by 15% and increased miR-29c and miR-200c expression by 3.9-fold and 2.2-fold, respectively. The mRNA expression of miR-29c targets (COL3A1, TGF-β3, CDK2, SPARC) and miR-200c targets (CDK2, FN1, TDO2), as well as PRL, E2F1, and EZH2, was significantly decreased. Protein abundance of collagen, COL3A1, FN1, CDK2, SPARC, and EZH2 was also reduced. IHC analysis of xenograft sections using the markers of Ki67 for cell proliferation and cleaved caspase 3 for apoptosis showed decreased cell proliferation and no changes in apoptosis in the XIST knockdown xenografts. This analysis also revealed decreased collagen and E2F1 staining nuclei in the XIST knockdown xenografts. These results indicate that downregulation of XIST in fibroids has beneficial therapeutic effects, by reducing tumor growth and the expression of genes involved in cell proliferation, inflammation, and extracellular matrix regulation.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- The Lundquist Institute for Biomedical Innovation, Torrance, California, USA
| | - Nhu Ton
- The Lundquist Institute for Biomedical Innovation, Torrance, California, USA
| | - Shawn Rysling
- The Lundquist Institute for Biomedical Innovation, Torrance, California, USA
| | - Omid Khorram
- The Lundquist Institute for Biomedical Innovation, Torrance, California, USA
- Department of Obstetrics/Gynecology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
2
|
Chuang TD, Ton N, Manrique N, Rysling S, Khorram O. Targeting the long non-coding RNA MIAT for the treatment of fibroids in an animal model. Clin Sci (Lond) 2024; 138:699-709. [PMID: 38817011 PMCID: PMC11166562 DOI: 10.1042/cs20240190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/03/2024] [Accepted: 05/30/2024] [Indexed: 06/01/2024]
Abstract
Our previous studies indicated that there is overexpression of MIAT in fibroids and MIAT is a sponge for the miR-29 family in these tumors. The objective of the present study was to determine if the knockdown of MIAT in fibroid xenografts will increase miR-29 levels and reduce the expression of genes targeted by this miRNA such as collagen and cell cycle regulatory proteins in a mouse model for fibroids. Ovariectomized CB-17 SCID/Beige mice bearing estrogen/progesterone pellets were implanted subcutaneously in the flank with equal weight of fibroid explants which had been transduced by lentivirus for either control (empty vector) or MIAT knockdown for four weeks (n=7). Knockdown of MIAT in fibroid xenografts resulted in a 30% reduction of tumor weight and a marked increase in miR-29a, -b, and -c levels in the xenografts. There was reduced cell proliferation and expression of cell cycle regulatory genes CCND1, CDK2, and E2F1 and no significant changes in apoptosis. The xenografts with MIAT knockdown expressed lower mRNA and protein levels of FN1, COL3A1, and TGF-β3, and total collagen protein. Targeting MIAT, which sponges the pro-fibrotic miR-29 family, is an effective therapy for fibroids by reducing cell proliferation and thereby, tumor growth and accumulation of ECM, which is a hallmark of these benign gynecologic tumors.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA, U.S.A
- The Lundquist Institute for Biomedical Innovation, Torrance, CA, U.S.A
| | - Nhu Ton
- The Lundquist Institute for Biomedical Innovation, Torrance, CA, U.S.A
| | - Nathaly Manrique
- The Lundquist Institute for Biomedical Innovation, Torrance, CA, U.S.A
| | - Shawn Rysling
- The Lundquist Institute for Biomedical Innovation, Torrance, CA, U.S.A
| | - Omid Khorram
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA, U.S.A
- The Lundquist Institute for Biomedical Innovation, Torrance, CA, U.S.A
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, U.S.A
| |
Collapse
|
3
|
Pereira IT, Gomes-Júnior R, Hansel-Frose A, França RSV, Liu M, Soliman HAN, Chan SSK, Dudley SC, Kyba M, Dallagiovanna B. Cardiac Development Long Non-Coding RNA ( CARDEL) Is Activated during Human Heart Development and Contributes to Cardiac Specification and Homeostasis. Cells 2024; 13:1050. [PMID: 38920678 PMCID: PMC11201801 DOI: 10.3390/cells13121050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Successful heart development depends on the careful orchestration of a network of transcription factors and signaling pathways. In recent years, in vitro cardiac differentiation using human pluripotent stem cells (hPSCs) has been used to uncover the intricate gene-network regulation involved in the proper formation and function of the human heart. Here, we searched for uncharacterized cardiac-development genes by combining a temporal evaluation of human cardiac specification in vitro with an analysis of gene expression in fetal and adult heart tissue. We discovered that CARDEL (CARdiac DEvelopment Long non-coding RNA; LINC00890; SERTM2) expression coincides with the commitment to the cardiac lineage. CARDEL knockout hPSCs differentiated poorly into cardiac cells, and hPSC-derived cardiomyocytes showed faster beating rates after controlled overexpression of CARDEL during differentiation. Altogether, we provide physiological and molecular evidence that CARDEL expression contributes to sculpting the cardiac program during cell-fate commitment.
Collapse
Affiliation(s)
- Isabela T. Pereira
- Basic Stem Cell Biology Laboratory, Instituto Carlos Chagas-FIOCRUZ-PR, Curitiba 81350-010, PR, Brazil; (R.G.-J.); (A.H.-F.); (R.S.V.F.); (B.D.)
| | - Rubens Gomes-Júnior
- Basic Stem Cell Biology Laboratory, Instituto Carlos Chagas-FIOCRUZ-PR, Curitiba 81350-010, PR, Brazil; (R.G.-J.); (A.H.-F.); (R.S.V.F.); (B.D.)
| | - Aruana Hansel-Frose
- Basic Stem Cell Biology Laboratory, Instituto Carlos Chagas-FIOCRUZ-PR, Curitiba 81350-010, PR, Brazil; (R.G.-J.); (A.H.-F.); (R.S.V.F.); (B.D.)
| | - Rhaíza S. V. França
- Basic Stem Cell Biology Laboratory, Instituto Carlos Chagas-FIOCRUZ-PR, Curitiba 81350-010, PR, Brazil; (R.G.-J.); (A.H.-F.); (R.S.V.F.); (B.D.)
| | - Man Liu
- Department of Medicine, Division of Cardiology, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (S.C.D.J.)
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA; (H.A.N.S.); (S.S.K.C.); (M.K.)
| | - Hossam A. N. Soliman
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA; (H.A.N.S.); (S.S.K.C.); (M.K.)
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Sunny S. K. Chan
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA; (H.A.N.S.); (S.S.K.C.); (M.K.)
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Samuel C. Dudley
- Department of Medicine, Division of Cardiology, University of Minnesota, Minneapolis, MN 55455, USA; (M.L.); (S.C.D.J.)
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA; (H.A.N.S.); (S.S.K.C.); (M.K.)
| | - Michael Kyba
- Lillehei Heart Institute, University of Minnesota, Minneapolis, MN 55455, USA; (H.A.N.S.); (S.S.K.C.); (M.K.)
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Bruno Dallagiovanna
- Basic Stem Cell Biology Laboratory, Instituto Carlos Chagas-FIOCRUZ-PR, Curitiba 81350-010, PR, Brazil; (R.G.-J.); (A.H.-F.); (R.S.V.F.); (B.D.)
| |
Collapse
|
4
|
Chuang TD, Ton N, Rysling S, Boos D, Khorram O. The Effect of Race/Ethnicity and MED12 Mutation on the Expression of Long Non-Coding RNAs in Uterine Leiomyoma and Myometrium. Int J Mol Sci 2024; 25:1307. [PMID: 38279317 PMCID: PMC10816284 DOI: 10.3390/ijms25021307] [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: 12/16/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
The objective of this study was to elucidate the expression of long non-coding RNA (lncRNA) in leiomyomas (Lyo) and paired myometrium (Myo) and explore the impact of race and MED12 mutation. Fold change analysis (Lyo/paired Myo) indicated the expression of 63 lncRNAs was significantly altered in the mutated group but not in the non-mutated Lyo. Additionally, 65 lncRNAs exhibited an over 1.5-fold change in the Black but not the White group. Fifteen differentially expressed lncRNAs identified with next-generation sequencing underwent qRT-PCR confirmation. Compared with Myo, the expression of TPTEP1, PART1, RPS10P7, MSC-AS1, SNHG12, CA3-AS1, LINC00337, LINC00536, LINC01436, LINC01449, LINC02433, and LINC02624 was significantly higher, while the expression of ZEB2-AS1, LINC00957, and LINC01186 was significantly lower. Comparison of normal Myo with diseased Myo showed significant differences in the expression of several lncRNAs. Analysis based on race and Lyo MED12 mutation status indicated a significantly higher expression of RPS10P7, SNHG12, LINC01449, LINC02433, and LINC02624 in Lyo from Black patients. The expression of TPTEP1, PART1, RPS10P7, MSC-AS1, LINC00337, LINC00536, LINC01436, LINC01449, LINC02433, and LINC02624 was higher, while LINC01186 was significantly lower in the MED12-mutated group. These results indicate that Lyo are characterized by aberrant lncRNA expression, which is further impacted by race and Lyo MED12 mutation status.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.B.)
| | - Nhu Ton
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.B.)
| | - Shawn Rysling
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.B.)
| | - Drake Boos
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.B.)
| | - Omid Khorram
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.B.)
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, CA 90024, USA
| |
Collapse
|
5
|
Sheng QJ, Tan Y, Zhang L, Wu ZP, Wang B, He XY. Heterogeneous graph framework for predicting the association between lncRNA and disease and case on uterine fibroid. Comput Biol Med 2023; 165:107331. [PMID: 37619322 DOI: 10.1016/j.compbiomed.2023.107331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023]
Abstract
Long non-coding RNAs (lncRNAs) play crucial regulatory roles in various cellular processes, including gene expression, chromatin remodeling, and protein localization. Dysregulation of lncRNAs has been linked to several diseases, making it essential to understand their functions in disease mechanisms and therapeutic strategies. However, traditional experimental methods for studying lncRNA function are time-consuming, expensive, and offer limited insights. In recent years, computational methods have emerged as valuable tools for predicting lncRNA functions and their associations with diseases. However, many existing methods focus on constructing separate networks for lncRNA and disease similarity, resulting in information loss and insufficient processing capacity for isolated nodes. To address this, we developed 'RGLD' by combining Random Walk with restarting (RWR), Graph Neural Network (GNN), and Graph Attention Networks (GAT) to predict lncRNA-disease associations in a heterogeneous network. RGLD achieved an impressive AUC of 0.88, outperforming other methods. It can also predict novel associations between lncRNAs and diseases. RGLD identified HOTAIR, MEG3, and PVT1 as lncRNAs associated with uterine fibroids. Biological experiments directly or indirectly verified the involvement of these three lncRNAs in uterine fibroids, validating the accuracy of RGLD's predictions. Furthermore, we extensively discussed the functions of the target genes regulated by these lncRNAs in uterine fibroids, providing evidence for their role in the development and progression of the disease.
Collapse
Affiliation(s)
- Qing-Jing Sheng
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tong Ji University, Shanghai, China; Shanghai Key Laboratory of Maternal and Fetal Medicine, Shanghai First Maternity and Infant Hospital, Shanghai, China
| | - Yuan Tan
- Department of Integrated Traditional Chinese Medicine (TCM) & Western Medicine, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Key Laboratory of Maternal and Fetal Medicine, Shanghai First Maternity and Infant Hospital, Shanghai, China
| | - Liyuan Zhang
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Zhi-Ping Wu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tong Ji University, Shanghai, China; Shanghai Key Laboratory of Maternal and Fetal Medicine, Shanghai First Maternity and Infant Hospital, Shanghai, China
| | - Beiying Wang
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tong Ji University, Shanghai, China; Shanghai Key Laboratory of Maternal and Fetal Medicine, Shanghai First Maternity and Infant Hospital, Shanghai, China
| | - Xiao-Ying He
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tong Ji University, Shanghai, China; Shanghai Key Laboratory of Maternal and Fetal Medicine, Shanghai First Maternity and Infant Hospital, Shanghai, China.
| |
Collapse
|
6
|
Chuang TD, Ton N, Rysling S, Quintanilla D, Boos D, Gao J, McSwiggin H, Yan W, Khorram O. The Influence of Race/Ethnicity on the Transcriptomic Landscape of Uterine Fibroids. Int J Mol Sci 2023; 24:13441. [PMID: 37686244 PMCID: PMC10487975 DOI: 10.3390/ijms241713441] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
The objective of this study was to determine if the aberrant expression of select genes could form the basis for the racial disparity in fibroid characteristics. The next-generation RNA sequencing results were analyzed as fold change [leiomyomas/paired myometrium, also known as differential expression (DF)], comparing specimens from White (n = 7) and Black (n = 12) patients. The analysis indicated that 95 genes were minimally changed in tumors from White (DF ≈ 1) but were significantly altered by more than 1.5-fold (up or down) in Black patients. Twenty-one novel genes were selected for confirmation in 69 paired fibroids by qRT-PCR. Among these 21, coding of transcripts for the differential expression of FRAT2, SOX4, TNFRSF19, ACP7, GRIP1, IRS4, PLEKHG4B, PGR, COL24A1, KRT17, MMP17, SLN, CCDC177, FUT2, MYO5B, MYOG, ZNF703, CDC25A, and CDCA7 was significantly higher, while the expression of DAB2 and CAV2 was significantly lower in tumors from Black or Hispanic patients compared with tumors from White patients. Western blot analysis revealed a greater differential expression of PGR-A and total progesterone (PGR-A and PGR-B) in tumors from Black compared with tumors from White patients. Collectively, we identified a set of genes uniquely expressed in a race/ethnicity-dependent manner, which could form the underlying mechanisms for the racial disparity in fibroids and their associated symptoms.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Nhu Ton
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Shawn Rysling
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Derek Quintanilla
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Drake Boos
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Jianjun Gao
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Hayden McSwiggin
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
| | - Wei Yan
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Omid Khorram
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA;
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA; (N.T.); (S.R.); (D.Q.); (D.B.); (J.G.); (H.M.); (W.Y.)
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at the University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
7
|
Song Y, Guo HL, Zhang MZ, Zhang ZL, Jin K, He QQ, Li H. lncRNA‑miRNA‑mRNA network in female offspring born from obese dams. Exp Ther Med 2023; 25:140. [PMID: 36845957 PMCID: PMC9947576 DOI: 10.3892/etm.2023.11839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 10/28/2022] [Indexed: 02/15/2023] Open
Abstract
Maternal obesity is associated with disturbance of lipid metabolism and obesity in offspring; however, the pathogenesis is still unclear. The present study elucidated the role of potential lipid metabolism-associated long non-coding RNA (lncRNA) and identified the pathways involved in mice born to obese dams. In the present study, maternal obesity was induced by feeding a high-fat diet for 10 weeks in female C57/BL6 mice, whereas control mice were fed a standard diet. All female mice mated with healthy male mice and were allowed to deliver spontaneously. The results demonstrated that female offspring from obese dams presented a tendency to become overweight in the first 8 weeks after birth; however, maternal obesity did not significantly alter the body weight of male offspring. RNA-sequencing analysis was performed on female offspring liver at 3 weeks old. Significantly dysregulated lncRNAs and downstream targets in female offspring liver were identified using bioinformatics analysis. lncRNA, microRNA (miRNA or miR) and mRNA expression levels in liver and AML12 cells were assessed using reverse transcription-quantitative PCR. A total of 8 upregulated and 17 downregulated lncRNAs were demonstrated in offspring from obese dams and lncRNA Lockd was indicated to be a key dysregulated lncRNA. Competing endogenous RNA (ceRNA) models suggested that the lncRNA Lockd/miR-582-5p/Elovl5 pathway was key for lipid metabolism in the liver of offspring from obese dams. Finally, small interfering RNA and miRNA inhibitor transfection was used to evaluate the ceRNA models in AML12 cells. Taken together, the results of the present study indicated that lncRNA Lockd-miR-582-5p-Elovl5 network may be disrupted in lipid metabolism and lead to obesity in the offspring of obese dams. This research will provide new insights into the molecular mechanism of obesity and lipid metabolism disorder.
Collapse
Affiliation(s)
- Yong Song
- School of Public Health, Wuhan University, Wuhan, Hubei 430071, P.R. China,Institute of Preventive Medicine Information, Hubei Provincial Center for Disease Control and Prevention, Wuhan, Hubei 430079, P.R. China
| | - Hong-Lin Guo
- School of Public Administration, South Central University for Nationalities, Wuhan, Hubei 430074, P.R. China
| | - Min-Zhe Zhang
- School of Public Health, Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Ze-Lin Zhang
- Department of Clinical Nutrition, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| | - Ke Jin
- School of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, Hubei 430023, P.R. China
| | - Qi-Qiang He
- Shenzhen Research Institute, Wuhan University, Shenzhen, Guangdong 518000, P.R. China
| | - Hui Li
- Medical Department, Taixing People's Hospital, Taizhou, Jiangsu 225400, P.R. China,Correspondence to: Miss Hui Li, Medical Department, Taixing People's Hospital, 1 Changzheng Road, Taixing, Taizhou, Jiangsu 225400, P.R. China
| |
Collapse
|
8
|
Chuang TD, Gao J, Quintanilla D, McSwiggin H, Boos D, Yan W, Khorram O. Differential Expression of MED12-Associated Coding RNA Transcripts in Uterine Leiomyomas. Int J Mol Sci 2023; 24:ijms24043742. [PMID: 36835153 PMCID: PMC9960582 DOI: 10.3390/ijms24043742] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/15/2023] Open
Abstract
Recent studies have demonstrated that somatic MED12 mutations in exon 2 occur at a frequency of up to 80% and have a functional role in leiomyoma pathogenesis. The objective of this study was to elucidate the expression profile of coding RNA transcripts in leiomyomas, with and without these mutations, and their paired myometrium. Next-generation RNA sequencing (NGS) was used to systematically profile the differentially expressed RNA transcripts from paired leiomyomas (n = 19). The differential analysis indicated there are 394 genes differentially and aberrantly expressed only in the mutated tumors. These genes were predominantly involved in the regulation of extracellular constituents. Of the differentially expressed genes that overlapped in the two comparison groups, the magnitude of change in gene expression was greater for many genes in tumors bearing MED12 mutations. Although the myometrium did not express MED12 mutations, there were marked differences in the transcriptome landscape of the myometrium from mutated and non-mutated specimens, with genes regulating the response to oxygen-containing compounds being most altered. In conclusion, MED12 mutations have profound effects on the expression of genes pivotal to leiomyoma pathogenesis in the tumor and the myometrium which could alter tumor characteristics and growth potential.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA
| | - Jianjun Gao
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA
| | - Derek Quintanilla
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA
| | - Hayden McSwiggin
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA
| | - Drake Boos
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA
| | - Wei Yan
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90502, USA
| | - Omid Khorram
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
- The Lundquist Institute for Biomedical Innovation, Torrance, CA 90502, USA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA 90502, USA
- Correspondence: ; Tel.: +1-(310)-222-3867
| |
Collapse
|
9
|
A Possible Cause for the Differential Expression of a Subset of miRNAs in Mesenchymal Stem Cells Derived from Myometrium and Leiomyoma. Genes (Basel) 2022; 13:genes13071106. [PMID: 35885889 PMCID: PMC9319258 DOI: 10.3390/genes13071106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 12/01/2022] Open
Abstract
The aetiology of leiomyoma is debated; however, dysregulated progenitor cells or miRNAs appear to be involved. Previous profiling analysis of miRNA in healthy myometrium- (M-MSCs) and leiomyoma- (L-MSCs) derived mesenchymal stem cells (MSCs) identified 15 miRNAs differentially expressed between M-MSCs and L-MSCs. Here, we try to elucidate whether these differentially regulated 15 miRNAs arise as a conversion of M-MSCs along the differentiation process or whether they may originate from divergent cell commitment. To trace the origin of the dysregulation, a comparison was made of the expression of miRNAs previously identified as differentially regulated in M-MSCs and L-MSCs with that detected in MSCs from amniotic fluid (considered as a substitute for embryonic cells). The results do not allow for a foregone conclusion: the miRNAs converging to the adherens junction pathway showed a gradual change along the differentiation process, and the miRNAs which coincided with the other three pathways (ECM-receptor interaction, TGFβ and cell cycle) showed a complex, not linear, regulation and, therefore, a trend along the hypothetical differentiation process was not deduced. However, the role of miRNAs appears to be predominant in the onset of leiomyoma and may follow two different mechanisms (early commitment; exacerbation); furthermore, miRNAs can support the observed (epigenetic) predisposition.
Collapse
|
10
|
Chuang TD, Quintanilla D, Boos D, Khorram O. Differential Expression of Super-Enhancer-Associated Long Non-coding RNAs in Uterine Leiomyomas. Reprod Sci 2022; 29:2960-2976. [PMID: 35641855 PMCID: PMC9537225 DOI: 10.1007/s43032-022-00981-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/18/2022] [Indexed: 11/18/2022]
Abstract
Super-enhancer-associated long non-coding RNAs (SE-lncRNAs) are a specific set of lncRNAs transcribed from super-enhancer (SE) genomic regions. Recent studies have revealed that SE-lncRNAs play essential roles in tumorigenesis through the regulation of oncogenes. The objective of this study was to elucidate the expression profile of SE-lncRNAs with concurrent assessment of associated mRNAs in leiomyomas and paired myometrium. Arraystar SE-lncRNAs arrays were used to systematically profile the differentially expressed SE-lncRNAs along with the corresponding SE-regulated protein coding genes in eight leiomyomas and paired myometrium. The analysis indicated 7680 SE-lncRNAs were expressed, of which 721 SE-lncRNAs were overexpressed, while 247 SE-lncRNAs were underexpressed by 1.5-fold or greater in leiomyoma. Thirteen novel SE-lncRNAs and their corresponding protein coding genes were selected, and their expression was confirmed in eighty-one paired leiomyoma tissues by quantitative real-time PCR. The thirteen pairs of SE-lncRNAs and their corresponding protein coding genes included RP11-353N14.2/CBX4, SOCS2-AS1/SOCS2, RP1-170O19.14/HOXA11, CASC15/PRL, EGFLAM-AS1/EGFLAM, RP11-225H22/NEURL1, RP5-1086K13.1/CD58, AC092839.3/SPTBN1, RP11-69I8.3/CTGF, TM4SF1-AS1/TM4SF1, RP11-373D23/FOSL2, RP11-399K21.11/COMTD1, and CTB-113P19.1/SPARC. Among these SE-lncRNAs, the expression of SOCS2-AS1/SOCS2, RP11-353N14.2/CBX4, RP1-170O19.14/HOXA11, and RP11-225H22/NEURL1 was significantly higher in African Americans as compared with Caucasians. The expression of RP11-353N14.2/CBX4, SOCS2-AS1/SOCS2, CASC15/PRL, and CTB-113P19.1/SPARC was significantly higher in tumors with MED12-mutation-positive as compared with MED12-mutation-negative tumors. Collectively, our results indicate that the differential expression of SE in leiomyomas is another mechanism contributing to dysregulation of protein coding genes in leiomyomas and that race and MED12 mutation can influence the expression of a select group of SE.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, CA, 90502, USA
| | - Derek Quintanilla
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, CA, 90502, USA
| | - Drake Boos
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, CA, 90502, USA
| | - Omid Khorram
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, CA, 90502, USA.
| |
Collapse
|
11
|
Kim M, Kang D, Kwon MY, Lee HJ, Kim MJ. MicroRNAs as potential indicators of the development and progression of uterine leiomyoma. PLoS One 2022; 17:e0268793. [PMID: 35639702 PMCID: PMC9154092 DOI: 10.1371/journal.pone.0268793] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 05/07/2022] [Indexed: 11/24/2022] Open
Abstract
Recent studies demonstrated a significant role of several microRNAs (miRs) in the development of leiomyoma. Here, we investigated miR expression profiles using microarray and found a significantly higher expression of miRs in leiomyoma than in adjacent myometrium. We also confirmed the upregulation of five selected miRs including miR-181a-5p, 127-3p, 28-3p, 30b-5p and let-7c-5p in cellular proliferation, extracellular matrix turnover, and angiogenesis by RT-qPCR. Interestingly, the miRs showed a higher expression in cases of large leiomyoma or in patients with a history of transfusion due to anemia. We then analyzed the expression of the miR target molecules including Transforming Growth Factor Beta Receptor 2 (TGFBR2) and Insulin-like Growth Factor 2 mRNA Binding Protein 1 (IGF2BP1) via immunohistochemistry. TGFBR2 and IGF2BP1 were positively stained in 81% and 62.5% of leiomyoma tissues but not in adjacent myometrium. Both were more frequently positive in patients with ≥ 6 cm leiomyoma and mass effect. The mean expression levels of miR-181a-5p, 127-3p, 28-3p, 30b-5p and let-7c-5p were higher in cases with TGFBR2 and IGF2BP1 positive leiomyoma. We observed several miRs were overexpressed in leiomyoma tissues, and these results provide insight into the role of miRs in the development and progression of leiomyoma and underscore the need to validate their utility as diagnostic or therapeutic targets.
Collapse
Affiliation(s)
- Myungshin Kim
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Dain Kang
- Catholic Genetic Laboratory Center, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi Yeon Kwon
- Clinical Medicine Research, Bucheon St. Mary’s hospital, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Hee Jeong Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Min Jeong Kim
- Department of Obstetrics & Gynecology, Buheon St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
12
|
Navarro A, Bariani MV, Park HS, Zota AR, Al-Hendy A. Report of Exosomes Isolated from a Human Uterine Leiomyoma Cell Line and Their Impact on Endometrial Vascular Endothelial Cells. Pharmaceuticals (Basel) 2022; 15:577. [PMID: 35631403 PMCID: PMC9143402 DOI: 10.3390/ph15050577] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 11/29/2022] Open
Abstract
Uterine leiomyomas are the most common pelvic tumor in women of reproductive age; they cause irregular heavy menstrual bleeding leading to anemia and subsequent negative effects on quality of life. Exosomes have arisen as main players of disease progression in several illnesses, including a range of benign and malignant conditions; however, their role in leiomyomas' pathophysiology remains unknown. We investigated the effect of exosomes derived from human uterine leiomyoma tumor cells (HULM) and human myometrial cells (UTSM) on the behavior of human endometrial microvascular endothelial cells (HEMEC). HULM- and UTSM-derived exosomes were isolated and cocultured with HEMECs. Then, cell proliferation, mRNA expression, tube formation assay, and RNA-seq were performed. Treatment of HEMEC with HULM-derived exosomes increased cell proliferation by 60% compared to control untreated cells, upregulated C-MYC and VEGFA expression levels, and increased tube formation, length, and branching (markers of angiogenesis). Profiling of miRNA revealed that 84 miRNAs were significantly downregulated and 71 were upregulated in HULM-derived exosomes compared to UTSM-derived exosomes. These findings suggest that HULM-derived exosomes might have effects on HEMEC function, containing factors that enhance endometrial proliferation and angiogenesis, which may contribute to heavy menstrual bleeding. Further research on exosomes in uterine leiomyoma may identify possible novel biomarkers for treatment.
Collapse
Affiliation(s)
- Antonia Navarro
- Department of Obstetrics & Gynecology, University of Chicago, Chicago, IL 60637, USA; (A.N.); (M.V.B.); (H.-S.P.)
| | - Maria Victoria Bariani
- Department of Obstetrics & Gynecology, University of Chicago, Chicago, IL 60637, USA; (A.N.); (M.V.B.); (H.-S.P.)
| | - Hang-Soo Park
- Department of Obstetrics & Gynecology, University of Chicago, Chicago, IL 60637, USA; (A.N.); (M.V.B.); (H.-S.P.)
| | - Ami R. Zota
- Department of Environment Health, Milken School of Public Health, George Washington University, Washington, DC 20037, USA;
| | - Ayman Al-Hendy
- Department of Obstetrics & Gynecology, University of Chicago, Chicago, IL 60637, USA; (A.N.); (M.V.B.); (H.-S.P.)
| |
Collapse
|
13
|
Chuang TD, Quintanilla D, Boos D, Khorram O. Long Noncoding RNA MIAT Modulates the Extracellular Matrix Deposition in Leiomyomas by Sponging MiR-29 Family. Endocrinology 2021; 162:6365958. [PMID: 34491311 PMCID: PMC8459448 DOI: 10.1210/endocr/bqab186] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Indexed: 01/01/2023]
Abstract
The objective of this study was to determine the expression and functional role of a long noncoding RNA (lncRNA) MIAT (myocardial infarction-associated transcript) in leiomyoma pathogenesis. Leiomyoma compared with myometrium (n = 66) expressed significantly more MIAT that was independent of race/ethnicity and menstrual cycle phase but dependent on MED12 (mediator complex subunit 12) mutation status. Leiomyomas bearing the MED12 mutation expressed higher levels of MIAT and lower levels of microRNA 29 family (miR-29a, -b, and -c) compared with MED12 wild-type leiomyomas. Using luciferase reporter activity and RNA immunoprecipitation analysis, MIAT was shown to sponge the miR-29 family. In a 3-dimensional spheroid culture system, transient transfection of MIAT siRNA in leiomyoma smooth muscle cell (LSMC) spheroids resulted in upregulation of miR-29 family and downregulation of miR-29 targets, collagen type I (COL1A1), collagen type III (COL3A1), and TGF-β3 (transforming growth factor β-3). Treatment of LSMC spheroids with TGF-β3 induced COL1A1, COL3A1, and MIAT levels, but repressed miR-29 family expression. Knockdown of MIAT in LSMC spheroids blocked the effects of TGF-β3 on the induction of COL1A1 and COL3A1 expression. Collectively, these results underscore the physiological significance of MIAT in extracellular matrix accumulation in leiomyoma.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Ob/Gyn Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, CA 90502, USA
| | - Derek Quintanilla
- Department of Ob/Gyn Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, CA 90502, USA
| | - Drake Boos
- Department of Ob/Gyn Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, CA 90502, USA
| | - Omid Khorram
- Department of Ob/Gyn Harbor-UCLA Medical Center and The Lundquist Institute, Torrance, CA 90502, USA
- Correspondence: Omid Khorram, MD, PhD, Department of Ob/Gyn, Harbor-UCLA Medical Center, 1124 W. Carson St., Box 467, Torrance, CA 90502, USA.
| |
Collapse
|
14
|
Rooda I, Kaselt B, Liivrand M, Smolander OP, Salumets A, Velthut-Meikas A. Hsa-mir-548 family expression in human reproductive tissues. BMC Genom Data 2021; 22:40. [PMID: 34625017 PMCID: PMC8501715 DOI: 10.1186/s12863-021-00997-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/27/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Hsa-miR-548ba expressed in ovarian granulosa cells targets PTEN and LIFR, which are essential for ovarian follicle activation and growth. The expression pattern of hsa-miR-548ba correlates with its host gene follicle-stimulating hormone receptor (FSHR), and FSH has a positive influence on hsa-miR-548ba expression. However, hsa-miR-548ba is a member of a large hsa-mir-548 family with potentially overlapping targets. The current study aims to investigate the co-expression of hsa-mir-548 family members in FSHR-positive reproductive tissues and to explore the potential co-regulation of pathways. RESULTS For the above-described analysis, small RNA sequencing data from public data repositories were used. Sequencing results revealed that hsa-miR-548ba was expressed at the highest level in the ovarian granulosa cells and uterine myometrial samples together with another twelve and one hsa-miR-548 family members, respectively. Pathway enrichment analysis of microRNA targets in the ovarian samples revealed the hsa-miR-548ba and hsa-miR-548b-5p co-regulation of RAB geranylgeranylation in mural granulosa cells. Moreover, other hsa-mir-548 family members co-regulate pathways essential for ovarian functions (PIP3 activates AKT signalling and signalling by ERBB4). In addition to hsa-miR-548ba, hsa-miR-548o-3p is expressed in the myometrium, which separately targets the peroxisome proliferator-activated receptor alpha (PPARA) pathway. CONCLUSION This study reveals that hsa-mir-548 family members are expressed in variable combinations in the reproductive tract, where they potentially fulfil different regulatory roles. The results provide a reference for further studies of the hsa-mir-548 family role in the reproductive tract.
Collapse
Affiliation(s)
- Ilmatar Rooda
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia.
- Competence Centre on Health Technologies, Teaduspargi 13, 50411, Tartu, Estonia.
| | - Birgitta Kaselt
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Maria Liivrand
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Olli-Pekka Smolander
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| | - Andres Salumets
- Competence Centre on Health Technologies, Teaduspargi 13, 50411, Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 14186, Stockholm, Sweden
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, L. Puusepa St. 8, 50406, Tartu, Estonia
- Institute of Genomics, University of Tartu, Riia 23b, 51010, Tartu, Estonia
| | - Agne Velthut-Meikas
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, 12618, Tallinn, Estonia
| |
Collapse
|
15
|
Tong C, Wang C, Wang Y, Xiao X. TNRC6C-AS1 Promotes Thyroid Cancer Progression by Upregulating LPAR5 via miR-513c-5p. Cancer Manag Res 2021; 13:6141-6155. [PMID: 34393509 PMCID: PMC8354737 DOI: 10.2147/cmar.s312621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/19/2021] [Indexed: 12/21/2022] Open
Abstract
Background Considering the combined role of long non-coding RNA (lncRNAs)-microRNA (miRNA)-mRNA in tumorigenesis, the purpose of this study was to investigate how TNRC6C-AS1 regulates the expression of lysophosphatidic acid receptor 5 (LPAR5) by modulating miR-513c-5p, thus influencing the progression of thyroid cancer (THCA). Methods qRT-PCR and Western blotting were performed to detect the expression levels of TNRC6C-AS1, miR-513c-5p, and LPAR5 in THCA tissues and cell lines. The viability, proliferation, migration, and invasion were assessed using CCK-8, BrdU, wound healing, and transwell migration assays, respectively. Dual-luciferase reporter assay, RIP assay, and RNA pull-down assay were used to evaluate the relationship between TNRC6C-AS1, miR-513c-5p, and LPAR5. Results TNRC6C-AS1 was highly expressed in THCA tissues, and knockout of TNRC6C-AS1 reduced the viability, proliferation, migration, and invasion of THCA cells. TNRC6C-AS1 competitively adsorbed miR-513c-5p. In addition, the biological function of TNRC6C-AS1 was blocked by knocking down the thyroid cell line TNRC6C-AS1 with miR-513c-5p inhibitor transfection. LPAR5 is the target gene for miR-513c-5p, which has the ability to eliminate the influence of miR-513c-5p on THCA cells. Conclusion The TNRC6C-AS1/miR-513c-5p/LPAR5 axis is a novel signaling pathway that modulates THCA progression and may be a potential target for cancer therapy.
Collapse
Affiliation(s)
- Chuanming Tong
- Department of General Surgery, People's Hospital of Dongxihu District, Wuhan, Hubei, 430040, People's Republic of China
| | - Chuan Wang
- Department of General Surgery, People's Hospital of Dongxihu District, Wuhan, Hubei, 430040, People's Republic of China
| | - Yajie Wang
- Department of General Surgery, People's Hospital of Dongxihu District, Wuhan, Hubei, 430040, People's Republic of China
| | - Xiongsheng Xiao
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524000, People's Republic of China
| |
Collapse
|
16
|
Chuang TD, Quintanilla D, Boos D, Khorram O. Tryptophan catabolism is dysregulated in leiomyomas. Fertil Steril 2021; 116:1160-1171. [PMID: 34116832 DOI: 10.1016/j.fertnstert.2021.05.081] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To determine the expression and functional roles of indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO2) in leiomyoma. DESIGN Experimental study. SETTING Academic research laboratory. PATIENT(S) Women undergoing hysterectomy for leiomyoma. INTERVENTION(S) Blockade of IDO1 and TDO2. MAIN OUTCOME MEASURE(S) Expression of IDO1 and TDO2 in leiomyoma and the effects of their inhibitors on the extracellular matrix. RESULT(S) Leiomyoma expressed significantly higher levels of IDO1 and TDO2 messenger ribonucleic acid (mRNA; 60.3%, 35/58 pairs and 98.3%, 57/58 pairs, respectively) and protein (54%, 27/50 pairs and 92%, 46/50 pairs, respectively) as well as the enzyme activity marker kynurenine (78.3%, 36/46 pairs for IDO1/TDO2) compared with levels in matched myometrium. The expression of TDO2 but not IDO1 mRNA was significantly higher in fibroids from African American compared with that in Caucasian and Hispanic patients. The TDO2 but not the IDO1 protein and mRNA levels were more abundant in fibroids bearing the MED12 mutation compared with results in wild-type leiomyomas. Treatment of leiomyoma smooth muscle cell and myometrial smooth muscle cell spheroids with the TDO2 inhibitor 680C91 but not the IDO1 inhibitor epacadostat significantly repressed cell proliferation and the expression of collagen type I (COL1A1) and type III (COL3A1) in a dose-dependent manner; these effects were more pronounced in leiomyoma smooth muscle cells compared with myometrial smooth muscle cell spheroids. CONCLUSION(S) These results underscore the physiological significance of the tryptophan degradation pathway in the pathogenesis of leiomyomas and the potential utility of anti-TDO2 drugs for treatment of leiomyomas.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Obstetrics and Gynecology, Harbor-University of California-Los Angeles Medical Center and The Lundquist Institute, Torrance, California
| | - Derek Quintanilla
- Department of Obstetrics and Gynecology, Harbor-University of California-Los Angeles Medical Center and The Lundquist Institute, Torrance, California
| | - Drake Boos
- Department of Obstetrics and Gynecology, Harbor-University of California-Los Angeles Medical Center and The Lundquist Institute, Torrance, California
| | - Omid Khorram
- Department of Obstetrics and Gynecology, Harbor-University of California-Los Angeles Medical Center and The Lundquist Institute, Torrance, California.
| |
Collapse
|
17
|
Falahati Z, Mohseni-Dargah M, Mirfakhraie R. Emerging Roles of Long Non-coding RNAs in Uterine Leiomyoma Pathogenesis: a Review. Reprod Sci 2021; 29:1086-1101. [PMID: 33844188 DOI: 10.1007/s43032-021-00571-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/01/2021] [Indexed: 01/19/2023]
Abstract
Uterine leiomyoma (UL), as the most prevalent type of women's health disorders, is a benign tumor that originates from the smooth muscle cell layer of the uterus. A great number of associated complications are observed including infertility, miscarriage, bleeding, pain, dysmenorrhea, menorrhagia, and dyspareunia. Although the etiology of UL is largely undefined, environmental and genetic factors are witnessed to engage in the UL development. As long non-coding RNAs (lncRNAs) are involved in various types of cellular functions, in recent years, a great deal of attention has been drawn to them and their possible roles in UL pathogenesis. Moreover, they have illustrated their potential to be promising candidates for UL treatment. In this review paper, firstly, an overview of UL pathogenesis is presented. Then, the regulation of lncRNAs in UL and their possible mechanisms in cancer development are reviewed. Eventually, therapeutic approaches targeting lncRNAs in various cancers and UL are explored.
Collapse
Affiliation(s)
- Zahra Falahati
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan, Iran
| | - Masoud Mohseni-Dargah
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- School of Engineering, Faculty of Science and Engineering, Macquarie University, Sydney, Australia
| | - Reza Mirfakhraie
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Koodakyar St., Velenjak Ave, Chamran Highway, Tehran, Iran.
- Genomic Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Ciarmela P, Petraglia F. New epigenetic mechanism involved in leiomyoma formation. Fertil Steril 2020; 115:94-95. [PMID: 33272630 DOI: 10.1016/j.fertnstert.2020.09.143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 09/15/2020] [Indexed: 11/25/2022]
Affiliation(s)
- Pasquapina Ciarmela
- Department of Experimental and Clinical Medicine, Faculty of Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Felice Petraglia
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio," Obstetrics and Gynecology, University of Florence, Florence, Italy
| |
Collapse
|
19
|
Wang W, Zhou L, Wang J, Zhang X, Liu G. Circular RNA expression profiling identifies novel biomarkers in uterine leiomyoma. Cell Signal 2020; 76:109784. [DOI: 10.1016/j.cellsig.2020.109784] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 12/12/2022]
|
20
|
Chuang TD, Rehan A, Khorram O. Functional role of the long noncoding RNA X-inactive specific transcript in leiomyoma pathogenesis. Fertil Steril 2020; 115:238-247. [PMID: 33070965 DOI: 10.1016/j.fertnstert.2020.07.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 07/07/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To determine the expression and functional roles of a long noncoding RNA (lncRNA) X-inactive specific transcript (XIST) in leiomyoma. DESIGN Experimental study. SETTING Academic research laboratory. PATIENT(S) Women undergoing hysterectomy for leiomyoma. INTERVENTION(S) Overexpression and underexpression of XIST; blockade of specific protein 1 (SP1). MAIN OUTCOME MEASURE(S) Expression of XIST in leiomyoma and its effects on microRNA 29c (miR-29c), miR-200c, and their targets. RESULT(S) Leiomyoma expressed statistically significantly more XIST as compared with matched myometrium, independent of race/ethnicity and menstrual cycle phase. By use of a three-dimensional spheroid culture system, we found reduced XIST levels in leiomyoma smooth muscle cells (LSMC) after treatment with 17β-estradiol, progesterone, and their combination. The expression of XIST was down-regulated by treatment with the SP1-inhibitor mithramycin A and SP1 small interfering RNA. Knockdown of XIST resulted in inhibition of cell proliferation, up-regulation of miR-29c and miR-200c, and a concomitant inhibition of the target genes of these miRNAs, namely collagen type I (COL1A1), collagen type III (COL3A1), and fibronectin (FN1). By contrast, overexpression of XIST in myometrium smooth muscle cells repressed miR-29c and miR-200c, and induced COL1A1, COL3A1, and FN1 levels. By use of RNA immunoprecipitation analysis we confirmed XIST has sponge activity over miR-29c and miR-200c, which is more pronounced in leiomyoma as compared with myometrium. CONCLUSION(S) Our data demonstrate that increased expression of XIST in leiomyoma results in reduced expression of miR-29c and miR-200c with a consequent up-regulation of the genes targeted by these microRNAs including COL1A1, COL3A1, and FN1, which play key roles in extracellular matrix accumulation associated with fibroids.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, and Department of Obstetrics and Gynecology at Harbor-UCLA Medical Center, Torrance, California
| | - Anika Rehan
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, and Department of Obstetrics and Gynecology at Harbor-UCLA Medical Center, Torrance, California
| | - Omid Khorram
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, and Department of Obstetrics and Gynecology at Harbor-UCLA Medical Center, Torrance, California.
| |
Collapse
|
21
|
Deoxyelephantopin, a novel naturally occurring phytochemical impairs growth, induces G2/M arrest, ROS-mediated apoptosis and modulates lncRNA expression against uterine leiomyoma. Biomed Pharmacother 2020; 131:110751. [PMID: 33152917 DOI: 10.1016/j.biopha.2020.110751] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 09/04/2020] [Accepted: 09/07/2020] [Indexed: 12/22/2022] Open
Abstract
Deoxyelephantopin (DOE), a phytochemical, extracted and purified from Elephantopus scaber, has been shown to exhibit antitumor activities. Objective of the present study was to investigate anti-tumor and apoptosis-inducing properties of DOE against uterine leiomyoma (UL) and to explore their molecular mechanisms. Primary cell cultures from fresh UL tissue were established and maintained up to 12 passages. The cells exhibited continuous proliferation with 24 -h doubling time until 12 passages and was then subjected to molecular characterization. The growth inhibitory effect of DOE on UL cells was confirmed by colony formation, cellular senescence, AO/PI and DAPI staining. Fluorescent-activated cell sorting (FACS) assay for apoptosis and cell cycle arrest analysis revealed that DOE significantly (p < 0.05) inhibited the UL cell proliferation via cell cycle arrest at G2/M phase and induced apoptosis via ROS production by lowering mitochondrial membrane potential. Subsequently, the DOE induced ROS was markedly attenuated by co-treatment of N-Acetyl-Cysteine (NAC). Our quantitative RT-PCR and western blot results showed up-regulation of Bax, Caspase-3 and down-regulation of Bcl2, P53, αSMA, COL4A2, VEGF, PCNA, Cyclin B1 and oncogenic lncRNAs (H19, HOTAIR, BANCR and ROR) in DOE treated UL cells which further strengthen our findings. In conclusion, DOE inhibits growth of UL cells via cell cycle arrest at G2/M phase, induces ROS-dependent caspase-3-mediated mitochondrial intrinsic apoptotic pathway and down-regulation of oncogenic lncRNA in UL cells. Our findings suggest that DOE deserves for further systematic investigation in the uterine leiomyoma animal model as a novel apoptosis inducer for potential applications in the prevention or treatment of uterine leiomyoma.
Collapse
|
22
|
Chuang TD, Ansari A, Yu C, Sakurai R, Harb A, Liu J, Khorram O, Rehan VK. Mechanism underlying increased cardiac extracellular matrix deposition in perinatal nicotine-exposed offspring. Am J Physiol Heart Circ Physiol 2020; 319:H651-H660. [PMID: 32795172 DOI: 10.1152/ajpheart.00021.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although increased predisposition to cardiac fibrosis and cardiac dysfunction has been demonstrated in the perinatally nicotine-exposed heart, the underlying mechanisms remain unclear. With the use of a well-established rat model and cultured primary neonatal rat cardiac fibroblasts, the effect of perinatal nicotine exposure on offspring heart extracellular matrix deposition and the likely underlying mechanisms were investigated. Perinatal nicotine exposure resulted in increased collagen type I (COL1A1) and III (COL3A1) deposition along with a decrease in miR-29 family and an increase in long noncoding RNA myocardial infarction-associated transcript (MIAT) levels in offspring heart. Nicotine treatment of isolated primary neonatal rat cardiac fibroblasts suggested that these effects were mediated via nicotinic acetylcholine receptors including α7 and the induced collagens accumulation was reversed by a gain-of function of miR-29 family. Knockdown of MIAT resulted in increased miR-29 family and decreased COL1A1 and COL3A1 levels, suggesting nicotine-mediated MIAT induction as the underlying mechanism for nicotine-induced collagen deposition. Luciferase reporter assay and RNA immunoprecipitation studies showed an intense physical interaction between MIAT, miR-29 family, and argonaute 2, corroborating the mechanistic link between perinatal nicotine exposure and increased extracellular matrix deposition. Overall, perinatal nicotine exposure resulted in lower miR-29 family levels in offspring heart, while it elevated cardiac MIAT and collagen type I and III levels. These findings provide mechanistic basis for cardiac dysfunction in perinatal nicotine-exposed offspring and offer multiple novel potential therapeutic targets.NEW & NOTEWORTHY Using an established rat model and cultured primary neonatal cardiac fibroblasts, we show that nicotine mediated MIAT induction as the underlying mechanism for the excessive cardiac collagen deposition. These observations provide mechanistic basis for the increased predisposition to cardiac dysfunction following perinatal cigarette/nicotine exposure and offer novel potential therapeutic targets.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of and Obstetrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Aamir Ansari
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Celia Yu
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Reiko Sakurai
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Amir Harb
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Jie Liu
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Omid Khorram
- Department of and Obstetrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| | - Virender K Rehan
- Department of Pediatrics, Lundquist Institute for Biomedical Innovation at Harbor-University of California Los Angeles Medical Center, David Geffen School of Medicine at University of California Los Angeles, Torrance, California
| |
Collapse
|
23
|
Lu Q, Xu Q, Guo F, Lv Y, Song C, Feng M, Yu J, Zhang D, Cang J. Identification and characterization of long non-coding RNAs as competing endogenous RNAs in the cold stress response of Triticum aestivum. PLANT BIOLOGY (STUTTGART, GERMANY) 2020; 22:635-645. [PMID: 32249495 DOI: 10.1111/plb.13119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/24/2020] [Indexed: 05/11/2023]
Abstract
Long non-coding RNAs (lncRNAs) play important roles in plant development and stress responses. MicroRNAs (miRNAs) are involved in transcriptional and post-transcriptional gene regulation. It is not clear how lncRNA-mediated plant responses to cold stress and how lncRNAs, miRNAs and target mRNAs cooperate subject to the competing endogenous RNA (ceRNA). We interpreted the function of lncRNAs in the winter wheat cultivar Dongnongdongmai 1 (Dn1). A total of 9970 putative lncRNAs were initially identified from three Dn1 lncRNA libraries (5 °C, -10 °C and -25 °C) using high-throughput sequencing. Among the 14,626 genes detected via weighted gene co-expression network analysis, 7435 lncRNAs were co-expressed with 7191 mRNAs. We found six modules related to cold resistance in the lncRNA-mRNA weighted co-expression network, and the functions of mRNAs were similar in each module. Antioxidant systems and hormones played important roles in low-temperature responses. RNA sequencing analysis revealed that interactions between the 384 lncRNAs and 70 miRNAs were required for ceRNA activity. According to ceRNA activity, 225 lncRNAs, 60 miRNAs and 621 target mRNAs were involved in the regulatory networks of the cold stress response. Notably, a conserved region was found in the complementary regions of lncRNAs and miR164/408 but had reverse expression trends in the ceRNA network. Our results reveal possible roles of lncRNAs-mRNAs in the regulatory networks associated with tolerance to low temperature and provide useful information for more strategic use of genomic resources in wheat breeding.
Collapse
Affiliation(s)
- Q Lu
- College of Life Science, Northeast Agricultural University, Heilongjiang, China
| | - Q Xu
- College of Life Science, Northeast Agricultural University, Heilongjiang, China
| | - F Guo
- College of Life Science, Northeast Agricultural University, Heilongjiang, China
| | - Y Lv
- College of Life Science, Northeast Agricultural University, Heilongjiang, China
| | - C Song
- College of Life Science, Northeast Agricultural University, Heilongjiang, China
| | - M Feng
- College of Life Science, Northeast Agricultural University, Heilongjiang, China
| | - J Yu
- College of Life Science, Northeast Agricultural University, Heilongjiang, China
| | - D Zhang
- College of Life Science, Northeast Agricultural University, Heilongjiang, China
| | - J Cang
- College of Life Science, Northeast Agricultural University, Heilongjiang, China
| |
Collapse
|
24
|
Chuang TD, Khorram O. Cross-talk between miR-29c and transforming growth factor-β3 is mediated by an epigenetic mechanism in leiomyoma. Fertil Steril 2020; 112:1180-1189. [PMID: 31843095 DOI: 10.1016/j.fertnstert.2019.07.1324] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To determine the expression of miR-29c and its target gene transforming growth factor-β3 (TGF-β3) in leiomyoma and the mechanisms of their reciprocal regulation. DESIGN Experimental study. SETTING Academic research laboratory. PATIENT(S) Women undergoing hysterectomy for leiomyoma. INTERVENTION(S) Overexpression and underexpression of miR-29c; blockade of DNA methyltransferase 1 (DNMT1). MAIN OUTCOME MEASURE(S) The miR-29c and its target gene TGF-β3 in leiomyoma and the effects of TGF-β3 and blockade of DNMT1 on miR-29c expression. RESULT(S) Leiomyoma expressed significantly lower levels of miR-29c, but higher expression of TGF-β3 compared with matched myometrium. The expression of TGF-β3 and miR-29c were independent of race/ethnicity. Using 3' untranslated region luciferase reporter assay we confirmed that TGF-β3 is a direct target of miR-29c in leiomyoma smooth muscle cells (LSMCs). Gain-of-function of miR-29c in LSMCs inhibited the expression of TGF-β3 at protein and messenger RNA levels, whereas loss-of-function of miR-29c had the opposite effect. Treatment of LSMCs with TGF-β3 inhibited the expression of miR-29c, whereas it stimulated DNMT1 expression. Knockdown of DNMT1 through transfection with small interfering RNA significantly decreased the expression of TGF-β3, and induced miR-29c expression. Knockdown of DNMT1 also attenuated the inhibitory effect of TGF-β3 on miR-29c expression. Furthermore, we demonstrated that TGF-β3 increased the methylation level of miR-29c promoter in LSMCs. CONCLUSION(S) There is an inverse relationship in the expression of TGF-β3 and miR-29c in leiomyoma. The TGF-β3 is a direct target of miR-29c and inhibits the expression of miR-29c through an epigenetic mechanism. The cross-talk between miR-29c and TGF-β3 provides a feed forward mechanism of fibrosis in leiomyoma.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Obstetrics and Gynecology Harbor-University of California Los Angeles Medical Center and Los Angeles Biomedical Research Institute, Torrance, California
| | - Omid Khorram
- Department of Obstetrics and Gynecology Harbor-University of California Los Angeles Medical Center and Los Angeles Biomedical Research Institute, Torrance, California.
| |
Collapse
|
25
|
Tranilast induces MiR-200c expression through blockade of RelA/p65 activity in leiomyoma smooth muscle cells. Fertil Steril 2020; 113:1308-1318. [PMID: 32199621 DOI: 10.1016/j.fertnstert.2019.12.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/07/2019] [Accepted: 12/02/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVE To determine the mechanism by which tranilast induces miR-200c expression in leiomyoma smooth muscle cells (LSMCs). DESIGN Experimental study. SETTING Academic research laboratory. PATIENT(S) Women undergoing hysterectomy for leiomyoma. INTERVENTION(S) Blockade of RelA/p65. MAIN OUTCOME MEASURE(S) Effects of tranilast and blockade of RelA/p65 on miR-200c expression. RESULT(S) Tranilast, an inflammation inhibitor, dose-dependently induced miR-200c in LSMCs and myometrium smooth muscle cells (MSMCs), with a more profound effect in LSMCs than in MSMCs. The treatment of LSMCs with Bay 117082, an inhibitor of IκB phosphorylation, further enhanced miR-200c induction by tranilast. The knockdown of RelA/p65 by small interfering RNA also induced miR-200c expression in LSMCs. Although tranilast had no effect on total RelA/p65 protein levels in LSMCs, it significantly induced RelA/p65 phosphorylation at S536 while reducing its activity as well as its nuclear translocation. ChIP assay indicated that tranilast reduces the binding ability of RelA/p65 to miR-200c promoter, resulting in miR-200c induction. Tranilast also inhibited interleukin-8 (IL8) expression in LSMCs. The induction of miR-200c by tranilast partially mediates the inhibitory effect of tranilast on the expression of IL8 and cyclin-dependent kinase 2 in LSMCs. CONCLUSION(S) Induction of miR-200c by tranilast in LSMCs is mediated through a transcriptional mechanism involving inhibition of the nuclear factor κB signaling pathway. These results highlight the significance of inflammation in the pathogenesis of leiomyoma and the potential utility of antiinflammatory drugs for treatment of leiomyomas.
Collapse
|
26
|
Ali MA, Shaker OG, Alazrak M, AbdelHafez MN, Khalefa AA, Hemeda NF, Abdelmoktader A, Ahmed FA. Association analyses of a genetic variant in long non-coding RNA MEG3 with breast cancer susceptibility and serum MEG3 expression level in the Egyptian population. Cancer Biomark 2020; 28:49-63. [PMID: 32176630 DOI: 10.3233/cbm-191072] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND LncRNA MEG3 rs7158663 has been shown to confer cancer susceptibility, maybe through altering its gene expression level. OBJECTIVE We aimed to weigh the effect of rs7158663 on MEG3 serum level and breast cancer susceptibility. METHODS We genotyped rs7158663 G > A and measured serum MEG3 in 150 breast cancer, 95 fibroadenoma , and 154 controls by the TaqMan method. RESULTS The presence of rs7158663 G > A is a risk factor for breast cancer among fibroadenoma patients and controls, AA vs. GG genotypes (OR = 6.320, 95% CI = 2.587-15.439, P< 0.0001 when compared to controls and OR = 10.825, 95% CI = 1.929-60.742, P= 0.007 when compared to fibroadenoma). Decreased serum MEG3 was observed in breast cancer group when compared with fibroadenoma and/or controls [median (IQR) = 0.43 (0.27-0.55)] (P< 0.0001). However, increased serum MEG3 was noted in fibroadenoma group when compared with controls (P< 0.0001). A significance decreased serum MEG3 was found to be associated with mutant A allele than with wild G allele (P< 0.0001). The results showed that rs7158663 and lower MEG3 were significantly associated with patients with higher TNM staging and larger tumor size > 5 cm. CONCLUSION The presence of both rs7158663 and low MEG3 are diagnostic and unfavorable prognostic factors for BC patients.
Collapse
Affiliation(s)
- Marwa A Ali
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Olfat G Shaker
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Marwa N AbdelHafez
- Department of Medical Oncology, National Cancer Institute, Cairo University, Egypt
| | - Abeer A Khalefa
- Department of Physiology, Faculty of Medicine, Zagazig University, Egypt
| | - Nada F Hemeda
- Department of Genetics, Faculty of Agriculture, Fayoum University, Fayoum, Egypt
| | - Abdelrahman Abdelmoktader
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Fatma A Ahmed
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| |
Collapse
|
27
|
Anjum S, Sahar T, Nigam A, Wajid S. Transcriptome Analysis of mRNA in Uterine Leiomyoma Using Next-generation RNA Sequencing. Anticancer Agents Med Chem 2019; 19:1703-1718. [DOI: 10.2174/1871520619666190409102855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/18/2019] [Accepted: 03/22/2019] [Indexed: 12/14/2022]
Abstract
Background:
Uterine leiomyoma is a benign smooth muscle tumor of monoclonal nature in the
female reproductive tract and is one of the major health problems. More than 70% of the female population
suffers from uterine leiomyoma in their lifetime and in the advanced condition, it is associated with pregnancy
complications and infertility.
Objective:
Characterization and relative expression of mRNA transcripts through transcriptome profiling in
uterine leiomyoma and adjacent normal myometrium.
Methods:
Uterine leiomyoma tissue of an Indian female, age 32 years, with a family history of leiomyoma
(evident from mother’s hysterectomy for the same pathology) was used. Patient showed 9 multiple large lesions
appearing heterogeneously, deforming the uterine contour and causing distortion and splaying of the endometrial
cavity showing disease aggressiveness was taken for Next-generation sequencing (NGS) to develop
whole transcriptome profile along with the adjacent normal myometrium as control. The validation of the
relative expression of the selective transcripts was done using Real-Time PCR.
Results:
The transcriptome profile indicated 128 genes up-regulated and 98 down-regulated, with the Log2 fold
change ≥ 2 and P ≤ 0.05, highlighting the molecular network closely associated with focal adhesion, hyaluronan
and MAPK-signaling pathways. The mean relative fold change obtained from quantitative PCR as well as the
P-values of 10 selected transcripts evaluated from student’s t-test were as follows: BCAN: 7.93 fold (p-value
=0.0013); AAK1: 2.2 fold (p-value =0.0036); PCBP3: 3.4 fold (p-value =0.0197); MOV10L1: 3.4 fold (p-value
=0.0062); TWISTNB: 1.8 fold (p-value =0.006); TMSB15A: 2.1 fold (p-value =0.0023); SMAD1: 0.8 fold
(p-value =0.0112); ANXA1: 0.6 fold (p-value =0.0012); FOS: 0.6 fold (p-value =0.0191); SLFN11: 0.56 fold
(p-value =0.0001).
Conclusion:
The present study provides a roadmap, towards the analysis of genes and their roles in corresponding
pathways throwing light on their possible involvement in the pathology of the disease.
Collapse
Affiliation(s)
- Shadab Anjum
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Tahreem Sahar
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| | - Aruna Nigam
- Department of Obstetrics and Gynecology, HIMSR and HAH Centenary Hospital, Jamia Hamdard, New Delhi, 110062, India
| | - Saima Wajid
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
28
|
Li S, Zheng H, Chen L, Xu C, Qu X, Qin Z, Gao J, Li J, Liu J. Expression Profile and Potential Functions of Circulating Long Noncoding RNAs in Acute Ischemic Stroke in the Southern Chinese Han Population. Front Mol Neurosci 2019; 12:290. [PMID: 31849604 PMCID: PMC6895137 DOI: 10.3389/fnmol.2019.00290] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/13/2019] [Indexed: 12/24/2022] Open
Abstract
Background: Long noncoding RNAs (lncRNAs) have been confirmed to be associated with ischemic stroke (IS); however, their involvement still needs to be extensively explored. Therefore, we aimed to study the expression profile of lncRNAs and the potential roles and mechanisms of lncRNAs in the pathogenesis of acute ischemic stroke (AIS) in the Southern Chinese Han population. Methods: In this study, lncRNA and mRNA expression profiles in AIS were analyzed using high-throughput RNA sequencing (RNA-Seq) and validated using quantitative real-time polymerase chain reaction (qRT-PCR). Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment and network analyses were performed to predict the functions and interactions of the aberrantly expressed genes. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic value of lncRNAs in AIS. Results: RNA-Seq analysis showed that 428 lncRNAs and 957 mRNAs were significantly upregulated, while 791 lncRNAs and 4,263 mRNAs were downregulated in patients with AIS when compared with healthy controls. GO enrichment and KEGG pathway analyses of differentially expressed genes showed that the apoptosis, inflammatory, oxidative and calcium signaling pathways were potentially implicated in AIS pathology. The PCR results showed that the selected lncRNA-C14orf64 and lncRNA-AC136007.2 were significantly downregulated in AIS. ROC curve analysis showed that the area under the ROC curve (AUC) values of lncRNA-C14orf64 and lncRNA-AC136007.2 between AIS and healthy controls were 0.74 and 0.94, respectively. Conclusion: This study provides evidence of altered expression of lncRNAs and their potential functions in AIS. Our findings may facilitate pathological mechanistic studies of lncRNAs in AIS and provide potential diagnostic biomarkers and therapeutic targets for AIS.
Collapse
Affiliation(s)
- Shenghua Li
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huilei Zheng
- Department of Medical Examination and Health Management, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lan Chen
- Department of Internal Medicine, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chen Xu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiang Qu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhenxiu Qin
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinggui Gao
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jinpin Li
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jingli Liu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
29
|
Ning B, Yu D, Yu AM. Advances and challenges in studying noncoding RNA regulation of drug metabolism and development of RNA therapeutics. Biochem Pharmacol 2019; 169:113638. [PMID: 31518552 PMCID: PMC6802278 DOI: 10.1016/j.bcp.2019.113638] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 09/06/2019] [Indexed: 01/13/2023]
Abstract
Accumulating evidence has demonstrated that genome-derived noncoding RNAs (ncRNAs) play important roles in modulating inter-individual variations observed in drug metabolism and disposition by controlling the expression of genes coding drug metabolizing enzymes and transporters (DMETs) and relevant nuclear receptors (NRs). With the understanding of novel ncRNA regulatory mechanisms and significance in the control of disease initiation and progression, RNA-based therapies are under active investigation that may expand the druggable targets from conventional proteins to RNAs and the genome for the treatment of human diseases. Herein we provide an overview of research strategies, approaches and their limitations in biochemical and pharmacological studies pertaining to ncRNA functions in the regulation of drug and nutrient metabolism and disposition, and discussion on the promise and challenges in developing RNA therapeutics.
Collapse
Affiliation(s)
- Baitang Ning
- National Center for Toxicological Research (NCTR), US Food and Drug Administration, Jefferson, AR 72079, USA.
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao, China
| | - Ai-Ming Yu
- Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA.
| |
Collapse
|
30
|
Wu J, Chen L, Zheng C, Xu S, Gao Y, Wang J. Co-expression Network Analysis Revealing the Potential Regulatory Roles of lncRNAs in Alzheimer's Disease. Interdiscip Sci 2019; 11:645-654. [PMID: 30715720 DOI: 10.1007/s12539-019-00319-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 01/16/2023]
Abstract
Alzheimer's disease (AD) is one of the most common types of dementia among the elderly. Previous studies had revealed that the dysregulation of lncRNAs played important roles in human diseases, including AD. However, there is still a lack of comprehensive analysis of differently expressed long non-coding RNAs (lncRNAs) in different distinct regions related to AD. In present study, we identified a total of 678, 593, 941, 1445, 1179, 466 differently expressed lncRNAs that were found in entorhinal cortex (EC), middle temporal gyrus(MTG), hippocampus (HIP), superior frontal gyrus (SFG), posterior cingulate (PC), cortex and primary visual cortex (VCX) AD samples, respectively. Furthermore, we constructed lncRNA-mRNA co-expression networks in AD to explore the potential roles of these lncRNAs. Differentially expressed (DE) lncRNAs were involved in regulating metabolic process, respiratory electron transport chain and ATP metabolic process showed by GO analysis. Interestingly, KEGG analysis revealed these lncRNAs were associated with neurodegenerative disorders such as Alzheimer's disease, Huntington's disease and Parkinson's disease. Four lncRNAs (LOC100507557, LOC101929787, NEAT1, and JAZF1-AS1) were identified as key lncRNAs in AD progression and dysregulated in different distinct regions related to AD. Our study has uncovered several key lncRNAs in AD, which would give novel underlying therapeutic and prognostic targets for AD.
Collapse
Affiliation(s)
- Jiong Wu
- Department of Neurology, Zhejiang Hospital, Hangzhou, 310000, Zhejiang, China
| | - Linhui Chen
- Department of Neurology, Zhejiang Hospital, Hangzhou, 310000, Zhejiang, China
| | - Chaobo Zheng
- Department of Neurology, Zhejiang Hospital, Hangzhou, 310000, Zhejiang, China
| | - Shanhu Xu
- Department of Neurology, Zhejiang Hospital, Hangzhou, 310000, Zhejiang, China
| | - Yuhai Gao
- Department of Neurology, Zhejiang Hospital, Hangzhou, 310000, Zhejiang, China
| | - Junjun Wang
- Department of Neurology, Zhejiang Hospital, Hangzhou, 310000, Zhejiang, China.
| |
Collapse
|
31
|
Kim YJ, Kim YY, Shin JH, Kim H, Ku SY, Suh CS. Variation in MicroRNA Expression Profile of Uterine Leiomyoma with Endometrial Cavity Distortion and Endometrial Cavity Non-Distortion. Int J Mol Sci 2018; 19:E2524. [PMID: 30149651 PMCID: PMC6165274 DOI: 10.3390/ijms19092524] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/12/2018] [Accepted: 08/23/2018] [Indexed: 12/13/2022] Open
Abstract
The expression profile of microRNA (miRNA) in uterine leiomyoma (UL) cells is different from that in normal uterine myometrial (UM) cells. The effect of UL cells on uterine receptivity might vary according to their ability to distort the uterine endometrial cavity. However, the variation in miRNA expression profiles between endometrial cavity-distorting leiomyoma (ECDL) and endometrial cavity non-distorting leiomyoma (ECNDL) cells remains unknown. This study aimed to elucidate whether the expression profile of miRNAs in ECDL cells is dissimilar to that of ECNDL cells in uterus. Pelviscopic myomectomy was performed to obtain tissue samples of UL and their corresponding normal UM tissues (matched) from patients with UL (n = 26), among whom women with ECNDL and ECDL numbered 15 and 11, respectively. The relative expression of hsa-miR-15b, -29a, -29b, -29c, -197, and -200c as well as the candidate target genes in UL cells was compared to those in the matched UM cells using qRT-PCR to assess their ability to cause ECD. The spatial expression of miRNAs and target genes in the UL tissues was analyzed using in situ hybridization. Target gene expression was analyzed using qPCR after transfection with the mimics and inhibitors of miRNAs in UL cells. The relative expression level of miR-15b was upregulated, and the relative expression levels of miR-29a, -29b, -29c, -197, and -200c were downregulated in UL cells compared to those in UM cells. The relative expression levels of progesterone receptor, estrogen receptor, and matrix metalloproteinases (MMPs) were upregulated in UL cells compared to those in UM cells. The relative expression levels of miR-29c and -200c were downregulated, and the relative expression levels of estrogen receptor, MMPs and tissue inhibitors of metalloproteinases (TIMPs) were upregulated in ECDL cells compared to those in ECNDL cells. The expression profile of miRNAs in UL cells varied with respect to the occurrence or absence of endometrial cavity distortion. The biochemical properties of UL might be regulated by miRNAs in order to alter their effect on structural homeostasis of the uterus.
Collapse
Affiliation(s)
- Yong Jin Kim
- Department of Obstetrics and Gynecology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 110-744, Korea.
| | - Yoon Young Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul National University College of Medicine, 28 Yonkeun-dong, Chongno-gu, Seoul 110-744, Korea.
| | - Jung Ho Shin
- Department of Obstetrics and Gynecology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 110-744, Korea.
| | - Hoon Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul National University College of Medicine, 28 Yonkeun-dong, Chongno-gu, Seoul 110-744, Korea.
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul National University College of Medicine, 28 Yonkeun-dong, Chongno-gu, Seoul 110-744, Korea.
| | - Chang Suk Suh
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul National University College of Medicine, 28 Yonkeun-dong, Chongno-gu, Seoul 110-744, Korea.
| |
Collapse
|
32
|
Yang Q, Al-Hendy A. Non-coding RNAs: an important regulatory mechanism in pathogenesis of uterine fibroids. Fertil Steril 2018; 109:802-803. [PMID: 29778379 DOI: 10.1016/j.fertnstert.2018.02.120] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 01/25/2023]
Affiliation(s)
- Qiwei Yang
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, Illinois
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
33
|
Chuang TD, Xie Y, Yan W, Khorram O. Next-generation sequencing reveals differentially expressed small noncoding RNAs in uterine leiomyoma. Fertil Steril 2018; 109:919-929. [PMID: 29778390 PMCID: PMC6445395 DOI: 10.1016/j.fertnstert.2018.01.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/20/2018] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To determine the expression profile of small noncoding RNAs (sncRNAs) in leiomyoma, which has not been investigated to date. DESIGN Laboratory-based investigation. SETTING Academic center. PATIENT(S) Women undergoing hysterectomy for benign indications. INTERVENTION(S) Next-generation sequencing and screening of an sncRNA database with confirmatory analysis by quantitative reverse-transcription polymerase chain reaction (qRT-PCR). MAIN OUTCOME MEASURE(S) Expression profile of sncRNAs in leiomyoma and matched myometrium. RESULT(S) Screening our previously determined RNA sequencing data with the sncRNA database resulted in identification of 15 small nuclear (sn) RNAs, 284 small nucleolar (sno) RNAs, 98 Piwi-interacting (pi) RNAs, 152 transfer (t) RNAs, and 45 ribosomal (r) RNAs, of which 15 snoRNAs, 24 piRNAs, 7 tRNAs, and 6 rRNAs were differentially expressed at a 1.5-fold change cutoff in leiomyoma compared with myometrium. We selected 5 snoRNAs, 4 piRNAs, 1 tRNA, and 1 rRNA that were differentially expressed and confirmed their expression in paired tissues (n = 20) from both phases of the menstrual cycle with the use of qRT-PCR. The results indicated up-regulation of the snoRNAs (SNORD30, SNORD27, SNORA16A, SNORD46, and SNORD56) and down-regulation of the piRNAs (piR-1311, piR-16677, piR-20365, piR-4153), tRNA (TRG-GCC5-1), and rRNA (RNA5SP202) expression in leiomyoma compared with myometrium (P<.05). The pattern of expression of these sncRNAs was similar to RNA sequencing analysis, with no menstrual cycle-dependent differences detected except for SNORD30. Because Argonaute 2 (AGO2) is required for sncRNA-mediated gene silencing, we determined its expression and found greater abundance in leiomyoma. CONCLUSION(S) Our results provide the first evidence for the differential expression of additional classes of sncRNAs and AGO2 in leiomyoma, implicating their roles as a gene regulatory mechanism.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center and LA-Biomed Research Institute, Torrance, California
| | - Yeming Xie
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada
| | - Wei Yan
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, Nevada
| | - Omid Khorram
- Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center and LA-Biomed Research Institute, Torrance, California.
| |
Collapse
|
34
|
Maduro MR. Small Noncoding RNAs and Reproductive Physiology. Reprod Sci 2018; 25:633-634. [DOI: 10.1177/1933719118770258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
35
|
Chuang TD, Khorram O. Regulation of Cell Cycle Regulatory Proteins by MicroRNAs in Uterine Leiomyoma. Reprod Sci 2018; 26:250-258. [PMID: 29642801 DOI: 10.1177/1933719118768692] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The objective of this study was to determine whether miR-93, miR-29c, and miR-200c, which we previously reported to be downregulated in leiomyomas, target cell cycle regulatory proteins that influence cell proliferation. Based on TargetScan algorithm 3 cell cycle regulatory proteins namely, E2F transcription factor 1 (E2F1), Cyclin D1 (CCND1) and CDK2 which were predicted to be targets of these miRNAs were further analyzed. In 30 hysterectomy specimens, we found the expression of E2F1 and CCND1 messenger RNA (mRNA) was increased in leiomyoma as compared to matched myometrium, with no significant changes in CDK2 mRNA levels. There was a significant increase in the abundance of all 3 proteins in leiomyoma in comparison with matched myometrium. Using luciferase reporter assay, we demonstrated E2F1 and CCND1 are targets of miR-93 and CDK2 is a target of miR-29c and miR-200c. We confirmed these findings through transfection studies in which transfection of primary leiomyoma cells with miR-93 resulted in a significant decrease in the expression of E2F1 and CCND1 mRNA and protein levels, whereas knockdown of miR-93 had the opposite effect. Similarly, overexpression of miR-29c and miR-200c in leiomyoma cells inhibited the expression of CDK2 protein and mRNA, whereas knockdown of this microRNAs (miRNA) had the opposite effect. Transfection of miR-29c, miR-200c, and miR-93 in primary leiomyoma cells resulted in a time-dependent inhibition of cell proliferation and cell motility. These results collectively indicate that the 3 miRNAs known to be downregulated in fibroid tumors are critical in regulation of cell proliferation because of their effects on 3 key cell cycle regulatory proteins, which are overexpressed in uterine leiomyomas.
Collapse
Affiliation(s)
- Tsai-Der Chuang
- 1 Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center and LA-Biomed Research Institute, Torrance, CA, USA
| | - Omid Khorram
- 1 Department of Obstetrics and Gynecology, Harbor-UCLA Medical Center and LA-Biomed Research Institute, Torrance, CA, USA
| |
Collapse
|