1
|
Sun J, Teng M, Zhu W, Zhao X, Zhao L, Li Y, Zhang Z, Liu Y, Bi S, Wu F. MicroRNA and Gut Microbiota Alter Intergenerational Effects of Paternal Exposure to Polyethylene Nanoplastics. ACS NANO 2024; 18:18085-18100. [PMID: 38935618 DOI: 10.1021/acsnano.4c06298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Nanoplastics (NPs), as emerging contaminants, have been shown to cause testicular disorders in mammals. However, whether paternal inheritance effects on offspring health are involved in NP-induced reproductive toxicity remains unclear. In this study, we developed a mouse model where male mice were administered 200 nm polyethylene nanoparticles (PE-NPs) at a concentration of 2 mg/L through daily gavage for 35 days to evaluate the intergenerational effects of PE-NPs in an exclusive male-lineage transmission paradigm. We observed that paternal exposure to PE-NPs significantly affected growth phenotypes and sex hormone levels and induced histological damage in the testicular tissue of both F0 and F1 generations. In addition, consistent changes in sperm count, motility, abnormalities, and gene expression related to endoplasmic reticulum stress, sex hormone synthesis, and spermatogenesis were observed across paternal generations. The upregulation of microRNA (miR)-1983 and the downregulation of miR-122-5p, miR-5100, and miR-6240 were observed in both F0 and F1 mice, which may have been influenced by reproductive signaling pathways, as indicated by the RNA sequencing of testis tissues and quantitative real-time polymerase chain reaction findings. Furthermore, alterations in the gut microbiota and subsequent Spearman correlation analysis revealed that an increased abundance of Desulfovibrio (C21_c20) and Ruminococcus (gnavus) and a decreased abundance of Allobaculum were positively associated with spermatogenic dysfunction. These findings were validated in a fecal microbiota transplantation trial. Our results demonstrate that changes in miRNAs and the gut microbiota caused by paternal exposure to PE-NPs mediated intergenerational effects, providing deeper insights into mechanisms underlying the impact of paternal inheritance.
Collapse
Affiliation(s)
- Jiaqi Sun
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Miaomiao Teng
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Wentao Zhu
- Innovation Center of Pesticide Research, Department of Applied Chemistry, College of Science, China Agricultural University, Beijing 100193, China
| | - Xiaoli Zhao
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Lihui Zhao
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Yunxia Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Zixuan Zhang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Yunjie Liu
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agricultural, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Sheng Bi
- Department of Psychiatry and Behavioral Sciences, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Fengchang Wu
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| |
Collapse
|
2
|
Liao WL, Liu YF, Ying TH, Shieh JC, Hung YT, Lee HJ, Shen CY, Cheng CW. Inhibitory Effects of Ursolic Acid on the Stemness and Progression of Human Breast Cancer Cells by Modulating Argonaute-2. Int J Mol Sci 2022; 24:ijms24010366. [PMID: 36613808 PMCID: PMC9820512 DOI: 10.3390/ijms24010366] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
The stemness and metastasis of cancer cells are crucial features in determining cancer progression. Argonaute-2 (AGO2) overexpression was reported to be associated with microRNA (miRNA) biogenesis, supporting the self-renewal and differentiation characteristics of cancer stem cells (CSCs). Ursolic acid (UA), a triterpene compound, has multiple biological functions, including anticancer activity. In this study, we find that UA inhibits the proliferation of MDA-MB-231 and MCF-7 breast cancer cell lines using the CCK-8 assay. UA induced a significant decrease in the fraction of CSC in which it was examined by changes in the expression of stemness biomarkers, including the Nanog and Oct4 genes. UA altered invasion and migration capacities by significant decreases in the levels of epithelial-to-mesenchymal transition (EMT) proteins of slug and vimentin. Furthermore, the co-reduction in oncogenic miRNA levels (miR-9 and miR-221) was a result of the down-modulation in AGO2 in breast cancer cells in vitro. Mechanically, UA increases PTEN expression to inactivate the FAK/PI3K/Akt/mTOR signaling pathway and the decreased level of c-Myc in quantitative RT-PCR and Western blot imaging analyses. Our current understanding of the anticancer potential of UA in interrupting between EMT programming and the state of CSC suggests that UA can contribute to improvements in the clinical practice of breast cancer.
Collapse
Affiliation(s)
- Wen-Ling Liao
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 40433, Taiwan
- Center for Personalized Medicine, China Medical University Hospital, Taichung 40433, Taiwan
| | - Yu-Fan Liu
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Tsung-Ho Ying
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Jia-Ching Shieh
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Yueh-Tzu Hung
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Huei-Jane Lee
- Department of Biochemistry, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Chen-Yang Shen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Environmental Science, China Medical University, Taichung 40433, Taiwan
- Correspondence: (C.-Y.S.); (C.-W.C.); Tel.: +886-2-2789-9036 (C.-Y.S.); +886-4-2473-0022 (ext. 11677) (C.-W.C.); Fax: +886-2-2782-3047 (C.-Y.S.); +886-4-2372-3229 (C.-W.C.)
| | - Chun-Wen Cheng
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Correspondence: (C.-Y.S.); (C.-W.C.); Tel.: +886-2-2789-9036 (C.-Y.S.); +886-4-2473-0022 (ext. 11677) (C.-W.C.); Fax: +886-2-2782-3047 (C.-Y.S.); +886-4-2372-3229 (C.-W.C.)
| |
Collapse
|
3
|
Mazloom AR, Xu H, Reig-Palou J, Vasileva A, Román AC, Mulero-Navarro S, Lemischka IR, Sevilla A. Esrrb Regulates Specific Feed-Forward Loops to Transit From Pluripotency Into Early Stages of Differentiation. Front Cell Dev Biol 2022; 10:820255. [PMID: 35652095 PMCID: PMC9149258 DOI: 10.3389/fcell.2022.820255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/24/2022] [Indexed: 01/15/2023] Open
Abstract
Characterization of pluripotent states, in which cells can both self-renew or differentiate, with the irreversible loss of pluripotency, are important research areas in developmental biology. Although microRNAs (miRNAs) have been shown to play a relevant role in cellular differentiation, the role of miRNAs integrated into gene regulatory networks and its dynamic changes during these early stages of embryonic stem cell (ESC) differentiation remain elusive. Here we describe the dynamic transcriptional regulatory circuitry of stem cells that incorporate protein-coding and miRNA genes based on miRNA array expression and quantitative sequencing of short transcripts upon the downregulation of the Estrogen Related Receptor Beta (Esrrb). The data reveals how Esrrb, a key stem cell transcription factor, regulates a specific stem cell miRNA expression program and integrates dynamic changes of feed-forward loops contributing to the early stages of cell differentiation upon its downregulation. Together these findings provide new insights on the architecture of the combined transcriptional post-transcriptional regulatory network in embryonic stem cells.
Collapse
Affiliation(s)
- Amin R. Mazloom
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Huilei Xu
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jaume Reig-Palou
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Ana Vasileva
- Center for Radiological Research, Columbia University, New York, NY, United States
| | - Angel-Carlos Román
- Department of Biochemistry, Molecular Biology and Genetics, University of Extremadura, Badajoz, Spain
| | - Sonia Mulero-Navarro
- Department of Biochemistry, Molecular Biology and Genetics, University of Extremadura, Badajoz, Spain
| | - Ihor R. Lemischka
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ana Sevilla
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Institute of Biomedicine of the University of Barcelona (IBUB), Barcelona, Spain
- *Correspondence: Ana Sevilla,
| |
Collapse
|
4
|
Thiagalingam S. Epigenetic memory in development and disease: Unraveling the mechanism. Biochim Biophys Acta Rev Cancer 2020; 1873:188349. [PMID: 31982403 DOI: 10.1016/j.bbcan.2020.188349] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 01/14/2023]
Abstract
Epigenetic memory is an essential process of life that governs the inheritance of predestined functional characteristics of normal cells and the newly acquired properties of cells affected by cancer and other diseases from parental to progeny cells. Unraveling the molecular basis of epigenetic memory dictated by protein and RNA factors in conjunction with epigenetic marks that are erased and re-established during embryogenesis/development during the formation of somatic, stem and disease cells will have far reaching implications to our understanding of embryogenesis/development and various diseases including cancer. While there has been enormous progress made, there are still gaps in knowledge which includes, the identity of unique epigenetic memory factors (EMFs) and epigenome coding enzymes/co-factors/scaffolding proteins involved in the assembly of defined "epigenetic memorysomes" and the epigenome marks that constitute collections of gene specific epigenetic memories corresponding to specific cell types and physiological conditions. A better understanding of the molecular basis for epigenetic memory will play a central role in improving diagnostics and prognostics of disease states and aid the development of targeted therapeutics of complex diseases.
Collapse
Affiliation(s)
- Sam Thiagalingam
- Department of Medicine (Biomedical Genetics Section and Cancer Center), Department of Pathology & Laboratory Medicine, Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, United States of America.
| |
Collapse
|
5
|
Borden A, Kurian J, Nickoloff E, Yang Y, Troupes CD, Ibetti J, Lucchese AM, Gao E, Mohsin S, Koch WJ, Houser SR, Kishore R, Khan M. Transient Introduction of miR-294 in the Heart Promotes Cardiomyocyte Cell Cycle Reentry After Injury. Circ Res 2019; 125:14-25. [PMID: 30964391 PMCID: PMC6586499 DOI: 10.1161/circresaha.118.314223] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 04/05/2019] [Indexed: 02/06/2023]
Abstract
RATIONALE Embryonic heart is characterized of rapidly dividing cardiomyocytes required to build a working myocardium. Cardiomyocytes retain some proliferative capacity in the neonates but lose it in adulthood. Consequently, a number of signaling hubs including microRNAs are altered during cardiac development that adversely impacts regenerative potential of cardiac tissue. Embryonic stem cell cycle miRs are a class of microRNAs exclusively expressed during developmental stages; however, their effect on cardiomyocyte proliferation and heart function in adult myocardium has not been studied previously. OBJECTIVE To determine whether transient reintroduction of embryonic stem cell cycle miR-294 promotes cardiomyocyte cell cycle reentry enhancing cardiac repair after myocardial injury. METHODS AND RESULTS miR-294 is expressed in the heart during development, prenatal stages, lost in the neonate, and adult heart confirmed by qRT-PCR and in situ hybridization. Neonatal ventricular myocytes treated with miR-294 showed elevated expression of Ki67, p-histone H3, and Aurora B confirmed by immunocytochemistry compared with control cells. miR-294 enhanced oxidative phosphorylation and glycolysis in Neonatal ventricular myocytes measured by seahorse assay. Mechanistically, miR-294 represses Wee1 leading to increased activity of the cyclin B1/CDK1 complex confirmed by qRT-PCR and immunoblot analysis. Next, a doxycycline-inducible AAV9-miR-294 vector was delivered to mice for activating miR-294 in myocytes for 14 days continuously after myocardial infarction. miR-294-treated mice significantly improved left ventricular functions together with decreased infarct size and apoptosis 8 weeks after MI. Myocyte cell cycle reentry increased in miR-294 hearts analyzed by Ki67, pH3, and AurB (Aurora B kinase) expression parallel to increased small myocyte number in the heart. Isolated adult myocytes from miR-294 hearts showed increased 5-ethynyl-2'-deoxyuridine+ cells and upregulation of cell cycle markers and miR-294 targets 8 weeks after MI. CONCLUSIONS Ectopic transient expression of miR-294 recapitulates developmental signaling and phenotype in cardiomyocytes promoting cell cycle reentry that leads to augmented cardiac function in mice after myocardial infarction.
Collapse
Affiliation(s)
- Austin Borden
- Center for Metabolic Disease Research (CMDR), Temple University
| | - Justin Kurian
- Center for Metabolic Disease Research (CMDR), Temple University
| | - Emily Nickoloff
- Center for Translational Medicine (CTM), LKSOM, LKSOM, Temple University
| | - Yijun Yang
- Cardiovascular Research Institute (CVRC), Temple University
| | | | - Jessica Ibetti
- Center for Translational Medicine (CTM), LKSOM, LKSOM, Temple University
| | | | - Erhe Gao
- Center for Translational Medicine (CTM), LKSOM, LKSOM, Temple University
| | - Sadia Mohsin
- Cardiovascular Research Institute (CVRC), Temple University
- Department of Pharmacology, LKSOM, Temple University, LKSOM, Temple University
| | - Walter J Koch
- Center for Translational Medicine (CTM), LKSOM, LKSOM, Temple University
- Department of Pharmacology, LKSOM, Temple University, LKSOM, Temple University
| | - Steven R Houser
- Cardiovascular Research Institute (CVRC), Temple University
- Department of Physiology, LKSOM, Temple University
| | - Raj Kishore
- Center for Translational Medicine (CTM), LKSOM, LKSOM, Temple University
- Department of Pharmacology, LKSOM, Temple University, LKSOM, Temple University
| | - Mohsin Khan
- Center for Metabolic Disease Research (CMDR), Temple University
- Department of Physiology, LKSOM, Temple University
| |
Collapse
|
6
|
Moradi S, Braun T, Baharvand H. miR-302b-3p Promotes Self-Renewal Properties in Leukemia Inhibitory Factor-Withdrawn Embryonic Stem Cells. CELL JOURNAL 2018; 20:61-72. [PMID: 29308620 PMCID: PMC5761148 DOI: 10.22074/cellj.2018.4846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 01/07/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Embryonic stem cells (ESCs) are regulated by a gene regulatory circuitry composed of transcription factors, signaling pathways, metabolic mediators, and non-coding RNAs (ncRNAs). MicroRNAs (miRNAs) are short ncRNAs which play crucial roles in ESCs. Here, we explored the impact of miR-302b-3p on ESC self-renewal in the absence of leukemia inhibitory factor (LIF). MATERIALS AND METHODS In this experimental study, ESCs were cultured in the presence of 15% fetal bovine serum (FBS) and induced to differentiate by LIF removal. miR-302b-3p overexpression was performed by transient transfection of mature miRNA mimics. Cell cycle profiling was done using propidium iodide (PI) staining followed by flow cytometry. miRNA expression was quantified using a miR-302b-3p-specific TaqMan assay. Data were analyzed using t test, and a P<0.05 was considered statistically significant. RESULTS We observed that miR-302b-3p promoted the viability of both wild-type and LIF-withdrawn ESCs. It also increased ESC clonogenicity and alkaline phosphatase (AP) activity. The defective cell cycling of LIF-deprived ESCs was completely rescued by miR-302b-3p delivery. Moreover, miR-302b-3p inhibited the increased cell death rate induced by LIF removal. CONCLUSIONS miR-302b-3p, as a pluripotency-associated miRNA, promotes diverse features of ESC self-renewal in the absence of extrinsic LIF signals.
Collapse
Affiliation(s)
- Sharif Moradi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Thomas Braun
- Max-Planck Institute for Heart and Lung Research, Department of Cardiac Development and Remodelling, Bad Nauheim, Germany.
| | - Hossein Baharvand
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
7
|
Potential Role of Exosomes in Mending a Broken Heart: Nanoshuttles Propelling Future Clinical Therapeutics Forward. Stem Cells Int 2017; 2017:5785436. [PMID: 29163642 PMCID: PMC5662033 DOI: 10.1155/2017/5785436] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 09/13/2017] [Indexed: 02/06/2023] Open
Abstract
Stem cell transplantation therapy is a promising adjunct for regenerating damaged heart tissue; however, only modest improvements in cardiac function have been observed due to poor survival of transplanted cells in the ischemic heart. Therefore, there remains an unmet need for therapies that can aid in attenuating cardiac damage. Recent studies have demonstrated that exosomes released by stem cells could serve as a potential cell-free therapeutic for cardiac repair. These exosomes/nanoshuttles, once thought to be merely a method of waste disposal, have been shown to play a crucial role in physiological functions including short- and long-distance intercellular communication. In this review, we have summarized studies demonstrating the potential role of exosomes in improving cardiac function, attenuating cardiac fibrosis, stimulating angiogenesis, and modulating miRNA expression. Furthermore, exosomes carry an important cargo of miRNAs and proteins that could play an important role as a diagnostic marker for cardiovascular disease post-myocardial infarction. Although there is promising evidence from preclinical studies that exosomes released by stem cells could serve as a potential cell-free therapeutic for myocardial repair, there are several challenges that need to be addressed before exosomes could be fully utilized as off-the-shelf therapeutics for cardiac repair.
Collapse
|
8
|
Yuan K, Ai WB, Wan LY, Tan X, Wu JF. The miR-290-295 cluster as multi-faceted players in mouse embryonic stem cells. Cell Biosci 2017; 7:38. [PMID: 28794853 PMCID: PMC5547456 DOI: 10.1186/s13578-017-0166-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/01/2017] [Indexed: 01/04/2023] Open
Abstract
Increasing evidence indicates that embryonic stem cell specific microRNAs (miRNAs) play an essential role in the early development of embryo. Among them, the miR-290-295 cluster is the most highly expressed in the mouse embryonic stem cells and involved in various biological processes. In this paper, we reviewed the research progress of the function of the miR-290-295 cluster in embryonic stem cells. The miR-290-295 cluster is involved in regulating embryonic stem cell pluripotency maintenance, self-renewal, and reprogramming somatic cells to an embryonic stem cell-like state. Moreover, the miR-290-295 cluster has a latent pro-survival function in embryonic stem cells and involved in tumourigenesis and senescence with a great significance. Elucidating the interaction between the miR-290-295 cluster and other modes of gene regulation will provide us new ideas on the biology of pluripotent stem cells. In the near future, the broad prospects of the miRNA cluster will be shown in the stem cell field, such as altering cell identities with high efficiency through the transient introduction of tissue-specific miRNA cluster.
Collapse
Affiliation(s)
- Kai Yuan
- Institute of Organ Fibrosis and Targeted Drug Delivery, Medical College, China Three Gorges University, 8 Daxue Road, Xiling District, Yichang, 443002 China
| | - Wen-Bing Ai
- The Yiling Hospital of Yichang, 31 Donghu Road, Yi Ling District, Yichang, 443100 Hubei China
| | - Lin-Yan Wan
- Institute of Organ Fibrosis and Targeted Drug Delivery, Medical College, China Three Gorges University, 8 Daxue Road, Xiling District, Yichang, 443002 China.,The RenMin Hospital, China Three Gorges University, 31 Huti Subdistrict, Xi Ling District, Yichang, 443000 Hubei China
| | - Xiao Tan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, 8 Daxue Road, Xiling District, Yichang, 443002 China
| | - Jiang-Feng Wu
- Institute of Organ Fibrosis and Targeted Drug Delivery, Medical College, China Three Gorges University, 8 Daxue Road, Xiling District, Yichang, 443002 China
| |
Collapse
|
9
|
Gambardella G, Carissimo A, Chen A, Cutillo L, Nowakowski TJ, di Bernardo D, Blelloch R. The impact of microRNAs on transcriptional heterogeneity and gene co-expression across single embryonic stem cells. Nat Commun 2017; 8:14126. [PMID: 28102192 PMCID: PMC5253645 DOI: 10.1038/ncomms14126] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 12/01/2016] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs act posttranscriptionally to suppress multiple target genes within a cell population. To what extent this multi-target suppression occurs in individual cells and how it impacts transcriptional heterogeneity and gene co-expression remains unknown. Here we used single-cell sequencing combined with introduction of individual microRNAs. miR-294 and let-7c were introduced into otherwise microRNA-deficient Dgcr8 knockout mouse embryonic stem cells. Both microRNAs induce suppression and correlated expression of their respective gene targets. The two microRNAs had opposing effects on transcriptional heterogeneity within the cell population, with let-7c increasing and miR-294 decreasing the heterogeneity between cells. Furthermore, let-7c promotes, whereas miR-294 suppresses, the phasing of cell cycle genes. These results show at the individual cell level how a microRNA simultaneously has impacts on its many targets and how that in turn can influence a population of cells. The findings have important implications in the understanding of how microRNAs influence the co-expression of genes and pathways, and thus ultimately cell fate. MicroRNAs can posttranscriptionally repress multiple targets in a cell population. Here the authors use single-cell sequencing to investigate the effects of an individual miRNA on transcriptional heterogeneity and gene co-expression
Collapse
Affiliation(s)
| | | | - Amy Chen
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USA.,Department of Urology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Luisa Cutillo
- Telethon Institute of Genetics and Medicine, Pozzuoli, 80078 Naples, Italy
| | - Tomasz J Nowakowski
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USA
| | - Diego di Bernardo
- Telethon Institute of Genetics and Medicine, Pozzuoli, 80078 Naples, Italy.,Department of Chemical, Materials and Industrial Engineering, University of Naples 'Federico II', 80125 Naples, Italy
| | - Robert Blelloch
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, University of California, San Francisco, San Francisco, California 94143, USA.,Department of Urology, University of California, San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
10
|
Yang H, Liu CC, Wang CY, Zhang Q, An J, Zhang L, Hao DJ. Therapeutical Strategies for Spinal Cord Injury and a Promising Autologous Astrocyte-Based Therapy Using Efficient Reprogramming Techniques. Mol Neurobiol 2016; 53:2826-2842. [DOI: 10.1007/s12035-015-9157-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 03/19/2015] [Indexed: 01/01/2023]
|
11
|
Cruz-Santos MC, Aragón-Raygoza A, Espinal-Centeno A, Arteaga-Vázquez M, Cruz-Hernández A, Bako L, Cruz-Ramírez A. The Role of microRNAs in Animal Cell Reprogramming. Stem Cells Dev 2016; 25:1035-49. [PMID: 27224014 DOI: 10.1089/scd.2015.0359] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Our concept of cell reprogramming and cell plasticity has evolved since John Gurdon transferred the nucleus of a completely differentiated cell into an enucleated Xenopus laevis egg, thereby generating embryos that developed into tadpoles. More recently, induced expression of transcription factors, oct4, sox2, klf4, and c-myc has evidenced the plasticity of the genome to change the expression program and cell phenotype by driving differentiated cells to the pluripotent state. Beyond these milestone achievements, research in artificial cell reprogramming has been focused on other molecules that are different than transcription factors. Among the candidate molecules, microRNAs (miRNAs) stand out due to their potential to control the levels of proteins that are involved in cellular processes such as self-renewal, proliferation, and differentiation. Here, we review the role of miRNAs in the maintenance and differentiation of mesenchymal stem cells, epimorphic regeneration, and somatic cell reprogramming to induced pluripotent stem cells.
Collapse
Affiliation(s)
- María Concepción Cruz-Santos
- 1 Molecular and Developmental Complexity Group, Unidad de Genómica Avanzada (U.G.A.-LANGEBIO) CINVESTAV , Irapuato, México
| | - Alejandro Aragón-Raygoza
- 1 Molecular and Developmental Complexity Group, Unidad de Genómica Avanzada (U.G.A.-LANGEBIO) CINVESTAV , Irapuato, México
| | - Annie Espinal-Centeno
- 1 Molecular and Developmental Complexity Group, Unidad de Genómica Avanzada (U.G.A.-LANGEBIO) CINVESTAV , Irapuato, México
| | - Mario Arteaga-Vázquez
- 2 Laboratory of Epigenetics and Developmental Biology, Institute for Biotechnology and Applied Ecology (INBIOTECA) , Universidad Veracruzana, Xalapa, México
| | - Andrés Cruz-Hernández
- 3 Facultad of Chemistry, Autonomous University of Querétaro, Santiago de Querétaro, México
| | - Laszlo Bako
- 4 Department of Plant Physiology, Umeå University , Umeå, Sweden
| | - Alfredo Cruz-Ramírez
- 1 Molecular and Developmental Complexity Group, Unidad de Genómica Avanzada (U.G.A.-LANGEBIO) CINVESTAV , Irapuato, México
| |
Collapse
|
12
|
Lee YJ, Ramakrishna S, Chauhan H, Park WS, Hong SH, Kim KS. Dissecting microRNA-mediated regulation of stemness, reprogramming, and pluripotency. ACTA ACUST UNITED AC 2016; 5:2. [PMID: 27006752 PMCID: PMC4802578 DOI: 10.1186/s13619-016-0028-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 02/20/2016] [Indexed: 02/06/2023]
Abstract
Increasing evidence indicates that microRNAs (miRNAs), endogenous short non-coding RNAs 19–24 nucleotides in length, play key regulatory roles in various biological events at the post-transcriptional level. Embryonic stem cells (ESCs) represent a valuable tool for disease modeling, drug discovery, developmental studies, and potential cell-based therapies in regenerative medicine due to their unlimited self-renewal and pluripotency. Therefore, remarkable progress has been made in recent decades toward understanding the expression and functions of specific miRNAs in the establishment and maintenance of pluripotency. Here, we summarize the recent knowledge regarding the regulatory roles of miRNAs in self-renewal of pluripotent ESCs and during cellular reprogramming, as well as the potential role of miRNAs in two distinct pluripotent states (naïve and primed).
Collapse
Affiliation(s)
- Young Jin Lee
- iDream Research Center, MizMedi Women's Hospital, Seoul, 07639 South Korea
| | - Suresh Ramakrishna
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763 South Korea.,College of Medicine, Hanyang University, Seoul, South Korea
| | | | - Won Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon, 24341 South Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, School of Medicine, Kangwon National University, 1 Kangwondaehak-gil, Chuncheon-si, Gangwon-do 24341 South Korea.,Stem Cell Institute, Kangwon National University, Chuncheon, 24341 South Korea
| | - Kye-Seong Kim
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, 04763 South Korea.,College of Medicine, Hanyang University, Seoul, South Korea
| |
Collapse
|
13
|
Zhao ZG, Jin JY, Zhang AM, Zhang LP, Wang XX, Sun JG, Chen ZT. MicroRNA profile of tumorigenic cells during carcinogenesis of lung adenocarcinoma. J Cell Biochem 2015; 116:458-66. [PMID: 25359683 DOI: 10.1002/jcb.24999] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 10/20/2014] [Indexed: 12/19/2022]
Abstract
To obtain microRNA (miRNA) profile and clarify their biological function in tumorigenic Sca-1(+) CD34(+) cells during carcinogenesis of lung adenocarcinoma. After intranasal infection with recombinant Adeno-Cre viruses (AdV-Cre), lung adenocarcinoma was identified pathologically in Lox-stop-lox Kras (LSL-Kras) G12D mice. Sca-1(+) CD34(+) cells were sorted by flow cytometry and tested for tumor-initiating ability, self-renewal and tumorigenicity. MiRNA profiles were obtained using microarray and further confirmed by real-time RT-PCR (qRT-PCR). MiRNA functions were predicted bioinformatically, and miR-294 function was verified to explore its role in tumor migration and invasion. Lung adenocarcinoma was induced in LSL-Kras G12D mice within 30 days. In vivo, the tumorigenicity of Sca-1(+) CD34(+) cells was 25 times stronger than Sca-1(-) CD34(-) cells. During tumorigenesis of lung adenocarcinoma, the expression of 145 miRNAs in Sca-1(+) CD34(+) cells increased and 72 miRNAs decreased (P < 0.01). Four successively up-regulated miRNAs (miR-15a*, miR-203, miR-294 and miR-295*) and three successively down-regulated ones (miR-19b, miR-483 and miR-615-5p) were identified. Among them, miR-294 could constitutively bind to 3'-UTR of matrix metalloproteinase 3 (MMP3), and down-regulate MMP3 protein expression. MiR-294 also significantly inhibited migration and invasion of Lewis lung cancer cells. MiRNAs are characteristically expressed in tumor-initiating Sca-1(+) CD34(+) cells of lung adenocarcinoma, and may play important roles during the carcinogenesis of lung adenocarcinoma.
Collapse
Affiliation(s)
- Zhen-guo Zhao
- Cancer Institute of PLA, Xinqiao Hospital, Third Military Medical University, Chongqing, PR China
| | | | | | | | | | | | | |
Collapse
|
14
|
Wang A, He Q, Zhong Y. Systematically dissecting the global mechanism of miRNA functions in mouse pluripotent stem cells. BMC Genomics 2015; 16:490. [PMID: 26126859 PMCID: PMC4488055 DOI: 10.1186/s12864-015-1706-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 06/16/2015] [Indexed: 12/16/2022] Open
Abstract
Background MicroRNAs (miRNAs) critically modulate stem cell properties like pluripotency, but the fundamental mechanism remains largely unknown. Method This study systematically analyzes multiple-omics data and builds a systems physical network including genome-wide interactions between miRNAs and their targets to reveal the systems mechanism of miRNA functions in mouse pluripotent stem cells. Results Globally, miRNAs directly repress the pluripotent core factors during differentiation state. Surprisingly, during the pluripotent state, the top important miRNAs do not directly regulate the pluripotent core factors as previously thought, but they only directly target the pluripotent signal pathways and directly repress developmental processes. Furthermore, at the pluripotent state miRNAs predominately repress DNA methyltransferases, the core enzymes for DNA methylation. The decreasing methylation repressed by miRNAs in turn activates the top miRNAs and pluripotent core factors, creating an active circuit system to modulate pluripotency. Conclusion MiRNAs vary their functions with stem cell states. While miRNAs directly repress pluripotent core factors to facilitate differentiation during the differentiation state, they also help stem cells to maintain pluripotency by activating pluripotent cores through directly repressing DNA methylation systems and primarily inhibiting development in the pluripotent state. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1706-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anyou Wang
- School of medicine, University of California San Francisco, S-1268, Medical Sciences Building, 513 Parnassus Ave, San Francisco, CA, 94143, USA.
| | - Qianchuan He
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, USA.
| | - Yan Zhong
- Division of Gynecologic Oncology, Linyi Tumor Hospital, Shandong, 276000, China.
| |
Collapse
|
15
|
MiR-371-373 cluster acts as a tumor-suppressor-miR and promotes cell cycle arrest in unrestricted somatic stem cells. Tumour Biol 2015; 36:7765-74. [PMID: 25941115 DOI: 10.1007/s13277-015-3519-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/27/2015] [Indexed: 12/20/2022] Open
Abstract
Recent advances in small RNA research have implicated microRNAs (miRNAs) as important regulators of proliferation and development. The miR-371-373 cluster is prominently expressed in human embryonic stem cells (ESCs) and rapidly decreases after cell differentiation. MiR-371-373 cluster was investigated as one of the key factors of stem cell maintenance and pluripotency in unrestricted somatic stem cells (USSCs) using a lentivirus system. Gene expression showed a dual effect on proliferation, which revealed a transient cell cycle progression and consequent repression in pluripotency factors and cell cycle genes. Cell proliferation analysis with CFU, MTT, and DNA content assays further confirmed the dual effect of cluster after prolonged exposure. Analyzing the course of action, it seems that miR-371-373 cluster acts as an onco/tumor suppressor-miR. MiR371-373 cluster acts by modulating the function of these factors and limiting the excessive cell cycle propagation upon oncogenic stimuli to protect cells from replicative stress, but also activate CDK inhibitors and transcriptional repressors of the retinoblastoma family to cause cell cycle arrest. In contrast to the previous studies, we believe that miR-371-373 cluster functions as a self-renewal miRNA to induce and maintain the pluripotent state but also to potentially inhibit dysregulated proliferation through cell cycle arrest. It seems that miR-371-373 cluster presents with a dual effect in this cellular context which may possess different actions in various cells. This not only expands the basic knowledge of the cluster but may offer a great chance for therapeutic interventions.
Collapse
|
16
|
Khan M, Nickoloff E, Abramova T, Johnson J, Verma SK, Krishnamurthy P, Mackie AR, Vaughan E, Garikipati VNS, Benedict C, Ramirez V, Lambers E, Ito A, Gao E, Misener S, Luongo T, Elrod J, Qin G, Houser SR, Koch WJ, Kishore R. Embryonic stem cell-derived exosomes promote endogenous repair mechanisms and enhance cardiac function following myocardial infarction. Circ Res 2015; 117:52-64. [PMID: 25904597 DOI: 10.1161/circresaha.117.305990] [Citation(s) in RCA: 585] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 04/22/2015] [Indexed: 12/11/2022]
Abstract
RATIONALE Embryonic stem cells (ESCs) hold great promise for cardiac regeneration but are susceptible to various concerns. Recently, salutary effects of stem cells have been connected to exosome secretion. ESCs have the ability to produce exosomes, however, their effect in the context of the heart is unknown. OBJECTIVE Determine the effect of ESC-derived exosome for the repair of ischemic myocardium and whether c-kit(+) cardiac progenitor cells (CPCs) function can be enhanced with ESC exosomes. METHODS AND RESULTS This study demonstrates that mouse ESC-derived exosomes (mES Ex) possess ability to augment function in infarcted hearts. mES Ex enhanced neovascularization, cardiomyocyte survival, and reduced fibrosis post infarction consistent with resurgence of cardiac proliferative response. Importantly, mES Ex augmented CPC survival, proliferation, and cardiac commitment concurrent with increased c-kit(+) CPCs in vivo 8 weeks after in vivo transfer along with formation of bonafide new cardiomyocytes in the ischemic heart. miRNA array revealed significant enrichment of miR290-295 cluster and particularly miR-294 in ESC exosomes. The underlying basis for the beneficial effect of mES Ex was tied to delivery of ESC specific miR-294 to CPCs promoting increased survival, cell cycle progression, and proliferation. CONCLUSIONS mES Ex provide a novel cell-free system that uses the immense regenerative power of ES cells while avoiding the risks associated with direct ES or ES-derived cell transplantation and risk of teratomas. ESC exosomes possess cardiac regeneration ability and modulate both cardiomyocyte and CPC-based repair programs in the heart.
Collapse
Affiliation(s)
- Mohsin Khan
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Emily Nickoloff
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Tatiana Abramova
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Jennifer Johnson
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Suresh Kumar Verma
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Prasanna Krishnamurthy
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Alexander Roy Mackie
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Erin Vaughan
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Venkata Naga Srikanth Garikipati
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Cynthia Benedict
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Veronica Ramirez
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Erin Lambers
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Aiko Ito
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Erhe Gao
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Sol Misener
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Timothy Luongo
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - John Elrod
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Gangjian Qin
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Steven R Houser
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Walter J Koch
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.)
| | - Raj Kishore
- From the Center for Translational Medicine (M.K., E.N., J.J., S.K.V., P.K., E.V., V.N.S.G., C.B., E.G., T.L., J.E., W.J.K.), Cardiovascular Research Center (S.R.H.), Department of Physiology (S.R.H.), and Department of Pharmacology (T.L., W.J.K., R.K.), Temple University School of Medicine, Philadelphia, PA; and Feinberg Cardiovascular Research Institute, Northwestern University, Chicago, IL (T.A., A.R.M., E.V., V.R., E.L., A.I., S.M., G.Q., R.K.).
| |
Collapse
|
17
|
Suppression of epithelial-mesenchymal transition and apoptotic pathways by miR-294/302 family synergistically blocks let-7-induced silencing of self-renewal in embryonic stem cells. Cell Death Differ 2014; 22:1158-69. [PMID: 25501598 PMCID: PMC4572863 DOI: 10.1038/cdd.2014.205] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 10/27/2014] [Accepted: 11/03/2014] [Indexed: 01/01/2023] Open
Abstract
The embryonic stem cell (ESC)-enriched miR-294/302 family and the somatic cell-enriched let-7 family stabilizes the self-renewing and differentiated cell fates, respectively. The mechanisms underlying these processes remain unknown. Here we show that among many pathways regulated by miR-294/302, the combinatorial suppression of epithelial–mesenchymal transition (EMT) and apoptotic pathways is sufficient in maintaining the self-renewal of ESCs. The silencing of ESC self-renewal by let-7 was accompanied by the upregulation of several EMT regulators and the induction of apoptosis. The ectopic activation of either EMT or apoptotic program is sufficient in silencing ESC self-renewal. However, only combined but not separate suppression of the two programs inhibited the silencing of ESC self-renewal by let-7 and several other differentiation-inducing miRNAs. These findings demonstrate that combined repression of the EMT and apoptotic pathways by miR-294/302 imposes a synergistic barrier to the silencing of ESC self-renewal, supporting a model whereby miRNAs regulate complicated cellular processes through synergistic repression of multiple targets or pathways.
Collapse
|
18
|
Abstract
Embryonic stem cell maintenance, differentiation, and somatic cell reprogramming require the interplay of multiple pluripotency factors, epigenetic remodelers, and extracellular signaling pathways. RNA-binding proteins (RBPs) are involved in a wide range of regulatory pathways, from RNA metabolism to epigenetic modifications. In recent years we have witnessed more and more studies on the discovery of new RBPs and the assessment of their functions in a variety of biological systems, including stem cells. We review the current studies on RBPs and focus on those that have functional implications in pluripotency, differentiation, and/or reprogramming in both the human and mouse systems.
Collapse
|
19
|
Wickramasinghe S, Cánovas A, Rincón G, Medrano JF. RNA-Sequencing: A tool to explore new frontiers in animal genetics. Livest Sci 2014. [DOI: 10.1016/j.livsci.2014.06.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
20
|
Gruber AJ, Grandy WA, Balwierz PJ, Dimitrova YA, Pachkov M, Ciaudo C, Nimwegen EV, Zavolan M. Embryonic stem cell-specific microRNAs contribute to pluripotency by inhibiting regulators of multiple differentiation pathways. Nucleic Acids Res 2014; 42:9313-26. [PMID: 25030899 PMCID: PMC4132708 DOI: 10.1093/nar/gku544] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The findings that microRNAs (miRNAs) are essential for early development in many species and that embryonic miRNAs can reprogram somatic cells into induced pluripotent stem cells suggest that these miRNAs act directly on transcriptional and chromatin regulators of pluripotency. To elucidate the transcription regulatory networks immediately downstream of embryonic miRNAs, we extended the motif activity response analysis approach that infers the regulatory impact of both transcription factors (TFs) and miRNAs from genome-wide expression states. Applying this approach to multiple experimental data sets generated from mouse embryonic stem cells (ESCs) that did or did not express miRNAs of the ESC-specific miR-290-295 cluster, we identified multiple TFs that are direct miRNA targets, some of which are known to be active during cell differentiation. Our results provide new insights into the transcription regulatory network downstream of ESC-specific miRNAs, indicating that these miRNAs act on cell cycle and chromatin regulators at several levels and downregulate TFs that are involved in the innate immune response.
Collapse
Affiliation(s)
- Andreas J Gruber
- Biozentrum, University of Basel, Klingelberstrasse 50-70, CH-4056 Basel, Switzerland
| | - William A Grandy
- Biozentrum, University of Basel, Klingelberstrasse 50-70, CH-4056 Basel, Switzerland
| | - Piotr J Balwierz
- Biozentrum, University of Basel, Klingelberstrasse 50-70, CH-4056 Basel, Switzerland
| | - Yoana A Dimitrova
- Biozentrum, University of Basel, Klingelberstrasse 50-70, CH-4056 Basel, Switzerland
| | - Mikhail Pachkov
- Biozentrum, University of Basel, Klingelberstrasse 50-70, CH-4056 Basel, Switzerland
| | | | - Erik van Nimwegen
- Biozentrum, University of Basel, Klingelberstrasse 50-70, CH-4056 Basel, Switzerland
| | - Mihaela Zavolan
- Biozentrum, University of Basel, Klingelberstrasse 50-70, CH-4056 Basel, Switzerland
| |
Collapse
|
21
|
Abstract
Embryonic and induced pluripotent stem cells (ESCs and iPSCs) hold great promise for regenerative medicine. The therapeutic application of these cells requires an understanding of the molecular networks that regulate pluripotency, differentiation, and de-differentiation. Along with signaling pathways, transcription factors, and epigenetic regulators, microRNAs (miRNAs) are emerging as important regulators in the establishment and maintenance of pluripotency. These tiny RNAs control proliferation, survival, the cell cycle, and the pluripotency program of ESCs. In addition, they serve as barriers or factors to overcome barriers during the reprogramming process. Systematic screening for novel miRNAs that regulate the establishment and maintenance of pluripotent stem cells and further mechanistic investigations will not only shed new light on the biology of ESCs and iPSCs, but also help develop safe and efficient technologies to manipulate cell fate for regenerative medicine.
Collapse
|
22
|
Links between the oncoprotein YB-1 and small non-coding RNAs in breast cancer. PLoS One 2013; 8:e80171. [PMID: 24260353 PMCID: PMC3832415 DOI: 10.1371/journal.pone.0080171] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 10/07/2013] [Indexed: 12/21/2022] Open
Abstract
Background The nucleic acid-binding protein YB-1, a member of the cold-shock domain protein family, has been implicated in the progression of breast cancer and is associated with poor patient survival. YB-1 has sequence similarity to LIN28, another cold-shock protein family member, which has a role in the regulation of small noncoding RNAs (sncRNAs) including microRNAs (miRNAs). Therefore, to investigate whether there is an association between YB-1 and sncRNAs in breast cancer, we investigated whether sncRNAs were bound by YB-1 in two breast cancer cell lines (luminal A-like and basal cell-like), and whether the abundance of sncRNAs and mRNAs changed in response to experimental reduction of YB-1 expression. Results RNA-immunoprecipitation with an anti-YB-1 antibody showed that several sncRNAs are bound by YB-1. Some of these were bound by YB-1 in both breast cancer cell lines; others were cell-line specific. The small RNAs bound by YB-1 were derived from various sncRNA families including miRNAs such as let-7 and miR-320, transfer RNAs, ribosomal RNAs and small nucleolar RNAs (snoRNA). Reducing YB-1 expression altered the abundance of a number of transcripts encoding miRNA biogenesis and processing proteins but did not alter the abundance of mature or precursor miRNAs. Conclusions YB-1 binds to specific miRNAs, snoRNAs and tRNA-derived fragments and appears to regulate the expression of miRNA biogenesis and processing machinery. We propose that some of the oncogenic effects of YB-1 in breast cancer may be mediated through its interactions with sncRNAs.
Collapse
|
23
|
Diekmann U, Elsner M, Fiedler J, Thum T, Lenzen S, Naujok O. MicroRNA target sites as genetic tools to enhance promoter-reporter specificity for the purification of pancreatic progenitor cells from differentiated embryonic stem cells. Stem Cell Rev Rep 2013; 9:555-68. [PMID: 23111459 DOI: 10.1007/s12015-012-9416-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Pluripotent cells hold great promise for cell replacement therapies in regenerative medicine. All known protocols for directed in vitro differentiation of pluripotent cells did not yield pure populations complicating the characterization of the derived cells. In addition, the risk of tumor formation due to residual undifferentiated cells is a serious unresolved problem. In the present study the tissue-specific mouse Pdx1 promoter was used to control the expression of the reporter gene GFP2 in mouse ES cells in order to purify them via FACS during in vitro differentiation. The background fluorescence of transduced ES cells hampered the purification of Pdx1-positive cells due to a contaminating population of partially undifferentiated cells. MicroRNAs (mir) are important regulators of gene expression and were used to enhance promoter specificity during differentiation towards pancreatic progenitor cells. The mouse mmu-mir-294 was found to be mainly expressed during pluripotency, whereas the expression of the mir-302 cluster was increased during early differentiation. Integration of a microRNA target site for the mmu-mir-294 into the lentiviral vector reduced the background fluorescence specifically during pluripotency and permitted re-occurrence of GFP2 expression upon differentiation. A combination of the microRNA target site with the Pdx1 promoter fragment allowed the purification of pancreatic progenitors from differentiated ES cells. This population reflected an early pancreatic progenitor population without other contaminating cell lineages. In conclusion, microRNA target sites are efficient regulatory elements to control transgene expression and to enhance tissue specificity as presented in this study facilitating the sorting and purification of Pdx1-positive pancreatic progenitor cells.
Collapse
Affiliation(s)
- Ulf Diekmann
- Institute of Clinical Biochemistry, Hannover Medical School, 30625, Hannover, Germany
| | | | | | | | | | | |
Collapse
|
24
|
Meng W, Ye Z, Cui R, Perry J, Dedousi-Huebner V, Huebner A, Wang Y, Li B, Volinia S, Nakanishi H, Kim T, Suh SS, Ayers LW, Ross P, Croce CM, Chakravarti A, Jin VX, Lautenschlaeger T. MicroRNA-31 predicts the presence of lymph node metastases and survival in patients with lung adenocarcinoma. Clin Cancer Res 2013; 19:5423-33. [PMID: 23946296 DOI: 10.1158/1078-0432.ccr-13-0320] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE We conducted genome-wide miRNA-sequencing (miRNA-seq) in primary cancer tissue from patients of lung adenocarcinoma to identify markers for the presence of lymph node metastasis. EXPERIMENTAL DESIGN Markers for lymph node metastasis identified by sequencing were validated in a separate cohort using quantitative PCR. After additional validation in the The Cancer Genome Atlas (TCGA) dataset, functional characterization studies were conducted in vitro. RESULTS MiR-31 was upregulated in lung adenocarcinoma tissues from patients with lymph node metastases compared with those without lymph node metastases. We confirmed miR-31 to be upregulated in lymph node-positive patients in a separate patient cohort (P = 0.009, t test), and to be expressed at higher levels in adenocarcinoma tissue than in matched normal adjacent lung tissues (P < 0.0001, paired t test). MiR-31 was then validated as a marker for lymph node metastasis in an external validation cohort of 233 lung adenocarcinoma cases of the TCGA (P = 0.031, t test). In vitro functional assays showed that miR-31 increases cell migration, invasion, and proliferation in an ERK1/2 signaling-dependent manner. Notably, miR-31 was a significant predictor of survival in a multivariate cox regression model even when controlling for cancer staging. Exploratory in silico analysis showed that low expression of miR-31 is associated with excellent survival for T2N0 patients. CONCLUSIONS We applied miRNA-seq to study microRNomes in lung adenocarcinoma tissue samples for the first time and potentially identified a miRNA predicting the presence of lymph node metastasis and survival outcomes in patients of lung adenocarcinoma.
Collapse
Affiliation(s)
- Wei Meng
- Authors' Affiliations: Department of Molecular Medicine, The University of Texas Health Science Center, San Antonio, Texas; Department of Radiation Oncology, Department of Molecular Virology, Immunology and Medical Genetics; Comprehensive Cancer Center; Department of Pathology, College of Medicine; Department of Surgery, Division of Thoracic Surgery, The Ohio State University, Columbus, Ohio; and Yanbian University College of Medicine, Ji Lin, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Wang T, Shi SB, Sha HY. MicroRNAs in regulation of pluripotency and somatic cell reprogramming: small molecule with big impact. RNA Biol 2013; 10:1255-61. [PMID: 23921205 DOI: 10.4161/rna.25828] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
MicroRNAs (miRNAs), a group of small non-coding RNAs, have emerged as significant modulators in the establishment and generation of pluripotency, a developmental process that consists of complex cell-fate arrangements. The finding of embryonic stem cell (ESC) cycle-specific miRNAs reveals an important regulation scheme of pluripotency. Subsequent studies showed the ESC-enriched or ESC-depleted miRNAs can regulate induced pluripotent stem cells(iPSC). Moreover, miRNA profiling of iPSC and ESC may distinguish them from one another and facilitate the complex of regulatory network. The accumulative effects of miRNA action enable using miRNA alone to generate iPSCs. Despite the robustness of iPSC studies, further investigations are needed since miRNA may have more impact on induced pluripotency, and the roles of miRNAs in somatic cell nuclear transfer (SCNT), another approach toward cellular reprogramming, remains unclear. This point-of-view article will discuss miRNAs and their impact on the normal and induced pluripotency, as well as bring new insights on somatic cell reprogramming.
Collapse
Affiliation(s)
- Tian Wang
- State Key Laboratory for Medical Neurobiology; School of Basic Medical Sciences; Fudan University; Shanghai, China; Eight-year Medical Program; Zhongshan Hospital; Fudan University; Shanghai, China
| | | | | |
Collapse
|
26
|
Wang Y, Melton C, Li YP, Shenoy A, Zhang XX, Subramanyam D, Blelloch R. miR-294/miR-302 promotes proliferation, suppresses G1-S restriction point, and inhibits ESC differentiation through separable mechanisms. Cell Rep 2013; 4:99-109. [PMID: 23831024 DOI: 10.1016/j.celrep.2013.05.027] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 03/27/2013] [Accepted: 05/14/2013] [Indexed: 12/14/2022] Open
Abstract
The miR-294 and miR-302 microRNAs promote the abbreviated G1 phase of the embryonic stem cell (ESC) cell cycle and suppress differentiation induced by let-7. Here, we evaluated the role of the retinoblastoma (Rb) family proteins in these settings. Under normal growth conditions, miR-294 promoted the rapid G1-S transition independent of the Rb family. In contrast, miR-294 suppressed the further accumulation of cells in G1 in response to nutrient deprivation and cell-cell contact in an Rb-dependent fashion. We uncovered five additional miRNAs (miR-26a, miR-99b, miR-193, miR-199a-5p, and miR-218) that silenced ESC self-renewal in the absence of other miRNAs, all of which were antagonized by miR-294 and miR-302. Four of the six differentiation-inducing miRNAs induced an Rb-dependent G1 accumulation. However, all six still silenced self-renewal in the absence of the Rb proteins. These results show that the miR-294/miR-302 family acts through Rb-dependent and -independent pathways to regulate the G1 restriction point and the silencing of self-renewal, respectively.
Collapse
Affiliation(s)
- Yangming Wang
- Peking-Tsinghua Joint Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing 100871, China.
| | | | | | | | | | | | | |
Collapse
|
27
|
Fujii YR. The RNA gene information: retroelement-microRNA entangling as the RNA quantum code. Methods Mol Biol 2013; 936:47-67. [PMID: 23007498 DOI: 10.1007/978-1-62703-083-0_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
MicroRNA (miRNA) and retroelements may be a master of regulator in our life, which are evolutionally involved in the origin of species. To support the Darwinism from the aspect of molecular evolution process, it has tremendously been interested in the molecular information of naive RNA. The RNA wave model 2000 consists of four concepts that have altered from original idea of the miRNA genes for crosstalk among embryonic stem cells, their niche cells, and retroelements as a carrier vesicle of the RNA genes. (1) the miRNA gene as a mobile genetic element induces transcriptional and posttranscriptional silencing via networking-processes (no hierarchical architecture); (2) the RNA information supplied by the miRNA genes expands to intracellular, intercellular, intraorgan, interorgan, intraspecies, and interspecies under the cycle of life into the global environment; (3) the mobile miRNAs can self-proliferate; and (4) cells contain two types information as resident and genomic miRNAs. Based on RNA wave, we have developed an interest in investigation of the transformation from RNA information to quantum bits as physicochemical characters of RNA with the measurement of RNA electron spin. When it would have been given that the fundamental bases for the acquired characters in genetics can be controlled by RNA gene information, it may be available to apply for challenging against RNA gene diseases, such as stress-induced diseases.
Collapse
|
28
|
Regulation of stem cell populations by microRNAs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 786:329-51. [PMID: 23696365 DOI: 10.1007/978-94-007-6621-1_18] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
miRNAs are small non-coding RNAs that have emerged as crucial post-transcriptional regulators of gene expression. They are key players in various critical cellular processes such as proliferation, cell cycle progression, apoptosis and differentiation. Self-renewal capacity and differentiation potential are hallmarks of stem cells. The switch between self-renewal and differentiation requires rapid widespread changes in gene expression. Since miRNAs can repress the translation of many mRNA targets, they are good candidates to regulate cell fates. In the past few years, miRNAs have appeared as important new actors in stem cell development by regulating differentiation and maintenance of stem cells. In this chapter we will focus on the role of miRNAs in various stem cell populations. After an introduction on microRNA biogenesis, we will review the recent knowledge on miRNA expression and function in pluripotent cells and during the acquisition of stem cell fate. We will then briefly examine the role of miRNAs in adult and cancer stem cells.
Collapse
|
29
|
Large-scale identification of microRNA targets in murine Dgcr8-deficient embryonic stem cell lines. PLoS One 2012; 7:e41762. [PMID: 22912678 PMCID: PMC3422281 DOI: 10.1371/journal.pone.0041762] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Accepted: 06/25/2012] [Indexed: 01/04/2023] Open
Abstract
Small RNAs such as microRNAs play important roles in embryonic stem cell maintenance and differentiation. A broad range of microRNAs is expressed in embryonic stem cells while only a fraction of their targets have been identified. We have performed large-scale identification of embryonic stem cell microRNA targets using a murine embryonic stem cell line deficient in the expression of Dgcr8. These cells are heavily depleted for microRNAs, allowing us to reintroduce specific microRNA duplexes and identify refined target sets. We used deep sequencing of small RNAs, mRNA expression profiling and bioinformatics analysis of microRNA seed matches in 3' UTRs to identify target transcripts. Consequently, we have identified a network of microRNAs that converge on the regulation of several important cellular pathways. Additionally, our experiments have revealed a novel candidate for Dgcr8-independent microRNA genesis and highlighted the challenges currently facing miRNA annotation.
Collapse
|
30
|
MicroRNAs in embryonic stem cells. Epigenomics 2012. [DOI: 10.1017/cbo9780511777271.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
31
|
Karere GM, Glenn JP, VandeBerg JL, Cox LA. Differential microRNA response to a high-cholesterol, high-fat diet in livers of low and high LDL-C baboons. BMC Genomics 2012; 13:320. [PMID: 22809019 PMCID: PMC3536563 DOI: 10.1186/1471-2164-13-320] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 06/30/2012] [Indexed: 11/18/2022] Open
Abstract
Background Dysregulation of microRNA (miRNA) expression has been implicated in molecular genetic events leading to the progression and development of atherosclerosis. We hypothesized that miRNA expression profiles differ between baboons with low and high serum low-density lipoprotein cholesterol (LDL-C) concentrations in response to diet, and that a subset of these miRNAs regulate genes relevant to dyslipidemia and risk of atherosclerosis. Results Using Next Generation Illumina sequencing methods, we sequenced hepatic small RNA libraries from baboons differing in their LDL-C response to a high-cholesterol, high-fat (HCHF) challenge diet (low LDL-C, n = 3; high LDL-C, n = 3), resulting in 517 baboon miRNAs: 490 were identical to human miRNAs and 27 were novel. We compared miRNA expression profiles from liver biopsies collected before and after the challenge diet and observed that HCHF diet elicited expression of more miRNAs compared to baseline (chow) diet for both low and high LDL-C baboons. Eighteen miRNAs exhibited differential expression in response to HCHF diet in high LDL-C baboons compared to 10 miRNAs in low LDL-C baboons. We used TargetScan/Base tools to predict putative miRNA targets; miRNAs expressed in high LDL-C baboons had significantly more gene targets than miRNAs expressed in low LDL-C responders. Further, we identified miRNA isomers and other non-coding RNAs that were differentially expressed in response to the challenge diet in both high LDL-C and low LDL-C baboons. Conclusions We sequenced and annotated baboon liver miRNAs from low LDL-C and high LDL-C responders using high coverage Next Gen sequencing methods, determined expression changes in response to a HCHF diet challenge, and predicted target genes regulated by the differentially expressed miRNAs. The identified miRNAs will enrich the database for non-coding small RNAs including the extent of variation in these sequences. Further, we identified other small non-coding RNAs differentially expressed in response to diet. Our discovery of differentially expressed baboon miRNAs in response to a HCHF diet challenge that differ by LDL-C phenotype is a fundamental step in understating the role of non-coding RNAs in dyslipidemia.
Collapse
Affiliation(s)
- Genesio M Karere
- Department of Genetics, Texas Biomedical Research Institute, 7620 NW Loop 410, San Antonio, TX 78227-5301, USA
| | | | | | | |
Collapse
|
32
|
Paternal Benzo[a]pyrene Exposure Modulates MicroRNA Expression Patterns in the Developing Mouse Embryo. Int J Cell Biol 2012; 2012:407431. [PMID: 22548065 PMCID: PMC3324892 DOI: 10.1155/2012/407431] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 01/19/2012] [Indexed: 12/02/2022] Open
Abstract
Little attention has been given to how microRNA expression is affected by environmental contaminants exposure. We investigate the effects of paternal exposure to benzo[a]pyrene (B[a]P) on miRNA expression in the developing mouse embryo. Male mice were exposed to B[a]P (150 mg/kg i.p.), and their sperm was used four days later in in-vitro fertilization experiments. Twenty embryos each from 2-, 8-cell and the blastocyst stage were used for genome-wide miRNA expression profiling. Paternal exposure to B[a]P affected the expression of several miRNAs, and the target genes for some of the dysregulated miRNAs were enriched in many different pathways that are likely to be relevant for the developing mouse embryo. By linking the miRNA target genes to publicly available databases, we identified some miRNA target genes that may serve as global markers of B[a]P-mediated genotoxic stress. The dysregulated miRNAs may provide valuable knowledge about potential transgenerational effects of sublethal exposure to chemicals.
Collapse
|
33
|
Global microRNA level regulation of EGFR-driven cell-cycle protein network in breast cancer. Mol Syst Biol 2012; 8:570. [PMID: 22333974 PMCID: PMC3293631 DOI: 10.1038/msb.2011.100] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 12/12/2011] [Indexed: 12/22/2022] Open
Abstract
A genome-wide microRNA (miRNome) screen coupled with high-throughput monitoring of protein levels reveals complex, modular miRNA regulation of the EGFR-driven cell-cycle network, and identifies new miRNAs that can suppress breast cancer cell proliferation. ![]()
We interrogated, for the first time, a mammalian oncogenic signaling network with the miRNome and report the outputs at the protein level. Whole-genome microRNA (miRNA) effects on a given protein are generally mild, supporting a fine-tuning role for miRNAs, and these effects are dominated by sequence-matching mechanisms. We developed a novel network-analysis methodology with a bipartite graph model to identify proteins co-regulated by miRNAs. Besides the sequence-based mechanism, our results demonstrated that miRNAs simultaneously regulate several proteins belonging to the same functional module. We identified three miRNAs, miR-124, miR-147 and miR-193a-3p, as novel tumor suppressors that co-regulate EGFR-driven cell-cycle network proteins, and inhibit cell-cycle progression and proliferation in breast cancer. Our results demonstrate the potential to steer miRNA research toward the network level, underlining the need for systematic approaches before positioning miRNAs as drugs or drug targets.
The EGFR-driven cell-cycle pathway has been extensively studied due to its pivotal role in breast cancer proliferation and pathogenesis. Although several studies reported regulation of individual pathway components by microRNAs (miRNAs), little is known about how miRNAs coordinate the EGFR protein network on a global miRNA (miRNome) level. Here, we combined a large-scale miRNA screening approach with a high-throughput proteomic readout and network-based data analysis to identify which miRNAs are involved, and to uncover potential regulatory patterns. Our results indicated that the regulation of proteins by miRNAs is dominated by the nucleotide matching mechanism between seed sequences of the miRNAs and 3′-UTR of target genes. Furthermore, the novel network-analysis methodology we developed implied the existence of consistent intrinsic regulatory patterns where miRNAs simultaneously co-regulate several proteins acting in the same functional module. Finally, our approach led us to identify and validate three miRNAs (miR-124, miR-147 and miR-193a-3p) as novel tumor suppressors that co-target EGFR-driven cell-cycle network proteins and inhibit cell-cycle progression and proliferation in breast cancer.
Collapse
|
34
|
Studholme DJ. Deep sequencing of small RNAs in plants: applied bioinformatics. Brief Funct Genomics 2011; 11:71-85. [PMID: 22184332 DOI: 10.1093/bfgp/elr039] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Small RNAs, including microRNA and short-interfering RNAs, play important roles in plants. In recent years, developments in sequencing technology have enabled the large-scale discovery of sRNAs in various cells, tissues and developmental stages and in response to various stresses. This review describes the bioinformatics challenges to analysing these large datasets of short-RNA sequences and some of the solutions to those challenges.
Collapse
|
35
|
|