1
|
Wang Y, Zhang Z, Li H, Wang M, Qiu Y, Lu L. miR-29b-3p regulates cardiomyocytes pyroptosis in CVB3-induced myocarditis through targeting DNMT3A. Cell Mol Biol Lett 2024; 29:55. [PMID: 38643118 PMCID: PMC11031889 DOI: 10.1186/s11658-024-00576-8] [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: 01/03/2024] [Accepted: 04/08/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Viral myocarditis (VMC) is a disease resulting from viral infection, which manifests as inflammation of myocardial cells. Until now, the treatment of VMC is still a great challenge for clinicians. Increasing studies indicate the participation of miR-29b-3p in various diseases. According to the transcriptome sequencing analysis, miR-29b-3p was markedly upregulated in the viral myocarditis model. The purpose of this study was to investigate the role of miR-29b-3p in the progression of VMC. METHODS We used CVB3 to induce primary cardiomyocytes and mice to establish a model of viral myocarditis. The purity of primary cardiomyocytes was identified by immunofluorescence. The cardiac function of mice was detected by Vevo770 imaging system. The area of inflammatory infiltration in heart tissue was shown by hematoxylin and eosin (H&E) staining. The expression of miR-29b-3p and DNMT3A was detected by quantitative real time polymerase chain reaction (qRT-PCR). The expression of a series of pyroptosis-related proteins was detected by western blot. The role of miR-29b-3p/DNMT3A in CVB3-induced pyroptosis of cardiomyocytes was studied in this research. RESULTS Our data showed that the expression of miR-29b-3p was upregulated in CVB3-induced cardiomyocytes and heart tissues in mice. To explore the function of miR-29b-3p in CVB3-induced VMC, we conducted in vivo experiments by knocking down the expression of miR-29b-3p using antagomir. We then assessed the effects on mice body weight, histopathology changes, myocardial function, and cell pyroptosis in heart tissues. Additionally, we performed gain/loss-of-function experiments in vitro to measure the levels of pyroptosis in primary cardiomyocytes. Through bioinformatic analysis, we identified DNA methyltransferases 3A (DNMT3A) as a potential target gene of miR-29b-3p. Furthermore, we found that the expression of DNMT3A can be modulated by miR-29b-3p during CVB3 infection. CONCLUSIONS Our results demonstrate a correlation between the expression of DNMT3A and CVB3-induced pyroptosis in cardiomyocytes. These findings unveil a previously unidentified mechanism by which CVB3 induces cardiac injury through the regulation of miR-29b-3p/DNMT3A-mediated pyroptosis.
Collapse
Affiliation(s)
- Ya Wang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, Hubei, People's Republic of China
| | - Zhengyang Zhang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, Hubei, People's Republic of China
| | - Hui Li
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, Hubei, People's Republic of China
| | - Min Wang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, Hubei, People's Republic of China
| | - Yuting Qiu
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, Hubei, People's Republic of China
| | - Lili Lu
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, College of Medicine, Wuhan University of Science and Technology, Wuhan, 430065, Hubei, People's Republic of China.
| |
Collapse
|
2
|
Zhang B, Yan Z, Gao Y, Li J, Wang Z, Wang P, Yang Q, Huang X, Gun S. Integrated analysis of miRNA and mRNA expression profiles in testes of Landrace and Hezuo boars. Front Vet Sci 2022; 9:942669. [PMID: 36330159 PMCID: PMC9622794 DOI: 10.3389/fvets.2022.942669] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/27/2022] [Indexed: 11/21/2022] Open
Abstract
Precocious puberty is closely related to testicular development and spermatogenesis, and there is increasing evidence that miRNAs are involved in regulation of testicular development and spermatogenesis. However, little is known about the regulation of microRNAs (miRNAs) during precocious maturation in Hezuo (HZ) boars. In this study, serum Testosterone (T), Estradiol (E2), Follicle-stimulating hormone (FSH) and Luteinizing hormone (LH) levels were detected in HZ and Landrace (LC) boars in the postnatal period at 30, 90, 120, 180, and 240 days, and the testes of HZ and LC boars at 30 and 120 days were used for histological observation. In addition, we performed small RNA-Seq to identify miRNA at sexual immaturity (30-days-old) and maturity (120-days-old) of HZ boar testis (using LC boar as control) to reveal the key miRNA in regulation of precocious puberty. Hormone assay results showed that high levels of T, E2, FSH, and LH may be related to precocious sexual maturity of HZ boars, and that FSH may play an important function before sexual maturity. Histological observation showed that HZ boars developed earlier than LC boars and had reached sexual maturity at 120 days. Small RNA-Seq yielded a total of 359 exist miRNAs, 767 known miRNAs and 322 novel miRNAs in 12 samples; 549, 468, 133, and 247 differentially expressed (DE) miRNAs were identified between Ha vs. Hb, La vs. Lb, Ha vs. La, and Hb vs. Lb (log2 fold change >1 and p < 0.05). Enrichment analysis showed that target genes of these DE miRNAs were enriched in many gene ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathways (such as PI3K-Akt, Hippo and Rap1 signaling pathways) were related to testicular development and spermatogenesis. Further screening, some miRNAs (such as ssc-miR-29b, ssc-miR-199b, ssc-miR-383, ssc-miR-149, ssc-miR-615, and ssc-miR-370) were possibly associated with precocious puberty. These results provide new light on miRNA regulatory mechanisms involved in precocious puberty.
Collapse
Affiliation(s)
- Bo Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Zunqiang Yan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Yi Gao
- Jilin Rongtai Agricultural Development Co., Ltd., Changchun, China
| | - Jiyou Li
- Gansu General Station of Animal Husbandry Technology Extension, Lanzhou, China
| | - Zike Wang
- Gansu General Station of Animal Husbandry Technology Extension, Lanzhou, China
| | - Pengfei Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Qiaoli Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Xiaoyu Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, China
- Gansu Research Center for Swine Production Engineering and Technology, Lanzhou, China
- *Correspondence: Shuangbao Gun
| |
Collapse
|
3
|
Zhao H, Zhang L, Li Q, Zhao Z, Duan Y, Huang Z, Ke H, Liu C, Li H, Liu L, Du J, Wei Z, Mou C, Zhou J. Integrated analysis of the miRNA and mRNA expression profiles in Leiocassis longirostris at gonadal maturation. Funct Integr Genomics 2022; 22:655-667. [PMID: 35467220 DOI: 10.1007/s10142-022-00857-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 03/09/2022] [Accepted: 03/22/2022] [Indexed: 11/04/2022]
Abstract
Leiocassis longirostris is a commercially important fish species that shows a sexually dimorphic growth pattern. A lack of molecular data from the gonads of this species has hindered research and selective breeding efforts. In this study, we conducted a comprehensive analysis of the expression profile of miRNA and mRNA to explore their regulatory roles in the gonadal maturation stage of L. longirostris. We identified 60 differentially expressed miRNAs and 20,752 differentially expressed genes by sequencing. A total of 90 miRNAs and 21 target genes involved in gonad development and sex determination were identified. Overall, the results of this study enhance our understanding of the molecular mechanisms underlying sex determination and differentiation and provide valuable genomic information for the selective breeding of L. longirostris.
Collapse
Affiliation(s)
- Han Zhao
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Lu Zhang
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Qiang Li
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Zhongmeng Zhao
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Yuanliang Duan
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Zhipeng Huang
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Hongyu Ke
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Chao Liu
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Huadong Li
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Lu Liu
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Jun Du
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Zhen Wei
- Leiocassis Longirostris Foundation Seed Farm, Sichuan Province, China
| | - Chengyan Mou
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China
| | - Jian Zhou
- Fisheries Institute, Sichuan Academy of Agricultural Sciences, Chengdu, 611731, Sichuan, China.
| |
Collapse
|
4
|
Ramakrishna NB, Murison K, Miska EA, Leitch HG. Epigenetic Regulation during Primordial Germ Cell Development and Differentiation. Sex Dev 2021; 15:411-431. [PMID: 34847550 DOI: 10.1159/000520412] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/10/2021] [Indexed: 11/19/2022] Open
Abstract
Germline development varies significantly across metazoans. However, mammalian primordial germ cell (PGC) development has key conserved landmarks, including a critical period of epigenetic reprogramming that precedes sex-specific differentiation and gametogenesis. Epigenetic alterations in the germline are of unique importance due to their potential to impact the next generation. Therefore, regulation of, and by, the non-coding genome is of utmost importance during these epigenomic events. Here, we detail the key chromatin changes that occur during mammalian PGC development and how these interact with the expression of non-coding RNAs alongside broader epitranscriptomic changes. We identify gaps in our current knowledge, in particular regarding epigenetic regulation in the human germline, and we highlight important areas of future research.
Collapse
Affiliation(s)
- Navin B Ramakrishna
- Wellcome/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Genome Institute of Singapore, A*STAR, Biopolis, Singapore, Singapore
| | - Keir Murison
- MRC London Institute of Medical Sciences, London, United Kingdom
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
| | - Eric A Miska
- Wellcome/CRUK Gurdon Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Trust Genome Campus, Cambridge, United Kingdom
| | - Harry G Leitch
- MRC London Institute of Medical Sciences, London, United Kingdom
- Institute of Clinical Sciences, Imperial College London, London, United Kingdom
- Centre for Paediatrics and Child Health, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
5
|
Ben Maamar M, Nilsson EE, Skinner MK. Epigenetic transgenerational inheritance, gametogenesis and germline development†. Biol Reprod 2021; 105:570-592. [PMID: 33929020 PMCID: PMC8444706 DOI: 10.1093/biolre/ioab085] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/12/2021] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
One of the most important developing cell types in any biological system is the gamete (sperm and egg). The transmission of phenotypes and optimally adapted physiology to subsequent generations is in large part controlled by gametogenesis. In contrast to genetics, the environment actively regulates epigenetics to impact the physiology and phenotype of cellular and biological systems. The integration of epigenetics and genetics is critical for all developmental biology systems at the cellular and organism level. The current review is focused on the role of epigenetics during gametogenesis for both the spermatogenesis system in the male and oogenesis system in the female. The developmental stages from the initial primordial germ cell through gametogenesis to the mature sperm and egg are presented. How environmental factors can influence the epigenetics of gametogenesis to impact the epigenetic transgenerational inheritance of phenotypic and physiological change in subsequent generations is reviewed.
Collapse
Affiliation(s)
- Millissia Ben Maamar
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Eric E Nilsson
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| | - Michael K Skinner
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
6
|
Wang Y, Liu X, Yu L, Hong X, Zhao J, Zhu J, Yuan J, Li W, Zhu X. Identification and analysis of novel microRNAs provide insights to reproductive capacity of the cultured Asian yellow pond turtle Mauremys mutica. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2021; 40:100890. [PMID: 34404014 DOI: 10.1016/j.cbd.2021.100890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/24/2021] [Accepted: 08/03/2021] [Indexed: 10/20/2022]
Abstract
The crucial roles of miRNAs in regulating animal growth, development, and disease resistance have been extensively reported, but their roles in relation to the reproductive capacity of aquatic animals (numbers of eggs laid and hatchlings), especially reptiles, remain unclear. In this study, high-throughput sequencing technology was used to screen miRNAs related to reproductive capacity based on the construction of a cDNA library of ovaries from higher-fecundity (HF) and lower-fecundity (LF) M. mutica. The results showed that 15,767,494 (93.98%) and 14,137,621 (94.17%) high-quality reads were obtained from the HF and LF groups, respectively. We screened 131 miRNAs that were differentially expressed between the HF and LF groups, of which 78 were upregulated and 53 were downregulated compared with the M. mutica reference genome. GO and KEGG pathway enrichment analyses of the target genes of differentially expressed miRNAs revealed significant differences in the enrichment frequencies of genes associated with ATP binding and proteolysis between the HF and LF groups, while the tricarboxylic acid cycle, glucagon signaling pathway and vitamin B6 metabolic pathway were shown to potentially help determine reproductive capacity. Ten miRNAs were verified by qRT-PCR to confirm the reliability and accuracy of the sequencing results, and a miRNA-mRNA target gene interaction network was constructed. These results will further our understanding of the regulatory mechanism of miRNAs in regards to turtle reproductive capacity.
Collapse
Affiliation(s)
- Yakun Wang
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong 510380, PR China
| | - Xiaoli Liu
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong 510380, PR China
| | - Lingyun Yu
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong 510380, PR China
| | - Xiaoyou Hong
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong 510380, PR China
| | - Jian Zhao
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong 510380, PR China
| | - Junxian Zhu
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong 510380, PR China
| | - Ju Yuan
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong 510380, PR China; College of Fisheries and Life Science, Shanghai Ocean University, Shanghai 201306, PR China
| | - Wei Li
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong 510380, PR China.
| | - Xinping Zhu
- Key Laboratory of Tropical & Subtropical Fishery Resource Application & Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong 510380, PR China.
| |
Collapse
|
7
|
LncRNAs induce oxidative stress and spermatogenesis by regulating endoplasmic reticulum genes and pathways. Aging (Albany NY) 2021; 13:13764-13787. [PMID: 34001678 PMCID: PMC8202879 DOI: 10.18632/aging.202971] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 11/08/2020] [Indexed: 12/17/2022]
Abstract
Oligozoospermia or low sperm count is a leading cause of male infertility worldwide. Despite decades of work on non-coding RNAs (ncRNAs) as regulators of spermatogenesis, fertilization, and male fertility, the literature on the function of long non-coding RNAs (lncRNAs) in human oligozoospermia is scarce. We integrated lncRNA and mRNA sequencing data from 12 human normozoospermic and oligozoospermic samples and comprehensively analyzed the function of differentially expressed lncRNAs (DE lncRNAs) and mRNAs (DE mRNAs) in male infertility. The target genes of DE lncRNAs were identified using a Gaussian graphical model. Gene ontology terms and Kyoto Encyclopedia of Genes and Genomes pathways were primarily enriched in protein transport and localization to the endoplasmic reticulum (ER). The lncRNA–mRNA co-expression network revealed cis- and trans-regulated target genes of lncRNAs. The transcriptome data implicated DE lncRNAs and DE mRNAs and their target genes in the accumulation of unfolded proteins in sperm ER, PERK-EIF2 pathway-induced ER stress, oxidative stress, and sperm cell apoptosis in individuals with oligozoospermia. These findings suggest that the identified lncRNAs and pathways could serve as effective therapeutic targets for male infertility.
Collapse
|
8
|
CEA, CA 15-3, and miRNA expression as potential biomarkers in canine mammary tumors. Chromosome Res 2021; 29:175-188. [PMID: 33638118 DOI: 10.1007/s10577-021-09652-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/26/2021] [Accepted: 02/02/2021] [Indexed: 10/22/2022]
Abstract
The most often detected tumor in intact bitches is mammary tumors and represents a significant clinical problem throughout the world. Mammary neoplasms in canine have heterogeneous morphology, so the choice of the most appropriate biomarker is the biggest challenge in CMT detection. We performed a retrospective analysis and evaluated the canine cancer antigens and miRNA expression profiles as potential biomarkers. Sixty dogs based on histological examination divided into three groups, viz., dogs with a benign mammary tumor, malignant mammary tumor, and control/healthy. The CA 15-3 was found more sensitive than CEA but detection of both will increase sensitivity. miR-21 expression differed significantly in all three groups. miR-29b expression differed significantly between the control and benign group and control and malignant group. The miR-21 overexpression and miR-29b downregulation with CMT are associated with clinical stage and can be used as non-invasive diagnostic and prognostic biomarkers. Hence, evaluation of CA 15-3 along with CEA would be a non-invasive technique for detecting canine mammary tumors. Evaluation of deregulated circulating miR-21 could be a valuable prognostic marker for early detection of mammary tumors in canines while miR-29b can add sensitivity in the detection of the canine mammary tumors if evaluated with miR-21.
Collapse
|
9
|
Alexandri C, Daniel A, Bruylants G, Demeestere I. The role of microRNAs in ovarian function and the transition toward novel therapeutic strategies in fertility preservation: from bench to future clinical application. Hum Reprod Update 2020; 26:174-196. [PMID: 32074269 DOI: 10.1093/humupd/dmz039] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/02/2019] [Accepted: 10/01/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND New therapeutic approaches in oncology have converted cancer from a certain death sentence to a chronic disease. However, there are still challenges to be overcome regarding the off-target toxicity of many of these treatments. Oncological therapies can lead to future infertility in women. Given this negative impact on long-term quality of life, fertility preservation is highly recommended. While gamete and ovarian tissue cryopreservation are the usual methods offered, new pharmacological-based options aiming to reduce ovarian damage during oncological treatment are very attractive. In this vein, advances in the field of transcriptomics and epigenomics have brought small noncoding RNAs, called microRNAs (miRNAs), into the spotlight in oncology. MicroRNAs also play a key role in follicle development as regulators of follicular growth, atresia and steroidogenesis. They are also involved in DNA damage repair responses and they can themselves be modulated during chemotherapy. For these reasons, miRNAs may be an interesting target to develop new protective therapies during oncological treatment. This review summarizes the physiological role of miRNAs in reproduction. Considering recently developed strategies based on miRNA therapy in oncology, we highlight their potential interest as a target in fertility preservation and propose future strategies to make the transition from bench to clinic. OBJECTIVE AND RATIONALE How can miRNA therapeutic approaches be used to develop new adjuvant protective therapies to reduce the ovarian damage caused by cytotoxic oncological treatments? SEARCH METHODS A systematic search of English language literature using PubMed and Google Scholar databases was performed through to 2019 describing the role of miRNAs in the ovary and their use for diagnosis and targeted therapy in oncology. Personal data illustrate miRNA therapeutic strategies to target the gonads and reduce chemotherapy-induced follicular damage. OUTCOMES This review outlines the importance of miRNAs as gene regulators and emphasizes the fact that insights in oncology can inspire new adjuvant strategies in the field of onco-fertility. Recent improvements in nanotechnology offer the opportunity for drug development using next-generation miRNA-nanocarriers. WIDER IMPLICATIONS Although there are still some barriers regarding the immunogenicity and toxicity of these treatments and there is still room for improvement concerning the specific delivery of miRNAs into the ovaries, we believe that, in the future, miRNAs can be developed as powerful and non-invasive tools for fertility preservation.
Collapse
Affiliation(s)
- C Alexandri
- Research Laboratory in Human Reproduction, Faculty of Medicine, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - A Daniel
- Research Laboratory in Human Reproduction, Faculty of Medicine, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium.,Université de Tours, Faculty of Science and Technology, 37200 Tours, France
| | - G Bruylants
- Engineering of Molecular NanoSystems, Ecole Polytechnique de Bruxelles, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium
| | - I Demeestere
- Research Laboratory in Human Reproduction, Faculty of Medicine, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium.,Fertility Clinic, CUB-Erasme, 1070 Brussels, Belgium
| |
Collapse
|
10
|
Wang Z, Dong S, Chen Q, Chen C, Xu Z, Dong Z. Analysis of the MicroRNA Profile by Sequencing in Ovarian Granular Cells from Women Suffering Fluorosis with Reproductive Dysfunction. Biol Trace Elem Res 2020; 197:101-106. [PMID: 31838738 DOI: 10.1007/s12011-019-01990-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 11/20/2019] [Indexed: 10/25/2022]
Abstract
Excessive intake of fluoride may cause female reproductive dysfunction but pathological mechanism is unclear. The miRNAs in follicular fluid are a class of small non-coding RNAs from granulosa cells. The aim of this study is to examine the differential expressions of miRNAs in ovarian granulosa cells from women suffering from fluorosis and infertility. Five fluorosis women suffering infertility and 5 non-fluorosis (normal ovarian secretory function control) women were included as two groups. These two groups were indexed by serum and urine fluoride (F) levels as high F group and control group. The concentrations of estradiol (E2), progesterone (P), human chorionic gonadotropin (HCG), follicle-stimulating hormone (FSH), and luteinizing hormone (LH) were measured by ELISA kits. The characteristics of menstruation from each woman were collected. The total RNA was isolated from granulosa cells for sequencing. The validation was completed by RT-qPCR. There was no significant difference between the two groups on age, the levels of E2, FSH, P, HCG, and LH in serum, and the characteristics of menstruation. A total of 37 miRNAs were upregulated and 40 miRNAs downregulated in follicular fluid from granulosa cells in the high F group compared with that in the control group. The expression of hsa-miR-29b increased in fluorosis patients identified by the miRNA sequence analysis. The distinct expression patterns of miRNAs in women requiring IVF from fluorosis area provide a new direction for molecular mechanism of fluorosis caused reproductive dysfunction.
Collapse
Affiliation(s)
- Zhihong Wang
- Department of Geriatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Siyuan Dong
- Guipei class 55, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Qun Chen
- Institute of Endemic Diseases, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission of the People's Republic of China, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, People's Republic of China.
| | - Chen Chen
- Endocrinology, School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Zhao Xu
- School of science, Xi'an Jiaotong University, Xi'an, China
| | - Zhaoheng Dong
- Shandong Shenghua New Material Technology Co., Ltd., Yantai, Shandong, China
| |
Collapse
|
11
|
Hui Y, Zhang Y, Wang K, Pan C, Chen H, Qu L, Song X, Lan X. Goat DNMT3B: An indel mutation detection, association analysis with litter size and mRNA expression in gonads. Theriogenology 2020; 147:108-115. [PMID: 32122684 DOI: 10.1016/j.theriogenology.2020.02.025] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 02/10/2020] [Accepted: 02/16/2020] [Indexed: 12/20/2022]
Abstract
DNA methyltransferase 3β (DNMT3B) is a gene encoding a de novo methylation enzyme that is required for DNA methylation during mammalian embryo development. Previous genome-wide association analysis suggested DNMT3B is a candidate gene for goat fertility, but there is no study on the effect of DNMT3B on litter size in goat. The aim of this study was to identify possible insertion/deletion (indel) mutations associated with litter size. Seven putative indels were designed to study their association with litter size, but just one 11-bp insertion variant of intron 22 (the last intron) was found in healthy female Shaanbei white cashmere goats (SBWC goats) (n = 1534). Statistical analysis showed that the 11-bp insertion was related to the first-born litter size (P < 0.01) and the goats with the deletion/deletion genotype had a higher average first-born litter size (P < 0.01). In addition, the expression profile of the DNMT3B mRNA in goat was detected, which revealed significant differences in DNMT3B mRNA expression in the gonads. Additionally, the results of western blotting revealed that the ovaries of mothers of multi-lamb (MML) had a higher level of DNMT3B protein than the ovaries of mothers of single-lamb (MSL). Furthermore, the mRNA of DNMT3B was widely expressed in male goats. Differences in mRNA expression levels were observed in the ovaries of MSL and MML. These findings indicated that the 11-bp indel in DNMT3B was significantly associated with first-born litter size, which can be used for marker-assisted selection (MAS) of goats for breeding.
Collapse
Affiliation(s)
- Yiqing Hui
- College of Animals Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling, Shaanxi, 712100, P.R. China.
| | - Yanghai Zhang
- College of Animals Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling, Shaanxi, 712100, P.R. China.
| | - Ke Wang
- College of Animals Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling, Shaanxi, 712100, P.R. China.
| | - Chuanying Pan
- College of Animals Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling, Shaanxi, 712100, P.R. China.
| | - Hong Chen
- College of Animals Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling, Shaanxi, 712100, P.R. China.
| | - Lei Qu
- Shaanxi Provincial Engineering and Technology Research Center of Cashmere Goats, Yulin University, Yulin, Shaanxi, 719000, PR China; College of Life Sciences, Yulin University, Yulin, Shaanxi, 719000, PR China.
| | - Xiaoyue Song
- Shaanxi Provincial Engineering and Technology Research Center of Cashmere Goats, Yulin University, Yulin, Shaanxi, 719000, PR China; College of Life Sciences, Yulin University, Yulin, Shaanxi, 719000, PR China.
| | - Xianyong Lan
- College of Animals Science and Technology, Northwest A&F University, No.22 Xinong Road, Yangling, Shaanxi, 712100, P.R. China.
| |
Collapse
|
12
|
Ambeskovic M, Ilnytskyy Y, Kiss D, Currie C, Montina T, Kovalchuk I, Metz GAS. Ancestral stress programs sex-specific biological aging trajectories and non-communicable disease risk. Aging (Albany NY) 2020; 12:3828-3847. [PMID: 32087063 PMCID: PMC7066928 DOI: 10.18632/aging.102848] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/05/2020] [Indexed: 12/18/2022]
Abstract
The incidence of non-communicable diseases (NCDs) is rising globally but their causes are generally not understood. Here we show that cumulative ancestral stress leads to premature aging and raises NCD risk in a rat population. This longitudinal study revealed that cumulative multigenerational prenatal stress (MPS) across four generations (F0-F3) raises age- and sex-dependent adverse health outcomes in F4 offspring. MPS accelerated biological aging processes and exacerbated sex-specific incidences of respiratory and kidney diseases, inflammatory processes and tumors. Unbiased deep sequencing of frontal cortex revealed that MPS altered expression of microRNAs and their target genes involved in synaptic plasticity, stress regulation, immune function and longevity. Multi-layer top-down deep learning metabolite enrichment analysis of urine markers revealed altered metabolic homeodynamics in MPS males. Thus, peripheral metabolic signatures may provide sensitive biomarkers of stress vulnerability and disease risk. Programming by MPS appears to be a significant determinant of lifetime mental health trajectories, physical wellbeing and vulnerability to NCDs through altered epigenetic regulation.
Collapse
Affiliation(s)
- Mirela Ambeskovic
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge T1K 3M4, Alberta, Canada
| | - Yaroslav Ilnytskyy
- Department of Biological Sciences, University of Lethbridge, Lethbridge T1K 3M4, Alberta, Canada
| | - Douglas Kiss
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge T1K 3M4, Alberta, Canada
| | - Cheryl Currie
- Faculty of Health Sciences, University of Lethbridge, Lethbridge T1K 3M4, Alberta, Canada
| | - Tony Montina
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge T1K 3M4, Alberta, Canada
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge T1K 3M4, Alberta, Canada
| | - Gerlinde A S Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge T1K 3M4, Alberta, Canada
| |
Collapse
|
13
|
Kamalidehghan B, Habibi M, Afjeh SS, Shoai M, Alidoost S, Almasi Ghale R, Eshghifar N, Pouresmaeili F. The Importance of Small Non-Coding RNAs in Human Reproduction: A Review Article. APPLICATION OF CLINICAL GENETICS 2020; 13:1-11. [PMID: 32021379 PMCID: PMC6956659 DOI: 10.2147/tacg.s207491] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 12/04/2019] [Indexed: 12/14/2022]
Abstract
Background MicroRNAs (miRNA) play a key role in the regulation of gene expression through the translational suppression and control of post-transcriptional modifications. Aim Previous studies demonstrated that miRNAs conduct the pathways involved in human reproduction including maintenance of primordial germ cells (PGCs), spermatogenesis, oocyte maturation, folliculogenesis and corpus luteum function. The association of miRNA expression with infertility, polycystic ovary syndrome (PCOS), premature ovarian failure (POF), and repeated implantation failure (RIF) was previously revealed. Furthermore, there are evidences of the importance of miRNAs in embryonic development and implantation. Piwi-interacting RNAs (piRNAs) and miRNAs play an important role in the post-transcriptional regulatory processes of germ cells. Indeed, the investigation of small RNAs including miRNAs and piRNAs increase our understanding of the mechanisms involved in fertility. In this review, the current knowledge of microRNAs in embryogenesis and fertility is discussed. Conclusion Further research is necessary to provide new insights into the application of small RNAs in the diagnosis and therapeutic approaches to infertility.
Collapse
Affiliation(s)
- Behnam Kamalidehghan
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Habibi
- Central Laboratory, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara S Afjeh
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Shoai
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - Saeideh Alidoost
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Rouzbeh Almasi Ghale
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nahal Eshghifar
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran and Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farkhondeh Pouresmaeili
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Zhao W, Cheng L, Quek C, Bellingham SA, Hill AF. Novel miR-29b target regulation patterns are revealed in two different cell lines. Sci Rep 2019; 9:17449. [PMID: 31767948 PMCID: PMC6877611 DOI: 10.1038/s41598-019-53868-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 11/06/2019] [Indexed: 12/26/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs that regulate gene or protein expression by targeting mRNAs and triggering either translational repression or mRNA degradation. Distinct expression levels of miRNAs, including miR-29b, have been detected in various biological fluids and tissues from a large variety of disease models. However, how miRNAs "react" and function in different cellular environments is still largely unknown. In this study, the regulation patterns of miR-29b between human and mouse cell lines were compared for the first time. CRISPR/Cas9 gene editing was used to stably knockdown miR-29b in human cancer HeLa cells and mouse fibroblast NIH/3T3 cells with minimum off-targets. Genome editing revealed mir-29b-1, other than mir-29b-2, to be the main source of generating mature miR-29b. The editing of miR-29b decreased expression levels of its family members miR-29a/c via changing the tertiary structures of surrounding nucleotides. Comparing transcriptome profiles of human and mouse cell lines, miR-29b displayed common regulation pathways involving distinct downstream targets in macromolecular complex assembly, cell cycle regulation, and Wnt and PI3K-Akt signalling pathways; miR-29b also demonstrated specific functions reflecting cell characteristics, including fibrosis and neuronal regulations in NIH/3T3 cells and tumorigenesis and cellular senescence in HeLa cells.
Collapse
Affiliation(s)
- Wenting Zhao
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
- Department of Biochemistry and Molecular Biology, Bio21 Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Lesley Cheng
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - Camelia Quek
- Department of Biochemistry and Molecular Biology, Bio21 Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Shayne A Bellingham
- Department of Biochemistry and Molecular Biology, Bio21 Institute, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
15
|
Khalid M, Abdollahi M. Epigenetic modifications associated with pathophysiological effects of lead exposure. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2019; 37:235-287. [PMID: 31402779 DOI: 10.1080/10590501.2019.1640581] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Lead (Pb) exposure during different stages of development has demonstrated dose, duration, sex, and tissue-specific pathophysiological outcomes due to altered epigenetic regulation via (a) DNA methylation, (b) histone modifications, (c) miRNAs, and (d) chromatin accessibility. Pb-induced alteration of epigenetic regulation causes neurotoxic and extra-neurotoxic pathophysiological outcomes. Neurotoxic effects of Pb include dysfunction of memory and learning, behavioral disorder, attention deficit hyperactivity disorder, autism spectrum disorder, aging, Alzheimer's disease, tauopathy, and neurodegeneration. Extra-neurotoxic effects of Pb include altered body weight, metabolic disorder, cardiovascular disorders, hematopoietic disorder, and reproductive impairment. Pb exposure either early in life or at any stage of development results in undesirable pathophysiological outcomes that tends to sustain and maintain for a lifetime.
Collapse
Affiliation(s)
- Madiha Khalid
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Movahed E, Soleimani M, Hosseini S, Akbari Sene A, Salehi M. Aberrant expression of miR-29a/29b and methylation level of mouse embryos after in vitro fertilization and vitrification at two-cell stage. J Cell Physiol 2019; 234:18942-18950. [PMID: 30916357 DOI: 10.1002/jcp.28534] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/24/2019] [Accepted: 03/05/2019] [Indexed: 12/17/2022]
Abstract
Proper epigenetic modifications during preimplantation embryo development are important for a successful pregnancy. We aim to investigate the putative influence of in vitro fertilization (IVF) and vitrification on DNA methylation in mouse preimplantation embryos. The study groups consisted of blastocyst-derived vitrified two-cell embryos, nonvitrified embryos, and a control group of in vivo derived blastocysts. We assessed developmental competence, global DNA methylation, relative expression levels of miR-29a/29b, and their target genes, Dnmt3a/3b. Vitrified embryos had a lower developmental rate as compared with nonvitrified embryos. There was no significant decrease in blastocyst cell numbers among studied groups, whereas there was a steady decline in DNA methylation after IVF and vitrification. The levels of miR-29a/29b upregulated in the experimental groups as compared with the control group. IVF and vitrification caused Dnmt3a/3b downregulations in blastocysts. The results of this study have suggested that a relationship exists between IVF and embryo vitrification with methylation interruptions in the blastocysts.
Collapse
Affiliation(s)
- Elham Movahed
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mansooreh Soleimani
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Hosseini
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azade Akbari Sene
- IVF Department, Shahid Akbar-Abadi Hospital IVF Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Salehi
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Singh S, Shyam S, Sah S, Singh MK, Palta P. Treatment of Buffalo ( Bubalus bubalis) Somatic Cell Nuclear Transfer Embryos with MicroRNA-29b Mimic Improves Their Quality, Reduces DNA Methylation, and Changes Gene Expression Without Affecting Their Developmental Competence. Cell Reprogram 2019; 21:210-219. [PMID: 31199675 DOI: 10.1089/cell.2019.0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
microRNA-29b (miR-29b) plays an important role in controlling DNA methylation in cells. We investigated its role during early embryonic development in buffalo embryos produced by somatic cell nuclear transfer (SCNT) and in vitro fertilization (IVF). miR-29b expression was highest at the 2-cell stage, decreased (p < 0.001) at the 4-cell stage, and remained low thereafter at the 8-cell, morula, and blastocyst stages, showing a similar pattern in cloned and IVF embryos. Treatment of reconstructed embryos with miR-29b mimic for 1 hour after 1 hour of electrofusion increased (p < 0.05) the total cell number and decreased (p < 0.05) the levels of apoptosis and DNA methylation compared with controls. It also increased (p < 0.05) the ratio of inner cell mass:trophectoderm cell numbers of blastocysts compared with controls to the levels observed in IVF blastocysts. However, the blastocyst rate was not affected by treatment with miR-29b mimic (29.0% ± 2.0% vs. 27.0% ± 2.0% for controls). The treatment decreased (p < 0.001) the expression of epigenetic-related genes, DNMT3A and DNMT3B, but not DNMT1, and increased (p < 0.05) that of pluripotency- (NANOG, OCT4, and SOX2) and development-related genes (FGF4 and GLUT1) in blastocysts compared with controls. Our results suggest that miR-29b mimic treatment of reconstructed embryos improves the quality, reduces the level of apoptosis and DNA methylation, and changes gene expression in SCNT blastocysts without affecting the blastocyst rate.
Collapse
Affiliation(s)
- Shikha Singh
- Animal Biotechnology Center, ICAR-National Dairy Research Institute, Karnal, India
| | - Songyukta Shyam
- Animal Biotechnology Center, ICAR-National Dairy Research Institute, Karnal, India
| | - Shrutika Sah
- Animal Biotechnology Center, ICAR-National Dairy Research Institute, Karnal, India
| | - Manoj K Singh
- Animal Biotechnology Center, ICAR-National Dairy Research Institute, Karnal, India
| | - Prabhat Palta
- Animal Biotechnology Center, ICAR-National Dairy Research Institute, Karnal, India
| |
Collapse
|
18
|
Robles V, Valcarce DG, Riesco MF. Non-coding RNA regulation in reproduction: Their potential use as biomarkers. Noncoding RNA Res 2019; 4:54-62. [PMID: 31193491 PMCID: PMC6531869 DOI: 10.1016/j.ncrna.2019.04.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 12/13/2022] Open
Abstract
Non-coding RNAs (ncRNAs) are crucial regulatory elements in most biological processes and reproduction is also controlled by them. The different types of ncRNAs, as well as the high complexity of these regulatory pathways, present a complex scenario; however, recent studies have shed some light on these questions, discovering the regulatory function of specific ncRNAs on concrete reproductive biology processes. This mini review will focus on the role of ncRNAs in spermatogenesis and oogenesis, and their potential use as biomarkers for reproductive diseases or for reproduction success.
Collapse
Affiliation(s)
- Vanesa Robles
- Spanish Institute of Oceanography (IEO) Santander, Spain
- MODCELL GROUP, Department of Molecular Biology, Universidad de León, 24071, León, Spain
- Corresponding author. Planta de Cultivos el Bocal, IEO, Barrio Corbanera, Monte, Santander, 39012, Spain.
| | | | | |
Collapse
|
19
|
Portelli SS, Robertson EN, Malecki C, Liddy KA, Hambly BD, Jeremy RW. Epigenetic influences on genetically triggered thoracic aortic aneurysm. Biophys Rev 2018; 10:1241-1256. [PMID: 30267337 PMCID: PMC6233334 DOI: 10.1007/s12551-018-0460-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 09/17/2018] [Indexed: 12/14/2022] Open
Abstract
Genetically triggered thoracic aortic aneurysms (TAAs) account for 30% of all TAAs and can result in early morbidity and mortality in affected individuals. Epigenetic factors are now recognised to influence the phenotype of many genetically triggered conditions and have become an area of interest because of the potential for therapeutic manipulation. Major epigenetic modulators include DNA methylation, histone modification and non-coding RNA. This review examines epigenetic modulators that have been significantly associated with genetically triggered TAAs and their potential utility for translation to clinical practice.
Collapse
Affiliation(s)
- Stefanie S Portelli
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia.
| | - Elizabeth N Robertson
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Cassandra Malecki
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Kiersten A Liddy
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Brett D Hambly
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Richmond W Jeremy
- Discipline of Pathology and Bosch Institute, The University of Sydney, Sydney, NSW, 2006, Australia
- Cardiology Department, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| |
Collapse
|
20
|
Reza AMMT, Choi YJ, Han SG, Song H, Park C, Hong K, Kim JH. Roles of microRNAs in mammalian reproduction: from the commitment of germ cells to peri-implantation embryos. Biol Rev Camb Philos Soc 2018; 94:415-438. [PMID: 30151880 PMCID: PMC7379200 DOI: 10.1111/brv.12459] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/25/2018] [Accepted: 07/27/2018] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRNAs) are active regulators of numerous biological and physiological processes including most of the events of mammalian reproduction. Understanding the biological functions of miRNAs in the context of mammalian reproduction will allow a better and comparative understanding of fertility and sterility in male and female mammals. Herein, we summarize recent progress in miRNA‐mediated regulation of mammalian reproduction and highlight the significance of miRNAs in different aspects of mammalian reproduction including the biogenesis of germ cells, the functionality of reproductive organs, and the development of early embryos. Furthermore, we focus on the gene expression regulatory feedback loops involving hormones and miRNA expression to increase our understanding of germ cell commitment and the functioning of reproductive organs. Finally, we discuss the influence of miRNAs on male and female reproductive failure, and provide perspectives for future studies on this topic.
Collapse
Affiliation(s)
- Abu Musa Md Talimur Reza
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Yun-Jung Choi
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Sung Gu Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, 05029, Republic of Korea
| | - Hyuk Song
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Chankyu Park
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Kwonho Hong
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Centre (SRC), Konkuk University, Seoul, 143-701, Republic of Korea
| |
Collapse
|
21
|
Khazayel S, Mokarram P, Mohammadi Z, Ramezani F, Dayong Z. Derivative of Stevioside; CPUK02; Restores ESR1 Gene Methylation in MDA-MB 231. Asian Pac J Cancer Prev 2018; 19:2117-2123. [PMID: 30139210 PMCID: PMC6171390 DOI: 10.22034/apjcp.2018.19.8.2117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background: CPUK02 (15-Oxosteviol benzyl ester) is a new ent-kaurenoid derivative of stevioside and exhibits strong anti-cancer activity. Nowadays, the pattern of epigenetic in cancer has been topic of many studies and DNA methylation targeting represents a relevant strategy for cancer treatment. Since, no study conducted to this mechanism, we attempt to evaluate whether CPUK02 induce its anti-cancer effects via alteration the level of mRNA DNMT3B, DNMT3A expression and ESR1 methylation pattern in breast cancer cells line. Methods: MCF-7 (ER +) and MDA-MB231 (ER-) cell lines were treated for 24, 48 hours with 1 µM CPUK02 and 5-AZA-CdR (DNA methyltransferase inhibitor). Quantitative expression of DNMT3B and DNMT3A genes and ESR1 promoter methylation was assessed by Real-Time PCR and MS-PCR, respectively. Results: CPUK02 restored ESR1 promoter unmethylated allele in MDA-MB 231 cells. Also treatment with CPUK02 decreased the expression of both DNMT3A and DNMT3B genes like 5-AZA. The expression of DNMT genes were diminished by half compared with control cells. Conclusions: These results showed that CPUK02 has an anticancer effect on MDA-MB 231 cells which this effect can be done through several pathways.
Collapse
Affiliation(s)
- Saeed Khazayel
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran. ,
| | | | | | | | | |
Collapse
|
22
|
Règue-Guyon M, Lanfumey L, Mongeau R. Neuroepigenetics of Neurotrophin Signaling: Neurobiology of Anxiety and Affective Disorders. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2018; 158:159-193. [DOI: 10.1016/bs.pmbts.2018.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
23
|
Cardiovascular Risk Factors and Markers. BIOMATHEMATICAL AND BIOMECHANICAL MODELING OF THE CIRCULATORY AND VENTILATORY SYSTEMS 2018. [PMCID: PMC7123062 DOI: 10.1007/978-3-319-89315-0_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cardiovascular risk is assessed for the prediction and appropriate management of patients using collections of identified risk markers obtained from clinical questionnaire information, concentrations of certain blood molecules (e.g., N-terminal proB-type natriuretic peptide fragment and soluble receptors of tumor-necrosis factor-α and interleukin-2), imaging data using various modalities, and electrocardiographic variables, in addition to traditional risk factors.
Collapse
|
24
|
Wang H, An X, Yu H, Zhang S, Tang B, Zhang X, Li Z. MiR-29b/TET1/ZEB2 signaling axis regulates metastatic properties and epithelial-mesenchymal transition in breast cancer cells. Oncotarget 2017; 8:102119-102133. [PMID: 29254230 PMCID: PMC5731940 DOI: 10.18632/oncotarget.22183] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/27/2017] [Indexed: 12/21/2022] Open
Abstract
MiR-29b has been reported to be both a suppressor and a promoter in breast cancer (BC) cells proliferation and metastasis. Significant efforts have been made to explain the seemingly contradictory effects of miR-29b on BC, but no answer has yet been clearly verified. In this study, we overexpressed and knocked down miR-29b in BC cell lines, modulated expression of its downstream target gene TET1 and downregulated a downstream target gene of TET1, ZEB2, to explore the regulatory mechanism of miR-29b in BC cell proliferation, migration and epithelial-mesenchymal transition (EMT). Our results showed lower expression of miR-29b in BC samples and cell lines. Functional assays showed that miR-29b overexpression resulted in a higher cell proliferation, greater colony formation, higher migration rate and EMT. A dual luciferase assay identified TET1 as a direct target of miR-29b. As the promoting effects of miR-29b in the proliferation and metastasis of MDA-MB-231 and MCF-7, knockdown of TET1 also led to increased proliferation, colony formation, invasion and EMT. Further, we found that TET1 bound to the promoter of ZEB2, and siTET1 enhanced ZEB2 expression. Disruption of ZEB2 expression inhibited BC cells proliferation, colony formation and invasion. Our results establish the miR-29b/TET1/ZEB2 pathway in BC cell proliferation, migration and provide a theoretical basis for further research on the molecular mechanisms and new clinical treatments for BC.
Collapse
Affiliation(s)
- Hua Wang
- The First Bethune Hospital, Jilin University, Changchun, Jilin 130021, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| | - Xinglan An
- The First Bethune Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Hao Yu
- College of Animal Science, Jilin University, Changchun, Jilin 130062, China
| | - Sheng Zhang
- The First Bethune Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Bo Tang
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| | - Xueming Zhang
- College of Veterinary Medicine, Jilin University, Changchun, Jilin 130062, China
| | - Ziyi Li
- The First Bethune Hospital, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
25
|
Gao S, Li C, Xu Y, Chen S, Zhao Y, Chen L, Jiang Y, Liu Z, Fan R, Sun L, Wang F, Zhu X, Zhang J, Zhou X. Differential expression of microRNAs in TM3 Leydig cells of mice treated with brain-derived neurotrophic factor. Cell Biochem Funct 2017; 35:364-371. [DOI: 10.1002/cbf.3283] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/14/2017] [Accepted: 07/07/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Shan Gao
- College of Animal Sciences; Jilin University; Jilin China
| | - Chunjin Li
- College of Animal Sciences; Jilin University; Jilin China
| | - Ying Xu
- Reproductive Medical Center; The Second Hospital of Jilin University; Changchun China
| | - Shuxiong Chen
- College of Animal Sciences; Jilin University; Jilin China
| | - Yun Zhao
- College of Animal Sciences; Jilin University; Jilin China
| | - Lu Chen
- College of Animal Sciences; Jilin University; Jilin China
| | - Yanwen Jiang
- College of Animal Sciences; Jilin University; Jilin China
| | - Zhuo Liu
- College of Animal Sciences; Jilin University; Jilin China
| | - Rong Fan
- College of Animal Sciences; Jilin University; Jilin China
| | - Liting Sun
- College of Animal Sciences; Jilin University; Jilin China
| | - Fengge Wang
- College of Animal Sciences; Jilin University; Jilin China
| | - Xiaoling Zhu
- College of Animal Sciences; Jilin University; Jilin China
| | - Jing Zhang
- College of Animal Sciences; Jilin University; Jilin China
| | - Xu Zhou
- College of Animal Sciences; Jilin University; Jilin China
| |
Collapse
|
26
|
Lu H, Lei X, Liu J, Klaassen C. Regulation of hepatic microRNA expression by hepatocyte nuclear factor 4 alpha. World J Hepatol 2017; 9:191-208. [PMID: 28217257 PMCID: PMC5295159 DOI: 10.4254/wjh.v9.i4.191] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 10/02/2016] [Accepted: 12/02/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To uncover the role of hepatocyte nuclear factor 4 alpha (HNF4α) in regulating hepatic expression of microRNAs.
METHODS Microarray and real-time PCR were used to determine hepatic expression of microRNAs in young-adult mice lacking Hnf4α expression in liver (Hnf4α-LivKO). Integrative genomics viewer software was used to analyze the public chromatin immunoprecipitation-sequencing datasets for DNA-binding of HNF4α, RNA polymerase-II, and histone modifications to loci of microRNAs in mouse liver and human hepatoma cells. Dual-luciferase reporter assay was conducted to determine effects of HNF4α on the promoters of mouse and human microRNAs as well as effects of microRNAs on the untranslated regions (3’UTR) of two genes in human hepatoma cells.
RESULTS Microarray data indicated that most microRNAs remained unaltered by Hnf4α deficiency in Hnf4α-LivKO mice. However, certain liver-predominant microRNAs were down-regulated similarly in young-adult male and female Hnf4α-LivKO mice. The down-regulation of miR-101, miR-192, miR-193a, miR-194, miR-215, miR-802, and miR-122 as well as induction of miR-34 and miR-29 in male Hnf4α-LivKO mice were confirmed by real-time PCR. Analysis of public chromatin immunoprecipitation-sequencing data indicates that HNF4α directly binds to the promoters of miR-101, miR-122, miR-194-2/miR-192 and miR-193, which is associated with histone marks of active transcription. Luciferase reporter assay showed that HNF4α markedly activated the promoters of mouse and human miR-101b/miR-101-2 and the miR-194/miR-192 cluster. Additionally, miR-192 and miR-194 significantly decreased activities of luciferase reporters for the 3’UTR of histone H3F3 and chromodomain helicase DNA binding protein 1 (CHD1), respectively, suggesting that miR-192 and miR-194 might be important in chromosome remodeling through directly targeting H3F3 and CHD1.
CONCLUSION HNF4α is essential for hepatic basal expression of a group of liver-enriched microRNAs, including miR-101, miR-192, miR-193a, miR-194 and miR-802, through which HNF4α may play a major role in the post-transcriptional regulation of gene expression and maintenance of the epigenome in liver.
Collapse
|
27
|
Wang JY, Xiao L, Wang JY. Posttranscriptional regulation of intestinal epithelial integrity by noncoding RNAs. WILEY INTERDISCIPLINARY REVIEWS-RNA 2016; 8. [PMID: 27704722 DOI: 10.1002/wrna.1399] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 08/26/2016] [Accepted: 08/31/2016] [Indexed: 12/24/2022]
Abstract
Maintenance of the gut epithelial integrity under stressful environments requires epithelial cells to rapidly elicit changes in gene expression patterns to regulate their survival, adapt to stress, and keep epithelial homeostasis. Disruption of the intestinal epithelial integrity occurs commonly in patients with various critical illnesses, leading to the translocation of luminal toxic substances and bacteria to the blood stream. Recently, noncoding RNAs (ncRNAs) have emerged as a novel class of master regulators of gene expression and are fundamentally involved in many aspects of gut mucosal regeneration, protection, and epithelial barrier function. Here, we highlight the roles of several intestinal epithelial tissue-specific microRNAs, including miR-222, miR-29b, miR-503, and miR-195, and long ncRNAs such as H19 and SPRY4-IT1 in the regulation of cell proliferation, apoptosis, migration, and cell-to-cell interactions and also further analyze the mechanisms through which ncRNAs and their interactions with RNA-binding proteins modulate the stability and translation of target mRNAs. WIREs RNA 2017, 8:e1399. doi: 10.1002/wrna.1399 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Jun-Yao Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA
| | - Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.,Baltimore Veterans Affairs Medical Center, Baltimore, MD, USA.,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
28
|
Miao N, Wang X, Hou Y, Feng Y, Gong Y. Identification of male-biased microRNA-107 as a direct regulator for nuclear receptor subfamily 5 group A member 1 based on sexually dimorphic microRNA expression profiling from chicken embryonic gonads. Mol Cell Endocrinol 2016; 429:29-40. [PMID: 27036932 DOI: 10.1016/j.mce.2016.03.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/08/2016] [Accepted: 03/27/2016] [Indexed: 12/15/2022]
Abstract
Several studies indicate that sexual dimorphic microRNAs (miRNAs) in chicken gonads are likely to have important roles in sexual development, but a more global understanding of the roles of miRNAs in sexual differentiation is still needed. To this end, we performed miRNA expression profiling in chicken gonads at embryonic day 5.5 (E5.5). Among the sex-biased miRNAs validated by qRT-PCR, twelve male-biased and six female-biased miRNAs were consistent with the sequencing results. Bioinformatics analysis revealed that some sex-biased miRNAs were potentially involved in gonadal development. Further functional analysis found that over-expression of miR-107 directly inhibited nuclear receptor subfamily 5 group A member 1 (NR5a1), and its downstream cytochrome P450 family 19 subfamily A, polypeptide 1 (CYP19A1). However, anti-Mullerian hormone (AMH) was not directly or indirectly regulated by miR-107. Overall results indicate that miR-107 may specifically mediate avian ovary-development by post-transcriptional regulation of NR5a1 and CYP19A1 in estrogen signaling pathways.
Collapse
Affiliation(s)
- Nan Miao
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xin Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yue Hou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yanping Feng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| | - Yanzhang Gong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
29
|
Masoud AM, Bihaqi SW, Machan JT, Zawia NH, Renehan WE. Early-Life Exposure to Lead (Pb) Alters the Expression of microRNA that Target Proteins Associated with Alzheimer’s Disease. J Alzheimers Dis 2016; 51:1257-64. [DOI: 10.3233/jad-151018] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Anwar M. Masoud
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
- Biochemical Technology Program, Faculty of Applied Science, Thamar University, Thamar, Yemen
| | - Syed W. Bihaqi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail, Kingdom of Saudi Arabia
| | - Jason T. Machan
- Lifespan Biostatistics Core and Departments of Orthopaedics and Surgery, Warren Alpert Medical School, Brown University, Providence RI, USA
| | - Nasser H. Zawia
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI, USA
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| | - William E. Renehan
- Interdisciplinary Neuroscience Program, University of Rhode Island, Kingston, RI, USA
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, USA
| |
Collapse
|
30
|
Horsburgh S, Todryk S, Toms C, Moran CN, Ansley L. Exercise-conditioned plasma attenuates nuclear concentrations of DNA methyltransferase 3B in human peripheral blood mononuclear cells. Physiol Rep 2015; 3:3/12/e12621. [PMID: 26660547 PMCID: PMC4760429 DOI: 10.14814/phy2.12621] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
DNA methylation is modifiable by acute and chronic exercise. DNA methyltransferases (DNMT) catalyze this process; however, there is a lack of literature concerning the specific mechanisms by which exercise‐induced modifications occur. Interleukin 6 (IL‐6) stimulation of various cell lines has been shown to augment DNMT expression and nuclear translocation, which suggests a possible pathway by which exercise is able to elicit changes in epigenetic enzymes. The present study sought to elucidate the response of the de novo methyltransferases DNMT3A and DNMT3B to circulatory factors found in plasma isolated from whole blood before and after 120‐min of treadmill running at an intensity of 60% of individual velocity at V˙O2max (vV˙O2max) interspersed with 30‐sec sprints at 90% of vV˙O2max every 10‐min. Peripheral blood mononuclear cells (PBMCs) isolated from a resting participant were incubated with plasma isolated from exercising participants (n = 10) or recombinant IL‐6 (rIL‐6), followed by nuclear protein extraction and quantification of DNMT3A and DNMT3B concentrations. Nuclear concentrations of DNMT3B significantly decreased following the experimental protocol (P = 0.03), with no change observed in DNMT3A (P = 0.514).Various concentrations of rIL‐6 caused an elevation in both DNMT3A and DNMT3B nuclear concentration compared with the blank control. The conflicting results between exercising and rIL‐6 conditions suggests that IL‐6 does regulate DNMT nuclear transport, however, other plasma mediators may also exert significant influence on the nuclear concentrations of these enzymes.
Collapse
Affiliation(s)
- Steven Horsburgh
- Faculty of Health and Life Sciences, Northumbria University, Newcastle Upon Tyne, England
| | - Stephen Todryk
- Faculty of Health and Life Sciences, Northumbria University, Newcastle Upon Tyne, England
| | - Christopher Toms
- Research Department, British College of Osteopathic Medicine, London, England
| | - Colin N Moran
- Health and Exercise Sciences Research Group, University of Stirling, Stirling, Scotland
| | - Les Ansley
- Faculty of Health and Life Sciences, Northumbria University, Newcastle Upon Tyne, England
| |
Collapse
|
31
|
MicroRNAs: From Female Fertility, Germ Cells, and Stem Cells to Cancer in Humans. Stem Cells Int 2015; 2016:3984937. [PMID: 26664407 PMCID: PMC4655303 DOI: 10.1155/2016/3984937] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/19/2015] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs are a family of naturally occurring small noncoding RNA molecules that play an important regulatory role in gene expression. They are suggested to regulate a large proportion of protein encoding genes by mediating the translational suppression and posttranscriptional control of gene expression. Recent findings show that microRNAs are emerging as important regulators of cellular differentiation and dedifferentiation, and are deeply involved in developmental processes including human preimplantation development. They keep a balance between pluripotency and differentiation in the embryo and embryonic stem cells. Moreover, it became evident that dysregulation of microRNA expression may play a fundamental role in progression and dissemination of different cancers including ovarian cancer. The interest is still increased by the discovery of exosomes, that is, cell-derived vesicles, which can carry different proteins but also microRNAs between different cells and are involved in cell-to-cell communication. MicroRNAs, together with exosomes, have a great potential to be used for prognosis, therapy, and biomarkers of different diseases including infertility. The aim of this review paper is to summarize the existent knowledge on microRNAs related to female fertility and cancer: from primordial germ cells and ovarian function, germinal stem cells, oocytes, and embryos to embryonic stem cells.
Collapse
|
32
|
The Roles of miR-26, miR-29, and miR-203 in the Silencing of the Epigenetic Machinery during Melanocyte Transformation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:634749. [PMID: 26618174 PMCID: PMC4649077 DOI: 10.1155/2015/634749] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 10/12/2015] [Indexed: 12/31/2022]
Abstract
The epigenetic marks located throughout the genome exhibit great variation between normal and transformed cancer cells. While normal cells contain hypomethylated CpG islands near gene promoters and hypermethylated repetitive DNA, the opposite pattern is observed in cancer cells. Recently, it has been reported that alteration in the microenvironment of melanocyte cells, such as substrate adhesion blockade, results in the selection of anoikis-resistant cells, which have tumorigenic characteristics. Melanoma cells obtained through this model show an altered epigenetic pattern, which represents one of the first events during the melanocytes malignant transformation. Because microRNAs are involved in controlling components of the epigenetic machinery, the aim of this work was to evaluate the potential association between the expression of miR-203, miR-26, and miR-29 family members and the genes Dnmt3a, Dnmt3b, Mecp2, and Ezh2 during cells transformation. Our results show that microRNAs and their validated or predicted targets are inversely expressed, indicating that these molecules are involved in epigenetic reprogramming. We also show that miR-203 downregulates Dnmt3b in mouse melanocyte cells. In addition, treatment with 5-aza-CdR promotes the expression of miR-26 and miR-29 in a nonmetastatic melanoma cell line. Considering the occurrence of CpG islands near the miR-26 and miR-29 promoters, these data suggest that they might be epigenetically regulated in cancer.
Collapse
|
33
|
Bhin J, Jeong HS, Kim JS, Shin JO, Hong KS, Jung HS, Kim C, Hwang D, Kim KS. PGC-Enriched miRNAs Control Germ Cell Development. Mol Cells 2015; 38:895-903. [PMID: 26442865 PMCID: PMC4625071 DOI: 10.14348/molcells.2015.0146] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 06/25/2015] [Accepted: 07/08/2015] [Indexed: 01/21/2023] Open
Abstract
Non-coding microRNAs (miRNAs) regulate the translation of target messenger RNAs (mRNAs) involved in the growth and development of a variety of cells, including primordial germ cells (PGCs) which play an essential role in germ cell development. However, the target mRNAs and the regulatory networks influenced by miRNAs in PGCs remain unclear. Here, we demonstrate a novel miRNAs control PGC development through targeting mRNAs involved in various cellular pathways. We reveal the PGC-enriched expression patterns of nine miRNAs, including miR-10b, -18a, -93, -106b, -126-3p, -127, -181a, -181b, and -301, using miRNA expression analysis along with mRNA microarray analysis in PGCs, embryonic gonads, and postnatal testes. These miRNAs are highly expressed in PGCs, as demonstrated by Northern blotting, miRNA in situ hybridization assay, and miRNA qPCR analysis. This integrative study utilizing mRNA microarray analysis and miRNA target prediction demonstrates the regulatory networks through which these miRNAs regulate their potential target genes during PGC development. The elucidated networks of miRNAs disclose a coordinated molecular mechanism by which these miRNAs regulate distinct cellular pathways in PGCs that determine germ cell development.
Collapse
Affiliation(s)
- Jinhyuk Bhin
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 790-784,
Korea
| | - Hoe-Su Jeong
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, Seoul 133-791,
Korea
| | - Jong Soo Kim
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, Seoul 133-791,
Korea
| | - Jeong Oh Shin
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University, College of Dentistry, Seoul 120-752,
Korea
| | - Ki Sung Hong
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, Seoul 133-791,
Korea
| | - Han-Sung Jung
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Oral Science Research Center, BK21 PLUS Project, Yonsei University, College of Dentistry, Seoul 120-752,
Korea
| | - Changhoon Kim
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, Seoul 133-791,
Korea
| | - Daehee Hwang
- Department of Chemical Engineering, Pohang University of Science and Technology, Pohang 790-784,
Korea
- Department of New Biology and Center for Plant Aging Research, Institute for Basic Science, Daegu Gyeongbuk Institute of Science & Technology, Daegu 711-873,
Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Department of Biomedical Science, Hanyang University, Seoul 133-791,
Korea
| |
Collapse
|
34
|
Soubry A. Epigenetic inheritance and evolution: A paternal perspective on dietary influences. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2015; 118:79-85. [DOI: 10.1016/j.pbiomolbio.2015.02.008] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 01/18/2015] [Accepted: 02/23/2015] [Indexed: 12/23/2022]
|
35
|
Tu J, Ng SH, Luk ACS, Liao J, Jiang X, Feng B, Lun Mak KK, Rennert OM, Chan WY, Lee TL. MicroRNA-29b/Tet1 regulatory axis epigenetically modulates mesendoderm differentiation in mouse embryonic stem cells. Nucleic Acids Res 2015; 43:7805-22. [PMID: 26130713 PMCID: PMC4652748 DOI: 10.1093/nar/gkv653] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 06/13/2015] [Indexed: 12/03/2022] Open
Abstract
Ten eleven translocation (Tet) family-mediated DNA oxidation on 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) represents a novel epigenetic modification that regulates dynamic gene expression during embryonic stem cells (ESCs) differentiation. Through the role of Tet on 5hmC regulation in stem cell development is relatively defined, how the Tet family is regulated and impacts on ESCs lineage development remains elusive. In this study, we show non-coding RNA regulation on Tet family may contribute to epigenetic regulation during ESCs differentiation, which is suggested by microRNA-29b (miR-29b) binding sites on the Tet1 3′ untranslated region (3′ UTR). We demonstrate miR-29b increases sharply after embyoid body (EB) formation, which causes Tet1 repression and reduction of cellular 5hmC level during ESCs differentiation. Importantly, we show this miR-29b/Tet1 regulatory axis promotes the mesendoderm lineage formation both in vitro and in vivo by inducing the Nodal signaling pathway and repressing the key target of the active demethylation pathway, Tdg. Taken together, our findings underscore the contribution of small non-coding RNA mediated regulation on DNA demethylation dynamics and the differential expressions of key mesendoderm regulators during ESCs lineage specification. MiR-29b could potentially be applied to enrich production of mesoderm and endoderm derivatives and be further differentiated into desired organ-specific cells.
Collapse
Affiliation(s)
- Jiajie Tu
- Reproduction, Development and Endocrinology Program, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Shuk Han Ng
- Reproduction, Development and Endocrinology Program, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Alfred Chun Shui Luk
- Reproduction, Development and Endocrinology Program, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Jinyue Liao
- Reproduction, Development and Endocrinology Program, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xiaohua Jiang
- Stem Cell and Regeneration Program, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Bo Feng
- Stem Cell and Regeneration Program, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Kingston King Lun Mak
- Stem Cell and Regeneration Program, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China
| | - Owen M Rennert
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Wai-Yee Chan
- Reproduction, Development and Endocrinology Program, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Tin-Lap Lee
- Reproduction, Development and Endocrinology Program, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, China Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
36
|
McGinnis LK, Luense LJ, Christenson LK. MicroRNA in Ovarian Biology and Disease. Cold Spring Harb Perspect Med 2015; 5:a022962. [PMID: 25986593 DOI: 10.1101/cshperspect.a022962] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
MicroRNAs (miRNAs) are posttranscriptional gene regulatory molecules that show regulated expression within ovarian tissue. Most research investigating miRNAs in the ovary has relied exclusively on in vitro analyses. In this review, we highlight those few studies in which investigators have illustrated an in vivo effect of miRNAs on ovarian function. We also provide a synopsis of how these small noncoding RNAs can impact ovarian disease. miRNAs have great potential as novel diagnostic biomarkers for the detection of ovarian disease and in the assisted reproductive technologies (ART) for selection of healthy viable oocytes and embryos.
Collapse
Affiliation(s)
- Lynda K McGinnis
- Department Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Lacey J Luense
- Epigenetics Program, Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Lane K Christenson
- Department Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas 66160
| |
Collapse
|
37
|
Revay T, Quach AT, Maignel L, Sullivan B, King WA. Copy number variations in high and low fertility breeding boars. BMC Genomics 2015; 16:280. [PMID: 25888238 PMCID: PMC4404230 DOI: 10.1186/s12864-015-1473-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 03/20/2015] [Indexed: 01/17/2023] Open
Abstract
Background In this study we applied the extreme groups/selective genotyping approach for identifying copy number variations in high and low fertility breeding boars. The fertility indicator was the calculated Direct Boar Effect on litter size (DBE) that was obtained as a by-product of the national genetic evaluation for litter size (BLUP). The two groups of animals had DBE values at the upper (high fertility) and lower (low fertility) end of the distribution from a population of more than 38,000 boars. Animals from these two diverse phenotypes were genotyped with the Porcine SNP60K chip and compared by several approaches in order to prove the feasibility of our CNV analysis and to identify putative markers of fertility. Results We have identified 35 CNVRs covering 36.5 Mb or ~1.3% of the porcine genome. Among these 35 CNVRs, 14 were specific to the high fertility group, while 19 CNVRs were specific to the low fertility group which overlap with 137 QTLs of various reproductive traits. The identified 35 CNVRs encompassed 50 genes, among them 40 were specific to the low fertility group, seven to the high fertility group, while three were found in regions that were present in both groups but with opposite gain/loss status. A functional analysis of several databases revealed that the genes found in CNVRs from the low fertility group have been significantly enriched in members of the innate immune system, Toll-like receptor and RIG-I-like receptor signaling and fatty acid oxidation pathways. Conclusions We have demonstrated that our analysis pipeline could identify putative CNV markers of fertility, especially in case of low fertility boars. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1473-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tamas Revay
- University of Guelph, Ontario Veterinary College, Department of Biomedical Sciences, 50 Stone Rd E, Guelph, ON, N1G 2W1, Canada.
| | - Anh T Quach
- University of Guelph, Ontario Veterinary College, Department of Biomedical Sciences, 50 Stone Rd E, Guelph, ON, N1G 2W1, Canada.
| | - Laurence Maignel
- Canadian Centre for Swine Improvement Inc. (CCSI), Central Experimental Farm, Building #75, 960 Carling Avenue, Ottawa, ON, K1A 0C6, Canada.
| | - Brian Sullivan
- Canadian Centre for Swine Improvement Inc. (CCSI), Central Experimental Farm, Building #75, 960 Carling Avenue, Ottawa, ON, K1A 0C6, Canada.
| | - W Allan King
- University of Guelph, Ontario Veterinary College, Department of Biomedical Sciences, 50 Stone Rd E, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
38
|
Papadopoulou AS, Serneels L, Achsel T, Mandemakers W, Callaerts-Vegh Z, Dooley J, Lau P, Ayoubi T, Radaelli E, Spinazzi M, Neumann M, Hébert SS, Silahtaroglu A, Liston A, D'Hooge R, Glatzel M, De Strooper B. Deficiency of the miR-29a/b-1 cluster leads to ataxic features and cerebellar alterations in mice. Neurobiol Dis 2015; 73:275-88. [DOI: 10.1016/j.nbd.2014.10.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 09/05/2014] [Accepted: 10/01/2014] [Indexed: 12/20/2022] Open
|
39
|
Molecular characterization of exosomes and their microRNA cargo in human follicular fluid: bioinformatic analysis reveals that exosomal microRNAs control pathways involved in follicular maturation. Fertil Steril 2014; 102:1751-61.e1. [DOI: 10.1016/j.fertnstert.2014.08.005] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 08/04/2014] [Accepted: 08/05/2014] [Indexed: 12/15/2022]
|
40
|
Domingo-Gonzalez R, Wilke CA, Huang SK, Laouar Y, Brown JP, Freeman CM, Curtis JL, Yanik GA, Moore BB. Transforming growth factor-β induces microRNA-29b to promote murine alveolar macrophage dysfunction after bone marrow transplantation. Am J Physiol Lung Cell Mol Physiol 2014; 308:L86-95. [PMID: 25361568 DOI: 10.1152/ajplung.00283.2014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is complicated by pulmonary infections that manifest posttransplantation. Despite engraftment, susceptibility to infections persists long after reconstitution. Previous work using a murine bone marrow transplant (BMT) model implicated increased cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2) in promoting impaired alveolar macrophage (AM) responses. However, mechanisms driving COX-2 overexpression remained elusive. Previously, transforming growth factor-β (TGF-β) signaling after BMT was shown to promote hypomethylation of the COX-2 gene. Here, we provide mechanistic insight into how this occurs and show that TGF-β induces microRNA (miR)-29b while decreasing DNA methyltransferases (DNMT)1, DNMT3a, and DNMT3b in AMs after BMT. De novo DNMT3a and DNMT3b were decreased upon transient transfection of miR-29b, resulting in decreased methylation of the COX-2 promoter and induction of COX-2. As a consequence, miR-29b-driven upregulation of COX-2 promoted AM dysfunction, and transfection of BMT AMs with a miR-29b inhibitor rescued the bacterial-killing defect. MiR-29b-mediated defects in BMT AMs were dependent on increased levels of PGE2, as miR-29b-transfected AMs treated with a novel E prostanoid receptor 2 antagonist abrogated the impaired bacterial killing. We also demonstrate that patients that have undergone HSCT exhibit increased miR-29b; thus these studies highlight miR-29b in driving defective AM responses and identify this miRNA as a potential therapeutic target.
Collapse
Affiliation(s)
| | - Carol A Wilke
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Steven K Huang
- Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan
| | - Yasmina Laouar
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Jeanette P Brown
- Research Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| | | | - Jeffrey L Curtis
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan; Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan; Medical Service, VA Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Gregory A Yanik
- Department of Pediatrics, Division of Hematology-Oncology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Bethany B Moore
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, Michigan; Pulmonary and Critical Care Medicine Division, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan;
| |
Collapse
|
41
|
Pourrajab F, Babaei Zarch M, BaghiYazdi M, Hekmatimoghaddam S, Zare-Khormizi MR. MicroRNA-based system in stem cell reprogramming; differentiation/dedifferentiation. Int J Biochem Cell Biol 2014; 55:318-28. [PMID: 25150833 DOI: 10.1016/j.biocel.2014.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 08/09/2014] [Accepted: 08/11/2014] [Indexed: 12/26/2022]
Abstract
Stem cells (SCs) have self-renew ability and give rise to committed progenitors of a single or multiple lineages. Elucidating the genetic circuits that govern SCs to self-renew and to differentiate is essential to understand the roles of SCs and promise of these cells in regenerative medicine. MicroRNAs are widespread agents playing critical roles in regulatory networks of transcriptional expression and have been strongly linked with SCs for simultaneous maintenance of pluripotency properties such as self-renewal. This review aims to provide state-of-the-art presentations on microRNA-dependent molecular mechanisms contribute to the maintenance of pluripotency. Understanding the microRNA signature interactions, in conjunction with cell signaling, is critical for development of improved strategies to reprogram differentiated cells or direct differentiation of pluripotent cells.
Collapse
Affiliation(s)
- Fatemeh Pourrajab
- School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran; Department of Clinical Biochemistry and Molecular Biology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | | | - Mohammad BaghiYazdi
- School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | | | | |
Collapse
|
42
|
Xiao L, Wang JY. RNA-binding proteins and microRNAs in gastrointestinal epithelial homeostasis and diseases. Curr Opin Pharmacol 2014; 19:46-53. [PMID: 25063919 DOI: 10.1016/j.coph.2014.07.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/08/2014] [Accepted: 07/09/2014] [Indexed: 10/25/2022]
Abstract
The epithelium of gastrointestinal (GI) mucosa is a rapidly self-renewing tissue in the body, and its homeostasis is preserved through strict regulation of cell proliferation and apoptosis. Epithelial cells originate from a small number of pluripotent stem cells, which divide to either renew themselves or become committed crypt cells. RNA-binding proteins (RBPs) and microRNAs (miRNAs) regulate gene expression at the posttranscriptional level and are recently shown to modulate GI mucosal growth and repair after injury. Here we highlight the roles of RBPs HuR, CUG-binding protein 1, AU-binding factor 1, and several GI epithelial-specific miRNAs in gut mucosal homeostasis and diseases and also further analyze the mechanisms through which RBPs and miRNAs modulate the stability and translation of target mRNAs.
Collapse
Affiliation(s)
- Lan Xiao
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, USA; Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
| | - Jian-Ying Wang
- Cell Biology Group, Department of Surgery, University of Maryland School of Medicine, USA; Department of Pathology, University of Maryland School of Medicine, USA; Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA.
| |
Collapse
|
43
|
Abstract
MicroRNAs (miRNAs) are transcriptional and posttranscriptional regulators involved in nearly all known biological processes in distant eukaryotic clades. Their discovery and functional characterization have broadened our understanding of biological regulatory mechanisms in animals and plants. They show both evolutionary conserved and unique features across Metazoa. Here, we present the current status of the knowledge about the role of miRNA in development, growth, and physiology of teleost fishes, in comparison to other vertebrates. Infraclass Teleostei is the most abundant group among vertebrate lineage. Fish are an important component of aquatic ecosystems and human life, being the prolific source of animal proteins worldwide and a vertebrate model for biomedical research. We review miRNA biogenesis, regulation, modifications, and mechanisms of action. Specific sections are devoted to the role of miRNA in teleost development, organogenesis, tissue differentiation, growth, regeneration, reproduction, endocrine system, and responses to environmental stimuli. Each section discusses gaps in the current knowledge and pinpoints the future directions of research on miRNA in teleosts.
Collapse
Affiliation(s)
| | - Igor Babiak
- Faculty of Aquaculture and Biosciences, University of Nordland, Bodø, Norway
| |
Collapse
|
44
|
Kalani A, Kamat PK, Familtseva A, Chaturvedi P, Muradashvili N, Narayanan N, Tyagi SC, Tyagi N. Role of microRNA29b in blood-brain barrier dysfunction during hyperhomocysteinemia: an epigenetic mechanism. J Cereb Blood Flow Metab 2014; 34:1212-22. [PMID: 24802332 PMCID: PMC4083388 DOI: 10.1038/jcbfm.2014.74] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 03/04/2014] [Accepted: 03/25/2014] [Indexed: 12/22/2022]
Abstract
Although blood-brain barrier (BBB) integrity is maintained by the cross-talk of endothelial cells, junction proteins, and neurogliovascular network, the epigenetic mechanisms behind BBB permeability are largely unknown. We are reporting for the first time miR29b-mediated regulation of BBB, which is a novel mechanism underlying BBB integrity. We hypothesize that miR29b regulates BBB dysfunction by regulating DNMT3b, which consequently regulates the levels of metalloproteinases, that can eat up the membrane and junction proteins leading to leaky vasculature. In addition, 5'-azacytidine (5'-aza) was used to test its efficacy on BBB permeability. Blood-brain barrier disruption model was created by using homocysteine, and in the models miR29b was identified to be most affected, by using microRNA RT(2)-qPCR array. MiR29b mimics and inhibitors also confirmed that miR29b regulates the levels DNMT3b and MMP9. In hyperhomocysteinemic cystathionine-β-synthase deficient (CBS(+/-)) mice with high brain vessel permeability, miR29b levels were also high as compared with wild-type (WT) mice. Interestingly, 5'-aza improved BBB permeability by decreasing the expression of miR29b. In conclusion, our data suggested miR29b-mediated regulation of BBB dysfunction through DNMT3b and MMP9. It also potentiates the use of microRNAs as candidates for future epigenetic therapies in the improvement of BBB integrity.
Collapse
Affiliation(s)
- Anuradha Kalani
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Pradip K Kamat
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Anastasia Familtseva
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Pankaj Chaturvedi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Nino Muradashvili
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Nithya Narayanan
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Suresh C Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Neetu Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
45
|
Kollinerova S, Vassanelli S, Modriansky M. The role of miR-29 family members in malignant hematopoiesis. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2014; 158:489-501. [PMID: 24993745 DOI: 10.5507/bp.2014.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 05/27/2014] [Indexed: 12/31/2022] Open
Abstract
AIMS MicroRNAs of the miR-29 family members were one of the first microRNAs identified as possible therapeutic agents in malignant hematopoiesis. The aim of our review is to summarize the current state of knowledge on miR-29 family members. METHODS We performed literature searches involving miR-29 family members and their relationship to individual hematological malignancies, namely acute myeloid leukemia (AML), chronic lymphoblastic leukemia (CLL) and chronic myeloid leukemia (CML). We also searched for subgroups of hematological malignancies, e.g. multiple myeloma, that are regarded as members of the acute or chronic types of leukemias. RESULTS A number of genes appear to be regulated by miR-29 family members in various physiological and pathological situations. In our view regulation of Tcl-1, Mcl-1 and DNA methyltransferases is relevant in case of hematological malignancies, hence these are the focus of this review. miR-29 family members also function during normal T-cell and B-cell development. CONCLUSION MiR-29 family members appear to govern some general features in commonly heterogenous hematological malignancies and therefore form a potential target for treatment.
Collapse
Affiliation(s)
- Sona Kollinerova
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic
| | | | | |
Collapse
|
46
|
Real FM, Sekido R, Lupiáñez DG, Lovell-Badge R, Jiménez R, Burgos M. A microRNA (mmu-miR-124) prevents Sox9 expression in developing mouse ovarian cells. Biol Reprod 2013; 89:78. [PMID: 23946534 DOI: 10.1095/biolreprod.113.110957] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
In mammals, sex differentiation depends on gonad development, which is controlled by two groups of sex-determining genes that promote one gonadal sex and antagonize the opposite one. SOX9 plays a key role during testis development in all studied vertebrates, whereas it is kept inactive in the XX gonad at the critical time of sex determination, otherwise, ovary-to-testis gonadal sex reversal occurs. However, molecular mechanisms underlying repression of Sox9 at the beginning of ovarian development, as well as other important aspects of gonad organogenesis, remain largely unknown. Because there is indirect evidence that micro-RNAs (miRNA) are necessary for testicular function, the possible involvement of miRNAs in mammalian sex determination deserved further research. Using microarray technology, we have identified 22 miRNAs showing sex-specific expression in the developing gonads during the critical period of sex determination. Bioinformatics analyses led to the identification of miR-124 as the candidate gene for ovarian development. We knocked down or overexpressed miR-124 in primary gonadal cell cultures and observed that miR-124 is sufficient to induce the repression of both SOX9 translation and transcription in ovarian cells. Our results provide the first evidence of the involvement of a miRNA in the regulation of the gene controlling gonad development and sex determination. The miRNA microarray data reported here will help promote further research in this field, to unravel the role of other miRNAs in the genetic control of mammalian sex determination.
Collapse
Affiliation(s)
- Francisca M Real
- Departamento de Genética e Instituto de Biotecnología, Universidad de Granada, Centro de Investigación Biomédica, Granada, Spain
| | | | | | | | | | | |
Collapse
|
47
|
Starlard-Davenport A, Kutanzi K, Tryndyak V, Word B, Lyn-Cook B. Restoration of the methylation status of hypermethylated gene promoters by microRNA-29b in human breast cancer: A novel epigenetic therapeutic approach. J Carcinog 2013; 12:15. [PMID: 23961262 PMCID: PMC3746452 DOI: 10.4103/1477-3163.115720] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 12/13/2012] [Indexed: 01/08/2023] Open
Abstract
It is well established that transcriptional silencing of critical tumor-suppressor genes by DNA methylation is a fundamental component in the initiation of breast cancer. However, the involvement of microRNAs (miRNAs) in restoring abnormal DNA methylation patterns in breast cancer is not well understood. Therefore, we investigated whether miRNA-29b, due to its complimentarity to the 3’- untranslated region of DNA methyltransferase 3A (DNMT3A) and DNMT3B, could restore normal DNA methylation patterns in human breast cancers and breast cancer cell lines. We demonstrated that transfection of pre-miRNA-29b into less aggressive MCF-7 cells, but not MDA-MB-231 mesenchymal cells, inhibited cell proliferation, decreased DNMT3A and DNMT3B messenger RNA (mRNA), and decreased promoter methylation status of ADAM23 , CCNA1, CCND2, CDH1, CDKN1C, CDKN2A, HIC1, RASSF1, SLIT2, TNFRSF10D, and TP73 tumor-suppressor genes. Using methylation polymerase chain reaction (PCR) arrays and real-time PCR, we also demonstrated that the methylation status of several critical tumor-suppressor genes increased as stage of breast disease increased, while miRNA-29b mRNA levels were significantly decreased in breast cancers versus normal breast. This increase in methylation status was accompanied by an increase in DNMT1 and DNMT3B mRNA in advanced stage of human breast cancers and in MCF-7, MDA-MB-361, HCC70, Hs-578T, and MDA-MB-231 breast cancer cells as compared to normal breast specimens and MCF-10-2A, a non-tumorigenic breast cell line, respectively. Our findings highlight the potential for a new epigenetic approach in improving breast cancer therapy by targeting DNMT3A and DNMT3B through miRNA-29b in non-invasive epithelial breast cancer cells.
Collapse
|
48
|
Guenatri M, Duffié R, Iranzo J, Fauque P, Bourc'his D. Plasticity in Dnmt3L-dependent and -independent modes of de novo methylation in the developing mouse embryo. Development 2013; 140:562-72. [DOI: 10.1242/dev.089268] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A stimulatory DNA methyltransferase co-factor, Dnmt3L, has evolved in mammals to assist the process of de novo methylation, as genetically demonstrated in the germline. The function of Dnmt3L in the early embryo remains unresolved. By combining developmental and genetic approaches, we find that mouse embryos begin development with a maternal store of Dnmt3L, which is rapidly degraded and does not participate in embryonic de novo methylation. A zygotic-specific promoter of Dnmt3l is activated following gametic methylation loss and the potential recruitment of pluripotency factors just before implantation. Importantly, we find that zygotic Dnmt3L deficiency slows down the rate of de novo methylation in the embryo by affecting methylation density at some, but not all, genomic sequences. Dnmt3L is not strictly required, however, as methylation patterns are eventually established in its absence, in the context of increased Dnmt3A protein availability. This study proves that the postimplantation embryo is more plastic than the germline in terms of DNA methylation mechanistic choices and, importantly, that de novo methylation can be achieved in vivo without Dnmt3L.
Collapse
Affiliation(s)
- Mounia Guenatri
- INSERM U934/UMR3215, Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - Rachel Duffié
- INSERM U934/UMR3215, Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - Julian Iranzo
- INSERM U934/UMR3215, Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - Patricia Fauque
- INSERM U934/UMR3215, Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | - Déborah Bourc'his
- INSERM U934/UMR3215, Institut Curie, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| |
Collapse
|
49
|
Guo X, Liu Q, Wang G, Zhu S, Gao L, Hong W, Chen Y, Wu M, Liu H, Jiang C, Kang J. microRNA-29b is a novel mediator of Sox2 function in the regulation of somatic cell reprogramming. Cell Res 2013; 23:142-56. [PMID: 23266889 PMCID: PMC3541656 DOI: 10.1038/cr.2012.180] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2012] [Revised: 09/07/2012] [Accepted: 11/23/2012] [Indexed: 12/29/2022] Open
Abstract
Fibroblasts can be reprogrammed into induced pluripotent stem cells (iPSCs) by the application of Yamanaka factors (OSKM), but the mechanisms underlying this reprogramming remain poorly understood. Here, we report that Sox2 directly regulates endogenous microRNA-29b (miR-29b) expression during iPSC generation and that miR-29b expression is required for OSKM- and OSK-mediated reprogramming. Mechanistic studies show that Dnmt3a and Dnmt3b are in vivo targets of miR-29b and that Dnmt3a and Dnmt3b expression is inversely correlated with miR-29b expression during reprogramming. Moreover, the effect of miR-29b on reprogramming can be blocked by Dnmt3a or Dnmt3b overexpression. Further experiments indicate that miR-29b-DNMT signaling is significantly involved in the regulation of DNA methylation-related reprogramming events, such as mesenchymal-to-epithelial transition (MET) and Dlk1-Dio3 region transcription. Thus, our studies not only reveal that miR-29b is a novel mediator of reprogramming factor Sox2 but also provide evidence for a multistep mechanism in which Sox2 drives a miR-29b-DNMT signaling axis that regulates DNA methylation-related events during reprogramming.
Collapse
Affiliation(s)
- Xudong Guo
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Qidong Liu
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Guiying Wang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Songcheng Zhu
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Longfei Gao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Wujun Hong
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Yafang Chen
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Minjuan Wu
- Department of Embryology and Histology, Second Military Medical University, Shanghai 200433, China
| | - Houqi Liu
- Department of Embryology and Histology, Second Military Medical University, Shanghai 200433, China
| | - Cizhong Jiang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Jiuhong Kang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Health Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| |
Collapse
|
50
|
Abstract
One of the most important and evolutionarily conserved strategies to control gene expression in higher metazoa is posttranscriptional regulation via small regulatory RNAs such as microRNAs (miRNAs), endogenous small interfering RNAs (endo-siRNAs), and piwi-interacting RNAs (piRNAs). Primordial germ cells, which are defined by their totipotent potential and noted for their dependence on posttranscriptional regulation by RNA-binding proteins, rely on these small regulatory RNAs for virtually every aspect of their development, including specification, migration, and differentiation into competent gametes. Here, we review current knowledge of the roles miRNAs, endo-siRNAs, and piRNAs play at all stages of germline development in various organisms, focusing on studies in the mouse.
Collapse
Affiliation(s)
- Matthew S Cook
- Department of Urology, University of California, San Francisco, California, USA.
| | | |
Collapse
|