1
|
Ranjan R, Ma B, Gleason RJ, Liao Y, Bi Y, Davis BEM, Yang G, Clark M, Mahajan V, Condon M, Broderick NA, Chen X. Modulating DNA Polα Enhances Cell Reprogramming Across Species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.19.613993. [PMID: 39345551 PMCID: PMC11429986 DOI: 10.1101/2024.09.19.613993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
As a fundamental biological process, DNA replication ensures the accurate copying of genetic information. However, the impact of this process on cellular plasticity in multicellular organisms remains elusive. Here, we find that reducing the level or activity of a replication component, DNA Polymerase α (Polα), facilitates cell reprogramming in diverse stem cell systems across species. In Drosophila male and female germline stem cell lineages, reducing Polα levels using heterozygotes significantly enhances fertility of both sexes, promoting reproductivity during aging without compromising their longevity. Consistently, in C. elegans the pola heterozygous hermaphrodites exhibit increased fertility without a reduction in lifespan, suggesting that this phenomenon is conserved. Moreover, in male germline and female intestinal stem cell lineages of Drosophila, polα heterozygotes exhibit increased resistance to tissue damage caused by genetic ablation or pathogen infection, leading to enhanced regeneration and improved survival during post-injury recovery, respectively. Additionally, fine tuning of an inhibitor to modulate Polα activity significantly enhances the efficiency of reprogramming human embryonic fibroblasts into induced pluripotent cells. Together, these findings unveil novel roles of a DNA replication component in regulating cellular reprogramming potential, and thus hold promise for promoting tissue health, facilitating post-injury rehabilitation, and enhancing healthspan.
Collapse
Affiliation(s)
- Rajesh Ranjan
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Baltimore, MD 21218, USA
| | - Binbin Ma
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Baltimore, MD 21218, USA
| | - Ryan J. Gleason
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yijun Liao
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Yingshan Bi
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Brendon E. M. Davis
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Guanghui Yang
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Baltimore, MD 21218, USA
| | - Maggie Clark
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Vikrant Mahajan
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | - Madison Condon
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
| | | | - Xin Chen
- Department of Biology, The Johns Hopkins University, Baltimore, MD 21218, USA
- Howard Hughes Medical Institute, Department of Biology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Baltimore, MD 21218, USA
| |
Collapse
|
2
|
Farina AR, Cappabianca LA, Zelli V, Sebastiano M, Mackay AR. Mechanisms involved in selecting and maintaining neuroblastoma cancer stem cell populations, and perspectives for therapeutic targeting. World J Stem Cells 2021; 13:685-736. [PMID: 34367474 PMCID: PMC8316860 DOI: 10.4252/wjsc.v13.i7.685] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/09/2021] [Accepted: 04/14/2021] [Indexed: 02/06/2023] Open
Abstract
Pediatric neuroblastomas (NBs) are heterogeneous, aggressive, therapy-resistant embryonal tumours that originate from cells of neural crest (NC) origin and in particular neuroblasts committed to the sympathoadrenal progenitor cell lineage. Therapeutic resistance, post-therapeutic relapse and subsequent metastatic NB progression are driven primarily by cancer stem cell (CSC)-like subpopulations, which through their self-renewing capacity, intermittent and slow cell cycles, drug-resistant and reversibly adaptive plastic phenotypes, represent the most important obstacle to improving therapeutic outcomes in unfavourable NBs. In this review, dedicated to NB CSCs and the prospects for their therapeutic eradication, we initiate with brief descriptions of the unique transient vertebrate embryonic NC structure and salient molecular protagonists involved NC induction, specification, epithelial to mesenchymal transition and migratory behaviour, in order to familiarise the reader with the embryonic cellular and molecular origins and background to NB. We follow this by introducing NB and the potential NC-derived stem/progenitor cell origins of NBs, before providing a comprehensive review of the salient molecules, signalling pathways, mechanisms, tumour microenvironmental and therapeutic conditions involved in promoting, selecting and maintaining NB CSC subpopulations, and that underpin their therapy-resistant, self-renewing metastatic behaviour. Finally, we review potential therapeutic strategies and future prospects for targeting and eradication of these bastions of NB therapeutic resistance, post-therapeutic relapse and metastatic progression.
Collapse
Affiliation(s)
- Antonietta Rosella Farina
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Lucia Annamaria Cappabianca
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Veronica Zelli
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Michela Sebastiano
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy
| | - Andrew Reay Mackay
- Department of Applied Clinical and Biotechnological Sciences, University of L'Aquila, L'Aquila 67100, AQ, Italy.
| |
Collapse
|
3
|
Warrier S, Patil M, Bhansali S, Varier L, Sethi G. Designing precision medicine panels for drug refractory cancers targeting cancer stemness traits. Biochim Biophys Acta Rev Cancer 2020; 1875:188475. [PMID: 33188876 DOI: 10.1016/j.bbcan.2020.188475] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 02/06/2023]
Abstract
Cancer is one amongst the major causes of death today and cancer biology is one of the most well researched fields in medicine. The driving force behind cancer is considered to be a minor subpopulation of cells, the cancer stem cells (CSCs). Similar to other stem cells, these cells are self-renewing and proliferating but CSCs are also difficult to target by chemo- or radio-therapies. Cancer stem cells are known to be present in most of the cancer subgroups such as carcinoma, sarcoma, myeloma, leukemia, lymphomas and mixed cancer types. There is a wide gamut of factors attributed to the stemness of cancers, ranging from dysregulated signaling pathways, and activation of enzymes aiding immune evasion, to conducive tumor microenvironment, to name a few. The defining outcome of the increased presence of CSCs is tumor metastasis and relapse. Predictive medicine approach based on the plethora of CSC markers would be a move towards precision medicine to specifically identify CSC-rich tumors. In this review, we discuss the cancer subtypes and the role of different CSC specific markers in these varying subtypes. We also categorize the CSC markers based their defining trait contributing to stemness. This review thus provides a comprehensive approach to catalogue a predictive set of markers to identify the resistant and refractory cancer stem cell population within different tumor subtypes, so as to facilitate better prognosis and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India; Cuor Stem Cellutions Pvt Ltd, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India.
| | - Manasi Patil
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
| | - Sanyukta Bhansali
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore 560 065, India
| | | | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117 600, Singapore
| |
Collapse
|
4
|
Lu T, Yang Y, Li Z, Lu S. MicroRNA-214-3p inhibits the stem-like properties of lung squamous cell cancer by targeting YAP1. Cancer Cell Int 2020; 20:413. [PMID: 32863772 PMCID: PMC7450582 DOI: 10.1186/s12935-020-01506-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023] Open
Abstract
Background Emerging evidence reveals that microRNAs (miRNAs) play a crucial role in tumor progression, but the underlying mechanism of microRNAs in lung squamous cell cancer (LSCC) remains unclear. Method Western-blotting and quantitative real-time PCR (q-PCR) were carried out to detect mRNA and protein expression. Cell proliferation was evaluated by Cell Counting Kit-8 (CCK-8), colony-forming assay or sphere-forming assay, respectively. Results MiR-214-3p was markedly de-regulated in LSCC tissues and was inversely related to the level of Yes-associated protein1 (YAP1), which is the core transcription regulator of the Hippo signaling pathway. Kaplan–Meier survival curves illustrated that patients with high miR-214-3p expression demonstrated more favorable clinical outcomes. MiR-214-3p overexpression (OE) repressed proliferation and cancer stem-like cells (CSCs) properties in vitro and in vivo xenograft mouse model. Mechanistically, luciferase activity assay revealed that miR-214-3p directly targets YAP1 by specifically binding on the 3′ UTR of YAP1. Conclusion MiR-214-3p plays a pivotal role in CSCs properties by targeting YAP1, which provides a potential treatment strategy for LSCC patients.
Collapse
Affiliation(s)
- Tingting Lu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 Huaihai West Road, Shanghai, 200030 People's Republic of China
| | - Ying Yang
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 Huaihai West Road, Shanghai, 200030 People's Republic of China
| | - Ziming Li
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 Huaihai West Road, Shanghai, 200030 People's Republic of China
| | - Shun Lu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, 241 Huaihai West Road, Shanghai, 200030 People's Republic of China
| |
Collapse
|
5
|
Expression profile of microRNAs in the testes of patients with Klinefelter syndrome. Sci Rep 2020; 10:11470. [PMID: 32651451 PMCID: PMC7351945 DOI: 10.1038/s41598-020-68294-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 06/18/2020] [Indexed: 02/07/2023] Open
Abstract
Klinefelter syndrome (KS) is the most common sex chromosome aneuploidy. A distinctive characteristic of KS is oligozoospermia. Despite multiple studies that have described the natural history of the degenerative process of germ cells in patients with KS, the molecular mechanisms that initiate this process are not well characterized. MicroRNA (miRNA)-mediated post-transcriptional control mechanisms have been increasingly recognized as important regulators of spermatogenesis; however, only a few studies have evaluated the role of miRNAs in the gonadal failure of these patients. Here, we describe a differential expression profile for the miRNAs in testicular tissue samples taken from KS patients. We analysed testicular tissue samples from 4 KS patients and 5 control patients (obstructive azoospermia) through next-generation sequencing, which can provide information about the mechanisms involved in the degeneration of germ cells. A distinctive differential expression profile was identified for 166 miRNAs in the KS patients: 66 were upregulated, and 100 were downregulated. An interactome analysis was performed for 7 of the upregulated and the 20 downregulated miRNAs. The results showed that the target genes are involved in the development, proliferation, and differentiation processes of spermatogenesis, which may explain their role in the development of infertility. This is the first report of a miRNA expression profile generated from testicular tissue samples of KS patients.
Collapse
|
6
|
Conti I, Varano G, Simioni C, Laface I, Milani D, Rimondi E, Neri LM. miRNAs as Influencers of Cell-Cell Communication in Tumor Microenvironment. Cells 2020; 9:cells9010220. [PMID: 31952362 PMCID: PMC7016744 DOI: 10.3390/cells9010220] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/13/2020] [Accepted: 01/14/2020] [Indexed: 12/14/2022] Open
Abstract
microRNAs (miRNAs) are small noncoding RNAs that regulate gene expression at the posttranscriptional level, inducing the degradation of the target mRNA or translational repression. MiRNAs are involved in the control of a multiplicity of biological processes, and their absence or altered expression has been associated with a variety of human diseases, including cancer. Recently, extracellular miRNAs (ECmiRNAs) have been described as mediators of intercellular communication in multiple contexts, including tumor microenvironment. Cancer cells cooperate with stromal cells and elements of the extracellular matrix (ECM) to establish a comfortable niche to grow, to evade the immune system, and to expand. Within the tumor microenvironment, cells release ECmiRNAs and other factors in order to influence and hijack the physiological processes of surrounding cells, fostering tumor progression. Here, we discuss the role of miRNAs in the pathogenesis of multicomplex diseases, such as Alzheimer’s disease, obesity, and cancer, focusing on the contribution of both intracellular miRNAs, and of released ECmiRNAs in the establishment and development of cancer niche. We also review growing evidence suggesting the use of miRNAs as novel targets or potential tools for therapeutic applications.
Collapse
Affiliation(s)
- Ilaria Conti
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Gabriele Varano
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Carolina Simioni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Ilaria Laface
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Daniela Milani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Erika Rimondi
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
| | - Luca M. Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, 44121 Ferrara, Italy; (I.C.); (G.V.); (C.S.); (I.L.); (D.M.); (E.R.)
- LTTA—Electron Microscopy Center, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: ; Tel.: +39-0532-455940
| |
Collapse
|
7
|
Kaid C, Assoni A, Marçola M, Semedo-Kuriki P, Bortolin RH, Carvalho VM, Okamoto OK. Proteome and miRNome profiling of microvesicles derived from medulloblastoma cell lines with stem-like properties reveals biomarkers of poor prognosis. Brain Res 2020; 1730:146646. [PMID: 31917138 DOI: 10.1016/j.brainres.2020.146646] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 11/23/2019] [Accepted: 01/02/2020] [Indexed: 01/13/2023]
Abstract
Primary central nervous system (CNS) tumors are the most common deadly childhood cancer. Several patients with medulloblastoma experience local or metastatic recurrences after standard treatment, a condition associated with very poor prognosis. Current neuroimaging techniques do not accurately detect residual stem-like medulloblastoma cells promoting tumor relapses. In attempt to identify candidate tumor markers that could be circulating in blood or cerebrospinal (CSF) fluid of patients, we evaluated the proteome and miRNome content of extracellular microvesicles (MVs) released by highly-aggressive stem-like medulloblastoma cells overexpressing the pluripotent factor OCT4A. These cells display enhanced tumor initiating capability and resistance to chemotherapeutic agents. A common set of 464 proteins and 10 microRNAs were exclusively detected in MVs of OCT4A-overexpressing cells from four distinct medulloblastoma cell lines, DAOY, CHLA-01-MED, D283-MED, and USP13-MED. The interactome mapping of these exclusive proteins and miRNAs revealed ERK, PI3K/AKT/mTOR, EGF/EGFR, and stem cell self-renewal as the main oncogenic signaling pathways altered in these aggressive medulloblastoma cells. Of these MV cargos, four proteins (UBE2M, HNRNPCL2, HNRNPCL3, HNRNPCL4) and five miRNAs (miR-4449, miR-500b, miR-3648, miR-1291, miR-3607) have not been previously reported in MVs from normal tissues and in CSF. These proteins and miRNAs carried within MVs might serve as biomarkers of aggressive stem-like medulloblastoma cells to improve clinical benefit by helping refining diagnosis, patient stratification, and early detection of relapsed disease.
Collapse
Affiliation(s)
- Carolini Kaid
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Rua do Matão 277, CEP: 05508-090, Cidade Universitária, São Paulo, SP, Brazil
| | - Amanda Assoni
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Rua do Matão 277, CEP: 05508-090, Cidade Universitária, São Paulo, SP, Brazil
| | - Marina Marçola
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Rua do Matão 277, CEP: 05508-090, Cidade Universitária, São Paulo, SP, Brazil
| | - Patricia Semedo-Kuriki
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Rua do Matão 277, CEP: 05508-090, Cidade Universitária, São Paulo, SP, Brazil
| | - Raul Hernandes Bortolin
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Brazil
| | | | - Oswaldo Keith Okamoto
- Centro de Pesquisa sobre o Genoma Humano e Células-Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Rua do Matão 277, CEP: 05508-090, Cidade Universitária, São Paulo, SP, Brazil; Hemotherapy and Cellular Therapy Department, Hospital Israelita Albert Einstein, Sao Paulo, SP, Brazil.
| |
Collapse
|
8
|
Hu J, Wang J. From embryonic stem cells to induced pluripotent stem cells-Ready for clinical therapy? Clin Transplant 2019; 33:e13573. [PMID: 31013374 DOI: 10.1111/ctr.13573] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 04/18/2019] [Indexed: 01/08/2023]
Abstract
Embryonic stem cells and induced pluripotent stem cells have increasingly important roles in many different fields of research and medicine. Major areas of impact include improved in vitro disease models, drug screening, and the development of cell-based clinical therapies. Here, we review the generation and uses of embryonic stem cells compared to induced pluripotent stem cells and discuss their advantages and limitations. We also evaluate the feasibility of clinical therapies and the future prospects for induced pluripotent cell-based treatments.
Collapse
Affiliation(s)
- Jing Hu
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jimei Wang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
9
|
Asadzadeh Z, Mansoori B, Mohammadi A, Aghajani M, Haji‐Asgarzadeh K, Safarzadeh E, Mokhtarzadeh A, Duijf PHG, Baradaran B. microRNAs in cancer stem cells: Biology, pathways, and therapeutic opportunities. J Cell Physiol 2018; 234:10002-10017. [DOI: 10.1002/jcp.27885] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/13/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Student Research Committee, Tabriz University of Medical Sciences Tabriz Iran
| | - Ali Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Marjan Aghajani
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | | | - Elham Safarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
- Department of Microbiology & Immunology Faculty of Medicine, Ardabil University of Medical Sciences Ardabil Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| | - Pascal H. G. Duijf
- Translational Research Institute, University of Queensland Diamantina Institute, The University of Queensland Brisbane Queensland Australia
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences Tabriz Iran
| |
Collapse
|
10
|
Wang H, Unternaehrer JJ. Epithelial-mesenchymal Transition and Cancer Stem Cells: At the Crossroads of Differentiation and Dedifferentiation. Dev Dyn 2018; 248:10-20. [DOI: 10.1002/dvdy.24678] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/29/2018] [Accepted: 09/27/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Hanmin Wang
- Division of Biochemistry, Department of Basic Sciences; Loma Linda University; Loma Linda California
| | - Juli J. Unternaehrer
- Division of Biochemistry, Department of Basic Sciences; Loma Linda University; Loma Linda California
| |
Collapse
|
11
|
Malla S, Melguizo-Sanchis D, Aguilo F. Steering pluripotency and differentiation with N 6-methyladenosine RNA modification. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:394-402. [PMID: 30412796 DOI: 10.1016/j.bbagrm.2018.10.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/21/2018] [Accepted: 10/27/2018] [Indexed: 11/15/2022]
Abstract
Chemical modifications of RNA provide a direct and rapid way to modulate the existing transcriptome, allowing the cells to adapt rapidly to the changing environment. Among these modifications, N6-methyladenosine (m6A) has recently emerged as a widely prevalent mark of messenger RNA in eukaryotes, linking external stimuli to an intricate network of transcriptional, post-transcriptional and translational processes. m6A modification modulates a broad spectrum of biochemical processes, including mRNA decay, translation and splicing. Both m6A modification and the enzymes that control m6A metabolism are essential for normal development. In this review, we summarized the most recent findings on the role of m6A modification in maintenance of the pluripotency of embryonic stem cells (ESCs), cell fate specification, the reprogramming of somatic cells into induced pluripotent stem cells (iPSCs), and differentiation of stem and progenitor cells. This article is part of a Special Issue entitled: mRNA modifications in gene expression control edited by Dr. Soller Matthias and Dr. Fray Rupert.
Collapse
Affiliation(s)
- Sandhya Malla
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, SE-90185 Umeå, Sweden; Department of Medical Biosciences, Umeå University, SE-901 85 Umeå, Sweden
| | - Dario Melguizo-Sanchis
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, SE-90185 Umeå, Sweden; Department of Medical Biosciences, Umeå University, SE-901 85 Umeå, Sweden
| | - Francesca Aguilo
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, SE-90185 Umeå, Sweden; Department of Medical Biosciences, Umeå University, SE-901 85 Umeå, Sweden.
| |
Collapse
|
12
|
Sun Y, Kuek V, Liu Y, Tickner J, Yuan Y, Chen L, Zeng Z, Shao M, He W, Xu J. MiR-214 is an important regulator of the musculoskeletal metabolism and disease. J Cell Physiol 2018; 234:231-245. [PMID: 30076721 DOI: 10.1002/jcp.26856] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 05/10/2018] [Indexed: 12/21/2022]
Abstract
MiR-214 belongs to a family of microRNA (small, highly conserved noncoding RNA molecules) precursors that play a pivotal role in biological functions, such as cellular function, tissue development, tissue homeostasis, and pathogenesis of diseases. Recently, miR-214 emerged as a critical regulator of musculoskeletal metabolism. Specifically, miR-214 can mediate skeletal muscle myogenesis and vascular smooth muscle cell proliferation, migration, and differentiation. MiR-214 also modulates osteoblast function by targeting specific molecular pathways and the expression of various osteoblast-related genes; promotes osteoclast activity by targeting phosphatase and tensin homolog (Pten); and mediates osteoclast-osteoblast intercellular crosstalk via an exosomal miRNA paracrine mechanism. Importantly, dysregulation in miR-214 expression is associated with pathological bone conditions such as osteoporosis, osteosarcoma, multiple myeloma, and osteolytic bone metastasis of breast cancer. This review discusses the cellular targets of miR-214 in bone, the molecular mechanisms governing the activities of miR-214 in the musculoskeletal system, and the putative role of miR-214 in skeletal diseases. Understanding the biology of miR-214 could potentially lead to the development of miR-214 as a possible biomarker and a therapeutic target for musculoskeletal diseases.
Collapse
Affiliation(s)
- Youqiang Sun
- The Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.,Division of Pathology and Laboratory Medicine, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia.,The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Vincent Kuek
- Division of Pathology and Laboratory Medicine, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Yuhao Liu
- The Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.,Division of Pathology and Laboratory Medicine, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia.,The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jennifer Tickner
- Division of Pathology and Laboratory Medicine, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Yu Yuan
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, Guangdong, China
| | - Leilei Chen
- The Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.,The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhikui Zeng
- The Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.,The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Min Shao
- The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.,Department of Orthopedics, Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei He
- The Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.,The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jiake Xu
- Division of Pathology and Laboratory Medicine, School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia.,The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
13
|
Yan Y, Yang X, Li TT, Gu KL, Hao J, Zhang Q, Wang Y. Significant differences of function and expression of microRNAs between ground state and serum-cultured pluripotent stem cells. J Genet Genomics 2017; 44:179-189. [PMID: 28411033 DOI: 10.1016/j.jgg.2017.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 01/16/2017] [Accepted: 01/16/2017] [Indexed: 01/08/2023]
Abstract
Serum- and 2i-cultured embryonic stem cells (ESCs) show different epigenetic landscapes and transcriptomic profiles. The difference in the function and expression of microRNAs (miRNAs) between these two states remains unclear. Here, we showed that 2i- and serum-cultured ESCs exhibited distinctive miRNA expression profiles with >100 miRNAs differentially expressed, and the expression changes were largely due to transcriptional regulation. We further characterized the function of miRNAs differentially expressed under two conditions and found that ESCs exhibited higher degree of dependency on miRNAs for rapid proliferation; since Dgcr8-/- or Dicer1-/- but not wild-type ESCs showed slower growth rate and more accumulation in the G1 phase under 2i than serum condition. More interestingly, introduction of various self-renewal-silencing miRNAs in wild-type or Dgcr8-/- ESCs failed to silence the self-renewal in 2i medium, but regained the ability to silence the self-renewal upon the addition of serum. Our findings reveal significant differences in the expression and function of miRNAs between serum- and 2i-cultured ESCs.
Collapse
Affiliation(s)
- Ying Yan
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Xi Yang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Ting-Ting Li
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100871, China
| | - Kai-Li Gu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Jing Hao
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Qiang Zhang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China
| | - Yangming Wang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing 100871, China.
| |
Collapse
|
14
|
The Role of RNA Interference in Stem Cell Biology: Beyond the Mutant Phenotypes. J Mol Biol 2017; 429:1532-1543. [PMID: 28118980 DOI: 10.1016/j.jmb.2017.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/13/2017] [Accepted: 01/16/2017] [Indexed: 01/01/2023]
Abstract
Complex gene regulation systems ensure the maintenance of cellular identity during early development in mammals. Eukaryotic small RNAs have emerged as critical players in RNA interference (RNAi) by mediating gene silencing during embryonic stem cell self-renewal. Most of the proteins involved in the biogenesis of small RNAs are essential for proliferation and differentiation into the three germ layers of mouse embryonic stem cells. In the last decade, new functions for some RNAi proteins, independent of their roles in RNAi pathways, have been demonstrated in different biological systems. In parallel, new concepts in stem cell biology have emerged. Here, we review and integrate the current understanding of how RNAi proteins regulate stem cell identity with the new advances in the stem cell field and the recent non-canonical functions of the RNAi proteins. Finally, we propose a reevaluation of all RNAi mutant phenotypes, as non-canonical (small non-coding RNA independent) functions may contribute to the molecular mechanisms governing mouse embryonic stem cells commitment.
Collapse
|
15
|
Bodak M, Cirera-Salinas D, Yu J, Ngondo RP, Ciaudo C. Dicer, a new regulator of pluripotency exit and LINE-1 elements in mouse embryonic stem cells. FEBS Open Bio 2017; 7:204-220. [PMID: 28174687 PMCID: PMC5292673 DOI: 10.1002/2211-5463.12174] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/18/2016] [Accepted: 12/05/2016] [Indexed: 12/18/2022] Open
Abstract
A gene regulation network orchestrates processes ensuring the maintenance of cellular identity and genome integrity. Small RNAs generated by the RNAse III DICER have emerged as central players in this network. Moreover, deletion of Dicer in mice leads to early embryonic lethality. To better understand the underlying mechanisms leading to this phenotype, we generated Dicer‐deficient mouse embryonic stem cells (mESCs). Their detailed characterization revealed an impaired differentiation potential, and incapacity to exit from the pluripotency state. We also observed a strong accumulation of LINE‐1 (L1s) transcripts, which was translated at protein level and led to an increased L1s retrotransposition. Our findings reveal Dicer as a new essential player that sustains mESCs self‐renewal and genome integrity by controlling L1s regulation.
Collapse
Affiliation(s)
- Maxime Bodak
- Department of Biology RNAi and Genome Integrity IMHS Swiss Federal Institute of Technology Zurich Zurich Switzerland; Life Science Zurich Graduate School Molecular Life Science Program University of Zurich Switzerland
| | - Daniel Cirera-Salinas
- Department of Biology RNAi and Genome Integrity IMHS Swiss Federal Institute of Technology Zurich Zurich Switzerland
| | - Jian Yu
- Department of Biology RNAi and Genome Integrity IMHS Swiss Federal Institute of Technology Zurich Zurich Switzerland; Life Science Zurich Graduate School Molecular and Translational Biomedicine Program University of Zurich Switzerland
| | - Richard P Ngondo
- Department of Biology RNAi and Genome Integrity IMHS Swiss Federal Institute of Technology Zurich Zurich Switzerland
| | - Constance Ciaudo
- Department of Biology RNAi and Genome Integrity IMHS Swiss Federal Institute of Technology Zurich Zurich Switzerland
| |
Collapse
|
16
|
Agrawal R, Dale TP, Al-Zubaidi MA, Benny Malgulwar P, Forsyth NR, Kulshreshtha R. Pluripotent and Multipotent Stem Cells Display Distinct Hypoxic miRNA Expression Profiles. PLoS One 2016; 11:e0164976. [PMID: 27783707 PMCID: PMC5081191 DOI: 10.1371/journal.pone.0164976] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 10/04/2016] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs are reported to have a crucial role in the regulation of self-renewal and differentiation of stem cells. Hypoxia has been identified as a key biophysical element of the stem cell culture milieu however, the link between hypoxia and miRNA expression in stem cells remains poorly understood. We therefore explored miRNA expression in hypoxic human embryonic and mesenchymal stem cells (hESCs and hMSCs). A total of 50 and 76 miRNAs were differentially regulated by hypoxia (2% O2) in hESCs and hMSCs, respectively, with a negligible overlap of only three miRNAs. We found coordinate regulation of precursor and mature miRNAs under hypoxia suggesting their regulation mainly at transcriptional level. Hypoxia response elements were located upstream of 97% of upregulated hypoxia regulated miRNAs (HRMs) suggesting hypoxia-inducible-factor (HIF) driven transcription. HIF binding to the candidate cis-elements of specific miRNAs under hypoxia was confirmed by Chromatin immunoprecipitation coupled with qPCR. Role analysis of a subset of upregulated HRMs identified linkage to reported inhibition of differentiation while a downregulated subset of HRMs had a putative role in the promotion of differentiation. MiRNA-target prediction correlation with published hypoxic hESC and hMSC gene expression profiles revealed HRM target genes enriched in the cytokine:cytokine receptor, HIF signalling and pathways in cancer. Overall, our study reveals, novel and distinct hypoxia-driven miRNA signatures in hESCs and hMSCs with the potential for application in optimised culture and differentiation models for both therapeutic application and improved understanding of stem cell biology.
Collapse
Affiliation(s)
- Rahul Agrawal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi, India-110016
| | - Tina P. Dale
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, University of Keele, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire, ST4 7QB, United Kingdom
| | - Mohammed A. Al-Zubaidi
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, University of Keele, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire, ST4 7QB, United Kingdom
- College of Pharmacy, Al-Mustansiriyah University, Baghdad, Iraq
| | - Prit Benny Malgulwar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India-110029
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, University of Keele, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire, ST4 7QB, United Kingdom
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi, India-110016
| |
Collapse
|
17
|
AK048794 maintains the mouse embryonic stem cell pluripotency by functioning as an miRNA sponge for miR-592. Biochem J 2016; 473:3639-3654. [PMID: 27520307 DOI: 10.1042/bcj20160540] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 08/12/2016] [Indexed: 11/17/2022]
Abstract
MiR-592 has been identified as a neural-enriched microRNA, plays an important role in mNPCs differentiation, could induce astrogliogenesis differentiation arrest or/and enhance neurogenesis in vitro Previous studies showed that long noncoding RNAs (lncRNAs) were involved in the neuronal development and activity. To investigate the role of miR-592 in neurogenesis, we described the expression profile of lncRNAs in miR-592 knockout mouse embryonic stem cells (mESCs) and the corresponding normal mESCs by microarray. By the microarray analysis and luciferase reporter assays, we demonstrated that lncRNA - AK048794, regulated by transcription factor GATA1, functioned as a competing endogenous RNA (ceRNA) for miR-592 and led to the de-repression of its endogenous target FAM91A1, which is involved in mESC pluripotency maintenance. Taken together, these observations imply that AK048794 modulated the expression of multiple genes involved in mESC pluripotency maintenance by acting as a ceRNA for miR-592, which may build up the link between the regulatory miRNA network and mESC pluripotency.
Collapse
|
18
|
Zhu CM, Yan F. Functions of large intergenic non-coding RNA-regulator of reprogramming. Shijie Huaren Xiaohua Zazhi 2016; 24:331-337. [DOI: 10.11569/wcjd.v24.i3.331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In the human genome, there are a large number of non-coding RNAs (ncRNAs). These ncRNAs have attracted considerable attention in recent years. Large intergenic ncRNA-regulator of reprogramming (lincRNA-ROR), a newly identified lincRNA, was initially found to regulate the process of reprogramming. Studies have indicated that lincRNA-ROR has an important role in induced pluripotent stem cells (iPSCs), DNA damage, and oxidative stress. Besides, it has been shown to be dysregulated in many types of cancer, including breast cancer and hepatocellular carcinoma. ROR functions as a regulatory molecule in a wide variety of biological processes. However, its mechanism of action remains unclear. In this review, we will focus on the research background, functions, and characteristics of ROR, as well as its regulatory mechanisms and its association with cancers.
Collapse
|
19
|
Hinton A, Hunter SE, Afrikanova I, Jones GA, Lopez AD, Fogel GB, Hayek A, King CC. sRNA-seq analysis of human embryonic stem cells and definitive endoderm reveals differentially expressed microRNAs and novel IsomiRs with distinct targets. Stem Cells 2015; 32:2360-72. [PMID: 24805944 DOI: 10.1002/stem.1739] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 03/30/2014] [Accepted: 04/09/2014] [Indexed: 11/06/2022]
Abstract
MicroRNAs (miRNAs) are noncoding, regulatory RNAs expressed dynamically during differentiation of human embryonic stem cells (hESCs) into defined lineages. Mapping developmental expression of miRNAs during transition from pluripotency to definitive endoderm (DE) should help to elucidate the mechanisms underlying lineage specification and ultimately enhance differentiation protocols. In this report, next generation sequencing was used to build upon our previous analysis of miRNA expression in human hESCs and DE. From millions of sequencing reads, 747 and 734 annotated miRNAs were identified in pluripotent and DE cells, respectively, including 77 differentially expressed miRNAs. Among these, four of the top five upregulated miRNAs were previously undetected in DE. Furthermore, the stem-loop for miR-302a, an important miRNA for both hESCs self-renewal and endoderm specification, produced several highly expressed miRNA species (isomiRs). Overall, isomiRs represented >10% of sequencing reads in >40% of all detected stem-loop arms, suggesting that the impact of these abundant miRNA species may have been overlooked in previous studies. Because of their relative abundance, the role of differential isomiR targeting was studied using the miR-302 cluster as a model system. A miRNA mimetic for miR-302a-5p, but not miR-302a-5p(+3), decreased expression of orthodenticle homeobox 2 (OTX2). Conversely, isomiR 302a-5p(+3) selectively decreased expression of tuberous sclerosis protein 1, but not OTX2, indicating nonoverlapping specificity of miRNA processing variants. Taken together, our characterization of miRNA expression, which includes novel miRNAs and isomiRs, helps establish a foundation for understanding the role of miRNAs in DE formation and selective targeting by isomiRs.
Collapse
Affiliation(s)
- Andrew Hinton
- Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, California, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
MicroRNA dynamics during human embryonic stem cell differentiation to pancreatic endoderm. Gene 2015; 574:359-70. [PMID: 26297998 DOI: 10.1016/j.gene.2015.08.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 08/12/2015] [Indexed: 11/23/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that have emerged as critical regulators of human embryonic stem cell (hESC) pluripotency and differentiation. Despite the wealth of information about the role individual that miRNAs play in these two processes, there has yet to be a large-scale temporal analysis of the dynamics of miRNA expression as hESCs move from pluripotency into defined lineages. In this report, we used Next Generation Sequencing (NGS) to map temporal expression of miRNAs over ten 24-hour intervals as pluripotent cells were differentiated into pancreatic endoderm. Of the 2042 known human miRNAs, 694 had non-zero expression on all 11 days. Of these 694 miRNAs, 494 showed statistically significant changes in expression during differentiation. Clusters of miRNAs were identified, each displaying unique expression profiles distributed over multiple days. Selected miRNAs associated with pluripotency/differentiation (miR-302/367 and miR-371/372/373) and development/growth (miR-21, miR-25, miR-103, miR-9, and miR-92a) were found to have distinct expression profiles correlated with changes in media used to drive the differentiation process. Taken together, the clustering of miRNAs to identify expression dynamics that occur over longer periods of time (days vs. hours) provides unique insight into specific stages of differentiation. Major shifts in defined stages of hESC differentiation appear to be heavily dependent upon changes in external environmental factors, rather than intrinsic conditions in the cells.
Collapse
|
21
|
El-Badawy A, El-Badri N. Regulators of pluripotency and their implications in regenerative medicine. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2015; 8:67-80. [PMID: 25960670 PMCID: PMC4410894 DOI: 10.2147/sccaa.s80157] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The ultimate goal of regenerative medicine is to replace damaged tissues with new functioning ones. This can potentially be accomplished by stem cell transplantation. While stem cell transplantation for blood diseases has been increasingly successful, widespread application of stem cell therapy in the clinic has shown limited results. Despite successful efforts to refine existing methodologies and to develop better ones for reprogramming, clinical application of stem cell therapy suffers from issues related to the safety of the transplanted cells, as well as the low efficiency of reprogramming technology. Better understanding of the underlying mechanism(s) involved in pluripotency should accelerate the clinical application of stem cell transplantation for regenerative purposes. This review outlines the main decision-making factors involved in pluripotency, focusing on the role of microRNAs, epigenetic modification, signaling pathways, and toll-like receptors. Of special interest is the role of toll-like receptors in pluripotency, where emerging data indicate that the innate immune system plays a vital role in reprogramming. Based on these data, we propose that nongenetic mechanisms for reprogramming provide a novel and perhaps an essential strategy to accelerate application of regenerative medicine in the clinic.
Collapse
Affiliation(s)
- Ahmed El-Badawy
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine, Zewail City of Science and Technology, Giza, Egypt
| |
Collapse
|
22
|
Kapinas K, Kim H, Mandeville M, Martin-Buley LA, Croce CM, Lian JB, van Wijnen AJ, Stein JL, Altieri DC, Stein GS. microRNA-mediated survivin control of pluripotency. J Cell Physiol 2015; 230:63-70. [PMID: 24891298 DOI: 10.1002/jcp.24681] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 05/20/2014] [Indexed: 01/25/2023]
Abstract
Understanding the mechanisms that sustain pluripotency in human embryonic stem cells (hESCs) is an active area of research that may prove useful in regenerative medicine and will provide fundamental information relevant to development and cancer. hESCs and cancer cells share the unique ability to proliferate indefinitely and rapidly. Because the protein survivin is uniquely overexpressed in virtually all human cancers and in hESCs, we sought to investigate its role in supporting the distinctive capabilities of these cell types. Results presented here suggest that survivin contributes to the maintenance of pluripotency and that post-transcriptional control of survivin isoform expression is selectively regulated by microRNAs. miR-203 has been extensively studied in human tumors, but has not been characterized in hESCs. We show that miR-203 expression and activity is consistent with the expression and subcellular localization of survivin isoforms that in turn modulate expression of the Oct4 and Nanog transcription factors to sustain pluripotency. This study contributes to understanding of the complex regulatory mechanisms that govern whether hESCs proliferate or commit to lineages.
Collapse
|
23
|
Nair R, Santos L, Awasthi S, von Erlach T, Chow LW, Bertazzo S, Stevens MM. Extracellular Vesicles Derived from Preosteoblasts Influence Embryonic Stem Cell Differentiation. Stem Cells Dev 2014; 23:1625-35. [DOI: 10.1089/scd.2013.0633] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Rekha Nair
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Lívia Santos
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Siddhant Awasthi
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | - Thomas von Erlach
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Lesley W. Chow
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Sergio Bertazzo
- Department of Materials, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| | - Molly M. Stevens
- Department of Materials, Imperial College London, London, United Kingdom
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Institute of Biomedical Engineering, Imperial College London, London, United Kingdom
| |
Collapse
|
24
|
Bhaskaran M, Mohan M. MicroRNAs: history, biogenesis, and their evolving role in animal development and disease. Vet Pathol 2014; 51:759-74. [PMID: 24045890 PMCID: PMC4013251 DOI: 10.1177/0300985813502820] [Citation(s) in RCA: 423] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The discovery of microRNAs (miRNAs) in 1993 followed by developments and discoveries in small RNA biology have redefined the biological landscape by significantly altering the longstanding dogmas that defined gene regulation. These small RNAs play a significant role in modulation of an array of physiological and pathological processes ranging from embryonic development to neoplastic progression. Unique miRNA signatures of various inherited, metabolic, infectious, and neoplastic diseases have added a new dimension to the studies that look at their pathogenesis and highlight their potential to be reliable biomarkers. Also, altering miRNA functionality and the development of novel in vivo delivery systems to achieve targeted modulation of specific miRNA function are being actively pursued as novel approaches for therapeutic intervention in many diseases. Here we review the current body of knowledge on the role of miRNAs in development and disease and discuss future implications.
Collapse
Affiliation(s)
- M Bhaskaran
- Infectious Disease Aerobiology, Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - M Mohan
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| |
Collapse
|
25
|
Du J, Wu Y, Ai Z, Shi X, Chen L, Guo Z. Mechanism of SB431542 in inhibiting mouse embryonic stem cell differentiation. Cell Signal 2014; 26:2107-16. [PMID: 24949833 DOI: 10.1016/j.cellsig.2014.06.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/09/2014] [Accepted: 06/09/2014] [Indexed: 12/22/2022]
Abstract
SB431542 (SB) is an established small molecular inhibitor that specifically binds to the ATP binding domains of the activin receptor-like kinase receptors, ALK5, ALK4 and ALK7, and thus specifically inhibits Smad2/3 activation and blocks TGF-β signal transduction. SB maintains the undifferentiated state of mouse embryonic stem cells. However, the way of SB in maintaining the undifferentiated state of mouse embryonic stem cells remains unclear. Considering that SB could not maintain embryonic stem cells pluripotency when leukemia inhibitory factor was withdrawn, we sought to identify the mechanism of SB on pluripotent maintenance. Transcripts regulated by SB, including message RNAs and small non-coding RNAs were examined through microarray and deep-sequence experiments. After examination, Western blot analysis, and quantitative real-time PCR verification, we found that SB regulated the transcript expressions related to self-renewal and differentiation. SB mainly functioned by inhibiting differentiation. The key pluripotent factors expression were not significantly affected by SB, and intrinsic differentiation-related transcripts including fibroblast growth factor family members, were significantly down-regulated by SB. Moreover, SB could partially inhibit the retinoic acid response to neuronal differentiation of mouse embryonic stem cells.
Collapse
Affiliation(s)
- Juan Du
- College of Life Sciences, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China
| | - Yongyan Wu
- College of Veterinary Medicine, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China
| | - Zhiying Ai
- College of Life Sciences, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China
| | - Xiaoyan Shi
- College of Life Sciences, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China
| | - Linlin Chen
- College of Life Sciences, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China
| | - Zekun Guo
- College of Veterinary Medicine, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China; Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, 3 Taicheng Road, Yangling 712100, Shaanxi, China.
| |
Collapse
|
26
|
Ryazansky SS, Mikhaleva EA, Olenkina OV. Essential functions of microRNAs in animal reproductive organs. Mol Biol 2014. [DOI: 10.1134/s0026893314030182] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
27
|
Lou CH, Shao A, Shum EY, Espinoza JL, Huang L, Karam R, Wilkinson MF. Posttranscriptional control of the stem cell and neurogenic programs by the nonsense-mediated RNA decay pathway. Cell Rep 2014; 6:748-64. [PMID: 24529710 DOI: 10.1016/j.celrep.2014.01.028] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 12/11/2013] [Accepted: 01/21/2014] [Indexed: 11/19/2022] Open
Abstract
The mechanisms dictating whether a cell proliferates or differentiates have undergone intense scrutiny, but they remain poorly understood. Here, we report that UPF1, a central component in the nonsense-mediated RNA decay (NMD) pathway, plays a key role in this decision by promoting the proliferative, undifferentiated cell state. UPF1 acts, in part, by destabilizing the NMD substrate encoding the TGF-β inhibitor SMAD7 and stimulating TGF-β signaling. UPF1 also promotes the decay of mRNAs encoding many other proteins that oppose the proliferative, undifferentiated cell state. Neural differentiation is triggered when NMD is downregulated by neurally expressed microRNAs (miRNAs). This UPF1-miRNA circuitry is highly conserved and harbors negative feedback loops that act as a molecular switch. Our results suggest that the NMD pathway collaborates with the TGF-β signaling pathway to lock in the stem-like state, a cellular state that is stably reversed when neural differentiation signals that induce NMD-repressive miRNAs are received.
Collapse
Affiliation(s)
- Chih H Lou
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093-0695, USA
| | - Ada Shao
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093-0695, USA
| | - Eleen Y Shum
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093-0695, USA
| | - Josh L Espinoza
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093-0695, USA
| | - Lulu Huang
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093-0695, USA
| | - Rachid Karam
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093-0695, USA
| | - Miles F Wilkinson
- Department of Reproductive Medicine, School of Medicine, University of California, San Diego, La Jolla, CA 92093-0695, USA; Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA 92093-0695, USA.
| |
Collapse
|
28
|
Meng X, Leslie P, Zhang Y, Dong J. Stem cells in a three-dimensional scaffold environment. SPRINGERPLUS 2014; 3:80. [PMID: 24570851 PMCID: PMC3931863 DOI: 10.1186/2193-1801-3-80] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 01/31/2014] [Indexed: 02/08/2023]
Abstract
Stem cells have emerged as important players in the generation and maintenance of many tissues. However, the accurate in vitro simulation of the native stem cell niche remains difficult due at least in part to the lack of a comprehensive definition of the critical factors of the stem cell niche based on in vivo models. Three-dimensional (3D) cell culture systems have allowed the development of useful models for investigating stem cell physiology particularly with respect to their ability to sense and generate mechanical force in response to their surrounding environment. We review the use of 3D culture systems for stem cell culture and discuss the relationship between stem cells and 3D growth matrices including the roles of the extracellular matrix, scaffolds, soluble factors, cell-cell interactions and shear stress effects within this environment. We also discuss the potential for novel methods that mimic the native stem cell niche in vitro as well as the current associated challenges.
Collapse
Affiliation(s)
- Xuan Meng
- Hospital & Institute of Hepatobiliary Surgery, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853 China ; Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7512 USA ; Hospital & Institute of Hepatobiliary Surgery, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853 China
| | - Patrick Leslie
- Hospital & Institute of Hepatobiliary Surgery, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853 China ; Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7512 USA ; Hospital & Institute of Hepatobiliary Surgery, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853 China
| | - Yanping Zhang
- Hospital & Institute of Hepatobiliary Surgery, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853 China ; Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7512 USA ; Department of Pharmacology, School of Medicine, University of North Carolina, Chapel Hill, NC 27599-7512 USA
| | - Jiahong Dong
- Hospital & Institute of Hepatobiliary Surgery, Chinese PLA General Hospital, Fuxing Road 28, Haidian District, Beijing, 100853 China
| |
Collapse
|
29
|
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
|
30
|
MicroRNAs are involved in the self-renewal and differentiation of cancer stem cells. Acta Pharmacol Sin 2013; 34:1374-80. [PMID: 24122008 DOI: 10.1038/aps.2013.134] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Accepted: 08/22/2013] [Indexed: 12/23/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules, whose primary function is to regulate gene expression at the post-transcriptional/translational levels. MiRNAs play crucial roles in normal biological processes and are commonly dys-regulated in human diseases. Stem cells are regarded as the "mother" cells of all types of differentiated cells that comprise tissues and organs of the body. A novel hypothesis proposes that tumors are composed of heterogeneous cells derived from cancer stem cells, which have self-renewal and differentiation capabilities similar to those of normal stem cells. Cancer stem cells have been isolated and characterized from various tumors. Given recent studies supporting the critical regulatory roles of miRNAs in the self-renewal and differentiation of cancer stem cells, better understanding the functions of miRNAs will provide invaluable insights into the prevention of tumorigenesis and tumor progression. In this review, we will summarize the research progress in the study of miRNAs involved in the self-renewal and differentiation of cancer stem cells.
Collapse
|
31
|
Gómez-Cabello D, Adrados I, Gamarra D, Kobayashi H, Takatsu Y, Takatsu K, Gil J, Palmero I. DGCR8-mediated disruption of miRNA biogenesis induces cellular senescence in primary fibroblasts. Aging Cell 2013; 12:923-31. [PMID: 23773483 DOI: 10.1111/acel.12117] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2013] [Indexed: 12/21/2022] Open
Abstract
The regulation of gene expression by microRNAs (miRNAs) is critical for normal development and physiology. Conversely, miRNA function is frequently impaired in cancer, and other pathologies, either by aberrant expression of individual miRNAs or dysregulation of miRNA synthesis. Here, we have investigated the impact of global disruption of miRNA biogenesis in primary fibroblasts of human or murine origin, through the knockdown of DGCR8, an essential mediator of the synthesis of canonical miRNAs. We find that the inactivation of DGCR8 in these cells results in a dramatic antiproliferative response, with the acquisition of a senescent phenotype. Senescence triggered by DGCR8 loss is accompanied by the upregulation of the cell-cycle inhibitor p21CIP1. We further show that a subset of senescence-associated miRNAs with the potential to target p21CIP1 is downregulated during DGCR8-mediated senescence. Interestingly, the antiproliferative response to miRNA biogenesis disruption is retained in human tumor cells, irrespective of p53 status. In summary, our results show that defective synthesis of canonical microRNAs results in cell-cycle arrest and cellular senescence in primary fibroblasts mediated by specific miRNAs, and thus identify global miRNA disruption as a novel senescence trigger.
Collapse
Affiliation(s)
| | - Isabel Adrados
- Instituto de Investigaciones Biomédicas ‘Alberto Sols’ CSIC-UAM; Madrid; Spain
| | - David Gamarra
- Instituto de Investigaciones Biomédicas ‘Alberto Sols’ CSIC-UAM; Madrid; Spain
| | - Hikaru Kobayashi
- Instituto de Investigaciones Biomédicas ‘Alberto Sols’ CSIC-UAM; Madrid; Spain
| | - Yoshihiro Takatsu
- Cell Proliferation Group; MRC Clinical Sciences Centre; Imperial College London; London; UK
| | - Kyoko Takatsu
- Cell Proliferation Group; MRC Clinical Sciences Centre; Imperial College London; London; UK
| | - Jesús Gil
- Cell Proliferation Group; MRC Clinical Sciences Centre; Imperial College London; London; UK
| | - Ignacio Palmero
- Instituto de Investigaciones Biomédicas ‘Alberto Sols’ CSIC-UAM; Madrid; Spain
| |
Collapse
|
32
|
The Lin28/let-7a/c-Myc pathway plays a role in non-muscle invasive bladder cancer. Cell Tissue Res 2013; 354:533-41. [PMID: 24036903 DOI: 10.1007/s00441-013-1715-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 07/19/2013] [Indexed: 12/23/2022]
Abstract
We investigate the role of the Lin28/let-7a/c-Myc pathway in non-muscle invasive bladder cancer (NMIBC). Using RT-PCR, western blot and immunohistochemistry techniques, the levels of pre-let-7a, let-7a, Lin28 and c-Myc RNA and/or proteins were determined in samples of normal bladder tissue and bladder cancer. Expression of pre-let-7a was found to be negatively correlated with the pathological grade of bladder cancer, while let-7a showed a positive correlation with bladder cancer pathological grade. Expression of Lin28 RNA and protein was not significantly different between normal bladder tissue and low-grade transitional cell carcinoma of bladder (TCC) but the expression levels in high-grade TCC were remarkably increased. Expression of c-Myc RNA and protein was significantly higher in bladder cancer samples in comparison to normal bladder tissue without correlation with cancer differentiation. Expression of all the above RNAs and proteins showed no significant difference in Ta and T1 stages. The Lin28/let-7a/c-Myc pathway plays an important role in NMIBC. In particular, expression levels of let-7a correlate with the degree of cancer differentiation but not cancer stage.
Collapse
|
33
|
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
|
34
|
Hsu DM, Agarwal S, Benham A, Coarfa C, Trahan DN, Chen Z, Stowers PN, Courtney AN, Lakoma A, Barbieri E, Metelitsa LS, Gunaratne P, Kim ES, Shohet JM. G-CSF receptor positive neuroblastoma subpopulations are enriched in chemotherapy-resistant or relapsed tumors and are highly tumorigenic. Cancer Res 2013; 73:4134-46. [PMID: 23687340 DOI: 10.1158/0008-5472.can-12-4056] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Neuroblastoma is a neural crest-derived embryonal malignancy, which accounts for 13% of all pediatric cancer mortality, primarily due to tumor recurrence. Therapy-resistant cancer stem cells are implicated in tumor relapse, but definitive phenotypic evidence of the existence of these cells has been lacking. In this study, we define a highly tumorigenic subpopulation in neuroblastoma with stem cell characteristics, based on the expression of CSF3R, which encodes the receptor for granulocyte colony-stimulating factor (G-CSF). G-CSF receptor positive (aka G-CSFr(+) or CD114(+)) cells isolated from a primary tumor and the NGP cell line by flow cytometry were highly tumorigenic and capable of both self-renewal and differentiation to progeny cells. CD114(+) cells closely resembled embryonic and induced pluripotent stem cells with respect to their profiles of cell cycle, miRNA, and gene expression. In addition, they reflect a primitive undifferentiated neuroectodermal/neural crest phenotype revealing a developmental hierarchy within neuroblastoma tumors. We detected this dedifferentiated neural crest subpopulation in all established neuroblastoma cell lines, xenograft tumors, and primary tumor specimens analyzed. Ligand activation of CD114 by the addition of exogenous G-CSF to CD114(+) cells confirmed intact STAT3 upregulation, characteristic of G-CSF receptor signaling. Together, our data describe a novel distinct subpopulation within neuroblastoma with enhanced tumorigenicity and a stem cell-like phenotype, further elucidating the complex heterogeneity of solid tumors such as neuroblastoma. We propose that this subpopulation may represent an additional target for novel therapeutic approaches to this aggressive pediatric malignancy.
Collapse
Affiliation(s)
- Danielle M Hsu
- Division of Pediatric Surgery, Michael E DeBakey Department of Surgery, Section of Hematology-Oncology, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kramer AS, Harvey AR, Plant GW, Hodgetts SI. Systematic Review of Induced Pluripotent Stem Cell Technology as a Potential Clinical Therapy for Spinal Cord Injury. Cell Transplant 2013; 22:571-617. [DOI: 10.3727/096368912x655208] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Transplantation therapies aimed at repairing neurodegenerative and neuropathological conditions of the central nervous system (CNS) have utilized and tested a variety of cell candidates, each with its own unique set of advantages and disadvantages. The use and popularity of each cell type is guided by a number of factors including the nature of the experimental model, neuroprotection capacity, the ability to promote plasticity and guided axonal growth, and the cells' myelination capability. The promise of stem cells, with their reported ability to give rise to neuronal lineages to replace lost endogenous cells and myelin, integrate into host tissue, restore functional connectivity, and provide trophic support to enhance and direct intrinsic regenerative ability, has been seen as a most encouraging step forward. The advent of the induced pluripotent stem cell (iPSC), which represents the ability to “reprogram” somatic cells into a pluripotent state, hails the arrival of a new cell transplantation candidate for potential clinical application in therapies designed to promote repair and/or regeneration of the CNS. Since the initial development of iPSC technology, these cells have been extensively characterized in vitro and in a number of pathological conditions and were originally reported to be equivalent to embryonic stem cells (ESCs). This review highlights emerging evidence that suggests iPSCs are not necessarily indistinguishable from ESCs and may occupy a different “state” of pluripotency with differences in gene expression, methylation patterns, and genomic aberrations, which may reflect incomplete reprogramming and may therefore impact on the regenerative potential of these donor cells in therapies. It also highlights the limitations of current technologies used to generate these cells. Moreover, we provide a systematic review of the state of play with regard to the use of iPSCs in the treatment of neurodegenerative and neuropathological conditions. The importance of balancing the promise of this transplantation candidate in the light of these emerging properties is crucial as the potential application in the clinical setting approaches. The first of three sections in this review discusses (A) the pathophysiology of spinal cord injury (SCI) and how stem cell therapies can positively alter the pathology in experimental SCI. Part B summarizes (i) the available technologies to deliver transgenes to generate iPSCs and (ii) recent data comparing iPSCs to ESCs in terms of characteristics and molecular composition. Lastly, in (C) we evaluate iPSC-based therapies as a candidate to treat SCI on the basis of their neurite induction capability compared to embryonic stem cells and provide a summary of available in vivo data of iPSCs used in SCI and other disease models.
Collapse
Affiliation(s)
- Anne S. Kramer
- Spinal Cord Repair Laboratory, School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Western Australia
| | - Alan R. Harvey
- Spinal Cord Repair Laboratory, School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Western Australia
| | - Giles W. Plant
- Stanford Partnership for Spinal Cord Injury and Repair, Stanford Institute for Neuro-Innovation and Translational Neurosciences, Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Stuart I. Hodgetts
- Spinal Cord Repair Laboratory, School of Anatomy, Physiology and Human Biology, The University of Western Australia, Perth, Western Australia
| |
Collapse
|
36
|
Endogenous miRNA Sponge lincRNA-RoR Regulates Oct4, Nanog, and Sox2 in Human Embryonic Stem Cell Self-Renewal. Dev Cell 2013; 25:69-80. [DOI: 10.1016/j.devcel.2013.03.002] [Citation(s) in RCA: 618] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 01/23/2013] [Accepted: 03/01/2013] [Indexed: 12/12/2022]
|
37
|
Garg M. MicroRNAs, stem cells and cancer stem cells. World J Stem Cells 2012; 4:62-70. [PMID: 22993663 PMCID: PMC3443713 DOI: 10.4252/wjsc.v4.i7.62] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 04/18/2012] [Accepted: 04/25/2012] [Indexed: 02/06/2023] Open
Abstract
This review discusses the various regulatory characteristics of microRNAs that are capable of generating widespread changes in gene expression via post translational repression of many mRNA targets and control self-renewal, differentiation and division of cells. It controls the stem cell functions by controlling a wide range of pathological and physiological processes, including development, differentiation, cellular proliferation, programmed cell death, oncogenesis and metastasis. Through either mRNA cleavage or translational repression, miRNAs alter the expression of their cognate target genes; thereby modulating cellular pathways that affect the normal functions of stem cells, turning them into cancer stem cells, a likely cause of relapse in cancer patients. This present review further emphasizes the recent discoveries on the functional analysis of miRNAs in cancer metastasis and implications on miRNA based therapy using miRNA replacement or anti-miRNA technologies in specific cancer stem cells that are required to establish their efficacy in controlling tumorigenic potential and safe therapeutics.
Collapse
Affiliation(s)
- Minal Garg
- Minal Garg, Department of Biochemistry, University of Lucknow, Lucknow 226007, India
| |
Collapse
|
38
|
MicroRNA regulation of Cbx7 mediates a switch of Polycomb orthologs during ESC differentiation. Cell Stem Cell 2012; 10:33-46. [PMID: 22226354 PMCID: PMC3277884 DOI: 10.1016/j.stem.2011.12.004] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 10/11/2011] [Accepted: 12/02/2011] [Indexed: 12/20/2022]
Abstract
The Polycomb Group (PcG) of chromatin modifiers regulates pluripotency and differentiation. Mammalian genomes encode multiple homologs of the Polycomb repressive complex 1 (PRC1) components, including five orthologs of the Drosophila Polycomb protein (Cbx2, Cbx4, Cbx6, Cbx7, and Cbx8). We have identified Cbx7 as the primary Polycomb ortholog of PRC1 complexes in embryonic stem cells (ESCs). The expression of Cbx7 is downregulated during ESC differentiation, preceding the upregulation of Cbx2, Cbx4, and Cbx8, which are directly repressed by Cbx7. Ectopic expression of Cbx7 inhibits differentiation and X chromosome inactivation and enhances ESC self-renewal. Conversely, Cbx7 knockdown induces differentiation and derepresses lineage-specific markers. In a functional screen, we identified the miR-125 and miR-181 families as regulators of Cbx7 that are induced during ESC differentiation. Ectopic expression of these miRNAs accelerates ESC differentiation via regulation of Cbx7. These observations establish a critical role for Cbx7 and its regulatory miRNAs in determining pluripotency.
Collapse
|
39
|
Camahort R, Cowan CA. Cbx proteins help ESCs walk the line between self-renewal and differentiation. Cell Stem Cell 2012; 10:4-6. [PMID: 22226347 DOI: 10.1016/j.stem.2011.12.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The Polycomb repressive complexes (PRC) regulate self-renewal and differentiation in embryonic stem cells (ESCs). In this issue of Cell Stem Cell, Morey et al. (2012) and O'Loghlen et al. (2012) report that dynamic interchange of PRC subunits modulates the balance between self-renewal and lineage commitment in ESCs.
Collapse
Affiliation(s)
- Raymond Camahort
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | | |
Collapse
|
40
|
Abstract
Recent months have seen rapid advances in the field of transdifferentiation, specifically in the conversion of fibroblasts to neurons. Most surprising is the observation that the ability to drive these transitions is not limited to transcription factors, but that they can be promoted by microRNAs as well. Indeed, in one case, microRNAs alone induced the transdifferentiation of fibroblasts to neuron-like cells, albeit at a low efficiency. Here, we review this rapidly advancing field, discuss possible mechanisms underlying microRNA-induced transdifferentiation and the potential for microRNAs to drive such transitions to any cell type of interest in vitro and in vivo.
Collapse
Affiliation(s)
- Archana Shenoy
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences Department of Urology, 35 Medical Center Way, Pod B 1018, University of California, San Francisco, San Francisco, CA. 94143 - 0667 USA
| | | |
Collapse
|
41
|
Cassar PA, Stanford WL. Integrating post-transcriptional regulation into the embryonic stem cell gene regulatory network. J Cell Physiol 2012; 227:439-49. [PMID: 21503874 DOI: 10.1002/jcp.22787] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Stem cell behavior is orchestrated as a multilayered, concert of gene regulatory mechanisms collectively referred to as the gene regulatory network (GRN). Via cooperative mechanisms, transcriptional, epigenetic, and post-transcriptional regulators activate and repress gene expression to finely regulate stem cell self-renewal and commitment. Due to their tractability, embryonic stem cells (ESCs) serve as the model stem cell to dissect the complexities of the GRN, and discern its relation to stem cell fate. By way of high-throughput genomic analysis, targets of individual gene regulators have been established in ESCs. The compilation of these discrete networks has revealed convergent, multi-dimensional gene regulatory mechanisms involving transcription factors, epigenetic modifiers, non-coding RNA (ncRNA), and RNA-binding proteins. Here we highlight the seminal genomic studies that have shaped our understanding of the ESC GRN and describe alternate post-transcriptional gene regulatory mechanisms that require in depth analyses to draft networks that fully model ESC behavior.
Collapse
Affiliation(s)
- Paul A Cassar
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
42
|
Babiarz JE, Ravon M, Sridhar S, Ravindran P, Swanson B, Bitter H, Weiser T, Chiao E, Certa U, Kolaja KL. Determination of the human cardiomyocyte mRNA and miRNA differentiation network by fine-scale profiling. Stem Cells Dev 2012; 21:1956-65. [PMID: 22050602 DOI: 10.1089/scd.2011.0357] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To gain insight into the molecular regulation of human heart development, a detailed comparison of the mRNA and miRNA transcriptomes across differentiating human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes and biopsies from fetal, adult, and hypertensive human hearts was performed. Gene ontology analysis of the mRNA expression levels of the hiPSCs differentiating into cardiomyocytes revealed 3 distinct groups of genes: pluripotent specific, transitional cardiac specification, and mature cardiomyocyte specific. Hierarchical clustering of the mRNA data revealed that the transcriptome of hiPSC cardiomyocytes largely stabilizes 20 days after initiation of differentiation. Nevertheless, analysis of cells continuously cultured for 120 days indicated that the cardiomyocytes continued to mature toward a more adult-like gene expression pattern. Analysis of cardiomyocyte-specific miRNAs (miR-1, miR-133a/b, and miR-208a/b) revealed an miRNA pattern indicative of stem cell to cardiomyocyte specification. A biostatistitical approach integrated the miRNA and mRNA expression profiles revealing a cardiomyocyte differentiation miRNA network and identified putative mRNAs targeted by multiple miRNAs. Together, these data reveal the miRNA network in human heart development and support the notion that overlapping miRNA networks re-enforce transcriptional control during developmental specification.
Collapse
Affiliation(s)
- Joshua E Babiarz
- Nonclinical Safety, Hoffmann-La Roche, Nutley, New Jersey 07110, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Hinton A, Hunter S, Reyes G, Fogel GB, King CC. From pluripotency to islets: miRNAs as critical regulators of human cellular differentiation. ADVANCES IN GENETICS 2012; 79:1-34. [PMID: 22989764 DOI: 10.1016/b978-0-12-394395-8.00001-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNAs) actively regulate differentiation as pluripotent cells become cells of pancreatic endocrine lineage, including insulin-producing β cells. The process is dynamic; some miRNAs help maintain pluripotency, while others drive cell fate decisions. Here, we survey the current literature and describe the biological role of selected miRNAs in maintenance of both mouse and human embryonic stem cell (ESC) pluripotency. Subsequently, we review the increasing evidence that miRNAs act at selected points in differentiation to regulate decisions about early cell fate (definitive endoderm and mesoderm), formation of pancreatic precursor cells, endocrine cell function, as well as epithelial to mesenchymal transition.
Collapse
Affiliation(s)
- Andrew Hinton
- Pediatric Diabetes Research Center, University of California, San Diego, La Jolla, CA, USA
| | | | | | | | | |
Collapse
|
44
|
MicroRNAs regulating cell pluripotency and vascular differentiation. Vascul Pharmacol 2011; 55:69-78. [PMID: 21854874 DOI: 10.1016/j.vph.2011.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 07/26/2011] [Accepted: 08/02/2011] [Indexed: 12/12/2022]
Abstract
Human embryonic stem cells (hESC) offer broad potential for regenerative medicine owing to their capacity for self renewal, exponential scale up and differentiation into any cell type in the adult body. hESC have been proposed as a potentially unlimited source for the generation of transplantable, healthy, functional vascular cells for repair of ischemic tissues. To optimally harness this potential necessitates precise control over biological processes that govern maintenance, pluripotency and cell differentiation including signalling cascades, gene expression profiles and epigenetic modification. Such control may be elicited by microRNAs, which are powerful negative regulators of gene expression. Here, we review the role for miRNAs in both the maintenance of pluripotency and differentiation of cells to a cardiovascular lineage including endothelial cells, vascular smooth muscle cells and cardiomyocytes and put this into context for regenerative medicine in the cardiovascular system.
Collapse
|
45
|
Babiarz JE, Hsu R, Melton C, Thomas M, Ullian EM, Blelloch R. A role for noncanonical microRNAs in the mammalian brain revealed by phenotypic differences in Dgcr8 versus Dicer1 knockouts and small RNA sequencing. RNA (NEW YORK, N.Y.) 2011; 17:1489-501. [PMID: 21712401 PMCID: PMC3153973 DOI: 10.1261/rna.2442211] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 05/11/2011] [Indexed: 05/24/2023]
Abstract
Noncanonical microRNAs (miRNAs) and endogenous small interfering RNAs (endo-siRNAs) are distinct subclasses of small RNAs that bypass the DGCR8/DROSHA Microprocessor but still require DICER1 for their biogenesis. What role, if any, they have in mammals remains unknown. To identify potential functional properties for these subclasses, we compared the phenotypes resulting from conditional deletion of Dgcr8 versus Dicer1 in post-mitotic neurons. The loss of Dicer1 resulted in an earlier lethality, more severe structural abnormalities, and increased apoptosis relative to that from Dgcr8 loss. Deep sequencing of small RNAs from the hippocampus and cortex of the conditional knockouts and control littermates identified multiple noncanonical microRNAs that were expressed at high levels in the brain relative to other tissues, including mirtrons and H/ACA snoRNA-derived small RNAs. In contrast, we found no evidence for endo-siRNAs in the brain. Taken together, our findings provide evidence for a diverse population of highly expressed noncanonical miRNAs that together are likely to play important functional roles in post-mitotic neurons.
Collapse
Affiliation(s)
- Joshua E. Babiarz
- 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
| | - Ruby Hsu
- Department of Ophthalmology and Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - Collin Melton
- 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
- Program in Biomedical Sciences, University of California, San Francisco, San Francisco, California 94143, USA
| | - Molly Thomas
- Program in Biomedical Sciences, University of California, San Francisco, San Francisco, California 94143, USA
| | - Erik M. Ullian
- Department of Ophthalmology and Physiology, University of California, San Francisco, San Francisco, California 94143, USA
| | - 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
|
46
|
Subramanyam D, Blelloch R. From microRNAs to targets: pathway discovery in cell fate transitions. Curr Opin Genet Dev 2011; 21:498-503. [PMID: 21636265 DOI: 10.1016/j.gde.2011.04.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2011] [Accepted: 04/27/2011] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRNAs) are 22 nt non-coding RNAs that regulate expression of downstream targets by messenger RNA (mRNA) destabilization and translational inhibition. A large number of eukaryotic mRNAs are targeted by miRNAs, with many individual mRNAs being targeted by multiple miRNAs. Further, a single miRNA can target hundreds of mRNAs, making these small RNAs powerful regulators of cell fate decisions. Such regulation by miRNAs has been observed in the maintenance of the embryonic stem cell (ESC) cell cycle and during ESC differentiation. MiRNAs can also promote the dedifferentiation of somatic cells to induced pluripotent stem cells. During this process they target multiple downstream genes, which represent important nodes of key cellular processes. Here, we review these findings and discuss how miRNAs may be used as tools to discover novel pathways that are involved in cell fate transitions using dedifferentiation of somatic cells to induced pluripotent stem cells as a case study.
Collapse
Affiliation(s)
- Deepa Subramanyam
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Center for Reproductive Sciences, Department of Urology, University of California San Francisco, San Francisco, CA, USA
| | | |
Collapse
|
47
|
Kopecky B, Fritzsch B. Regeneration of Hair Cells: Making Sense of All the Noise. Pharmaceuticals (Basel) 2011; 4:848-879. [PMID: 21966254 PMCID: PMC3180915 DOI: 10.3390/ph4060848] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 06/04/2011] [Accepted: 06/08/2011] [Indexed: 12/17/2022] Open
Abstract
Hearing loss affects hundreds of millions of people worldwide by dampening or cutting off their auditory connection to the world. Current treatments for sensorineural hearing loss (SNHL) with cochlear implants are not perfect, leaving regenerative medicine as the logical avenue to a perfect cure. Multiple routes to regeneration of damaged hair cells have been proposed and are actively pursued. Each route not only requires a keen understanding of the molecular basis of ear development but also faces the practical limitations of stem cell regulation in the delicate inner ear where topology of cell distribution is essential. Improvements in our molecular understanding of the minimal essential genes necessary for hair cell formation and recent advances in stem cell manipulation, such as seen with inducible pluripotent stem cells (iPSCs) and epidermal neural crest stem cells (EPI-NCSCs), have opened new possibilities to advance research in translational stem cell therapies for individuals with hearing loss. Despite this, more detailed network maps of gene expression are needed, including an appreciation for the roles of microRNAs (miRs), key regulators of transcriptional gene networks. To harness the true potential of stem cells for hair cell regeneration, basic science and clinical medicine must work together to expedite the transition from bench to bedside by elucidating the full mechanisms of inner ear hair cell development, including a focus on the role of miRs, and adapting this knowledge safely and efficiently to stem cell technologies.
Collapse
Affiliation(s)
- Benjamin Kopecky
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
- Medical Scientist Training Program, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
48
|
Ambros V. MicroRNAs and developmental timing. Curr Opin Genet Dev 2011; 21:511-7. [PMID: 21530229 DOI: 10.1016/j.gde.2011.04.003] [Citation(s) in RCA: 234] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 04/01/2011] [Indexed: 12/22/2022]
Abstract
MicroRNAs regulate temporal transitions in gene expression associated with cell fate progression and differentiation throughout animal development. Genetic analysis of developmental timing in the nematode Caenorhabditis elegans identified two evolutionarily conserved microRNAs, lin-4/mir-125 and let-7, that regulate cell fate progression and differentiation in C. elegans cell lineages. MicroRNAs perform analogous developmental timing functions in other animals, including mammals. By regulating cell fate choices and transitions between pluripotency and differentiation, microRNAs help to orchestrate developmental events throughout the developing animal, and to play tissue homeostasis roles important for disease, including cancer.
Collapse
Affiliation(s)
- Victor Ambros
- UMass Medical School, Molecular Medicine, 373 Plantation St, Worcester, MA 01605, USA.
| |
Collapse
|