1
|
Liu R, Zhang H, Xin J, Xie S, Jiao F, Li Y, Chu M, Qiu J, Yan Y. Novel circular RNA hsa_circ_0036683 suppresses proliferation and migration by mediating the miR-4664-3p/CDK2AP2 axis in non-small cell lung cancer. Thorac Cancer 2024; 15:1929-1945. [PMID: 39113208 PMCID: PMC11462936 DOI: 10.1111/1759-7714.15396] [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: 04/08/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND The aim of the present study was to investigate the function of novel circular RNA hsa_circ_0036683 (circ-36683) in non-small cell lung cancer (NSCLC). METHODS RNA sequencing was used to screen out differentially expressed miRNAs. Expression levels of miR-4664-3p and circ-36683 were evaluated in lung carcinoma cells and tissues by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The effects of miR-4664-3p and circ-36683 on proliferation and migration were assessed using cell counting kit-8 (CCK-8), wound healing and transwell migration assays and xenograft experiments. The targeting relationship of circ-36683/miR-4664-3p/CDK2AP2 was assessed by luciferase reporter assays, western blot, qRT-PCR and argonaute2-RNA immunoprecipitation (AGO2 RIP). Co-immunoprecipitation (Co-IP), 5-ethynyl-2'-deoxyuridine (EdU) staining and CCK-8 were used to validate the indispensable role of CDK2AP2 in suppressing cell proliferation as a result of CDK2AP1 overexpression. RESULTS By RNA sequencing, miR-4664-3p was screened out as an abnormally elevated miRNA in NSCLC tissues. Transfection of miR-4664-3p could promote cell proliferation, migration and xenograft tumor growth. As a target of miR-4664-3p, CDK2AP2 expression was downregulated by miR-4664-3p transfection and CDK2AP2 overexpression could abolish the proliferation promotion resulting from miR-4664-3p elevation. Circ-36683, derived from back splicing of ABHD2 pre-mRNA, was attenuated in NSCLC tissue and identified as a sponge of miR-4664-3p. The functional study revealed that circ-36683 overexpression suppressed cell proliferation, migration and resulted in G0/G1 phase arrest. More importantly, the antioncogenic function of circ-36683 was largely dependent on the miR-4664-3p/CDK2AP2 axis, through which circ-36683 could upregulate the expression of p53/p21/p27 and downregulate the expression of CDK2/cyclin E1. CONCLUSION The present study revealed the antioncogenic role of circ-36683 in suppressing cell proliferation and migration and highlighted that targeting the circ-36683/miR-4664-3p/CDK2AP2 axis is a promising strategy for the intervention of NSCLC.
Collapse
Affiliation(s)
- Rui Liu
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiChina
| | - Han Zhang
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiChina
| | - Jiaxuan Xin
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiChina
| | - Shu‐yang Xie
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiChina
| | - Fei Jiao
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiChina
| | - You‐Jie Li
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiChina
| | - Meng‐yuan Chu
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiChina
| | - Junming Qiu
- Yantai Affiliated Hospital of Binzhou Medical UniversityYantaiChina
| | - Yun‐fei Yan
- Department of Biochemistry and Molecular BiologyBinzhou Medical UniversityYantaiChina
| |
Collapse
|
2
|
Chen Y, Li M, Wu Y. The occurrence and development of induced pluripotent stem cells. Front Genet 2024; 15:1389558. [PMID: 38699229 PMCID: PMC11063328 DOI: 10.3389/fgene.2024.1389558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
The ectopic expression of four transcription factors, Oct3/4, Sox2, Klf4, and c-Myc (OSKM), known as "Yamanaka factors," can reprogram or stimulate the production of induced pluripotent stem cells (iPSCs). Although OSKM is still the gold standard, there are multiple ways to reprogram cells into iPSCs. In recent years, significant progress has been made in improving the efficiency of this technology. Ten years after the first report was published, human pluripotent stem cells have gradually been applied in clinical settings, including disease modeling, cell therapy, new drug development, and cell derivation. Here, we provide a review of the discovery of iPSCs and their applications in disease and development.
Collapse
Affiliation(s)
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
3
|
Gu J, Li Y, Tian Y, Zhang Y, Cheng Y, Tang Y. Noncanonical functions of microRNAs in the nucleus. Acta Biochim Biophys Sin (Shanghai) 2024; 56:151-161. [PMID: 38167929 PMCID: PMC10984876 DOI: 10.3724/abbs.2023268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 11/03/2023] [Indexed: 01/05/2024] Open
Abstract
MicroRNAs (miRNAs) are small noncoding RNAs (ncRNAs) that play their roles in the regulation of physiological and pathological processes. Originally, it was assumed that miRNAs only modulate gene expression posttranscriptionally in the cytoplasm by inducing target mRNA degradation. However, with further research, evidence shows that mature miRNAs also exist in the cell nucleus, where they can impact gene transcription and ncRNA maturation in several ways. This review provides an overview of novel models of nuclear miRNA functions. Some of the models remain to be verified by experimental evidence, and more details of the miRNA regulation network remain to be discovered in the future.
Collapse
Affiliation(s)
- Jiayi Gu
- College of Basic Medical SciencesShanghai Jiao Tong University School of MedicineShanghai200001China
| | - Yuanan Li
- College of Basic Medical SciencesShanghai Jiao Tong University School of MedicineShanghai200001China
| | - Youtong Tian
- College of Basic Medical SciencesShanghai Jiao Tong University School of MedicineShanghai200001China
| | - Yehao Zhang
- College of Basic Medical SciencesShanghai Jiao Tong University School of MedicineShanghai200001China
| | - Yongjun Cheng
- Department of Rheumatologythe First People’s Hospital of WenlingWenling317500China
| | - Yuanjia Tang
- Shanghai Institute of Rheumatology/Department of RheumatologyRenji HospitalShanghai Jiao Tong University School of MedicineShanghai200001China
- State Key Laboratory of Oncogenes and Related GenesShanghai Cancer InstituteRenji HospitalShanghai200031China
| |
Collapse
|
4
|
Ghosh A, Som A. Network analysis of transcriptomic data uncovers molecular signatures and the interplay of mRNAs, lncRNAs, and miRNAs in human embryonic stem cells. Differentiation 2024; 135:100738. [PMID: 38008592 DOI: 10.1016/j.diff.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/30/2023] [Accepted: 11/09/2023] [Indexed: 11/28/2023]
Abstract
Growing evidence has shown that besides the protein coding genes, the non-coding elements of the genome are indispensable for maintaining the property of self-renewal in human embryonic stem cells and in cell fate determination. However, the regulatory mechanisms and the landscape of interactions between the coding and non-coding elements is poorly understood. In this work, we used weighted gene co-expression network analysis (WGCNA) on transcriptomic data retrieved from RNA-seq and small RNA-seq experiments and reconstructed the core human pluripotency network (called PluriMLMiNet) consisting of 375 mRNA, 57 lncRNA and 207 miRNAs. Furthermore, we derived networks specific to the naïve and primed states of human pluripotency (called NaiveMLMiNet and PrimedMLMiNet respectively) that revealed a set of molecular markers (RPS6KA1, ZYG11A, ZNF695, ZNF273, and NLRP2 for naive state, and RAB34, TMEM178B, PTPRZ1, USP44, KIF1A and LRRN1 for primed state) which can be used to distinguish the pluripotent state from the non-pluripotent state and also to identify the intra-pluripotency states (i.e., naïve and primed state). The lncRNA DANT1 was found to be a crucial as it formed a bridge between the naive and primed state-specific networks. Analysis of the genes neighbouring DANT1 suggested its possible role as a competing endogenous RNA (ceRNA) for the induction and maintenance of human pluripotency. This was computationally validated by predicting the missing DANT1-miRNA interactions to complete the ceRNA circuit. Here we first report that DANT1 might harbour binding sites for miRNAs hsa-miR-30c-2-3p, hsa-miR-210-3p and hsa-let-7b-5p which may influence pluripotency.
Collapse
Affiliation(s)
- Arindam Ghosh
- Centre of Bioinformatics, Institute of Interdisciplinary Studies, University of Allahabad, Prayagraj, 211002, India; Institute of Biomedicine, University of Eastern Finland, FI-70210, Kuopio, Finland.
| | - Anup Som
- Centre of Bioinformatics, Institute of Interdisciplinary Studies, University of Allahabad, Prayagraj, 211002, India.
| |
Collapse
|
5
|
Paloviita P, Vuoristo S. The non-coding genome in early human development - Recent advancements. Semin Cell Dev Biol 2022; 131:4-13. [PMID: 35177347 DOI: 10.1016/j.semcdb.2022.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 12/14/2022]
Abstract
Not that long ago, the human genome was discovered to be mainly non-coding, that is comprised of DNA sequences that do not code for proteins. The initial paradigm that non-coding is also non-functional was soon overturned and today the work to uncover the functions of non-coding DNA and RNA in human early embryogenesis has commenced. Early human development is characterized by large-scale changes in genomic activity and the transcriptome that are partly driven by the coordinated activation and repression of repetitive DNA elements scattered across the genome. Here we provide examples of recent novel discoveries of non-coding DNA and RNA interactions and mechanisms that ensure accurate non-coding activity during human maternal-to-zygotic transition and lineage segregation. These include studies on small and long non-coding RNAs, transposable element regulation, and RNA tailing in human oocytes and early embryos. High-throughput approaches to dissect the non-coding regulatory networks governing early human development are a foundation for functional studies of specific genomic elements and molecules that has only begun and will provide a wider understanding of early human embryogenesis and causes of infertility.
Collapse
Affiliation(s)
- Pauliina Paloviita
- Department of Obstetrics and Gynaecology, University of Helsinki, 00014 Helsinki, Finland
| | - Sanna Vuoristo
- Department of Obstetrics and Gynaecology, University of Helsinki, 00014 Helsinki, Finland.
| |
Collapse
|
6
|
Hasani Fard AH, Kamalipour F, Mazaheri Z, Hosseini SJ. The Effect of miR-106b-5p Expression in The Production of iPS-Like Cells from Mice SSCs during The Formation of Teratoma and The Three Embryonic Layers. CELL JOURNAL 2022; 24:442-448. [PMID: 36093803 PMCID: PMC9468720 DOI: 10.22074/cellj.2022.8147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Indexed: 11/24/2022]
Abstract
<strong>Objective:</strong> According to the mounting data, microRNAs (miRNAs) may play a key role in reprogramming. miR-106b<br />is considered as an enhancer in reprogramming efficiency. Based on induced pluripotent stem cells (iPSCs), cell treatments have a huge amount of potential. One of the main concerns about using iPSCs in therapeutic settings is the possibility of tumor formation. It is hypothesized that a procedure that can reprogram cells with less genetic manipulation reduces the possibility of tumorigenicity.<br /><strong>Materials and Methods:</strong> In this experimental study, miR-106b-5p transduced by pLV-miRNA vector into mice isolated spermatogonial stem cells (SSCs) to achieve iPS-like cells. Then the transduced cells were cultured in specific conditions to study the formation of three germ layers. The tumorigenicity of these iPS-like cells was investigated by transplantation into male BALB/C mice.<br /><strong>Results:</strong> We show that SSCs can be successfully reprogrammed into induced iPS-like cells by pLV-miRNA vector to transduce the hsa-mir-106b-5p into SSCs and generating osteogenic, neural and hepatoblast lineage cells in vitro as a result of pluripotency. Although these iPS-like cells are pluripotent, they cannot form palpable tumors in vivo.<br /><strong>Conclusion:</strong> These results demonstrate that infection of hsa-mir-106b-5p into SSCs can reprogram them into iPSCs<br />and advanced germ cell lineages without tumorigenicity. Also, a novel approach for studying the generation of iPSCs<br />and the application of iPS or iPS-like cells in regenerative medicine is presented.
Collapse
Affiliation(s)
- Amir Hossein Hasani Fard
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Kamalipour
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohreh Mazaheri
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Jalil Hosseini
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran,P.O.Box: 19899/34148Men’s Health and Reproductive Health Research CenterShahid Beheshti University of
Medical SciencesTehranIran
| |
Collapse
|
7
|
Hasani Fard AH, Valizadeh M, Mazaheri Z, Hosseini SJ. MiR-106b-5p Regulates the Reprogramming of Spermatogonial Stem Cells into iPSC (Induced Pluripotent Stem Cell)-Like Cells. IRANIAN BIOMEDICAL JOURNAL 2022; 26:291-300. [PMID: 35791490 PMCID: PMC9432470 DOI: 10.52547/ibj.3594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 01/05/2022] [Indexed: 11/18/2022]
Abstract
Background Recent years have brought notable progress in raising the efficiency of the reprogramming technique so that approaches have evolved from known transgenic factors to only a few miRNAs. Nevertheless, there is a poor understanding of both the key factors and biological networks underlying this reprogramming. The present study aimed to investigate the potential of miR-106b-5p in regulating spermatogonial stem cells (SSCs) to induced pluripotent stem cell (iPSC)-like cells. Methods We used SSCs because pluripotency is inducible in SSCs under defined culture conditions, and they have a few issues compared to other adult stem cells. As both signaling and post-transcriptional gene controls are critical for pluripotency regulation, we traced the expression of Oct-4, Sox-2, Klf-4, c-Myc, and Nanog (OSKMN). Besides, we considered miR-106b-5p targets using bioinformatic methods. Results Our results showed that transfected SSCs with miR-106b-5p increased the expression of the OSKMN factors, which was significantly more than negative control groups. Moreover, using the functional miRNA enrichment analysis, online tools, and databases, we predicted that miR-106b-5p targeted a signaling pathway gene named MAPK1/ERK2, related to regulating stem cell pluripotency. Conclusion Together, our data suggest that miR-106b-5p regulates the reprogramming of SSCs into iPSC-like cells. Furthermore, noteworthy progress in the in vitro development of SSCs indicates promise reservoirs and opportunities for future clinical trials.
Collapse
Affiliation(s)
- Amir Hossein Hasani Fard
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmoud Valizadeh
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohreh Mazaheri
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Jalil Hosseini
- Men’s Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Sun X, Zhang X, Yang L, Dong B. A microRNA Cluster-Lefty Pathway is Required for Cellulose Synthesis During Ascidian Larval Metamorphosis. Front Cell Dev Biol 2022; 10:835906. [PMID: 35372357 PMCID: PMC8965075 DOI: 10.3389/fcell.2022.835906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Synthesis of cellulose and formation of tunic structure are unique traits in the tunicate animal group. However, the regulatory mechanism of tunic formation remains obscure. Here, we identified a novel microRNA cluster of three microRNAs, including miR4018a, miR4000f, and miR4018b in Ciona savignyi. In situ hybridization and promoter assays showed that miR4018a/4000f/4018b cluster was expressed in the mesenchymal cells in the larval trunk, and the expression levels were downregulated during the later tailbud stage and larval metamorphosis. Importantly, overexpression of miR4018a/4000f/4018b cluster in mesenchymal cells abolished the cellulose synthesis in Ciona larvae and caused the loss of tunic cells in metamorphic larvae, indicating the regulatory roles of miR4018a/4000f/4018b cluster in cellulose synthesis and mesenchymal cell differentiation into tunic cells. To elucidate the molecular mechanism, we further identified the target genes of miR4018a/4000f/4018b cluster using the combination approaches of TargetScan prediction and RNA-seq data. Left-right determination factor (Lefty) was confirmed as one of the target genes after narrow-down screening and an experimental luciferase assay. Furthermore, we showed that Lefty was expressed in the mesenchymal and tunic cells, indicating its potentially regulatory roles in mesenchymal cell differentiation and tunic formation. Notably, the defects in tunic formation and loss of tunic cells caused by overexpression of miR4018a/4000f/4018b cluster could be restored when Lefty was overexpressed in Ciona larvae, suggesting that miR4018a/4000f/4018b regulated the differentiation of mesenchymal cells into tunic cells through the Lefty signaling pathway during ascidian metamorphosis. Our findings, thus, reveal a novel microRNA-Lefty molecular pathway that regulates mesenchymal cells differentiating into tunic cells required for the tunic formation in tunicate species.
Collapse
Affiliation(s)
- Xueping Sun
- Sars Fang Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Xiaoming Zhang
- Sars Fang Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Likun Yang
- Sars Fang Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Bo Dong
- Sars Fang Centre, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China.,Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, China
| |
Collapse
|
9
|
Zhu Y, Li J, Liu H, Song Z, Yang Q, Lu C, Chen W. Circular RNA, hsa_circRNA_102049, promotes colorectal cancer cell migration and invasion via binding and suppressing miRNA-455-3p. Exp Ther Med 2022; 23:244. [PMID: 35222721 PMCID: PMC8815054 DOI: 10.3892/etm.2022.11169] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 04/28/2021] [Indexed: 11/25/2022] Open
Abstract
Colorectal cancer (CRC) is the second most prevalent malignant gastrointestinal tumor type worldwide, displaying poor prognosis. Accumulating studies have reported the significance of circular RNAs (circRNAs) and microRNAs (miRNAs) in CRC carcinogenesis and development. At present, the functions and mechanisms of action underlying the circular RNA, hsa_circRNA_102049, in CRC are not completely understood. The present study aimed to establish the involvement of hsa_circRNA_102049 in CRC, as well as the associated mechanisms. The expression levels of hsa_circRNA_102049 and miRNA-455-3p were measured in CRC cell lines and tissues via reverse transcription-quantitative PCR. CRC progression was evaluated by performing Cell Counting Kit-8, flow cytometry, wound healing and Transwell invasion assays. The results demonstrated that hsa_circRNA_102049 was highly expressed in both CRC tissues and cell lines, which was associated with enhanced CRC cell proliferation, migration and invasion. Furthermore, miR-455-3p expression was downregulated in CRC cells and served as a target of has_circRNA_102049, which was validated by performing the dual luciferase reporter assay. hsa_circRNA_102049 knockdown significantly increased miR-455-3p expression, which was significantly reversed by co-transfection with the miR-455-3p inhibitor. Notably, miRNA-455-3p overexpression alleviated hsa_circRNA_102049-mediated induction of CRC cell proliferation, migration and invasion. The present study clearly demonstrated that miRNA-455-3p was a target of hsa_circRNA_102049. Moreover, the results indicated that the circular RNA, hsa_circRNA_102049, may function as a tumor promoter in CRC via directly sponging miRNA-455-3p.
Collapse
Affiliation(s)
- Yuandong Zhu
- Department of Colorectal and Anal Surgery, Yiwu Central Hospital, Yiwu, Zhejiang 322000, P.R. China
| | - Jianjion Li
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Haiyuan Liu
- Department of Colorectal and Anal Surgery, Yiwu Central Hospital, Yiwu, Zhejiang 322000, P.R. China
| | - Zhengming Song
- Department of Colorectal and Anal Surgery, Yiwu Central Hospital, Yiwu, Zhejiang 322000, P.R. China
| | - Qinghua Yang
- Department of Colorectal and Anal Surgery, Yiwu Central Hospital, Yiwu, Zhejiang 322000, P.R. China
| | - Chengdong Lu
- Department of Colorectal and Anal Surgery, Yiwu Central Hospital, Yiwu, Zhejiang 322000, P.R. China
| | - Wenbin Chen
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
10
|
Oliver C, Annacondia ML, Wang Z, Jullien PE, Slotkin RK, Köhler C, Martinez G. The miRNome function transitions from regulating developmental genes to transposable elements during pollen maturation. THE PLANT CELL 2022; 34:784-801. [PMID: 34755870 PMCID: PMC8824631 DOI: 10.1093/plcell/koab280] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 11/04/2021] [Indexed: 06/13/2023]
Abstract
Animal and plant microRNAs (miRNAs) are essential for the spatio-temporal regulation of development. Together with this role, plant miRNAs have been proposed to target transposable elements (TEs) and stimulate the production of epigenetically active small interfering RNAs. This activity is evident in the plant male gamete containing structure, the male gametophyte or pollen grain. How the dual role of plant miRNAs, regulating both genes and TEs, is integrated during pollen development and which mRNAs are regulated by miRNAs in this cell type at a genome-wide scale are unknown. Here, we provide a detailed analysis of miRNA dynamics and activity during pollen development in Arabidopsis thaliana using small RNA and degradome parallel analysis of RNA end high-throughput sequencing. Furthermore, we uncover miRNAs loaded into the two main active Argonaute (AGO) proteins in the uninuclear and mature pollen grain, AGO1 and AGO5. Our results indicate that the developmental progression from microspore to mature pollen grain is characterized by a transition from miRNAs targeting developmental genes to miRNAs regulating TE activity.
Collapse
Affiliation(s)
- Cecilia Oliver
- Department of Plant Biology, Swedish University of Agricultural Sciences and Linnean Center for Plant Biology, Uppsala 75007, Sweden
| | - Maria Luz Annacondia
- Department of Plant Biology, Swedish University of Agricultural Sciences and Linnean Center for Plant Biology, Uppsala 75007, Sweden
| | - Zhenxing Wang
- Department of Plant Biology, Swedish University of Agricultural Sciences and Linnean Center for Plant Biology, Uppsala 75007, Sweden
- College of Horticulture and State Key Laboratory of Crop Genetics and Germplasm Enhancement, Nanjing Agricultural University, Nanjing 210095, China
- Key Laboratory of Landscaping, Ministry of Agriculture and Rural Affairs and Key Laboratory of Biology of Ornamental Plants in East China, National Forestry and Grassland Administration, Nanjing Agricultural University, Nanjing 210095, China
| | - Pauline E Jullien
- Institute of Plant Sciences, University of Bern, Bern 3013, Switzerland
| | - R Keith Slotkin
- Donald Danforth Plant Science Center, St. Louis, Missouri 63132, USA
- Division of Biological Sciences, University of Missouri Columbia, Columbia, Missouri 65201, USA
| | - Claudia Köhler
- Department of Plant Biology, Swedish University of Agricultural Sciences and Linnean Center for Plant Biology, Uppsala 75007, Sweden
- Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm 14476, Germany
| | | |
Collapse
|
11
|
Expression Profile of miRs in Mesial Temporal Lobe Epilepsy: Systematic Review. Int J Mol Sci 2022; 23:ijms23020951. [PMID: 35055144 PMCID: PMC8781102 DOI: 10.3390/ijms23020951] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/08/2022] [Accepted: 01/12/2022] [Indexed: 02/04/2023] Open
Abstract
Temporal lobe epilepsy (TLE) is one of the most common forms of focal epilepsy in children and adults. TLE is characterized by variable onset and seizures. Moreover, this form of epilepsy is often resistant to pharmacotherapy. The search for new mechanisms for the development of TLE may provide us with a key to the development of new diagnostic methods and a personalized approach to the treatment. In recent years, the role of non-coding ribonucleic acids (RNA) has been actively studied, among which microRNA (miR) is of the greatest interest. (1) Background: The purpose of the systematic review is to analyze the studies carried out on the role of miRs in the development of mesial TLE (mTLE) and update the existing knowledge about the biomarkers of this disease. (2) Methods: The search for publications was carried out in the databases PubMed, Springer, Web of Science, Clinicalkeys, Scopus, OxfordPress, Cochrane. The search was carried out using keywords and combinations. We analyzed publications for 2016–2021, including original studies in an animal model of TLE and with the participation of patients with TLE, thematic and systemic reviews, and Cochrane reviews. (3) Results: this thematic review showed that miR‒155, miR‒153, miR‒361‒5p, miR‒4668‒5p, miR‒8071, miR‒197‒5p, miR‒145, miR‒181, miR‒199a, miR‒1183, miR‒129‒2‒3p, miR‒143‒3p (upregulation), miR–134, miR‒0067835, and miR‒153 (downregulation) can be considered as biomarkers of mTLE. However, the roles of miR‒146a, miR‒142, miR‒106b, and miR‒223 are questionable and need further study. (4) Conclusion: In the future, it will be possible to consider previously studied miRs, which have high specificity and sensitivity in mTLE, as prognostic biomarkers (predictors) of the risk of developing this disease in patients with potentially epileptogenic structural damage to the mesial regions of the temporal lobe of the brain (congenital disorders of the neuronal migration and neurogenesis, brain injury, neuro-inflammation, tumor, impaired blood supply, neurodegeneration, etc.).
Collapse
|
12
|
Zeidler M, Kummer KK, Schöpf CL, Kalpachidou T, Kern G, Cader MZ, Kress M. NOCICEPTRA: Gene and microRNA Signatures and Their Trajectories Characterizing Human iPSC-Derived Nociceptor Maturation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2102354. [PMID: 34486248 PMCID: PMC8564443 DOI: 10.1002/advs.202102354] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Indexed: 05/07/2023]
Abstract
Nociceptors are primary afferent neurons serving the reception of acute pain but also the transit into maladaptive pain disorders. Since native human nociceptors are hardly available for mechanistic functional research, and rodent models do not necessarily mirror human pathologies in all aspects, human induced pluripotent stem cell-derived nociceptors (iDN) offer superior advantages as a human model system. Unbiased mRNA::microRNA co-sequencing, immunofluorescence staining, and qPCR validations, reveal expression trajectories as well as miRNA target spaces throughout the transition of pluripotent cells into iDNs. mRNA and miRNA candidates emerge as regulatory hubs for neurite outgrowth, synapse development, and ion channel expression. The exploratory data analysis tool NOCICEPTRA is provided as a containerized platform to retrieve experimentally determined expression trajectories, and to query custom gene sets for pathway and disease enrichments. Querying NOCICEPTRA for marker genes of cortical neurogenesis reveals distinct similarities and differences for cortical and peripheral neurons. The platform provides a public domain neuroresource to exploit the entire data sets and explore miRNA and mRNA as hubs regulating human nociceptor differentiation and function.
Collapse
Affiliation(s)
- Maximilian Zeidler
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| | - Kai K. Kummer
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| | - Clemens L. Schöpf
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| | | | - Georg Kern
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| | - M. Zameel Cader
- Weatherall Institute of Molecular MedicineUniversity of OxfordOxfordOX3 9DSUK
| | - Michaela Kress
- Institute of PhysiologyMedical University of InnsbruckInnsbruck6020Austria
| |
Collapse
|
13
|
Microrna analysis of human decidua mesenchymal stromal cells from preeclampsia patients. Placenta 2021; 115:12-19. [PMID: 34534911 DOI: 10.1016/j.placenta.2021.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 01/03/2023]
Abstract
INTRODUCTION In preeclampsia (PE), human decidua mesenchymal stromal cells (hDMSCs) are exposed to abnormally high levels of oxidative stress and inflammatory factors circulating in the maternal blood. MicroRNAs (miRNAs) have been shown to have a significant impact on the differentiation, maturation and function of mesenchymal stromal cells (MSCs). Our aim in the present study is firstly to investigate differentially expressed miRNA levels to be used as a biomarker in the early detection of PE and secondly to investigate whether those differentially expressed miRNAs in hDMSCs have an effect on the pathogenesis of PE. METHODS This study covers miRNA expression analysis of hDMSCs from 7 PE patient and 7 healthy pregnant women and is a preliminary study to investigate putative biomarkers. After cell culture and cell sorting, total RNA including miRNAs were isolated from hDMSCs. Let-7b-3p, let-7f-1-3p, miR-191-3p, miR-550a-5p, miR-33b-3p and miR-425-3p were used for miRNA analysis and U6 snRNA was used for normalization of the samples. MiRNA analysis was performed by droplet digital polymerase chain reaction (ddPCR) method and obtained results were evaluated statistically. RESULTS As a result of the analysis, it was observed that the levels of hsa-miR-33b-3p significantly (AUC: 0.93, p = 0.04, fold change: 4.5) increased in hDMSC of PE patients compared to healthy controls. However, let-7b-3p, let-7f-1-3p, miR-191-3p, miR-550a-5p, and miR-425-3p were not considered as significant because they did not meet the p < 0,05 requirement. DISCUSSION Within the scope of the study, it is predicted that miR-33b-3p (p = 0.004, AUC = 0.93) can be used as a biomarker in detecting PE.
Collapse
|
14
|
Xie D, Tong M, Xia B, Feng G, Wang L, Li A, Luo G, Wan H, Zhang Z, Zhang H, Yang YG, Zhou Q, Wang M, Wang XJ. Long noncoding RNA lnc-NAP sponges mmu-miR-139-5p to modulate Nanog functions in mouse ESCs and embryos. RNA Biol 2021; 18:875-887. [PMID: 32991228 PMCID: PMC8081037 DOI: 10.1080/15476286.2020.1827591] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/30/2020] [Accepted: 09/20/2020] [Indexed: 12/30/2022] Open
Abstract
The pluripotency of embryonic stem cells (ESCs) is controlled by a multilayer regulatory network, of which the key factors include core pluripotency genes Oct4, Sox2 and Nanog, and multiple microRNAs (miRNAs). Recently, long noncoding RNAs (lncRNAs) have been discovered as a class of new regulators for ESCs, and some lncRNAs could function as competing endogenous RNAs (ceRNAs) to regulate mRNAs by competitively binding to miRNAs. Here, we identify mmu-miR-139-5p as a new regulator for Nanog by targeting Nanog 3' untranslated region (UTR) to repress Nanog expression in mouse ESCs and embryos. Such regulation could be released by an ESC-specifically expressed ceRNA named lnc-NAP. The expression of lnc-NAP is activated by OCT4, SOX2, as well as NANOG through promoter binding. Downregulation of lnc-NAP reduces Nanog abundance, which leads to decreased pluripotency of mouse ESCs and embryonic lethality. These results reveal lnc-NAP as a new regulator for Nanog in mouse ESCs, and uncover a feed-forward regulatory loop of Nanog through the participation of lnc-NAP.
Collapse
MESH Headings
- 3' Untranslated Regions/genetics
- Animals
- Cell Differentiation/genetics
- Embryo, Mammalian/cytology
- Embryo, Mammalian/embryology
- Embryo, Mammalian/metabolism
- Embryonic Stem Cells/cytology
- Embryonic Stem Cells/metabolism
- Gene Expression Regulation, Developmental
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Inbred NOD
- Mice, SCID
- MicroRNAs/genetics
- Nanog Homeobox Protein/genetics
- Nanog Homeobox Protein/metabolism
- Octamer Transcription Factor-3/genetics
- Octamer Transcription Factor-3/metabolism
- Promoter Regions, Genetic/genetics
- Protein Binding
- RNA, Long Noncoding/genetics
- RNA-Seq/methods
- Reverse Transcriptase Polymerase Chain Reaction/methods
- SOXB1 Transcription Factors/genetics
- SOXB1 Transcription Factors/metabolism
- Mice
Collapse
Affiliation(s)
- Dongfang Xie
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Man Tong
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Baolong Xia
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Guihai Feng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Leyun Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ang Li
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Guanzheng Luo
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Haifeng Wan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zeyu Zhang
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Hao Zhang
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Yun-Gui Yang
- University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Genomic and Precision Medicine, Collaborative Innovation Center of Genetics and Development, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Meng Wang
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Xiu-Jie Wang
- Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Innovation Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
15
|
Wang Y, Wang Z, Xie K, Zhao X, Jiang X, Chen B, Han Y, Lu Y, Huang L, Zhang W, Yang Y, Shi P. High-Efficiency Cellular Reprogramming by Nanoscale Puncturing. NANO LETTERS 2020; 20:5473-5481. [PMID: 32520569 DOI: 10.1021/acs.nanolett.0c01979] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Induced pluripotent stem cells (iPSCs) bear great potential for disease modeling, drug discovery, and regenerative medicine; however, the wide adoption of iPSC for clinically relevant applications has been hindered by the extremely low reprogramming efficiency. Here, we describe a high-efficiency cellular reprogramming strategy by puncturing cells with an array of diamond nanoneedles, which is applied to temporally disrupt the cell membrane in a reversible and minimally invasive format. This method enables high-efficiency cytoplasmic delivery of mini-intronic plasmid vectors to initiate the conversion of human fibroblast cells to either primed iPSCs or naı̈ve iPSCs. The nanopuncturing operation is directly performed on cells in adherent culture without any cell lift-off and is completed within just 5 min. The treated cells are then cultured in feeder-free medium to achieve a reprogramming efficiency of 1.17 ± 0.28%, which is more than 2 orders of magnitude higher than the typical results from common methods involving plasmid delivery.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Zixun Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Kai Xie
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Xi Zhao
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Xuezhen Jiang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Bing Chen
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Ying Han
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Yang Lu
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong SAR, China
| | - Linfeng Huang
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Wenjun Zhang
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
- Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, Hong Kong SAR, China
| | - Yang Yang
- Functional Thin Films Research Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Peng Shi
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, China
- Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, Hong Kong SAR, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen 518057, China
| |
Collapse
|
16
|
Tumor microRNA profile and prognostic value for lymph node metastasis in oral squamous cell carcinoma patients. Oncotarget 2020; 11:2204-2215. [PMID: 32577165 PMCID: PMC7289532 DOI: 10.18632/oncotarget.27616] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/14/2020] [Indexed: 12/23/2022] Open
Abstract
Neck lymph node metastasis (LN+) is one of the most significant prognostic factors affecting 1-in-2 patients diagnosed with oral squamous cell carcinoma (OSCC). The different LN outcomes between clinico-pathologically similar primary tumors suggest underlying molecular signatures that could be associated with the risk of nodal disease development. MicroRNAs (miRNAs)are short non-coding molecules that regulate the expression of their target genes to maintain the balance of cellular processes. A plethora of evidence has indicated that aberrantly expressed miRNAs are involved in cancers with either an antitumor or oncogenic role. In this study, we characterized miRNA expression among OSCC fresh-frozen tumors with known outcomes of nodal disease (82 LN+, 76 LN0). We identified 49 differentially expressed miRNAs in tumors of the LN+ group. Using penalized lasso Cox regression, we identified a group of 10 miRNAs of which expression levels were highly associated with nodal-disease free survival. We further reported a 4-miRNA panel (miR-21-5p, miR-107, miR-1247-3p, and miR-181b-3p) with high accuracy in discriminating LN status, suggesting their potential application as prognostic biomarkers for nodal disease.
Collapse
|
17
|
Kolarz B, Ciesla M, Dryglewska M, Rosenthal AK, Majdan M. Hypermethylation of the miR-155 gene in the whole blood and decreased plasma level of miR-155 in rheumatoid arthritis. PLoS One 2020; 15:e0233897. [PMID: 32484820 PMCID: PMC7266293 DOI: 10.1371/journal.pone.0233897] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/14/2020] [Indexed: 11/18/2022] Open
Abstract
Objectives miR-155 plays a critical role in the inflammatory process and in diseases such as rheumatoid arthritis (RA). miR155 gene expression is regulated by its gene promoter region CpG island methylation. Previous studies have shown inconsistent changes in circulating levels of mir-155 in RA patients. The aims of our study were to evaluate miR-155 levels in plasma, to investigate its gene methylation level, and to correlate these levels with RA disease activity. Methods One hundred and twenty-five patients with RA, and 30 age and sex-matched healthy controls (HC) were enrolled. Whole blood and plasma samples were collected and stored at -80°C until analysis. DAS28 score at the time of the blood draw was used to assess RA disease activity. The methylation status of miR-155 host gene was determined in whole blood by quantitative real-time methylation-specific PCR (qPCR). miR-155 expression levels were evaluated by quantitative reverse transcription PCR. Results We found significantly lower circulating miR155 levels in RA patients compared to HC. Interestingly, the miR-155 gene methylation level was significantly higher in RA patients than in HC. miR-155 levels did not correlate with ACPA or RF positivity or disease activity. Conclusions We show here higher miR-155 methylation in whole blood and lower plasma miR155 expression in RA patients in comparison to HC. The evaluation of miR-155 host gene methylation status or miR155 plasma level might be a potentially useful marker in RA determination.
Collapse
Affiliation(s)
- Bogdan Kolarz
- College of Medical Sciences, University of Rzeszow, Rzeszow, Poland
- * E-mail:
| | - Marek Ciesla
- College of Medical Sciences, University of Rzeszow, Rzeszow, Poland
| | - Magdalena Dryglewska
- Department of Rheumatology and Connective Tissue Disease, Medical University of Lublin, Lublin, Poland
| | - Ann K. Rosenthal
- Division of Rheumatology, Department of Medicine, Medical College of Wisconsin, Wauwatosa, WI, United States of America
| | - Maria Majdan
- Department of Rheumatology and Connective Tissue Disease, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
18
|
Wang Y, Wang X, Jiang Y, Liu R, Cao D, Pan J, Luo Y. Identification of key miRNAs and genes for mouse retinal development using a linear model. Mol Med Rep 2020; 22:494-506. [PMID: 32319662 PMCID: PMC7248464 DOI: 10.3892/mmr.2020.11082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 04/01/2020] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) are upstream regulators of gene expression and are involved in several biological processes. The purpose of the present study was to obtain a detailed spatiotemporal miRNA expression profile in mouse retina, to identify one or more miRNAs that are key to mouse retinal development and to investigate the roles and mechanisms of these miRNAs. The miRNA expression pattern of the developing mouse retina was acquired from Locked Nucleic Acid microarrays. Data were processed to identify differentially expressed miRNAs (DE‑miRNAs) using the linear model in Python 3.6. Following bioinformatics analysis and reverse transcription‑quantitative polymerase chain reaction validation, 8 miRNAs (miR‑9‑5p, miR‑130a‑3p, miR‑92a‑3p, miR‑20a‑5p, miR‑93‑5p, miR‑9‑3p, miR‑709 and miR‑124) were identified as key DE‑miRNAs with low variability during mouse retinal development. Gene Ontology analysis revealed that the target genes of the DE‑miRNAs were enriched in cellular metabolic processes. Kyoto Encyclopedia of Genes and Genomes analysis demonstrated that the target genes of the DE‑miRNAs were significantly enriched in PI3K/AKT/mTOR, class O of forkhead box transcription factors, mitogen‑activated protein kinase (MAPK), neurotrophin and transforming growth factor (TGF)‑β signaling, as well as focal adhesion and the axon guidance pathway. PI3K, AKT, PTEN, MAPK1, Son of Sevenless, sphingosine‑1‑phosphate receptor 1, BCL‑2L11, TGF‑β receptor type 1/2 and integrin α (ITGA)/ITGAB, which are key components of the aforementioned pathways and were revealed to be target genes of several of the DE‑miRNAs. The present study used a linear model to identify several DE‑miRNAs, as well as their target genes and associated pathways, which may serve crucial roles in mouse retinal development. Therefore, the results obtained in the present study may provide the groundwork for further experiments.
Collapse
Affiliation(s)
- Yishen Wang
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Xiao Wang
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Yukang Jiang
- Department of Statistical Science, School of Mathematics, Southern China Research Center of Statistical Science, Sun Yat‑Sen University, Guangzhou, Guangdong 51027, P.R. China
| | - Ruyuan Liu
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Di Cao
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Jianying Pan
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| | - Yan Luo
- State Key Laboratory of Ophthalmology, Image Reading Center, Zhongshan Ophthalmic Center, Sun Yat‑Sen University, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
19
|
Li QV, Rosen BP, Huangfu D. Decoding pluripotency: Genetic screens to interrogate the acquisition, maintenance, and exit of pluripotency. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1464. [PMID: 31407519 PMCID: PMC6898739 DOI: 10.1002/wsbm.1464] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 05/31/2019] [Accepted: 07/17/2019] [Indexed: 01/25/2023]
Abstract
Pluripotent stem cells have the ability to unlimitedly self-renew and differentiate to any somatic cell lineage. A number of systems biology approaches have been used to define this pluripotent state. Complementary to systems level characterization, genetic screens offer a unique avenue to functionally interrogate the pluripotent state and identify the key players in pluripotency acquisition and maintenance, exit of pluripotency, and lineage differentiation. Here we review how genetic screens have helped us decode pluripotency regulation. We will summarize results from RNA interference (RNAi) based screens, discuss recent advances in CRISPR/Cas-based genetic perturbation methods, and how these advances have made it possible to more comprehensively interrogate pluripotency and differentiation through genetic screens. Such investigations will not only provide a better understanding of this unique developmental state, but may enhance our ability to use pluripotent stem cells as an experimental model to study human development and disease progression. Functional interrogation of pluripotency also provides a valuable roadmap for utilizing genetic perturbation to gain systems level understanding of additional cellular states, from later stages of development to pathological disease states. This article is categorized under: Developmental Biology > Stem Cell Biology and Regeneration Developmental Biology > Developmental Processes in Health and Disease Biological Mechanisms > Cell Fates.
Collapse
Affiliation(s)
- Qing V. Li
- Sloan Kettering Institute, 1275 York Avenue, New York, New York 10065, USA
- Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA
- These authors contributed equally
| | - Bess P. Rosen
- Sloan Kettering Institute, 1275 York Avenue, New York, New York 10065, USA
- Weill Graduate School of Medical Sciences at Cornell University, 1300 York Avenue, New York, New York 10065, USA
- These authors contributed equally
| | - Danwei Huangfu
- Sloan Kettering Institute, 1275 York Avenue, New York, New York 10065, USA
| |
Collapse
|
20
|
Piperigkou Z, Karamanos NK. Dynamic Interplay between miRNAs and the Extracellular Matrix Influences the Tumor Microenvironment. Trends Biochem Sci 2019; 44:1076-1088. [DOI: 10.1016/j.tibs.2019.06.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 06/11/2019] [Accepted: 06/12/2019] [Indexed: 12/19/2022]
|
21
|
Di Stefano B, Luo EC, Haggerty C, Aigner S, Charlton J, Brumbaugh J, Ji F, Rabano Jiménez I, Clowers KJ, Huebner AJ, Clement K, Lipchina I, de Kort MAC, Anselmo A, Pulice J, Gerli MFM, Gu H, Gygi SP, Sadreyev RI, Meissner A, Yeo GW, Hochedlinger K. The RNA Helicase DDX6 Controls Cellular Plasticity by Modulating P-Body Homeostasis. Cell Stem Cell 2019; 25:622-638.e13. [PMID: 31588046 DOI: 10.1016/j.stem.2019.08.018] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/19/2019] [Accepted: 08/29/2019] [Indexed: 01/02/2023]
Abstract
Post-transcriptional mechanisms have the potential to influence complex changes in gene expression, yet their role in cell fate transitions remains largely unexplored. Here, we show that suppression of the RNA helicase DDX6 endows human and mouse primed embryonic stem cells (ESCs) with a differentiation-resistant, "hyper-pluripotent" state, which readily reprograms to a naive state resembling the preimplantation embryo. We further demonstrate that DDX6 plays a key role in adult progenitors where it controls the balance between self-renewal and differentiation in a context-dependent manner. Mechanistically, DDX6 mediates the translational suppression of target mRNAs in P-bodies. Upon loss of DDX6 activity, P-bodies dissolve and release mRNAs encoding fate-instructive transcription and chromatin factors that re-enter the ribosome pool. Increased translation of these targets impacts cell fate by rewiring the enhancer, heterochromatin, and DNA methylation landscapes of undifferentiated cell types. Collectively, our data establish a link between P-body homeostasis, chromatin organization, and stem cell potency.
Collapse
Affiliation(s)
- Bruno Di Stefano
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA
| | - En-Ching Luo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA; Stem Cell Program, University of California San Diego, La Jolla, CA 92093, USA; Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Chuck Haggerty
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Stefan Aigner
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA; Stem Cell Program, University of California San Diego, La Jolla, CA 92093, USA; Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Jocelyn Charlton
- Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Justin Brumbaugh
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA
| | - Fei Ji
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | - Inés Rabano Jiménez
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA; Stem Cell Program, University of California San Diego, La Jolla, CA 92093, USA; Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Katie J Clowers
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Aaron J Huebner
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA
| | - Kendell Clement
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Inna Lipchina
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA
| | - Marit A C de Kort
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA
| | - Anthony Anselmo
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | - John Pulice
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA
| | - Mattia F M Gerli
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | - Hongcang Gu
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Ruslan I Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
| | - Alexander Meissner
- Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA; Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92093, USA; Stem Cell Program, University of California San Diego, La Jolla, CA 92093, USA; Institute for Genomic Medicine, University of California at San Diego, La Jolla, CA 92093, USA.
| | - Konrad Hochedlinger
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Cancer Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Harvard Stem Cell Institute, 1350 Massachusetts Avenue, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
22
|
Silvestro S, Bramanti P, Mazzon E. Role of miRNAs in Alzheimer's Disease and Possible Fields of Application. Int J Mol Sci 2019; 20:E3979. [PMID: 31443326 PMCID: PMC6720959 DOI: 10.3390/ijms20163979] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/07/2019] [Accepted: 08/14/2019] [Indexed: 01/02/2023] Open
Abstract
miRNAs (or microRNAs) are a class of single-stranded RNA molecules, responsible for post-transcriptional gene silencing through binding to the coding region as well as 3' and 5' untranslated region of target genes. About 70% of experimentally detectable miRNAs are expressed in the brain and some studies suggest that miRNAs are intimately involved in synaptic function and in specific signals during memory formation. More and more evidence demonstrates the possible involvement of miRNAs in Alzheimer's disease (AD). AD is the most common form of senile dementia, a disease that affects memory and cognitive functions. It is a neurodegenerative disorder characterized by loss of synapses, extracellular amyloid plaques composed of the amyloid-β peptide (Aβ), and intracellular aggregates of hyperphosphorylated TAU protein. This review aims to provide an overview of the in vivo studies of the last 5 years in the literature describing the role of the different miRNAs involved in AD. miRNAs hold huge potential as diagnostic and prognostic biomarkers and, at the same time, their modulation could be a potential therapeutic strategy against AD.
Collapse
Affiliation(s)
- Serena Silvestro
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| |
Collapse
|
23
|
Yakovlev AF. The Role of miRNA in Differentiation, Cell Proliferation, and Pathogenesis of Poultry Diseases. Russ J Dev Biol 2019. [DOI: 10.1134/s1062360419030081] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
24
|
Integrated Computational Analysis Highlights unique miRNA Signatures in the Subventricular Zone and Striatum of GM2 Gangliosidosis Animal Models. Int J Mol Sci 2019; 20:ijms20133179. [PMID: 31261761 PMCID: PMC6651736 DOI: 10.3390/ijms20133179] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/24/2019] [Accepted: 06/27/2019] [Indexed: 12/12/2022] Open
Abstract
This work explores for the first time the potential contribution of microRNAs (miRNAs) to the pathophysiology of the GM2 gangliosidosis, a group of Lysosomal Storage Diseases. In spite of the genetic origin of GM2 gangliosidosis, the cascade of events leading from the gene/protein defects to the cell dysfunction and death is not fully elucidated. At present, there is no cure for patients. Taking advantage of the animal models of two forms of GM2 gangliosidosis, Tay-Sachs (TSD) and Sandhoff (SD) diseases, we performed a microRNA screening in the brain subventricular zone (SVZ) and striatum (STR), which feature the neurogenesis and neurodegeneration states, respectively, in adult mutant mice. We found abnormal expression of a panel of miRNAs involved in lipid metabolism, CNS development and homeostasis, and neuropathological processes, highlighting region- and disease-specific profiles of miRNA expression. Moreover, by using a computational analysis approach, we identified a unique disease- (SD or TSD) and brain region-specific (SVZ vs. STR) miRNAs signatures of predicted networks potentially related to the pathogenesis of the diseases. These results may contribute to the understanding of GM2 gangliosidosis pathophysiology, with the aim of developing effective treatments.
Collapse
|
25
|
Frame FM, Maitland NJ. Epigenetic Control of Gene Expression in the Normal and Malignant Human Prostate: A Rapid Response Which Promotes Therapeutic Resistance. Int J Mol Sci 2019; 20:E2437. [PMID: 31108832 PMCID: PMC6566891 DOI: 10.3390/ijms20102437] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/24/2019] [Accepted: 04/29/2019] [Indexed: 12/12/2022] Open
Abstract
A successful prostate cancer must be capable of changing its phenotype in response to a variety of microenvironmental influences, such as adaptation to treatment or successful proliferation at a particular metastatic site. New cell phenotypes emerge by selection from the large, genotypically heterogeneous pool of candidate cells present within any tumor mass, including a distinct stem cell-like population. In such a multicellular model of human prostate cancer, flexible responses are primarily governed not only by de novo mutations but appear to be dominated by a combination of epigenetic controls, whose application results in treatment resistance and tumor relapse. Detailed studies of these individual cell populations have resulted in an epigenetic model for epithelial cell differentiation, which is also instructive in explaining the reported high and inevitable relapse rates of human prostate cancers to a multitude of treatment types.
Collapse
Affiliation(s)
- Fiona M Frame
- The Cancer Research Unit, Department of Biology, University of York, Heslington, York YO10 5DD, UK.
| | - Norman J Maitland
- The Cancer Research Unit, Department of Biology, University of York, Heslington, York YO10 5DD, UK.
| |
Collapse
|
26
|
Bo C, Wang J, Zhang H, Cao Y, Lu X, Wang T, Wang Y, Li S, Kong X, Sun X, Liu Z, Ning S, Wang L. Global pathway view analysis of microRNA clusters in myasthenia gravis. Mol Med Rep 2019; 19:2350-2360. [PMID: 30664201 DOI: 10.3892/mmr.2019.9845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 10/25/2018] [Indexed: 11/05/2022] Open
Abstract
The significant roles of microRNAs (miRNAs) in the pathogenesis of myasthenia gravis (MG) have been observed in numerous previous studies. The impact of miRNA clusters on immunity has been demonstrated in previous years; however, the regulation of miRNA clusters in MG remains to be elucidated. In the present study, 245 MG risk genes were collected and 99 MG risk pathways enriched by these genes were identified. A catalog of 126 MG risk miRNAs was then created; the MG risk miRNAs were located on each chromosome and a miRNA cluster was defined as a number of miRNAs with a relative distance of <6 kb on the same sub‑band, same band, same region and same chromosome. Furthermore, enrichment analyses were performed using the target genes of the MG risk miRNA clusters, and a number of risk pathways of each miRNA clusters were identified. As a result, 15 significant miRNA clusters associated with MG were identified. Additionally, the most significant pathways of the miRNA clusters were identified to be enriched on chromosomes 9, 19 and 22, characterized by immunity, infection and carcinoma, suggesting that the mechanism of MG may be associated with certain abnormalities of miRNA clusters on chromosomes 9, 19 and 22. The present study provides novel insight into a global pathway view of miRNA clusters in the pathogenesis of MG.
Collapse
Affiliation(s)
- Chunrui Bo
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jianjian Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Huixue Zhang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yuze Cao
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaoyu Lu
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Tianfeng Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yu Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Shuang Li
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaotong Kong
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xuesong Sun
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Zhaojun Liu
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Shangwei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Lihua Wang
- Department of Neurology, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
27
|
Liu Z, Zhang C, Skamagki M, Khodadadi-Jamayran A, Zhang W, Kong D, Chang CW, Feng J, Han X, Townes TM, Li H, Kim K, Zhao R. Elevated p53 Activities Restrict Differentiation Potential of MicroRNA-Deficient Pluripotent Stem Cells. Stem Cell Reports 2018; 9:1604-1617. [PMID: 29141234 PMCID: PMC5688240 DOI: 10.1016/j.stemcr.2017.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 12/21/2022] Open
Abstract
Pluripotent stem cells (PSCs) deficient for microRNAs (miRNAs), such as Dgcr8−/− or Dicer−/– embryonic stem cells (ESCs), contain no mature miRNA and cannot differentiate into somatic cells. How miRNA deficiency causes differentiation defects remains poorly understood. Here, we report that miR-302 is sufficient to enable neural differentiation of differentiation-incompetent Dgcr8−/− ESCs. Our data showed that miR-302 directly suppresses the tumor suppressor p53, which is modestly upregulated in Dgcr8−/− ESCs and serves as a barrier restricting neural differentiation. We demonstrated that direct inactivation of p53 by SV40 large T antigen, a short hairpin RNA against Trp53, or genetic ablation of Trp53 in Dgcr8−/− PSCs enables neural differentiation, while activation of p53 by the MDM2 inhibitor nutlin-3a in wild-type ESCs inhibits neural differentiation. Together, we demonstrate that a major function of miRNAs in neural differentiation is suppression of p53 and that modest activation of p53 blocks neural differentiation of miRNA-deficient PSCs. miR-302 enables neural differentiation of differentiation-incompetent Dgcr8−/− ESCs miR-302 directly suppresses p53 expression p53 inhibits neural differentiation of Dgcr8−/− and wild-type PSCs p53 may eliminate genetically defective embryos to save maternal resources
Collapse
Affiliation(s)
- Zhong Liu
- Department of Biochemistry and Molecular Genetics, Stem Cell Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Center for Individualized Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Maria Skamagki
- Cancer Biology and Genetics Program, Center for Cell Engineering, Center for Stem Cell Biology, Sloan-Kettering Institute, Cell and Developmental Biology Program, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Alireza Khodadadi-Jamayran
- Department of Biochemistry and Molecular Genetics, Stem Cell Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Wei Zhang
- Department of Biochemistry and Molecular Genetics, Stem Cell Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Dexin Kong
- Department of Biochemistry and Molecular Genetics, Stem Cell Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chia-Wei Chang
- Department of Biochemistry and Molecular Genetics, Stem Cell Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jingyang Feng
- Cook County Health and Hospital System, John H. Stroger Hospital, Chicago, IL 60612, USA
| | - Xiaosi Han
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tim M Townes
- Department of Biochemistry and Molecular Genetics, Stem Cell Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Center for Individualized Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Kitai Kim
- Cancer Biology and Genetics Program, Center for Cell Engineering, Center for Stem Cell Biology, Sloan-Kettering Institute, Cell and Developmental Biology Program, Weill Medical College of Cornell University, New York, NY 10065, USA.
| | - Rui Zhao
- Department of Biochemistry and Molecular Genetics, Stem Cell Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA; Gregory Fleming James Cystic Fibrosis Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
28
|
Ishikawa D, Diekmann U, Fiedler J, Just A, Thum T, Lenzen S, Naujok O. miRNome Profiling of Purified Endoderm and Mesoderm Differentiated from hESCs Reveals Functions of miR-483-3p and miR-1263 for Cell-Fate Decisions. Stem Cell Reports 2018; 9:1588-1603. [PMID: 29141233 PMCID: PMC5688239 DOI: 10.1016/j.stemcr.2017.10.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 10/12/2017] [Accepted: 10/12/2017] [Indexed: 12/20/2022] Open
Abstract
Pluripotent stem cells hold great promise for regenerative medicine since they can differentiate into all somatic cells. MicroRNAs (miRNAs) could be important for the regulation of these cell-fate decisions. Profiling of miRNAs revealed 19 differentially expressed miRNAs in the endoderm and 29 in the mesoderm when analyzing FACS-purified cells derived from human embryonic stem cells. The mesodermal-enriched miR-483-3p was identified as an important regulator for the generation of mesodermal PDGFRA+ paraxial cells. Repression of its target PGAM1 significantly increased the number of PDGFRA+ cells. Furthermore, miR-483-3p, miR-199a-3p, and miR-214-3p might also have functions for the mesodermal progenitors. The endoderm-specific miR-489-3p and miR-1263 accelerated and increased endoderm differentiation upon overexpression. KLF4 was identified as a target of miR-1263. RNAi-mediated downregulation of KLF4 partially mimicked miR-1263 overexpression. Thus, the effects of this miRNA were mediated by facilitating differentiation through destabilization of pluripotency along with other not yet defined targets.
Collapse
Affiliation(s)
- Daichi Ishikawa
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany; Department of Surgery, Tokushima University, 3-18-15, Kuramoto, Tokushima 770-8503, Japan
| | - Ulf Diekmann
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Jan Fiedler
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Annette Just
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany; National Heart and Lung Institute, Imperial College London, Sydney Street, London SW3 6NP, UK
| | - Sigurd Lenzen
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | - Ortwin Naujok
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany.
| |
Collapse
|
29
|
Cusumano P, Biscontin A, Sandrelli F, Mazzotta GM, Tregnago C, De Pittà C, Costa R. Modulation of miR-210 alters phasing of circadian locomotor activity and impairs projections of PDF clock neurons in Drosophila melanogaster. PLoS Genet 2018; 14:e1007500. [PMID: 30011269 PMCID: PMC6062148 DOI: 10.1371/journal.pgen.1007500] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 07/26/2018] [Accepted: 06/19/2018] [Indexed: 01/03/2023] Open
Abstract
Single microRNAs are usually associated with hundreds of putative target genes that can influence multiple phenotypic traits in Drosophila, ranging from development to behaviour. We investigated the function of Drosophila miR-210 in circadian behaviour by misexpressing it within circadian clock cells. Manipulation of miR-210 expression levels in the PDF (pigment dispersing factor) positive neurons affected the phase of locomotor activity, under both light-dark conditions and constant darkness. PER cyclical expression was not affected in clock neurons, however, when miR-210 was up-regulated, a dramatic alteration in the morphology of PDF ventral lateral neuron (LNv) arborisations was observed. The effect of miR-210 in shaping neuronal projections was confirmed in vitro, using a Drosophila neuronal cell line. A transcriptomic analysis revealed that miR-210 overexpression affects the expression of several genes belonging to pathways related to circadian processes, neuronal development, GTPases signal transduction and photoreception. Collectively, these data reveal the role of miR-210 in modulating circadian outputs in flies and guiding/remodelling PDF positive LNv arborisations and indicate that miR-210 may have pleiotropic effects on the clock, light perception and neuronal development. In recent years, the role of microRNAs in regulating the endogenous circadian clock and its rhythmic outputs for behaviour/physiology has been recognized. We have observed that depletion or over-expression of miR-210 in Drosophila melanogaster modulates the phase of locomotor activity, without affecting the molecular oscillation of the pacemaker neurons. Moreover, miR-210 over-expression dramatically alters the pattern of projections from the PDF-positive Lateral Neurons (LNvs). Differentially expressed genes detected in miR-210 over-expressing flies implicated circadian processes, neuronal development, and photoreception. Taken together, our findings indicate the involvement of miR-210 in modulating circadian output and remodelling the projections of PDF clock neurons, and suggest that miR-210 may have pleiotropic effects on clock, light perception and neuronal development.
Collapse
Affiliation(s)
- Paola Cusumano
- Department of Biology, University of Padova, Padova, Italy
| | | | | | | | - Claudia Tregnago
- Department of Women and Children’s Health, University of Padova, Padova, Italy
| | - Cristiano De Pittà
- Department of Biology, University of Padova, Padova, Italy
- * E-mail: (CD); (RC)
| | - Rodolfo Costa
- Department of Biology, University of Padova, Padova, Italy
- * E-mail: (CD); (RC)
| |
Collapse
|
30
|
Splicing factors as regulators of miRNA biogenesis – links to human disease. Semin Cell Dev Biol 2018; 79:113-122. [DOI: 10.1016/j.semcdb.2017.10.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 10/09/2017] [Accepted: 10/09/2017] [Indexed: 12/16/2022]
|
31
|
Li L, Miu KK, Gu S, Cheung HH, Chan WY. Comparison of multi-lineage differentiation of hiPSCs reveals novel miRNAs that regulate lineage specification. Sci Rep 2018; 8:9630. [PMID: 29941943 PMCID: PMC6018499 DOI: 10.1038/s41598-018-27719-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 06/07/2018] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are known to be crucial players in governing the differentiation of human induced pluripotent stem cells (hiPSCs). Despite their utter importance, identifying key lineage specifiers among the myriads of expressed miRNAs remains challenging. We believe that the current practice in mining miRNA specifiers via delineating dynamic fold-changes only is inadequate. Our study, therefore, provides evidence to pronounce "lineage specificity" as another important attribute to qualify for these lineage specifiers. Adopted hiPSCs were differentiated into representative lineages (hepatic, nephric and neuronal) over all three germ layers whilst the depicted miRNA expression changes compiled into an integrated atlas. We demonstrated inter-lineage analysis shall aid in the identification of key miRNAs with lineage-specificity, while these shortlisted candidates were collectively known as "lineage-specific miRNAs". Subsequently, we followed through the fold-changes along differentiation via computational analysis to identify miR-192 and miR-372-3p, respectively, as representative candidate key miRNAs for the hepatic and nephric lineages. Indeed, functional characterization validated that miR-192 and miR-372-3p regulate lineage differentiation via modulation of the expressions of lineage-specific genes. In summary, our presented miRNA atlas is a resourceful ore for the mining of key miRNAs responsible for lineage specification.
Collapse
Affiliation(s)
- Lu Li
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR
- School of Pharmacy, University of Southern California, Los Angeles, CA, USA
| | - Kai-Kei Miu
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR
| | - Shen Gu
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR
- M&H Genetics/Baylor Genetics Laboratories, Baylor College of Medicine, Houston, TX, USA
| | - Hoi-Hung Cheung
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR.
| | - Wai-Yee Chan
- CUHK-CAS GIBH Joint Research Laboratory on Stem Cell and Regenerative Medicine, School of Biomedical Sciences, the Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR.
| |
Collapse
|
32
|
López-Beas J, Capilla-González V, Aguilera Y, Mellado N, Lachaud CC, Martín F, Smani T, Soria B, Hmadcha A. miR-7 Modulates hESC Differentiation into Insulin-Producing Beta-like Cells and Contributes to Cell Maturation. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 12:463-477. [PMID: 30195784 PMCID: PMC6070677 DOI: 10.1016/j.omtn.2018.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 06/08/2018] [Accepted: 06/09/2018] [Indexed: 12/19/2022]
Abstract
Human pluripotent stem cells retain the extraordinary capacity to differentiate into pancreatic beta cells. For this particular lineage, more effort is still required to stress the importance of developing an efficient, reproducible, easy, and cost-effective differentiation protocol to obtain more mature, homogeneous, and functional insulin-secreting cells. In addition, microRNAs (miRNAs) have emerged as a class of small non-coding RNAs that regulate many cellular processes, including pancreatic differentiation. Some miRNAs are known to be preferentially expressed in islets. Of note, miR-375 and miR-7 are two of the most abundant pancreatic miRNAs, and they are necessary for proper pancreatic islet development. Here we provide new insight into specific miRNAs involved in pancreatic differentiation. We found that miR-7 is differentially expressed during the differentiation of human embryonic stem cells (hESCs) into a beta cell-like phenotype and that its modulation plays an important role in generating mature pancreatic beta cells. This strategy may be exploited to optimize the potential for in vitro differentiation of hESCs into insulin-producing beta-like cells for use in preclinical studies and future clinical applications as well as the prospective uses of miRNAs to improve this process.
Collapse
Affiliation(s)
- Javier López-Beas
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Sevilla, Spain
| | - Vivian Capilla-González
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Sevilla, Spain
| | - Yolanda Aguilera
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Sevilla, Spain
| | - Nuria Mellado
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Sevilla, Spain
| | - Christian C Lachaud
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Sevilla, Spain
| | - Franz Martín
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Sevilla, Spain; Centro de Investigación Biomédica en Red sobre Diabetes y Enfermedades Metabólicas-CIBERDEM, Madrid, Spain
| | - Tarik Smani
- Instituto de Biomedicina de Sevilla-IBiS, Universidad de Sevilla/HUVR/Junta de Andalucía/CSIC, Sevilla, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovaculares-CIBERCV, Madrid, Spain
| | - Bernat Soria
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Sevilla, Spain; Centro de Investigación Biomédica en Red sobre Diabetes y Enfermedades Metabólicas-CIBERDEM, Madrid, Spain
| | - Abdelkrim Hmadcha
- Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), University of Pablo de Olavide-University of Seville-CSIC, Sevilla, Spain; Centro de Investigación Biomédica en Red sobre Diabetes y Enfermedades Metabólicas-CIBERDEM, Madrid, Spain.
| |
Collapse
|
33
|
Sherstyuk VV, Medvedev SP, Ri MT, Vyatkin YV, Saik OV, Shtokalo DN, Zakian SM. The search for microRNAs potentially involved in the selfrenewal maintaining of laboratory rat pluripotent stem cells. Vavilovskii Zhurnal Genet Selektsii 2018. [DOI: 10.18699/vj18.345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Self-renewal of cultured pluripotent stem cells is a complex process, which includes multiple functional and regulatory levels. Transcription factors, their target genes, chromatin modifiers, signaling pathways, and regulatory noncoding RNAs are involved in the maintaining of self-renewal. Studies of molecular and genetic bases of maintaining self-renewal and pluripotency in cultured mammalian cells are important to understand processes in preimplantation embryogenesis and to develop efficient techniques to obtain pluripotent stem cell lines for experimental biology and medicine. MicroRNAs (miRNAs) play an important role in pluripotency maintaining and reprogramming. However, involvement of this class of noncoding RNAs and functions of individual molecules are poorly studied. The goal of this study was the search for the miRNAs potentially involved in the pluripotency maintaining and reprogramming of Rattus norvegicus cells. We analyzed the expression of miRNAs in rat embryonic stem cells, induced pluripotent stem cells and embryonic fibroblasts using bioinformatic methods and data obtained with next generation sequencing. The analysis of differential expression between groups of rat pluripotent cells and fibroblasts, and the analysis of experimentally confirmed target genes of differentially expressed known rat miRNAs revealed novel potential players of pluripotency maintaining and reprogramming processes. In addition, novel members of these processes were revealed among novel rat miRNAs. The use of bioinformatic and systems biology approaches is the first step, which is necessary for choosing candidates for the subsequent experimental studies. The results obtained substantially improve our understanding of the self-renewal regulation system of the laboratory rat, a popular biomedical object, and our knowledge about the system in mammals.
Collapse
Affiliation(s)
- V. V. Sherstyuk
- Institute of Cytology and Genetics SB RAS; E.N. Meshalkin National Medical Research Center, Ministry of Health of Russian Federation; Institute of Chemical Biology and Fundamental Medicine SB RAS; Novosibirsk State University
| | - S. P. Medvedev
- Institute of Cytology and Genetics SB RAS; E.N. Meshalkin National Medical Research Center, Ministry of Health of Russian Federation; Institute of Chemical Biology and Fundamental Medicine SB RAS; Novosibirsk State University
| | - M. T. Ri
- AcademGene LLC; St. Laurent Institute
| | - Y. V. Vyatkin
- Institute of Cytology and Genetics SB RAS; Novosibirsk State University; AcademGene LLC; St. Laurent Institute
| | - O. V. Saik
- Institute of Cytology and Genetics SB RAS
| | - D. N. Shtokalo
- Institute of Cytology and Genetics SB RAS; AcademGene LLC; St. Laurent Institute; A.P. Ershov Institute of Informatics Systems SB RAS
| | - S. M. Zakian
- Institute of Cytology and Genetics SB RAS; E.N. Meshalkin National Medical Research Center, Ministry of Health of Russian Federation; Institute of Chemical Biology and Fundamental Medicine SB RAS; Novosibirsk State University
| |
Collapse
|
34
|
Piperigkou Z, Götte M, Theocharis AD, Karamanos NK. Insights into the key roles of epigenetics in matrix macromolecules-associated wound healing. Adv Drug Deliv Rev 2018; 129:16-36. [PMID: 29079535 DOI: 10.1016/j.addr.2017.10.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/14/2017] [Accepted: 10/20/2017] [Indexed: 02/08/2023]
Abstract
Extracellular matrix (ECM) is a dynamic network of macromolecules, playing a regulatory role in cell functions, tissue regeneration and remodeling. Wound healing is a tissue repair process necessary for the maintenance of the functionality of tissues and organs. This highly orchestrated process is divided into four temporally overlapping phases, including hemostasis, inflammation, proliferation and tissue remodeling. The dynamic interplay between ECM and resident cells exerts its critical role in many aspects of wound healing, including cell proliferation, migration, differentiation, survival, matrix degradation and biosynthesis. Several epigenetic regulatory factors, such as the endogenous non-coding microRNAs (miRNAs), are the drivers of the wound healing response. microRNAs have pivotal roles in regulating ECM composition during wound healing and dermal regeneration. Their expression is associated with the distinct phases of wound healing and they serve as target biomarkers and targets for systematic regulation of wound repair. In this article we critically present the importance of epigenetics with particular emphasis on miRNAs regulating ECM components (i.e. glycoproteins, proteoglycans and matrix proteases) that are key players in wound healing. The clinical relevance of miRNA targeting as well as the delivery strategies designed for clinical applications are also presented and discussed.
Collapse
|
35
|
Tu J, Cao D, Li L, Cheung HH, Chan WY. MicroRNA profiling during directed differentiation of cortical interneurons from human-induced pluripotent stem cells. FEBS Open Bio 2018; 8:502-512. [PMID: 29632804 PMCID: PMC5881541 DOI: 10.1002/2211-5463.12377] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/19/2017] [Accepted: 12/23/2017] [Indexed: 01/21/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) are useful for modeling neuron development and related diseases. Cortical interneurons are essential players in neuropsychiatric diseases such as autism. miRNAs are a class of pivotal regulators in neural differentiation. Using a previously established model of cortical interneuron differentiation from human embryonic stem cells, we profiled miRNAs involved in differentiation from human iPSCs. A number of miRNAs were modulated in the differentiation process. This study captured the temporal in vitro neurogenesis from iPSCs to mature cortical interneurons. The specific miRNAs identified at each stage of differentiation are of potential use for drug discovery and prospective clinical applications.
Collapse
Affiliation(s)
- Jiajie Tu
- Ministry of Education Key Laboratory for Regenerative Medicine (CUHK-Jinan University) School of Biomedical Sciences Faculty of Medicine The Chinese University of Hong Kong Hong Kong SAR China.,CUHK-CAS Guangzhou Institute of Biomedicine and Health Joint Laboratory on Stem Cell and Regenerative Medicine School of Biomedical Sciences Faculty of Medicine The Chinese University of Hong Kong Hong Kong SAR China
| | - Dandan Cao
- Ministry of Education Key Laboratory for Regenerative Medicine (CUHK-Jinan University) School of Biomedical Sciences Faculty of Medicine The Chinese University of Hong Kong Hong Kong SAR China
| | - Lu Li
- CUHK-CAS Guangzhou Institute of Biomedicine and Health Joint Laboratory on Stem Cell and Regenerative Medicine School of Biomedical Sciences Faculty of Medicine The Chinese University of Hong Kong Hong Kong SAR China
| | - Hoi-Hung Cheung
- Ministry of Education Key Laboratory for Regenerative Medicine (CUHK-Jinan University) School of Biomedical Sciences Faculty of Medicine The Chinese University of Hong Kong Hong Kong SAR China.,CUHK-CAS Guangzhou Institute of Biomedicine and Health Joint Laboratory on Stem Cell and Regenerative Medicine School of Biomedical Sciences Faculty of Medicine The Chinese University of Hong Kong Hong Kong SAR China
| | - Wai-Yee Chan
- Ministry of Education Key Laboratory for Regenerative Medicine (CUHK-Jinan University) School of Biomedical Sciences Faculty of Medicine The Chinese University of Hong Kong Hong Kong SAR China.,CUHK-CAS Guangzhou Institute of Biomedicine and Health Joint Laboratory on Stem Cell and Regenerative Medicine School of Biomedical Sciences Faculty of Medicine The Chinese University of Hong Kong Hong Kong SAR China
| |
Collapse
|
36
|
miR-302/367-induced neurons reduce behavioral impairment in an experimental model of Alzheimer's disease. Mol Cell Neurosci 2018; 86:50-57. [DOI: 10.1016/j.mcn.2017.11.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 11/03/2017] [Accepted: 11/22/2017] [Indexed: 01/13/2023] Open
|
37
|
Generation of Induced Progenitor-like Cells from Mature Epithelial Cells Using Interrupted Reprogramming. Stem Cell Reports 2017; 9:1780-1795. [PMID: 29198829 PMCID: PMC5785620 DOI: 10.1016/j.stemcr.2017.10.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 10/26/2017] [Accepted: 10/27/2017] [Indexed: 01/17/2023] Open
Abstract
A suitable source of progenitor cells is required to attenuate disease or affect cure. We present an "interrupted reprogramming" strategy to generate "induced progenitor-like (iPL) cells" using carefully timed expression of induced pluripotent stem cell reprogramming factors (Oct4, Sox2, Klf4, and c-Myc; OSKM) from non-proliferative Club cells. Interrupted reprogramming allowed controlled expansion yet preservation of lineage commitment. Under clonogenic conditions, iPL cells expanded and functioned as a bronchiolar progenitor-like population to generate mature Club cells, mucin-producing goblet cells, and cystic fibrosis transmembrane conductance regulator (CFTR)-expressing ciliated epithelium. In vivo, iPL cells can repopulate CFTR-deficient epithelium. This interrupted reprogramming process could be metronomically applied to achieve controlled progenitor-like proliferation. By carefully controlling the duration of expression of OSKM, iPL cells do not become pluripotent, and they maintain their memory of origin and retain their ability to efficiently return to their original phenotype. A generic technique to produce highly specified populations may have significant implications for regenerative medicine.
Collapse
|
38
|
Gadd45a opens up the promoter regions of miR-295 facilitating pluripotency induction. Cell Death Dis 2017; 8:e3107. [PMID: 29022923 PMCID: PMC5682663 DOI: 10.1038/cddis.2017.497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 08/13/2017] [Accepted: 08/31/2017] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) play crucial roles in the establishment of pluripotent state by controlling pluripotent network. However, the molecular mechanisms controlling miRNAs during somatic cell reprogramming remain obscure. In this study, we show Gadd45a (growth arrest and DNA-damage-inducible protein 45a) enhances reprogramming by activating miR-295. Furthermore, we show that Gadd45a binds the promoter regions of miR-295. Nuclease accessibility assay indicates that Gadd45a opens the promoter regions of miR-295. Levels of H3K9Ac and H3K27Ac on the promoter regions of miR-295 were also increased. In conclusion, our results indicate that Gadd45a relaxes the promoter regions of miR-295 and promotes the expression of miR-295 during reprogramming, implying a concise mechanism of Gadd45a and miR-290 cluster cooperation in cell-fate determination.
Collapse
|
39
|
Long noncoding RNA CCAT1 functions as a ceRNA to antagonize the effect of miR-410 on the down-regulation of ITPKB in human HCT-116 and HCT-8 cells. Oncotarget 2017; 8:92855-92863. [PMID: 29190961 PMCID: PMC5696227 DOI: 10.18632/oncotarget.21612] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 08/26/2017] [Indexed: 01/17/2023] Open
Abstract
Colorectal cancer is one of the most common malignancies, which has seriously affected people's health. Abnormal expression of long non-coding RNAs and microRNAs are closely related to the process of occurrence, development, invasion and metastasis of colorectal cancer. However, the effect of lnc CCAT1 on human HCT-116/HCT-8 cells and its potential mechanism were investigated. In present study, differential expression of CCAT1, miR-410 and ITPKB were detected in colon cancer tissues and adjacent parts. Then the prediction programs were applied to predict the target genes of miR-410. The complementary bindings of miR-410 with lnc CCAT1 and ITPKB were assessed by luciferase assays. The interaction between LncRNA CCAT1 and miR-410 was analyzed. In addition, the mRNA and protein of ITPKB and apoptosis factors were examined in cells after miR-410 overexpression or silencing. Meanwhile, MTT and flow cytometer were used to detect the cells proliferation and apoptosis level. Results showed that CCAT1 and miR-410 were up-regulated in colon cancer tissues, but ITPKB was down-regulated. Lnc CCAT1 and ITPKB were predicted to be the targets of miR-410 and the prediction were verified by luciferase assays. The expression of lnc CCAT1 and ITPKB were inhibited by miR-410 in human HCT-116/HCT-8 cells. Meanwhile, lnc CCAT1 could lead to a decrease of miR-410. Furthermore, miR-410 overexpression could promote cell proliferation and reduce apoptosis. In summary, these data demonstrated that miR-410 could promote cell proliferation and reduce apoptosis by inhibiting ITPKB expression and the expression of lnc CCAT1 antagonized the effect of miR-410.
Collapse
|
40
|
Mechanism of human somatic reprogramming to iPS cell. J Transl Med 2017; 97:1152-1157. [PMID: 28530648 DOI: 10.1038/labinvest.2017.56] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/10/2017] [Accepted: 04/17/2017] [Indexed: 01/09/2023] Open
Abstract
Somatic reprogramming to induced pluripotent stem cells (iPSC) was realized in the year 2006 in mice, and in 2007 in humans, by transiently forced expression of a combination of exogenous transcription factors. Human and mouse iPSCs are distinctly reprogrammed into a 'primed' and a 'naïve' state, respectively. In the last decade, puzzle pieces of somatic reprogramming have been collected with difficulty. Collectively, dissecting reprogramming events and identification of the hallmark of sequentially activated/silenced genes have revealed mouse somatic reprogramming in fragments, but there is a long way to go toward understanding the molecular mechanisms of human somatic reprogramming, even with developing technologies. Recently, an established human intermediately reprogrammed stem cell (iRSC) line, which has paused reprogramming at the endogenous OCT4-negative/exogenous transgene-positive pre-MET (mesenchymal-to-epithelial-transition) stage can resume reprogramming into endogenous OCT4-positive iPSCs only by change of culture conditions. Genome-editing-mediated visualization of endogenous OCT4 activity with GFP in living iRSCs demonstrates the timing of OCT4 activation and entry to MET in the reprogramming toward iPSCs. Applications of genome-editing technology to pluripotent stem cells will reshape our approaches for exploring molecular mechanisms.
Collapse
|
41
|
Abstract
Epithelial-mesenchymal interactions are required to coordinate cell proliferation, patterning, and functional differentiation of multiple cell types in a developing organ. This exquisite coordination is dependent on various secreted molecules that provide developmental signals to mediate these tissue interactions. Recently, it was reported that mature mesenchymal-derived microRNAs (miRNAs) in the fetal mouse salivary gland are loaded into exosomes, and transported to the epithelium where they influence progenitor cell proliferation. The exosomal miRNAs regulated epithelial expression of genes involved in DNA methylation in progenitor cells to influence morphogenesis. Thus, exosomal miRNAs are mobile genetic signals that cross tissue boundaries within an organ. These findings raise many questions about how miRNA signals are initiated to coordinate organogenesis and whether they are master regulators of epithelial-mesenchymal interactions. The development of therapeutic applications using exosomal miRNAs for the regeneration of damaged adult organs is a promising area of research.
Collapse
Affiliation(s)
- Toru Hayashi
- a Department of Anatomical Science , Kitasato University School of Allied Health Sciences , Sagamihara , Kanagawa , Japan
| | - Matthew P Hoffman
- b Matrix and Morphogenesis Section , National Institute of Dental and Craniofacial Research, National Institutes of Health, DHHS , Bethesda , Maryland , USA
| |
Collapse
|
42
|
Nguyen PNN, Choo KB, Huang CJ, Sugii S, Cheong SK, Kamarul T. miR-524-5p of the primate-specific C19MC miRNA cluster targets TP53IPN1- and EMT-associated genes to regulate cellular reprogramming. Stem Cell Res Ther 2017; 8:214. [PMID: 28962647 PMCID: PMC5622517 DOI: 10.1186/s13287-017-0666-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 08/29/2017] [Accepted: 09/12/2017] [Indexed: 12/26/2022] Open
Abstract
Background Introduction of the transcription factors Oct4, Sox2, Klf4, and c-Myc (OSKM) is able to ‘reprogram’ somatic cells to become induced pluripotent stem cells (iPSCs). Several microRNAs (miRNAs) are known to enhance reprogramming efficiency when co-expressed with the OSKM factors. The primate-specific chromosome 19 miRNA cluster (C19MC) is essential in primate reproduction, development, and differentiation. miR-524-5p, a C19MC member, is highly homologous to the reprogramming miR-520d-5p; we also reported that miR-524-5p was expressed in iPSCs but not mesenchymal stem cells (MSCs). This study aimed to elucidate possible contributions of miR-524-5p to the reprogramming process. Methods A miR-524-5p precursor was introduced into human fibroblast HFF-1 in the presence of OSKM, and the relative number of embryonic stem cell (ESC)-like colonies that stained positively with alkaline phosphatase (AP) and Nanog were quantified to determine reprogramming efficiency. A miR-524-5p mimic was transfected to MSCs to investigate the effects of miR-524-5p on TP53INP1, ZEB2, and SMAD4 expression by real-time polymerase chain reaction (PCR) and Western blot. Direct gene targeting was confirmed by luciferase activity. A phylogenetic tree of TP53INP1 was constructed by the Clustal method. Contribution of miR-524-5p to cell proliferation and apoptosis was examined by cell counts, BrdU, MTT, and cell death assays, and pluripotency gene expression by real-time PCR. Results Co-expressing the miR-524 precursor with OSKM resulted in a two-fold significant increase in the number of AP- and Nanog-positive ESC-like colonies, indicating a role for miR-524-5p in reprogramming. The putative target, TP53INP1, showed an inverse expression relationship with miR-524-5p; direct TP53INP1 targeting was confirmed in luciferase assays. miR-524-5p-induced TP53INP1 downregulation enhanced cell proliferation, suppressed apoptosis, and upregulated the expression of pluripotency genes, all of which are critical early events of the reprogramming process. Interestingly, the TP53INP1 gene may have co-evolved late with the primate-specific miR-524-5p. miR-524-5p also promoted mesenchymal-to-epithelial transition (MET), a required initial event of reprogramming, by directly targeting the epithelial-to-mesenchymal transition (EMT)-related genes, ZEB2 and SMAD4. Conclusions Via targeting TP53INP1, ZEB2, and SMAD4, miR-524-5p contributes to the early stage of inducing pluripotency by promoting cell proliferation, inhibiting apoptosis, upregulating expression of pluripotency genes, and enhancing MET. Other C19MC miRNAs may have similar reprogramming functions. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0666-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Phan Nguyen Nhi Nguyen
- Centre for Stem Cell Research, Universiti Tunku Abdul Rahman, Sungai Long, Kajang, Selangor DE, Malaysia.,Department of Preclinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Bandar Sungai Long, Cheras, 43000, Kajang, Selangor DE, Malaysia
| | - Kong Bung Choo
- Centre for Stem Cell Research, Universiti Tunku Abdul Rahman, Sungai Long, Kajang, Selangor DE, Malaysia. .,Department of Preclinical Sciences, Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Bandar Sungai Long, Cheras, 43000, Kajang, Selangor DE, Malaysia.
| | - Chiu-Jung Huang
- Department of Animal Science, Chinese Culture University, Taipei, Taiwan.,Graduate Institute of Biotechnology, Chinese Culture University, Taipei, Taiwan
| | - Shigeki Sugii
- Singapore BioImaging Consortium A*Star, Singapore, Singapore.,Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Soon Keng Cheong
- Centre for Stem Cell Research, Universiti Tunku Abdul Rahman, Sungai Long, Kajang, Selangor DE, Malaysia.,Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long, Kajang, Selangor DE, Malaysia
| | - Tunku Kamarul
- Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning, Kuala Lumpur, Malaysia.,Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
43
|
Farzaneh M, Attari F, Khoshnam SE. Concise Review: LIN28/let-7 Signaling, a Critical Double-Negative Feedback Loop During Pluripotency, Reprogramming, and Tumorigenicity. Cell Reprogram 2017; 19:289-293. [PMID: 28846452 DOI: 10.1089/cell.2017.0015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
MicroRNAs (miRNAs) with 20-30 nucleotides have recently emerged as the multidimensional regulators of cell fate decisions. Recent improvement in high-throughput sequencing has highlighted the potential role of LIN28/let-7 regulatory network in several developmental events. It was proposed that this pathway might represent a functional signature in cell proliferation, transition between commitment and pluripotency, and regulation of cancer and tumorigenicity. LIN28/let-7 regulatory pathway is one of the excellent examples of the relationship between an miRNA and mRNAs. This review article highlights the potentials of LIN28/let-7 signaling in gene regulatory pathways during pluripotency, reprogramming, and tumorigenicity.
Collapse
Affiliation(s)
- Maryam Farzaneh
- 1 Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology , ACECR, Tehran, Iran
| | - Farnoosh Attari
- 2 Department of Animal Biology, School of Biology, College of Science, University of Tehran , Tehran, Iran
| | - Seyed Esmaeil Khoshnam
- 3 Department of Physiology, Faculty of Medicine, Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences , Ahvaz, Iran .,4 Student Research Committee, Ahvaz Jundishapur University of Medical Sciences , Ahvaz, Iran
| |
Collapse
|
44
|
Wang H, Xing D, Ren D, Feng W, Chen Y, Zhao Z, Xiao Z, Peng Z. MicroRNA‑643 regulates the expression of ZEB1 and inhibits tumorigenesis in osteosarcoma. Mol Med Rep 2017; 16:5157-5164. [PMID: 28849077 PMCID: PMC5647050 DOI: 10.3892/mmr.2017.7273] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 05/08/2017] [Indexed: 12/15/2022] Open
Abstract
Osteosarcoma is among the most malignant types of tumor worldwide and has become a leading contributor to tumor incidence, particularly in adolescents. Resistance to conventional treatment and the complexity of osteosarcoma tumorigenesis has resulted in high mortality rates. MicroRNAs are a class of noncoding RNAs, which regulate numerous biological processes. However, the involvement of miR‑643 in osteosarcoma remains to be elucidated. In the present study, reverse transcription‑quantitative polymerase chain reaction, luciferase reporter assay, invasion assay, viability assay, western blot analysis and in vivo implantation were performed to analyze the action of miR‑643 in osteosarcoma. The results demonstrated that miR‑643 inhibited the progression of osteosarcoma and acted as a potential tumor suppressor. The expression of miR‑643 was downregulated in osteosarcoma tissues and cell lines. In addition, miR‑643 transfection significantly impaired the proliferation and invasion of osteosarcoma cells. The present study also identified Zinc finger E‑box‑binding homeobox 1 (ZEB1) as a direct target of miR‑643, and the ectopic expression of ZEB1 counteracted the effect of miR‑643 transfection. A significant inverse correlation was also found between the expression of miR‑643 and ZEB1. A low expression of miR‑643 or a high expression of ZEB1 was associated with poor patient survival rates. The results of the present study suggested that the decreased expression of miR‑643 may be involved in the mechanism underlying the development of osteosarcoma. The intricate interactions between miR‑643 and ZEB1 may serve as a potential therapeutic target in osteosarcoma oncogenesis.
Collapse
Affiliation(s)
- Huan Wang
- Department of Hand Surgery One, The Puai Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Danmou Xing
- Department of Hand Surgery One, The Puai Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Dong Ren
- Department of Hand Surgery One, The Puai Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Wei Feng
- Department of Hand Surgery One, The Puai Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yan Chen
- Department of Hand Surgery One, The Puai Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhiming Zhao
- Department of Hand Surgery One, The Puai Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhihong Xiao
- Department of Hand Surgery One, The Puai Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Zhengren Peng
- Department of Hand Surgery One, The Puai Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|
45
|
Faiola F, Yin N, Fidalgo M, Huang X, Saunders A, Ding J, Guallar D, Dang B, Wang J. NAC1 Regulates Somatic Cell Reprogramming by Controlling Zeb1 and E-cadherin Expression. Stem Cell Reports 2017; 9:913-926. [PMID: 28781078 PMCID: PMC5599184 DOI: 10.1016/j.stemcr.2017.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 07/02/2017] [Accepted: 07/03/2017] [Indexed: 12/11/2022] Open
Abstract
Reprogramming somatic cells to induced pluripotent stem cells (iPSCs) is a long and inefficient process. A thorough understanding of the molecular mechanisms underlying reprogramming is paramount for efficient generation and safe application of iPSCs in medicine. While intensive efforts have been devoted to identifying reprogramming facilitators and barriers, a full repertoire of such factors, as well as their mechanistic actions, is poorly defined. Here, we report that NAC1, a pluripotency-associated factor and NANOG partner, is required for establishment of pluripotency during reprogramming. Mechanistically, NAC1 is essential for proper expression of E-cadherin by a dual regulatory mechanism: it facilitates NANOG binding to the E-cadherin promoter and fine-tunes its expression; most importantly, it downregulates the E-cadherin repressor ZEB1 directly via transcriptional repression and indirectly via post-transcriptional activation of the miR-200 miRNAs. Our study thus uncovers a previously unappreciated role for the pluripotency regulator NAC1 in promoting efficient somatic cell reprogramming. NAC1 is critical for efficient iPSC generation NAC1 facilitates NANOG binding to E-cadherin promoter NAC1 binds to Zeb1 promoter and represses its expression NAC1 binds to the miR-200 loci and indirectly activates E-cadherin expression
Collapse
Affiliation(s)
- Francesco Faiola
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Nuoya Yin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Miguel Fidalgo
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xin Huang
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Arven Saunders
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Junjun Ding
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Diana Guallar
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Baoyen Dang
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jianlong Wang
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
46
|
Tian L, Guo Z, Wang H, Liu X. MicroRNA-635 inhibits the malignancy of osteosarcoma by inducing apoptosis. Mol Med Rep 2017; 16:4829-4834. [DOI: 10.3892/mmr.2017.7127] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 04/07/2017] [Indexed: 11/05/2022] Open
|
47
|
Potassium as a pluripotency-associated element identified through inorganic element profiling in human pluripotent stem cells. Sci Rep 2017; 7:5005. [PMID: 28694442 PMCID: PMC5504050 DOI: 10.1038/s41598-017-05117-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/24/2017] [Indexed: 12/20/2022] Open
Abstract
Despite their well-known function in maintaining normal cell physiology, how inorganic elements are relevant to cellular pluripotency and differentiation in human pluripotent stem cells (hPSCs) has yet to be systematically explored. Using total reflection X-ray fluorescence (TXRF) spectrometry and inductively coupled plasma mass spectrometry (ICP-MS), we analyzed the inorganic components of human cells with isogenic backgrounds in distinct states of cellular pluripotency. The elemental profiles revealed that the potassium content of human cells significantly differs when their cellular pluripotency changes. Pharmacological treatment that alters cell membrane permeability to potassium affected the maintenance and establishment of cellular pluripotency via multiple mechanisms in bona fide hPSCs and reprogrammed cells. Collectively, we report that potassium is a pluripotency-associated inorganic element in human cells and provide novel insights into the manipulation of cellular pluripotency in hPSCs by regulating intracellular potassium.
Collapse
|
48
|
Song X, Xie Y, Liu Y, Shao M, Yang W. MicroRNA-492 overexpression exerts suppressive effects on the progression of osteosarcoma by targeting PAK7. Int J Mol Med 2017; 40:891-897. [DOI: 10.3892/ijmm.2017.3046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/24/2017] [Indexed: 11/06/2022] Open
|
49
|
Sherstyuk VV, Medvedev SP, Elisaphenko EA, Vaskova EA, Ri MT, Vyatkin YV, Saik OV, Shtokalo DN, Pokushalov EA, Zakian SM. Genome-wide profiling and differential expression of microRNA in rat pluripotent stem cells. Sci Rep 2017; 7:2787. [PMID: 28584262 PMCID: PMC5459850 DOI: 10.1038/s41598-017-02632-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 04/13/2017] [Indexed: 01/15/2023] Open
Abstract
MicroRNAs (miRNAs) constitute a class of small noncoding RNAs that plays an important role in the post-transcriptional regulation of gene expression. Much evidence has demonstrated that miRNAs are involved in regulating the human and mouse pluripotency. Nevertheless, to our knowledge, miRNAs in the pluripotent stem cells of one of the most commonly used model organisms - the Rattus norvegicus have not been studied. In the present study, we performed deep sequencing of small RNA molecules in the embryonic fibroblasts, embryonic stem cells, and induced pluripotent stem cells of laboratory rats. Bioinformatics analysis revealed 674 known miRNAs and 394 novel miRNA candidates in all of the samples. Expression of known pluripotency-associated miRNAs, such as the miR-290-295 and miR-183-96-182 clusters as well as members of the miR-200 family, was detected in rat pluripotent stem cells. Analysis of the targets of differentially expressed known and novel miRNAs showed their involvement in the regulation of pluripotency and the reprogramming process in rats. Bioinformatics and systems biology approaches identified potential pathways that are regulated by these miRNAs. This study contributes to our understanding of miRNAs in the regulation of pluripotency and cell reprogramming in the laboratory rat.
Collapse
Affiliation(s)
- Vladimir V Sherstyuk
- Federal Research Center Institute of Cytology and Genetics, the Siberian Branch of the Russian Academy of Sciences, 10 Lavrentyeva Ave., Novosibirsk, 630090, Russia.,Siberian Federal Biomedical Research Center, Ministry of Healthcare of the Russian Federation, 15 Rechkunovskaya St., Novosibirsk, 630055, Russia.,Institute of Chemical Biology and Fundamental Medicine, the Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., Novosibirsk, 630090, Russia.,Novosibirsk State University, 2 Pirogova St., Novosibirsk, 630090, Russia
| | - Sergey P Medvedev
- Federal Research Center Institute of Cytology and Genetics, the Siberian Branch of the Russian Academy of Sciences, 10 Lavrentyeva Ave., Novosibirsk, 630090, Russia.,Siberian Federal Biomedical Research Center, Ministry of Healthcare of the Russian Federation, 15 Rechkunovskaya St., Novosibirsk, 630055, Russia.,Institute of Chemical Biology and Fundamental Medicine, the Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., Novosibirsk, 630090, Russia.,Novosibirsk State University, 2 Pirogova St., Novosibirsk, 630090, Russia
| | - Evgeniy A Elisaphenko
- Federal Research Center Institute of Cytology and Genetics, the Siberian Branch of the Russian Academy of Sciences, 10 Lavrentyeva Ave., Novosibirsk, 630090, Russia.,Siberian Federal Biomedical Research Center, Ministry of Healthcare of the Russian Federation, 15 Rechkunovskaya St., Novosibirsk, 630055, Russia.,Institute of Chemical Biology and Fundamental Medicine, the Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., Novosibirsk, 630090, Russia
| | - Evgeniya A Vaskova
- Federal Research Center Institute of Cytology and Genetics, the Siberian Branch of the Russian Academy of Sciences, 10 Lavrentyeva Ave., Novosibirsk, 630090, Russia.,Siberian Federal Biomedical Research Center, Ministry of Healthcare of the Russian Federation, 15 Rechkunovskaya St., Novosibirsk, 630055, Russia.,Institute of Chemical Biology and Fundamental Medicine, the Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., Novosibirsk, 630090, Russia
| | - Maxim T Ri
- AcademGene LLC, 6 Lavrentyeva Ave., Novosibirsk, 630090, Russia.,St. Laurent Institute, 317 New Boston St., Woburn, MA, 01801, USA
| | - Yuri V Vyatkin
- Federal Research Center Institute of Cytology and Genetics, the Siberian Branch of the Russian Academy of Sciences, 10 Lavrentyeva Ave., Novosibirsk, 630090, Russia.,Novosibirsk State University, 2 Pirogova St., Novosibirsk, 630090, Russia.,AcademGene LLC, 6 Lavrentyeva Ave., Novosibirsk, 630090, Russia.,St. Laurent Institute, 317 New Boston St., Woburn, MA, 01801, USA
| | - Olga V Saik
- Federal Research Center Institute of Cytology and Genetics, the Siberian Branch of the Russian Academy of Sciences, 10 Lavrentyeva Ave., Novosibirsk, 630090, Russia
| | - Dmitry N Shtokalo
- Federal Research Center Institute of Cytology and Genetics, the Siberian Branch of the Russian Academy of Sciences, 10 Lavrentyeva Ave., Novosibirsk, 630090, Russia.,AcademGene LLC, 6 Lavrentyeva Ave., Novosibirsk, 630090, Russia.,St. Laurent Institute, 317 New Boston St., Woburn, MA, 01801, USA.,A.P.Ershov Institute of Informatics Systems, 6 Lavrentyeva Ave., Novosibirsk, 630090, Russia
| | - Evgeniy A Pokushalov
- Siberian Federal Biomedical Research Center, Ministry of Healthcare of the Russian Federation, 15 Rechkunovskaya St., Novosibirsk, 630055, Russia
| | - Suren M Zakian
- Federal Research Center Institute of Cytology and Genetics, the Siberian Branch of the Russian Academy of Sciences, 10 Lavrentyeva Ave., Novosibirsk, 630090, Russia. .,Siberian Federal Biomedical Research Center, Ministry of Healthcare of the Russian Federation, 15 Rechkunovskaya St., Novosibirsk, 630055, Russia. .,Institute of Chemical Biology and Fundamental Medicine, the Siberian Branch of the Russian Academy of Sciences, 8 Lavrentyeva Ave., Novosibirsk, 630090, Russia. .,Novosibirsk State University, 2 Pirogova St., Novosibirsk, 630090, Russia.
| |
Collapse
|
50
|
Cao Y, Wu T, Li D, Hu J, Lu H. MicroRNA‑336 directly targets Sox‑2 in osteosarcoma to inhibit tumorigenesis. Mol Med Rep 2017; 15:4217-4224. [PMID: 28440454 DOI: 10.3892/mmr.2017.6493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 02/02/2017] [Indexed: 11/06/2022] Open
Abstract
Previous evidence has suggested that microRNAs (miRNAs or miRs), which belong to a class of non‑coding RNAs, shape cellular processes by regulating gene expression. Abnormal expression of miRNAs has been associated with tumorigenesis in multiple cancers. However, the function of miR‑336 in osteosarcoma (OS) remains unknown. The experimental procedures used in the present study included flow cytometry, reverse transcription‑quantitative polymerase chain reaction, luciferase reporter assay, invasion assay, western blot analysis and in vivo implantation. The results of the present study demonstrated that miR‑336 may serve as a tumor suppressor in OS. Downregulation of miR‑336 was observed in human OS specimens as well as OS cell lines. In addition, a significant negative correlation between sex determining region Y‑box 2 (Sox‑2) expression and miR‑336 was demonstrated. miR‑336 was confirmed to target the 3'‑untranslated region of Sox‑2 to inhibit proliferation, migration and invasion of OS cells. Consistently, restoration of Sox‑2 expression counteracted the effect of miR‑336, and recovered the tumorigenic potential of OS cells. The present study established a novel association between miR‑336 and Sox‑2 in OS. This relationship between miR‑336 and Sox‑2 may lead to improved knowledge concerning OS progression and sheds light on potential novel therapeutic interventions for OS treatment.
Collapse
Affiliation(s)
- Yong Cao
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Tianding Wu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Dongzhe Li
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jianzhong Hu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Hongbin Lu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|