101
|
Long non-coding RNA: Classification, biogenesis and functions in blood cells. Mol Immunol 2019; 112:82-92. [DOI: 10.1016/j.molimm.2019.04.011] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/16/2019] [Accepted: 04/23/2019] [Indexed: 12/20/2022]
|
102
|
Rahmani E, Schweiger R, Rhead B, Criswell LA, Barcellos LF, Eskin E, Rosset S, Sankararaman S, Halperin E. Cell-type-specific resolution epigenetics without the need for cell sorting or single-cell biology. Nat Commun 2019; 10:3417. [PMID: 31366909 PMCID: PMC6668473 DOI: 10.1038/s41467-019-11052-9] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 06/17/2019] [Indexed: 02/07/2023] Open
Abstract
High costs and technical limitations of cell sorting and single-cell techniques currently restrict the collection of large-scale, cell-type-specific DNA methylation data. This, in turn, impedes our ability to tackle key biological questions that pertain to variation within a population, such as identification of disease-associated genes at a cell-type-specific resolution. Here, we show mathematically and empirically that cell-type-specific methylation levels of an individual can be learned from its tissue-level bulk data, conceptually emulating the case where the individual has been profiled with a single-cell resolution and then signals were aggregated in each cell population separately. Provided with this unprecedented way to perform powerful large-scale epigenetic studies with cell-type-specific resolution, we revisit previous studies with tissue-level bulk methylation and reveal novel associations with leukocyte composition in blood and with rheumatoid arthritis. For the latter, we further show consistency with validation data collected from sorted leukocyte sub-types.
Collapse
Affiliation(s)
- Elior Rahmani
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Regev Schweiger
- Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, 6997801, Israel
- MyHeritage Ltd., Or Yehuda, 6037606, Israel
| | - Brooke Rhead
- Computational Biology Graduate Group, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Lindsey A Criswell
- Russell/Engleman Rheumatology Research Center, Department of Medicine, University of California, San Francisco, San Francisco, CA, 94143, USA
| | - Lisa F Barcellos
- School of Public Health, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Eleazar Eskin
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Saharon Rosset
- Department of Statistics, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Sriram Sankararaman
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Eran Halperin
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Anesthesiology and Perioperative Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
103
|
Not Only Mutations Matter: Molecular Picture of Acute Myeloid Leukemia Emerging from Transcriptome Studies. JOURNAL OF ONCOLOGY 2019; 2019:7239206. [PMID: 31467542 PMCID: PMC6699387 DOI: 10.1155/2019/7239206] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/12/2019] [Indexed: 01/08/2023]
Abstract
The last two decades of genome-scale research revealed a complex molecular picture of acute myeloid leukemia (AML). On the one hand, a number of mutations were discovered and associated with AML diagnosis and prognosis; some of them were introduced into diagnostic tests. On the other hand, transcriptome studies, which preceded AML exome and genome sequencing, remained poorly translated into clinics. Nevertheless, gene expression studies significantly contributed to the elucidation of AML pathogenesis and indicated potential therapeutic directions. The power of transcriptomic approach lies in its comprehensiveness; we can observe how genome manifests its function in a particular type of cells and follow many genes in one test. Moreover, gene expression measurement can be combined with mutation detection, as high-impact mutations are often present in transcripts. This review sums up 20 years of transcriptome research devoted to AML. Gene expression profiling (GEP) revealed signatures distinctive for selected AML subtypes and uncovered the additional within-subtype heterogeneity. The results were particularly valuable in the case of AML with normal karyotype which concerns up to 50% of AML cases. With the use of GEP, new classes of the disease were identified and prognostic predictors were proposed. A plenty of genes were detected as overexpressed in AML when compared to healthy control, including KIT, BAALC, ERG, MN1, CDX2, WT1, PRAME, and HOX genes. High expression of these genes constitutes usually an unfavorable prognostic factor. Upregulation of FLT3 and NPM1 genes, independent on their mutation status, was also reported in AML and correlated with poor outcome. However, transcriptome is not limited to the protein-coding genes; other types of RNA molecules exist in a cell and regulate genome function. It was shown that microRNA (miRNA) profiles differentiated AML groups and predicted outcome not worse than protein-coding gene profiles. For example, upregulation of miR-10a, miR-10b, and miR-196b and downregulation of miR-192 were found as typical of AML with NPM1 mutation whereas overexpression of miR-155 was associated with FLT3-internal tandem duplication (FLT3-ITD). Development of high-throughput technologies and microarray replacement by next generation sequencing (RNA-seq) enabled uncovering a real variety of leukemic cell transcriptomes, reflected by gene fusions, chimeric RNAs, alternatively spliced transcripts, miRNAs, piRNAs, long noncoding RNAs (lncRNAs), and their special type, circular RNAs. Many of them can be considered as AML biomarkers and potential therapeutic targets. The relations between particular RNA puzzles and other components of leukemic cells and their microenvironment, such as exosomes, are now under investigation. Hopefully, the results of this research will shed the light on these aspects of AML pathogenesis which are still not completely understood.
Collapse
|
104
|
Ng M, Heckl D, Klusmann JH. The Regulatory Roles of Long Noncoding RNAs in Acute Myeloid Leukemia. Front Oncol 2019; 9:570. [PMID: 31338324 PMCID: PMC6629768 DOI: 10.3389/fonc.2019.00570] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 06/12/2019] [Indexed: 01/23/2023] Open
Abstract
In this post-genomic era, long noncoding RNAs (lncRNAs) are rapidly gaining recognition for their crucial roles across diverse biological processes and contexts. The human blood system is no exception, where dozens of lncRNAs have been established as regulators of normal and/or malignant hematopoiesis, and where ongoing works continue to uncover novel lncRNA functions. Our review focuses on lncRNAs that are involved in the pathogenesis of acute myeloid leukemia (AML) and the mechanisms through which they control gene expression in this disease context. We also comment on genome-wide sequencing or profiling studies that have implicated large sets of lncRNAs in AML pathophysiology.
Collapse
Affiliation(s)
- Michelle Ng
- Department of Pediatrics I, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Dirk Heckl
- Department of Pediatrics I, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Jan-Henning Klusmann
- Department of Pediatrics I, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
105
|
Papaioannou D, Nicolet D, Ozer HG, Mrózek K, Volinia S, Fadda P, Carroll AJ, Kohlschmidt J, Kolitz JE, Wang ES, Stone RM, Byrd JC, Garzon R, Bloomfield CD. Prognostic and Biologic Relevance of Clinically Applicable Long Noncoding RNA Profiling in Older Patients with Cytogenetically Normal Acute Myeloid Leukemia. Mol Cancer Ther 2019; 18:1451-1459. [PMID: 31164409 DOI: 10.1158/1535-7163.mct-18-1125] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/29/2019] [Accepted: 05/30/2019] [Indexed: 01/22/2023]
Abstract
We have previously shown that expression levels of 48 long noncoding RNAs (lncRNA) can generate a prognostic lncRNA score that independently associates with outcome of older patients with cytogenetically normal acute myeloid leukemia (CN-AML). However, the techniques used to identify and measure prognostic lncRNAs (i.e., RNA sequencing and microarrays) are not tailored for clinical testing. Herein, we report on an assay (based on the nCounter platform) that is designed to produce targeted measurements of prognostic lncRNAs in a clinically applicable manner. We analyzed a new cohort of 76 older patients with CN-AML and found that the nCounter assay yielded reproducible measurements and that the lncRNA score retained its prognostic value; patients with high lncRNA scores had lower complete remission (CR) rates (P = 0.009; 58% vs. 87%), shorter disease-free (P = 0.05; 3-year rates: 0% vs. 21%), overall (OS; P = 0.02, 3-year rates: 10% vs. 29%), and event-free survival (EFS; P = 0.002, 3-year rates: 0% vs. 18%) than patients with low lncRNA scores. In multivariable analyses, the lncRNA score independently associated with CR rates (P = 0.02), OS (P = 0.02), and EFS (P = 0.02). To gain biological insights, we examined our initial cohort of 71 older patients with CN-AML, previously analyzed with RNA sequencing. Genes involved in immune response and B-cell receptor signaling were enriched in patients with high lncRNA scores. We conclude that clinically applicable lncRNA profiling is feasible and potentially useful for risk stratification of older patients with CN-AML. Furthermore, we identify potentially targetable molecular pathways that are active in the high-risk patients with high lncRNA scores.
Collapse
Affiliation(s)
| | - Deedra Nicolet
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Alliance Statistics and Data Center, The Ohio State University, Columbus, Ohio
| | - Hatice G Ozer
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio
| | - Krzysztof Mrózek
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Stefano Volinia
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Paolo Fadda
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Andrew J Carroll
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jessica Kohlschmidt
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.,Alliance Statistics and Data Center, The Ohio State University, Columbus, Ohio
| | - Jonathan E Kolitz
- Monter Cancer Center, Hofstra Northwell School of Medicine, Lake Success, New York
| | - Eunice S Wang
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Richard M Stone
- Dana-Farber/Partners Cancer Care, Harvard University, Boston, Massachusetts
| | - John C Byrd
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Ramiro Garzon
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | |
Collapse
|
106
|
Long Non-Coding RNA and Acute Leukemia. Int J Mol Sci 2019; 20:ijms20030735. [PMID: 30744139 PMCID: PMC6387068 DOI: 10.3390/ijms20030735] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 10/20/2018] [Accepted: 10/22/2018] [Indexed: 12/19/2022] Open
Abstract
Acute leukemia (AL) is the main type of cancer in children worldwide. Mortality by this disease is high in developing countries and its etiology remains unanswered. Evidences showing the role of the long non-coding RNAs (lncRNAs) in the pathophysiology of hematological malignancies have increased drastically in the last decade. In addition to the contribution of these lncRNAs in leukemogenesis, recent studies have suggested that lncRNAs could be used as biomarkers in the diagnosis, prognosis, and therapeutic response in leukemia patients. The focus of this review is to describe the functional classification, biogenesis, and the role of lncRNAs in leukemogenesis, to summarize the evidence about the lncRNAs which are playing a role in AL, and how these genes could be useful as potential therapeutic targets.
Collapse
|
107
|
In-stem molecular beacon targeted to a 5'-region of tRNA inclusive of the D arm that detects mature tRNA with high sensitivity. PLoS One 2019; 14:e0211505. [PMID: 30695081 PMCID: PMC6351059 DOI: 10.1371/journal.pone.0211505] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/15/2019] [Indexed: 01/12/2023] Open
Abstract
Cellular functions are regulated by the up- and down-regulation and localization of RNA molecules. Therefore, many RNA detection methods have been developed to analyze RNA levels and localization. Molecular beacon (MB) is one of the major methods for quantitative RNA detection and analysis of RNA localization. Most oligonucleotide-based probes, including MB, are designed to target a long flexible region on the target RNA molecule, e.g., a single-stranded region. Recently, analyses of tRNA localization and levels became important, as it has been shown that environmental stresses and chemical reagents induce nuclear accumulation of tRNA and tRNA degradation in mammalian cells. However, tRNA is highly structured and does not harbor any long flexible regions. Hence, only a few methods are currently available for detecting tRNA. In the present study, we attempted to detect elongator tRNAMet (eMet) and initiator tRNAMet (iMet) by using an in-stem molecular beacon (ISMB), characterized by more effective quenching and significantly higher sensitivity than those of conventional MB. We found that ISMB1 targeted a 5′- region that includes the D arm of tRNA and that it detected eMet and iMet transcripts as well as mature eMet with high sensitivity. Moreover, the analysis revealed that the formation of the ISMB/tRNA transcript complex required more time than the formation of an ISMB/unstructured short RNA complex. These results suggest that ISMB-based tRNA detection can be a useful tool for various biological and medical studies.
Collapse
|
108
|
Tschumper RC, Shanafelt TD, Kay NE, Jelinek DF. Role of long non-coding RNAs in disease progression of early stage unmutated chronic lymphocytic leukemia. Oncotarget 2019; 10:60-75. [PMID: 30713603 PMCID: PMC6343752 DOI: 10.18632/oncotarget.26538] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 12/12/2018] [Indexed: 12/15/2022] Open
Abstract
Predicting disease progression in chronic lymphocytic leukemia (CLL) remains challenging particularly in patients with Rai Stage 0/I disease that have an unmutated immunoglobulin heavy chain variable region (UM IGHV). Even though patients with UM IGHV have a poor prognosis and generally require earlier treatment, not all UM IGHV patients experience more rapid disease progression with some remaining treatment free for many years. This observation suggests biologic characteristics other than known prognostic factors influence disease progression. Alterations in long non-coding RNA (lncRNA) expression levels have been implicated in diagnosis and prognosis of various cancers, however, their role in disease progression of early Rai stage UM CLL is unknown. Here we use microarray analysis to compare lncRNA and mRNA profiles of Rai 0/I UM IGHV patients who progressed in <2 years relative to patients who had not progressed for >5 years. Over 1,300 lncRNAs and 940 mRNAs were differentially expressed (fold change ≥ 2.0; p-value ≤ 0.05). Of interest, the differentially expressed lncRNAs T204050, NR_002947, and uc.436+, have known associated genes that have been linked to CLL. Thus, our study reveals differentially expressed lncRNAs in progressive early stage CLL requiring therapy versus indolent early Rai stage UM CLL. These lncRNAs have the potential to impact relevant biological processes and pathways that influence clinical outcome in CLL.
Collapse
Affiliation(s)
| | - Tait D Shanafelt
- Department of Hematology/Oncology, Stanford University, Stanford, CA, USA
| | - Neil E Kay
- Department of Internal Medicine, Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Diane F Jelinek
- Department of Immunology, Mayo Clinic, Scottsdale, AZ, USA.,Department of Internal Medicine, Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
109
|
Wu Z, Gao S, Zhao X, Chen J, Keyvanfar K, Feng X, Kajigaya S, Young NS. Long noncoding RNAs of single hematopoietic stem and progenitor cells in healthy and dysplastic human bone marrow. Haematologica 2018; 104:894-906. [PMID: 30545929 PMCID: PMC6518886 DOI: 10.3324/haematol.2018.208926] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 11/22/2018] [Indexed: 12/30/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are regulators of cell differentiation and development. The lncRNA transcriptome in human hematopoietic stem and progenitor cells is not comprehensively defined. We investigated lncRNAs in 979 human bone marrow-derived CD34+ cells by single cell RNA sequencing followed by de novo transcriptome reconstruction. We identified 3,173 lncRNAs in total, among which 2,365 were previously unknown, and we characterized lncRNA stem, differentiation, and maturation signatures. lncRNA expression exhibited high cell-to-cell variation, which was only apparent in single cell analysis. lncRNA expression followed a lineage-specific and highly dynamic pattern during early hematopoiesis. lncRNAs in hematopoietic cells closely correlated with protein-coding genes of known functions in the regulation of hematopoiesis and cell fate decisions, and the potential regulatory roles of lncRNAs in hematopoiesis were imputed by projection from protein-coding genes with a “guilt-by-association” approach. We characterized lncRNAs preferentially expressed in hematopoietic stem cells and in various downstream differentiated lineage progenitors. We also profiled lncRNA expression in single cells from patients with myelodysplastic syndromes and in aneuploid cells in particular. Our study provides a global view of lncRNAs in human hematopoietic stem and progenitor cells. We observed a highly ordered pattern of lncRNA expression and participation in regulation of early hematopoiesis, and coordinate aberrant messenger RNA and lncRNA transcriptomes in dysplastic hematopoiesis. (Registered at clinicaltrials.gov with identifiers: 00001620, 00001397)
Collapse
Affiliation(s)
- Zhijie Wu
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health
| | - Shouguo Gao
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health
| | - Xin Zhao
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health
| | - Jinguo Chen
- Trans-NIH Center for Human Immunology, Autoimmunity, and Inflammation, National Institutes of Health, Bethesda, MD, USA
| | - Keyvan Keyvanfar
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health
| | - Xingmin Feng
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health
| | - Sachiko Kajigaya
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health
| | - Neal S Young
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health
| |
Collapse
|
110
|
Wong NK, Huang CL, Islam R, Yip SP. Long non-coding RNAs in hematological malignancies: translating basic techniques into diagnostic and therapeutic strategies. J Hematol Oncol 2018; 11:131. [PMID: 30466456 PMCID: PMC6251105 DOI: 10.1186/s13045-018-0673-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/23/2018] [Indexed: 12/26/2022] Open
Abstract
Recent studies have revealed that non-coding regions comprise the vast majority of the human genome and long non-coding RNAs (lncRNAs) are a diverse class of non-coding RNAs that has been implicated in a variety of biological processes. Abnormal expression of lncRNAs has also been linked to different human diseases including cancers, yet the regulatory mechanisms and functional effects of lncRNAs are still ambiguous, and the molecular details also need to be confirmed. Unlike protein-coding gene, it is much more challenging to unravel the roles of lncRNAs owing to their unique and complex features such as functional diversity and low conservation among species, which greatly hamper their experimental characterization. In this review, we summarize and discuss both conventional and advanced approaches for the identification and functional characterization of lncRNAs related to hematological malignancies. In particular, the utility and advancement of clustered regularly interspaced short palindromic repeats (CRISPR)-Cas system as gene-editing tools are envisioned to facilitate the molecular dissection of lncRNAs via different knock-in/out strategies. Besides experimental considerations specific to lncRNAs, the roles of lncRNAs in the pathogenesis and progression of leukemia are also highlighted in the review. We expect that these insights may ultimately lead to clinical applications including development of biomarkers and novel therapeutic approaches targeting lncRNAs.
Collapse
Affiliation(s)
- Nonthaphat Kent Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Y9/F, Lee Shau Kee Building, Hung Hom, Hong Kong SAR, China
| | - Chien-Ling Huang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Y9/F, Lee Shau Kee Building, Hung Hom, Hong Kong SAR, China.
| | - Rashidul Islam
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Y9/F, Lee Shau Kee Building, Hung Hom, Hong Kong SAR, China
| | - Shea Ping Yip
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Y9/F, Lee Shau Kee Building, Hung Hom, Hong Kong SAR, China.
| |
Collapse
|
111
|
Szabó B, Murvai N, Abukhairan R, Schád É, Kardos J, Szeder B, Buday L, Tantos Á. Disordered Regions of Mixed Lineage Leukemia 4 (MLL4) Protein Are Capable of RNA Binding. Int J Mol Sci 2018; 19:ijms19113478. [PMID: 30400675 PMCID: PMC6274713 DOI: 10.3390/ijms19113478] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/01/2018] [Accepted: 11/02/2018] [Indexed: 01/11/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are emerging as important regulators of cellular processes and are extensively involved in the development of different cancers; including leukemias. As one of the accepted methods of lncRNA function is affecting chromatin structure; lncRNA binding has been shown for different chromatin modifiers. Histone lysine methyltransferases (HKMTs) are also subject of lncRNA regulation as demonstrated for example in the case of Polycomb Repressive Complex 2 (PRC2). Mixed Lineage Leukemia (MLL) proteins that catalyze the methylation of H3K4 have been implicated in several different cancers; yet many details of their regulation and targeting remain elusive. In this work we explored the RNA binding capability of two; so far uncharacterized regions of MLL4; with the aim of shedding light to the existence of possible regulatory lncRNA interactions of the protein. We demonstrated that both regions; one that contains a predicted RNA binding sequence and one that does not; are capable of binding to different RNA constructs in vitro. To our knowledge, these findings are the first to indicate that an MLL protein itself is capable of lncRNA binding.
Collapse
Affiliation(s)
- Beáta Szabó
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary.
| | - Nikoletta Murvai
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary.
| | - Rawan Abukhairan
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary.
| | - Éva Schád
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary.
| | - József Kardos
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Eötvös Loránd University, H-1117 Budapest, Hungary.
| | - Bálint Szeder
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary.
| | - László Buday
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary.
| | - Ágnes Tantos
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, H-1117 Budapest, Hungary.
| |
Collapse
|
112
|
Magilnick N, Boldin MP. Molecular Moirai: Long Noncoding RNA Mediators of HSC Fate. CURRENT STEM CELL REPORTS 2018; 4:158-165. [PMID: 30364527 DOI: 10.1007/s40778-018-0130-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Purpose of Review Hematopoiesis is an ordered developmental process that requires dynamic regulation to warrant proper response to physiological challenges and prevent malignancies. Long noncoding RNAs are emerging as key, multi-faceted regulators of gene expression. This review explores the function of lncRNAs in the control of HSC homeostasis and hematopoietic differentiation. Recent Findings Multiple lncRNAs have been implicated in maintaining HSC stemness and enabling progenitors to carry out the correct programs of lineage differentiation. Specific lncRNAs have been identified that regulate the differentiation of multipotent progenitors into terminally differentiated blood cells. These lncRNAs predominantly act by assisting master regulators that drive specific differentiation programs, either by enhancing or repressing the transcription of particular genomic loci. Summary Long noncoding RNAs contribute to the correct differentiation and maturation of various hematopoietic lineages by assisting with the activation of transcriptional programs in a time- and cell-dependent manner.
Collapse
|
113
|
Martinez-Soria N, McKenzie L, Draper J, Ptasinska A, Issa H, Potluri S, Blair HJ, Pickin A, Isa A, Chin PS, Tirtakusuma R, Coleman D, Nakjang S, Assi S, Forster V, Reza M, Law E, Berry P, Mueller D, Osborne C, Elder A, Bomken SN, Pal D, Allan JM, Veal GJ, Cockerill PN, Wichmann C, Vormoor J, Lacaud G, Bonifer C, Heidenreich O. The Oncogenic Transcription Factor RUNX1/ETO Corrupts Cell Cycle Regulation to Drive Leukemic Transformation. Cancer Cell 2018; 34:626-642.e8. [PMID: 30300583 PMCID: PMC6179967 DOI: 10.1016/j.ccell.2018.08.015] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 04/20/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
Oncogenic transcription factors such as the leukemic fusion protein RUNX1/ETO, which drives t(8;21) acute myeloid leukemia (AML), constitute cancer-specific but highly challenging therapeutic targets. We used epigenomic profiling data for an RNAi screen to interrogate the transcriptional network maintaining t(8;21) AML. This strategy identified Cyclin D2 (CCND2) as a crucial transmitter of RUNX1/ETO-driven leukemic propagation. RUNX1/ETO cooperates with AP-1 to drive CCND2 expression. Knockdown or pharmacological inhibition of CCND2 by an approved drug significantly impairs leukemic expansion of patient-derived AML cells and engraftment in immunodeficient murine hosts. Our data demonstrate that RUNX1/ETO maintains leukemia by promoting cell cycle progression and identifies G1 CCND-CDK complexes as promising therapeutic targets for treatment of RUNX1/ETO-driven AML.
Collapse
Affiliation(s)
- Natalia Martinez-Soria
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Lynsey McKenzie
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Julia Draper
- Cancer Research UK Manchester Institute, Manchester M20 4GJ, UK
| | - Anetta Ptasinska
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Hasan Issa
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Sandeep Potluri
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Helen J Blair
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Anna Pickin
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Asmida Isa
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Paulynn Suyin Chin
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Ricky Tirtakusuma
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Daniel Coleman
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Sirintra Nakjang
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Salam Assi
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Victoria Forster
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Mojgan Reza
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Ed Law
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Philip Berry
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Dorothee Mueller
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Cameron Osborne
- Department of Medical & Molecular Genetics, King's College London, London SE1 9RT, UK
| | - Alex Elder
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Simon N Bomken
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - Deepali Pal
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK
| | - James M Allan
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Gareth J Veal
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
| | - Peter N Cockerill
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Christian Wichmann
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, Ludwig-Maximilian University Hospital, Munich 80539, Germany
| | - Josef Vormoor
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK; Princess Maxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands
| | - Georges Lacaud
- Cancer Research UK Manchester Institute, Manchester M20 4GJ, UK
| | - Constanze Bonifer
- Institute for Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Olaf Heidenreich
- Wolfson Childhood Cancer Research Centre, Northern Institute for Cancer Research, Newcastle University, Brewery Lane, Newcastle upon Tyne NE1 7RU, UK; Princess Maxima Center for Pediatric Oncology, Utrecht 3584CS, the Netherlands.
| |
Collapse
|
114
|
Lee KH, Park HM, Son KH, Shin TJ, Cho JY. Transcriptome Signatures of Canine Mammary Gland Tumors and Its Comparison to Human Breast Cancers. Cancers (Basel) 2018; 10:cancers10090317. [PMID: 30205506 PMCID: PMC6162473 DOI: 10.3390/cancers10090317] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 08/27/2018] [Accepted: 09/04/2018] [Indexed: 01/22/2023] Open
Abstract
Breast cancer (BC)/mammary gland carcinoma (MGC) is the most frequently diagnosed and leading cause of cancer-related mortality in both women and canines. To better understand both canine MGC and human BC-specific genes, we sequenced RNAs obtained from eight pairs of carcinomas and adjacent normal tissues in dogs. By comprehensive transcriptome analysis, 351 differentially expressed genes (DEGs) were identified in overall canine MGCs. Based on the DEGs, comparative analysis revealed correlation existing among the three histological subtypes of canine MGC (ductal, simple, and complex) and four molecular subtypes of human BC (HER2+, ER+, ER&HER2+, and TNBC). Eight DEGs shared by all three subtypes of canine MGCs had been previously reported as cancer-associated genes in human studies. Gene ontology and pathway analyses using the identified DEGs revealed that the biological processes of cell proliferation, adhesion, and inflammatory responses are enriched in up-regulated MGC DEGs. In contrast, fatty acid homeostasis and transcription regulation involved in cell fate commitment were down-regulated in MGC DEGs. Moreover, correlations are demonstrated between upstream promoter transcripts and DEGs. Canine MGC- and subtype-enriched gene expression allows us to better understand both human BC and canine MGC, yielding new insight into the development of biomarkers and targets for both diseases.
Collapse
Affiliation(s)
- Kang-Hoon Lee
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| | - Hyoung-Min Park
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| | - Keun-Hong Son
- Department of Microbiology, College of Natural Sciences, Dankook University, Cheonan 31116, Korea.
| | - Tae-Jin Shin
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 Plus and Research Institute for Veterinary Science, School of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
115
|
Mao Y, Fu Z, Zhang Y, Dong L, Zhang Y, Zhang Q, Li X, Liu J. A seven-lncRNA signature predicts overall survival in esophageal squamous cell carcinoma. Sci Rep 2018; 8:8823. [PMID: 29891973 PMCID: PMC5995883 DOI: 10.1038/s41598-018-27307-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/25/2018] [Indexed: 12/11/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most common types of cancer and the leading causes of cancer-related mortality worldwide, especially in Eastern Asia. Here, we downloaded the microarray data of lncRNA expression profiles of ESCC patients from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) data sets and divided into training, validation and test set. The random survival forest (RSF) algorithm and Cox regression analysis were applied to identify a seven-lncRNA signature. Then the predictive ability of the seven-lncRNA signature was evaluated in the validation and test set using Kaplan-Meier test, time-dependent receiver operating characteristic (ROC) curves and dynamic area under curve (AUC). Stratified analysis and multivariate Cox regression also demonstrated the independence of the signature in prognosis prediction from other clinical factors. Besides, the predict accuracy of lncRNA signature was much better than that of tumor-node-metastasis (TNM) stage in all the three sets. LncRNA combined with TNM displayed better prognostic predict ability than either alone. The role of LINC00173 from the signature in modulating the proliferation and cell cycle of ESCC cells was also observed. These results indicated that this seven-lncRNA signature could be used as an independent prognostic biomarker for prognosis prediction of patients with ESCC.
Collapse
Affiliation(s)
- Yu Mao
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China.
| | - Zhanzhao Fu
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China
| | - Yunjie Zhang
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China
| | - Lixin Dong
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China
| | - Yanqiu Zhang
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China
| | - Qiang Zhang
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China
| | - Xin Li
- Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, Hebei, China
| | - Jia Liu
- Institute of basic medical sciences, Qilu Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
116
|
Hofmans M, Lammens T, Helsmoortel HH, Bresolin S, Cavé H, Flotho C, Hasle H, van den Heuvel-Eibrink MM, Niemeyer C, Stary J, Van Roy N, Van Vlierberghe P, Philippé J, De Moerloose B. The long non-coding RNA landscape in juvenile myelomonocytic leukemia. Haematologica 2018; 103:e501-e504. [PMID: 29858388 DOI: 10.3324/haematol.2018.189977] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Mattias Hofmans
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium .,Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Belgium
| | - Tim Lammens
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium.,Cancer Research Institute Ghent, Belgium
| | | | - Silvia Bresolin
- Department of Women and Child Health, University of Padova, Italy
| | - Hélène Cavé
- Department of Genetics, University Hospital of Robert Debré and Paris-Diderot University, Paris, France
| | - Christian Flotho
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University of Freiburg, Germany
| | - Henrik Hasle
- Department of Pediatrics, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - Marry M van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Dutch Childhood Oncology Group, The Hague, the Netherlands
| | - Charlotte Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, University of Freiburg, Germany
| | - Jan Stary
- Department of Pediatric Hematology/ Oncology, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Nadine Van Roy
- Cancer Research Institute Ghent, Belgium.,Center for Medical Genetics, Ghent University Hospital, Belgium
| | - Pieter Van Vlierberghe
- Cancer Research Institute Ghent, Belgium.,Center for Medical Genetics, Ghent University Hospital, Belgium
| | - Jan Philippé
- Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, Belgium.,Cancer Research Institute Ghent, Belgium
| | - Barbara De Moerloose
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Belgium.,Cancer Research Institute Ghent, Belgium
| |
Collapse
|
117
|
Grasedieck S, Mulaw M, Sperb N, Wessinger K, Rouhi A, Bommer M, Bromberg JEC, Otto M, Tumani H, von Arnim CAF, Mertens D, Kuchenbauer F. Comprehensive microRNA expression profiling in cerebrospinal fluid distinguishes between neurological disease classes. Neuropathol Appl Neurobiol 2018; 45:318-323. [PMID: 29679382 DOI: 10.1111/nan.12491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Sarah Grasedieck
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | - Medhanie Mulaw
- Institute of Experimental Cancer Research, Ulm University, Ulm, Germany
| | - Nadine Sperb
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | | | - Arefeh Rouhi
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany
| | - Martin Bommer
- Department of Hematology, Oncology, Palliative Care and Infectious Diseases, Alb-Fils-Kliniken, Göppingen, Germany
| | | | - Markus Otto
- Department of Neurology, Ulm University, Ulm, Germany
| | | | | | - Daniel Mertens
- Department of Internal Medicine III, Ulm University Hospital, Ulm, Germany.,DKFZ, Mechanisms of Leukemogenesis, Heidelberg, Germany
| | | |
Collapse
|
118
|
Li Y, Jing F, Ding Y, He Q, Zhong Y, Fan C. Long noncoding RNA CCAT1 polymorphisms are associated with the risk of colorectal cancer. Cancer Genet 2018; 222-223:13-19. [PMID: 29666003 DOI: 10.1016/j.cancergen.2018.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/18/2018] [Accepted: 02/13/2018] [Indexed: 12/27/2022]
Abstract
Colorectal cancer associated transcript 1 (CCAT1) is a novel long noncoding RNA, whose overexpression is evident in both early phase of tumorigenesis and later disease stages in colorectal cancer (CRC). No study has explored the relationship between CCAT1 polymorphisms and CRC risk. In the present study, a case-control study was conducted to investigate the association between CCAT1 polymorphisms and CRC risk in Chinese population. We identified that CCAT1 rs67085638 polymorphism was associated with an increased risk of CRC (OR = 1.72, 95%CI = 1.14-2.58, P = 0.009 in heterozygote codominant model; OR = 1.67, 95%CI = 1.13-2.47, P = 0.010 in dominant model). Moreover, CCAT1 rs7013433 polymorphism was associated with late clinical stage (OR = 1.82, 95%CI = 1.16-2.86, P = 0.009 in heterozygote codominant model; OR = 1.72, 95%CI = 1.13-2.63, P = 0.012 in dominant model). Our finding proposed a link between CCAT1 polymorphisms with CRC risk as well as different clinical stages.
Collapse
Affiliation(s)
- Yingjun Li
- Department of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Fangyuan Jing
- Department of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ye Ding
- Department of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qingfang He
- Department of Chronic Non-Communicable Diseases Control and Prevention, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, Zhejiang, China
| | - Yaohong Zhong
- Department of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chunhong Fan
- Department of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
119
|
Trino S, Lamorte D, Caivano A, Laurenzana I, Tagliaferri D, Falco G, Del Vecchio L, Musto P, De Luca L. MicroRNAs as New Biomarkers for Diagnosis and Prognosis, and as Potential Therapeutic Targets in Acute Myeloid Leukemia. Int J Mol Sci 2018; 19:ijms19020460. [PMID: 29401684 PMCID: PMC5855682 DOI: 10.3390/ijms19020460] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 02/07/2023] Open
Abstract
Acute myeloid leukemias (AML) are clonal disorders of hematopoietic progenitor cells which are characterized by relevant heterogeneity in terms of phenotypic, genotypic, and clinical features. Among the genetic aberrations that control disease development there are microRNAs (miRNAs). miRNAs are small non-coding RNAs that regulate, at post-transcriptional level, translation and stability of mRNAs. It is now established that deregulated miRNA expression is a prominent feature in AML. Functional studies have shown that miRNAs play an important role in AML pathogenesis and miRNA expression signatures are associated with chemotherapy response and clinical outcome. In this review we summarized miRNA signature in AML with different cytogenetic, molecular and clinical characteristics. Moreover, we reviewed the miRNA regulatory network in AML pathogenesis and we discussed the potential use of cellular and circulating miRNAs as biomarkers for diagnosis and prognosis and as therapeutic targets.
Collapse
MESH Headings
- Animals
- Antagomirs/genetics
- Antagomirs/metabolism
- Antagomirs/therapeutic use
- Biomarkers, Tumor/agonists
- Biomarkers, Tumor/antagonists & inhibitors
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Chromosome Aberrations
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/pathology
- Gene Expression Regulation, Leukemic
- Humans
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Mice
- MicroRNAs/agonists
- MicroRNAs/antagonists & inhibitors
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Molecular Targeted Therapy
- Oligoribonucleotides/genetics
- Oligoribonucleotides/metabolism
- Oligoribonucleotides/therapeutic use
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Prognosis
- Signal Transduction
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Stefania Trino
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Antonella Caivano
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Daniela Tagliaferri
- Biogem Scarl, Istituto di Ricerche Genetiche 'Gaetano Salvatore', 83031 Ariano Irpino, Italy.
| | - Geppino Falco
- Biogem Scarl, Istituto di Ricerche Genetiche 'Gaetano Salvatore', 83031 Ariano Irpino, Italy.
- Department of Biology, University of Naples Federico II, 80147 Naples, Italy.
| | - Luigi Del Vecchio
- CEINGE Biotecnologie Avanzate s.c.a r.l., 80147 Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, 80138 Naples, Italy.
| | - Pellegrino Musto
- Scientific Direction, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Potenza, Italy.
| | - Luciana De Luca
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| |
Collapse
|
120
|
Cui T, Zhang L, Huang Y, Yi Y, Tan P, Zhao Y, Hu Y, Xu L, Li E, Wang D. MNDR v2.0: an updated resource of ncRNA-disease associations in mammals. Nucleic Acids Res 2018; 46:D371-D374. [PMID: 29106639 PMCID: PMC5753235 DOI: 10.1093/nar/gkx1025] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/15/2017] [Accepted: 10/19/2017] [Indexed: 02/05/2023] Open
Abstract
Accumulating evidence suggests that diverse non-coding RNAs (ncRNAs) are involved in the progression of a wide variety of diseases. In recent years, abundant ncRNA-disease associations have been found and predicted according to experiments and prediction algorithms. Diverse ncRNA-disease associations are scattered over many resources and mammals, whereas a global view of diverse ncRNA-disease associations is not available for any mammals. Hence, we have updated the MNDR v2.0 database (www.rna-society.org/mndr/) by integrating experimental and prediction associations from manual literature curation and other resources under one common framework. The new developments in MNDR v2.0 include (i) an over 220-fold increase in ncRNA-disease associations enhancement compared with the previous version (including lncRNA, miRNA, piRNA, snoRNA and more than 1400 diseases); (ii) integrating experimental and prediction evidence from 14 resources and prediction algorithms for each ncRNA-disease association; (iii) mapping disease names to the Disease Ontology and Medical Subject Headings (MeSH); (iv) providing a confidence score for each ncRNA-disease association and (v) an increase of species coverage to six mammals. Finally, MNDR v2.0 intends to provide the scientific community with a resource for efficient browsing and extraction of the associations between diverse ncRNAs and diseases, including >260 000 ncRNA-disease associations.
Collapse
Affiliation(s)
- Tianyu Cui
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area and Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Lin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yan Huang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Ying Yi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Puwen Tan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yue Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Yongfei Hu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Liyan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area and Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- To whom correspondence should be addressed. Tel: +86 451 86699584; Fax: +86 451 86699584; . Correspondence may also be addressed to Enmin Li. Tel: +86 754 88900413; Fax: +86 754 88900847; . Correspondence may also be addressed to Liyan Xu. Tel: +86 754 88900460; Fax: +86 754 88900847;
| | - Enmin Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area and Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- To whom correspondence should be addressed. Tel: +86 451 86699584; Fax: +86 451 86699584; . Correspondence may also be addressed to Enmin Li. Tel: +86 754 88900413; Fax: +86 754 88900847; . Correspondence may also be addressed to Liyan Xu. Tel: +86 754 88900460; Fax: +86 754 88900847;
| | - Dong Wang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area and Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
- Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, China
- To whom correspondence should be addressed. Tel: +86 451 86699584; Fax: +86 451 86699584; . Correspondence may also be addressed to Enmin Li. Tel: +86 754 88900413; Fax: +86 754 88900847; . Correspondence may also be addressed to Liyan Xu. Tel: +86 754 88900460; Fax: +86 754 88900847;
| |
Collapse
|
121
|
Fallik N, Bar-Lavan Y, Greenshpan Y, Goldstein O, Grosch M, Drukker M, Gazit R. Neat1 in hematopoietic stem cells. Oncotarget 2017; 8:109575-109586. [PMID: 29312630 PMCID: PMC5752543 DOI: 10.18632/oncotarget.22729] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 11/10/2017] [Indexed: 12/26/2022] Open
Abstract
Hematopoietic Stem Cells (HSCs) generate blood and immune cells through a hierarchical process of differentiation. Genes that regulate this process are of great interest for understanding normal and also malignant hematopoiesis. Surprisingly, however, very little is known about long-non-coding RNAs (lncRNA) in HSCs. Neat1 is a lncRNA that plays a major role in the formation of sub-nuclear structures called paraspeckles, and was reported to regulate proliferation and differentiation in other cells types. We detected Neat1 expression using RNA-seq data and RT-qPCR in HSCs, progenitors and effector immune cells, by specific detection of its isoforms. Neat1 is highly expressed in stem and progenitor cells, yet it shows significant reduction in granulocytes. Microscopically, Neat1 is detected as sharp nuclear foci. Paraspeckle proteins NONO and PSPC1 are detected as aggregated nuclear foci in fresh primary hematopoietic cells, and in cultured cells. Induction of differentiation in vitro was found to enhance Neat1 expression. Taken together, our data demonstrate for the first time the expression of Neat1 and paraspeckles formation in HSCs and along hematopoiesis.
Collapse
Affiliation(s)
- Noam Fallik
- The Shraga Segal Department for Microbiology Immunology and Genetics, Faculty of Health Sciences, The Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Yael Bar-Lavan
- The Shraga Segal Department for Microbiology Immunology and Genetics, Faculty of Health Sciences, The Ben-Gurion University of the Negev, Be'er Sheva, Israel.,National Institute for Biotechnology in the Negev, The Ben-Gurion University of the Negev, Be'er Sheva, Israel.,Center for Regenerative Medicine and Stem Cells, The Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Yariv Greenshpan
- The Shraga Segal Department for Microbiology Immunology and Genetics, Faculty of Health Sciences, The Ben-Gurion University of the Negev, Be'er Sheva, Israel.,National Institute for Biotechnology in the Negev, The Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Oron Goldstein
- The Shraga Segal Department for Microbiology Immunology and Genetics, Faculty of Health Sciences, The Ben-Gurion University of the Negev, Be'er Sheva, Israel.,National Institute for Biotechnology in the Negev, The Ben-Gurion University of the Negev, Be'er Sheva, Israel.,Center for Regenerative Medicine and Stem Cells, The Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Markus Grosch
- Institute of Stem Cell Research, German Research Center for Environmental Health, Helmholtz Center Munich, Neuherberg, Germany
| | - Micha Drukker
- Institute of Stem Cell Research, German Research Center for Environmental Health, Helmholtz Center Munich, Neuherberg, Germany
| | - Roi Gazit
- The Shraga Segal Department for Microbiology Immunology and Genetics, Faculty of Health Sciences, The Ben-Gurion University of the Negev, Be'er Sheva, Israel.,National Institute for Biotechnology in the Negev, The Ben-Gurion University of the Negev, Be'er Sheva, Israel.,Center for Regenerative Medicine and Stem Cells, The Ben-Gurion University of the Negev, Be'er Sheva, Israel
| |
Collapse
|