101
|
Watson SF, Knol LI, Witteveldt J, Macias S. Crosstalk Between Mammalian Antiviral Pathways. Noncoding RNA 2019; 5:E29. [PMID: 30909383 PMCID: PMC6468734 DOI: 10.3390/ncrna5010029] [Citation(s) in RCA: 9] [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: 02/27/2019] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 12/15/2022] Open
Abstract
As part of their innate immune response against viral infections, mammals activate the expression of type I interferons to prevent viral replication and dissemination. An antiviral RNAi-based response can be also activated in mammals, suggesting that several mechanisms can co-occur in the same cell and that these pathways must interact to enable the best antiviral response. Here, we will review how the classical type I interferon response and the recently described antiviral RNAi pathways interact in mammalian cells. Specifically, we will uncover how the small RNA biogenesis pathway, composed by the nucleases Drosha and Dicer can act as direct antiviral factors, and how the type-I interferon response regulates the function of these. We will also describe how the factors involved in small RNA biogenesis and specific small RNAs impact the activation of the type I interferon response and antiviral activity. With this, we aim to expose the complex and intricate network of interactions between the different antiviral pathways in mammals.
Collapse
Affiliation(s)
- Samir F Watson
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK.
| | - Lisanne I Knol
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK.
| | - Jeroen Witteveldt
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK.
| | - Sara Macias
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3FL, UK.
| |
Collapse
|
102
|
Lee D, Park D, Park JH, Kim JH, Shin C. Poly(A)-specific ribonuclease sculpts the 3' ends of microRNAs. RNA (NEW YORK, N.Y.) 2019; 25:388-405. [PMID: 30591540 PMCID: PMC6380276 DOI: 10.1261/rna.069633.118] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 12/21/2018] [Indexed: 05/08/2023]
Abstract
The 3' ends of metazoan microRNAs (miRNAs) are initially defined by the RNase III enzymes during maturation, but subsequently experience extensive modifications by several enzymatic activities. For example, terminal nucleotidyltransferases (TENTs) elongate miRNAs by adding one or a few nucleotides to their 3' ends, which occasionally leads to differential regulation of miRNA stability or function. However, the catalytic entities that shorten miRNAs and the molecular consequences of such shortening are less well understood, especially in vertebrates. Here, we report that poly(A)-specific ribonuclease (PARN) sculpts the 3' ends of miRNAs in human cells. By generating PARN knockout cells and characterizing their miRNAome, we demonstrate that PARN digests the 3' extensions of miRNAs that are derived from the genome or attached by TENTs, thereby effectively reducing the length of miRNAs. Surprisingly, PARN-mediated shortening has little impact on miRNA stability, suggesting that this process likely operates to finalize miRNA maturation, rather than to initiate miRNA decay. PARN-mediated shortening is pervasive across most miRNAs and appears to be a conserved mechanism contributing to the 3' end formation of vertebrate miRNAs. Our findings add miRNAs to the expanding list of noncoding RNAs whose 3' end formation depends on PARN.
Collapse
Affiliation(s)
- Dooyoung Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Daechan Park
- Department of Biological Sciences, Ajou University, Suwon 16499, Republic of Korea
| | - June Hyun Park
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Jong Heon Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang 10408, Republic of Korea
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Chanseok Shin
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Agriculture and Life Sciences, and Plant Genomics and Breeding Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
103
|
Kwon SC, Baek SC, Choi YG, Yang J, Lee YS, Woo JS, Kim VN. Molecular Basis for the Single-Nucleotide Precision of Primary microRNA Processing. Mol Cell 2019; 73:505-518.e5. [DOI: 10.1016/j.molcel.2018.11.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/11/2018] [Accepted: 11/01/2018] [Indexed: 12/16/2022]
|
104
|
Park SK, Kee Y, Hwang BJ. Enhancement of gene knockdown efficiency by CNNC motifs in the intronic shRNA precursor. Genes Genomics 2019; 41:491-498. [PMID: 30656519 DOI: 10.1007/s13258-019-00783-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 01/03/2019] [Indexed: 11/28/2022]
Abstract
BACKGROUND Short hairpin RNAs (shRNAs) expressed from vectors have been used as an effective means of exploiting the RNA interference (RNAi) pathway in mammalian cells. Of several methods to express shRNA, a method of transcribing shRNAs embedded in microRNA precursors has been more widely used than the one that directly expresses shRNA from RNA polymerase III promoters because the microRNA precursor form of shRNA is known to cause lower levels of cytotoxicity and off-target effects than the overexpressed shRNAs from the RNA polymerase III promoters. OBJECTIVE We study the primary sequence features of microRNA precursors, which enhance their processing into mature form, helps design more potent shRNA precursors embedded in microRNA precursors. METHODS We measure the enhancement of gene knockdown efficiency by adding CNNC motifs in the 3' flanking region of shRNA precursor embedded in the human miR-30a microRNA precursor. RESULTS By systemically adding three CNNC motifs in the 3' flanking region of shRNA precursor, we found that addition of two CNNC motifs saturates their enhanced knockdown ability of shRNA and that the CNNC motif in the + 17 to + 20 from the drosha cleavage site is most important for the shRNA-mediated target gene knock down. We also did see little knockdown of target gene expression by the shRNA precursor lacking CNNC motif. CONCLUSION Since genetic studies generally require techniques that could reduce gene expression at different degrees, the findings in this study will allow us to use RNAi for genetic studies of reducing gene expression at different degrees.
Collapse
Affiliation(s)
- Seong Kyun Park
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Kangwon-do, Republic of Korea
| | - Yun Kee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Kangwon-do, Republic of Korea
| | - Byung Joon Hwang
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Kangwon-do, Republic of Korea.
| |
Collapse
|
105
|
Bofill-De Ros X, Kasprzak WK, Bhandari Y, Fan L, Cavanaugh Q, Jiang M, Dai L, Yang A, Shao TJ, Shapiro BA, Wang YX, Gu S. Structural Differences between Pri-miRNA Paralogs Promote Alternative Drosha Cleavage and Expand Target Repertoires. Cell Rep 2019; 26:447-459.e4. [PMID: 30625327 PMCID: PMC6369706 DOI: 10.1016/j.celrep.2018.12.054] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 11/11/2018] [Accepted: 12/11/2018] [Indexed: 12/16/2022] Open
Abstract
MicroRNA (miRNA) processing begins with Drosha cleavage, the fidelity of which is critical for downstream processing and mature miRNA target specificity. To understand how pri-miRNA sequence and structure influence Drosha cleavage, we studied the maturation of three pri-miR-9 paralogs, which encode the same mature miRNA but differ in the surrounding scaffold. We show that pri-miR-9-1 has a unique Drosha cleavage profile due to its distorted and flexible stem structure. Cleavage of pri-miR-9-1, but not pri-miR-9-2 or pri-miR-9-3, generates an alternative miR-9 with a shifted seed sequence that expands the scope of its target RNAs. Analyses of low-grade glioma patient samples indicate that the alternative-miR-9 has a potential role in tumor progression. Furthermore, we provide evidence that distortion of pri-miRNA stems induced by asymmetric internal loops correlates with Drosha cleavage at non-canonical sites. Our studies reveal that pri-miRNA paralogs can have distinct functions via differential Drosha processing.
Collapse
Affiliation(s)
- Xavier Bofill-De Ros
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Wojciech K Kasprzak
- Basic Science Program, RNA Biology Laboratory, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD 21702, USA
| | - Yuba Bhandari
- Protein-Nucleic Acid Interaction Section, Structural Biophysics Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Lixin Fan
- Small-Angle X-ray Scattering Core Facility, Center for Cancer Research of the National Cancer Institute, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21702, USA
| | - Quinn Cavanaugh
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Minjie Jiang
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Lisheng Dai
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Acong Yang
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Tie-Juan Shao
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Bruce A Shapiro
- RNA Structure and Design Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Yun-Xing Wang
- Protein-Nucleic Acid Interaction Section, Structural Biophysics Laboratory, National Cancer Institute, Frederick, MD 21702, USA
| | - Shuo Gu
- RNA Mediated Gene Regulation Section, RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA.
| |
Collapse
|
106
|
Abstract
MicroRNAs (miRNAs) are important regulators of gene expression that bind complementary target mRNAs and repress their expression. Precursor miRNA molecules undergo nuclear and cytoplasmic processing events, carried out by the endoribonucleases DROSHA and DICER, respectively, to produce mature miRNAs that are loaded onto the RISC (RNA-induced silencing complex) to exert their biological function. Regulation of mature miRNA levels is critical in development, differentiation, and disease, as demonstrated by multiple levels of control during their biogenesis cascade. Here, we will focus on post-transcriptional mechanisms and will discuss the impact of cis-acting sequences in precursor miRNAs, as well as trans-acting factors that bind to these precursors and influence their processing. In particular, we will highlight the role of general RNA-binding proteins (RBPs) as factors that control the processing of specific miRNAs, revealing a complex layer of regulation in miRNA production and function.
Collapse
Affiliation(s)
- Gracjan Michlewski
- Division of Infection and Pathway Medicine, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
- Zhejiang University-University of Edinburgh Institute, Zhejiang University, Zhejiang 314400, P.R. China
| | - Javier F Cáceres
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
| |
Collapse
|
107
|
Abstract
Adeno-associated viral vectors have emerged as an important tool for human gene therapy, having demonstrated high transduction efficiency in a broad range of target tissues, a good safety profile in animal models and human clinical trials, and prospective long-lasting gene expression. First discovered 20 years ago, RNA interference (RNAi) has become another important tool for human gene therapy, enabling scientists to move on from classical gene transfer to gene silencing approaches, or combinations thereof. In this chapter, we describe a simple step-by-step method that will allow gene silencing novices to design their own artificial miRNAs against a target of their choice, clone these miRNAs into an AAV-based vector, and rapidly screen for highly efficient artificial miRNAs. The described method takes into consideration recent advances in the field including miRNA processing from various cellular miRNA backbones, choice between polymerase II and III promoters, and the potential impact of these factors on toxicity as it relates to off-targeting and to saturation of the endogenous RNAi machinery.
Collapse
Affiliation(s)
- Florie Borel
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Christian Mueller
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA. .,Department of Pediatrics, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
108
|
Gutierrez-Camino Á, Umerez M, Lopez-Lopez E, Santos-Zorrozua B, Martin-Guerrero I, de Andoin NG, Ana S, Navajas A, Astigarraga I, Garcia-Orad A. Involvement of miRNA polymorphism in mucositis development in childhood acute lymphoblastic leukemia treatment. Pharmacogenomics 2018; 19:1403-1412. [DOI: 10.2217/pgs-2018-0113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Aim: Mucositis, linked to methotrexate, daunorubicin or cyclophosphamide, is a frequent childhood acute lymphoblastic leukemia (ALL) therapy side effect. miRNAs regulate the expression of pharmacokinetic/pharmacodynamic pathway genes. SNPs in miRNAs could affect their levels or function, and affect their pharmacokinetic/pharmacodynamic pathway target genes. Our aim was to determine the association between miRNA genetic variants targeting mucositis-related genes and mucositis-developing risk. Patients & methods: We analyzed 160 SNPs in 179 Spanish children with B-cell precursor ALL homogeneously treated with LAL/SHOP protocols. Results: We identified three SNPs in miR-4268, miR-4751 and miR-3117 associated with mucositis, diarrhea and vomiting, respectively. Conclusion: The effect of these SNPs on genes related to drug pharmacokinetics/pharmacodynamics could explain mucositis, diarrhea and vomiting development during ALL therapy.
Collapse
Affiliation(s)
- Ángela Gutierrez-Camino
- Department of Genetics, Physic Anthropology & Animal Physiology, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain
- BioCruces Health Research Institute, Barakaldo, 48903, Spain
| | - Maitane Umerez
- Department of Genetics, Physic Anthropology & Animal Physiology, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain
| | - Elixabet Lopez-Lopez
- Department of Genetics, Physic Anthropology & Animal Physiology, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain
- BioCruces Health Research Institute, Barakaldo, 48903, Spain
| | - Borja Santos-Zorrozua
- Department of Genetics, Physic Anthropology & Animal Physiology, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain
| | - Idoia Martin-Guerrero
- Department of Genetics, Physic Anthropology & Animal Physiology, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain
- BioCruces Health Research Institute, Barakaldo, 48903, Spain
| | - Nagore García de Andoin
- Department of Pediatrics, University Hospital Donostia, San Sebastian, 20014, Spain
- Department of Pediatrics, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain
| | - Sastre Ana
- Department of Oncohematology, University Hospital La Paz, Madrid, 28046, Spain
| | - Aurora Navajas
- BioCruces Health Research Institute, Barakaldo, 48903, Spain
- Department of Pediatrics, University Hospital Cruces, Barakaldo, 48903, Spain
| | - Itziar Astigarraga
- BioCruces Health Research Institute, Barakaldo, 48903, Spain
- Department of Pediatrics, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain
- Department of Pediatrics, University Hospital Cruces, Barakaldo, 48903, Spain
| | - Africa Garcia-Orad
- Department of Genetics, Physic Anthropology & Animal Physiology, University of the Basque Country, UPV/EHU, Leioa, 48940, Spain
- BioCruces Health Research Institute, Barakaldo, 48903, Spain
| |
Collapse
|
109
|
Kaadt E, Alsing S, Cecchi CR, Damgaard CK, Corydon TJ, Aagaard L. Efficient Knockdown and Lack of Passenger Strand Activity by Dicer-Independent shRNAs Expressed from Pol II-Driven MicroRNA Scaffolds. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 14:318-328. [PMID: 30654192 PMCID: PMC6348697 DOI: 10.1016/j.omtn.2018.11.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 11/21/2018] [Accepted: 11/21/2018] [Indexed: 12/21/2022]
Abstract
The expression of short hairpin RNAs (shRNAs) may result in unwanted activity from the co-processed passenger strand. Recent studies have shown that shortening the stem of conventional shRNAs abolishes passenger strand release. These Dicer-independent shRNAs, expressed from RNA polymerase III (Pol III) promoters, rely on Ago2 processing in resemblance to miR-451. Using strand-specific reporters, we tested two designs, and our results support the loss of passenger strand activity. We demonstrate that artificial primary microRNA (pri-miRNA) transcripts, expressed from Pol II promoters, can potently silence a gene of choice. Among six different scaffolds tested, miR-324 and miR-451 were readily re-targeted to direct efficient knockdown from either a CMV or a U1 snRNA promoter. Importantly, the miR-shRNAs have no passenger strand activity and remain active in Dicer-knockout cells. Our vectors are straightforward to design, as we replace the pre-miR-324 or -451 sequences with a Dicer-independent shRNA mimicking miR-451 with unpaired A-C nucleotides at the base. The use of Pol II promoters allows for controlled expression, while the inclusion of pri-miRNA sequences likely requires Drosha processing and, as such, mimics microRNA biogenesis. Since this improved and tunable system bypasses the requirement for Dicer activity and abolishes passenger strand activity completely, it will likely prove favorable in both research and therapeutic applications in terms of versatility and enhanced safety.
Collapse
Affiliation(s)
- Erik Kaadt
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Sidsel Alsing
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Claudia R Cecchi
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Thomas J Corydon
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark; Department of Ophthalmology, Aarhus University Hospital, 8000 Aarhus C, Denmark
| | - Lars Aagaard
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|
110
|
Nguyen HM, Nguyen TD, Nguyen TL, Nguyen TA. Orientation of Human Microprocessor on Primary MicroRNAs. Biochemistry 2018; 58:189-198. [DOI: 10.1021/acs.biochem.8b00944] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Huong Minh Nguyen
- Laboratory of Molecular Microbiology, Institute of Biotechnology, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Trung Duc Nguyen
- Division of Life Science, Hong Kong University of Science & Technology, Hong Kong, China
| | - Thuy Linh Nguyen
- Division of Life Science, Hong Kong University of Science & Technology, Hong Kong, China
| | - Tuan Anh Nguyen
- Division of Life Science, Hong Kong University of Science & Technology, Hong Kong, China
| |
Collapse
|
111
|
Chakraborty S, Islam MR, Ali MM, Nabi AHMN. Evolutionary Divergence of Brain-specific Precursor miRNAs Drives Efficient Processing and Production of Mature miRNAs in Human. Neuroscience 2018; 392:141-159. [PMID: 30273624 DOI: 10.1016/j.neuroscience.2018.09.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 08/09/2018] [Accepted: 09/07/2018] [Indexed: 12/18/2022]
Abstract
The hallmark of human evolution encompasses the dramatic increase in brain size and complexity. The intricate interplays of micro-RNAs (miRNAs) and their target genes are indispensable in brain development. Sequence divergence in distinct structural regions of Brain-specific precursor miRNAs (pre-miRNAs) and its consequence in the production of corresponding mature miRNAs in human are unknown. To address these questions, first we classified miRNAs into three categories based on tissue expression: Brain-specific (expressed exclusively in brain), Non-brain (expressed in Non-brain tissues) and Common (expressed in all tissues) and compared the sequence divergence of different structural regions (basal segment, lower and upper stem, internal and terminal loop) of categorized pre-miRNAs across human, non-human primates and rodents. Our analysis revealed that unpaired regions of Brain-specific pre-miRNAs in human bear traces of relatively high rate of evolutionary divergence compared to those in other species. Cross-tissue expression analysis unveiled the higher expression of the Brain-specific miRNAs in human compared to other species. Intriguingly, in human brain, expression levels of these miRNAs superseded the levels of the ubiquitously expressed "Common-miRNAs". Further analysis revealed that presence of certain motif and nucleotide preference in the Brain-specific pre-miRNAs may favor DROSHA and DICER to ameliorate miRNA processing. The higher processing efficiency of human Brain-specific miRNAs was reflected as an elevated production of corresponding mature miRNAs in the human brain. Finally, re-construction of gene-regulatory network uncovers different pathways driven by Brain-specific miRNAs that may contribute to the development of brain in human.
Collapse
Affiliation(s)
- Sajib Chakraborty
- Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh.
| | - Md Rezaul Islam
- Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh; Max Planck Research School for Neurosciences, Göttingen, Germany
| | - Md Muntasir Ali
- Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh; Unit of Microbiology, Belgian Nuclear Research Centre (SCK•CEN), 2400 Mol, Belgium
| | - A H M Nurun Nabi
- Department of Biochemistry and Molecular Biology, University of Dhaka, Bangladesh.
| |
Collapse
|
112
|
Oak N, Ghosh R, Huang KL, Wheeler DA, Ding L, Plon SE. Framework for microRNA variant annotation and prioritization using human population and disease datasets. Hum Mutat 2018; 40:73-89. [PMID: 30302893 DOI: 10.1002/humu.23668] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/05/2018] [Accepted: 10/08/2018] [Indexed: 11/10/2022]
Abstract
MicroRNA (miRNA) expression is frequently deregulated in human disease, in contrast, disease-associated miRNA mutations are understudied. We developed Annotative Database of miRNA Elements, ADmiRE, which combines multiple existing and new biological annotations to aid prioritization of causal miRNA variation. We annotated 10,206 mature (3,257 within seed region) miRNA variants from multiple large sequencing datasets including gnomAD (15,496 genomes; 123,136 exomes). The pattern of miRNA variation closely resembles protein-coding exonic regions, with no difference between intragenic and intergenic miRNAs (P = 0.56), and high confidence miRNAs demonstrate higher sequence constraint (P < 0.001). Conservation analysis across 100 vertebrates identified 765 highly conserved miRNAs that also have limited genetic variation in gnomAD. We applied ADmiRE to the TCGA PanCancerAtlas WES dataset containing over 10,000 individuals across 33 adult cancers and annotated 1,267 germline (rare in gnomAD) and 1,492 somatic miRNA variants. Several miRNA families with deregulated gene expression in cancer have low levels of both somatic and germline variants, e.g., let-7 and miR-10. In addition to known somatic miR-142 mutations in hematologic cancers, we describe novel somatic miR-21 mutations in esophageal cancers impacting downstream miRNA targets. Through the development of ADmiRE, we present a framework for annotation and prioritization of miRNA variation in disease datasets.
Collapse
Affiliation(s)
- Ninad Oak
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030.,Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX
| | - Rajarshi Ghosh
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030.,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030
| | - Kuan-Lin Huang
- Department of Medicine, Washington University in St. Louis, MO 63108.,McDonnel Genome Institute, Washington University in St. Louis, MO 63108
| | - David A Wheeler
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, MO 63108.,McDonnel Genome Institute, Washington University in St. Louis, MO 63108.,Department of Genetics, Washington University in St. Louis, MO 63108.,Siteman Cancer Center, Washington University in St. Louis, MO 63108
| | - Sharon E Plon
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030.,Texas Children's Cancer Center, Texas Children's Hospital, Houston, TX.,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030.,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
113
|
Treiber T, Treiber N, Meister G. Regulation of microRNA biogenesis and its crosstalk with other cellular pathways. Nat Rev Mol Cell Biol 2018; 20:5-20. [DOI: 10.1038/s41580-018-0059-1] [Citation(s) in RCA: 628] [Impact Index Per Article: 89.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
114
|
Zampetaki A, Albrecht A, Steinhofel K. Long Non-coding RNA Structure and Function: Is There a Link? Front Physiol 2018; 9:1201. [PMID: 30197605 PMCID: PMC6117379 DOI: 10.3389/fphys.2018.01201] [Citation(s) in RCA: 166] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 08/10/2018] [Indexed: 01/12/2023] Open
Abstract
RNA has emerged as the prime target for diagnostics, therapeutics and the development of personalized medicine. In particular, the non-coding RNAs (ncRNAs) that do not encode proteins, display remarkable biochemical versatility. They can fold into complex structures and interact with proteins, DNA and other RNAs, modulating the activity, DNA targets or partners of multiprotein complexes. Thus, ncRNAs confer regulatory plasticity and represent a new layer of epigenetic control that is dysregulated in disease. Intriguingly, for long non-coding RNAs (lncRNAs, >200 nucleotides length) structural conservation rather than nucleotide sequence conservation seems to be crucial for maintaining their function. LncRNAs tend to acquire complex secondary and tertiary structures and their functions only impose very subtle sequence constraints. In the present review we will discuss the biochemical assays that can be employed to determine the lncRNA structural configurations. The implications and challenges of linking function and lncRNA structure to design novel RNA therapeutic approaches will also be analyzed.
Collapse
Affiliation(s)
- Anna Zampetaki
- King's British Heart Foundation Centre, King's College London, London, United Kingdom
| | - Andreas Albrecht
- Faculty of Science and Technology, Middlesex University, London, United Kingdom
| | | |
Collapse
|
115
|
The inflammatory state of adipose tissue is not affected by the anti-inflammatory response of the A2a-adenosine system and miR-221/PTEN. Int J Biochem Cell Biol 2018; 100:42-48. [DOI: 10.1016/j.biocel.2018.04.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/26/2018] [Accepted: 04/30/2018] [Indexed: 12/22/2022]
|
116
|
Kim K, Nguyen TD, Li S, Nguyen TA. SRSF3 recruits DROSHA to the basal junction of primary microRNAs. RNA (NEW YORK, N.Y.) 2018; 24:892-898. [PMID: 29615481 PMCID: PMC6004053 DOI: 10.1261/rna.065862.118] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 03/29/2018] [Indexed: 05/22/2023]
Abstract
The Microprocessor complex, consisting of an RNase III DROSHA and the DGCR8 dimer, cleaves primary microRNA transcripts (pri-miRNAs) to initiate microRNA (miRNA) maturation. Pri-miRNAs are stem-loop RNAs, and ∼79% of them contain at least one of the three major and conserved RNA motifs, UG, UGU, and CNNC. We recently demonstrated that the basal UG and apical UGU motifs of pri-miRNAs interact with DROSHA and DGCR8, respectively. They help orient Microprocessor on pri-miRNA in a proper direction in which DROSHA and DGCR8 localize to the basal and apical pri-miRNA junctions, respectively. In addition, CNNC, located at ∼17 nucleotides (nt) from the Microprocessor cleavage site, interacts with SRSF3 (SRp20) to stimulate Microprocessor to process pri-miRNAs. The mechanism underlying this stimulation, however, is unknown. In this study, we discovered that SRSF3 recruits DROSHA to the basal junction in a CNNC-dependent manner, thereby enhancing Microprocessor activity. Furthermore, by generating various pri-miRNA substrates containing CNNC at different locations, we demonstrated that such stimulation only occurs when CNNC is located at ∼17 nt from the Microprocessor cleavage site. Our findings reveal the molecular mechanism of SRSF3 in pri-miRNA processing and support the previously proposed explanation for the highly conserved position of CNNC in SRSF3-enhanced pri-miRNA processing.
Collapse
Affiliation(s)
- Kijun Kim
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Trung Duc Nguyen
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
- HKUST Shenzhen Research Institute, Shenzhen 518057, China
| | - Shaohua Li
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
| | - Tuan Anh Nguyen
- Division of Life Science, Hong Kong University of Science and Technology, Hong Kong, China
- HKUST Shenzhen Research Institute, Shenzhen 518057, China
| |
Collapse
|
117
|
Structural basis for terminal loop recognition and stimulation of pri-miRNA-18a processing by hnRNP A1. Nat Commun 2018; 9:2479. [PMID: 29946118 PMCID: PMC6018666 DOI: 10.1038/s41467-018-04871-9] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 05/23/2018] [Indexed: 02/07/2023] Open
Abstract
Post-transcriptional mechanisms play a predominant role in the control of microRNA (miRNA) production. Recognition of the terminal loop of precursor miRNAs by RNA-binding proteins (RBPs) influences their processing; however, the mechanistic basis for how levels of individual or subsets of miRNAs are regulated is mostly unexplored. We previously showed that hnRNP A1, an RBP implicated in many aspects of RNA processing, acts as an auxiliary factor that promotes the Microprocessor-mediated processing of pri-mir-18a. Here, by using an integrative structural biology approach, we show that hnRNP A1 forms a 1:1 complex with pri-mir-18a where both RNA recognition motifs (RRMs) bind to cognate RNA sequence motifs in the terminal loop of pri-mir-18a. Terminal loop binding induces an allosteric destabilization of base-pairing in the pri-mir-18a stem that promotes its downstream processing. Our results highlight terminal loop RNA recognition by RBPs as a potential general principle of miRNA biogenesis and regulation. hnRNP A1 is an auxiliary factor that promotes the Microprocessor-mediated processing of pri-mir-18a, of the oncomiR-1 cluster. Here the authors employ an integrative structural biology approach and provide insights into the molecular mechanism of how hnRNP A1 facilitates pri-mir-18a biogenesis.
Collapse
|
118
|
A Network of Noncoding Regulatory RNAs Acts in the Mammalian Brain. Cell 2018; 174:350-362.e17. [PMID: 29887379 DOI: 10.1016/j.cell.2018.05.022] [Citation(s) in RCA: 446] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/23/2018] [Accepted: 05/10/2018] [Indexed: 01/23/2023]
Abstract
Noncoding RNAs (ncRNAs) play increasingly appreciated gene-regulatory roles. Here, we describe a regulatory network centered on four ncRNAs-a long ncRNA, a circular RNA, and two microRNAs-using gene editing in mice to probe the molecular consequences of disrupting key components of this network. The long ncRNA Cyrano uses an extensively paired site to miR-7 to trigger destruction of this microRNA. Cyrano-directed miR-7 degradation is much more effective than previously described examples of target-directed microRNA degradation, which come primarily from studies of artificial and viral RNAs. By reducing miR-7 levels, Cyrano prevents repression of miR-7-targeted mRNAs and enables accumulation of Cdr1as, a circular RNA known to regulate neuronal activity. Without Cyrano, excess miR-7 causes cytoplasmic destruction of Cdr1as in neurons, in part through enhanced slicing of Cdr1as by a second miRNA, miR-671. Thus, several types of ncRNAs can collaborate to establish a sophisticated regulatory network.
Collapse
|
119
|
Creugny A, Fender A, Pfeffer S. Regulation of primary microRNA processing. FEBS Lett 2018; 592:1980-1996. [PMID: 29683487 DOI: 10.1002/1873-3468.13067] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 04/12/2018] [Accepted: 04/16/2018] [Indexed: 12/28/2022]
Abstract
MicroRNAs (miRNAs) are evolutionarily conserved small regulatory RNAs that participate in the adjustment of many, if not all, fundamental biological processes. Molecular mechanisms involved in miRNA biogenesis and mode of action have been elucidated in the past two decades. Similar to many cellular pathways, miRNA processing and function can be globally or specifically regulated at several levels and by numerous proteins and RNAs. Given their role as fine-tuning molecules, it is essential for miRNA expression to be tightly regulated in order to maintain cellular homeostasis. Here, we review our current knowledge of the first step of their maturation occurring in the nucleus and how it can be specifically and dynamically modulated.
Collapse
Affiliation(s)
- Antoine Creugny
- Architecture and Reactivity of RNA, Institut de Biologie Moléculaire et Cellulaire du CNRS, Université de Strasbourg, France
| | - Aurélie Fender
- Architecture and Reactivity of RNA, Institut de Biologie Moléculaire et Cellulaire du CNRS, Université de Strasbourg, France
| | - Sébastien Pfeffer
- Architecture and Reactivity of RNA, Institut de Biologie Moléculaire et Cellulaire du CNRS, Université de Strasbourg, France
| |
Collapse
|
120
|
Abstract
MicroRNAs (miRNAs) are ∼22 nt RNAs that direct posttranscriptional repression of mRNA targets in diverse eukaryotic lineages. In humans and other mammals, these small RNAs help sculpt the expression of most mRNAs. This article reviews advances in our understanding of the defining features of metazoan miRNAs and their biogenesis, genomics, and evolution. It then reviews how metazoan miRNAs are regulated, how they recognize and cause repression of their targets, and the biological functions of this repression, with a compilation of knockout phenotypes that shows that important biological functions have been identified for most of the broadly conserved miRNAs of mammals.
Collapse
Affiliation(s)
- David P Bartel
- Howard Hughes Medical Institute and Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
121
|
Mou H, Zhong G, Gardner MR, Wang H, Wang YW, Cheng D, Farzan M. Conditional Regulation of Gene Expression by Ligand-Induced Occlusion of a MicroRNA Target Sequence. Mol Ther 2018; 26:1277-1286. [PMID: 29567311 PMCID: PMC5993935 DOI: 10.1016/j.ymthe.2018.02.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 02/20/2018] [Accepted: 02/22/2018] [Indexed: 11/03/2022] Open
Abstract
RNA switches that modulate gene expression with small molecules have a number of scientific and clinical applications. Here, we describe a novel class of small regulatory on switches based on the ability of a ligand-bound aptamer to promote stem formation between a microRNA target sequence (miR-T) and a complementary competing strand. Two on switch architectures employing this basic concept were evaluated, differing in the location of a tetracycline aptamer and the region of a miR-21 target sequence (miR-21-T) masked by its competing strand. Further optimizations of miR-21-T and its competing strand resulted in tetracycline-regulated on switches that induced luciferase expression by 19-fold in HeLa cells. A similar switch design based on miR-122-T afforded 7-fold regulation when placed in tandem, indicating that this approach can be extended to additional miR-T. Optimized on switches introduced into adeno-associated virus (AAV) vectors afforded 10-fold regulation of two antiviral proteins in AAV-transduced cells. Our data demonstrate that small-molecule-induced occlusion of a miR-T can be used to conditionally regulate gene expression in mammalian cells and suggest that regulatory switches built on this principle can be used to dose expression of an AAV transgene.
Collapse
Affiliation(s)
- Huihui Mou
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Guocai Zhong
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA.
| | - Matthew R Gardner
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Haimin Wang
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Yi-Wen Wang
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Dechun Cheng
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA; Department of Parasitology, Harbin Medical University, Harbin 150081, China
| | - Michael Farzan
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL 33458, USA.
| |
Collapse
|
122
|
Abstract
DROSHA is the catalytic subunit of the Microprocessor complex, which initiates microRNA (miRNA) maturation in the nucleus by recognizing and cleaving hairpin precursors embedded in primary transcripts. However, accumulating evidence suggests that not all hairpin substrates of DROSHA are associated with the generation of functional small RNAs. By targeting those hairpins, DROSHA regulates diverse aspects of RNA metabolism across the transcriptome, serves as a line of defense against the expression of potentially deleterious elements, and permits cell fate determination and differentiation. DROSHA is also versatile in the way that it executes these noncanonical functions, occasionally depending on its RNA-binding activity rather than its catalytic activity. Herein, we discuss the functional and mechanistic diversity of DROSHA beyond the miRNA biogenesis pathway in light of recent findings.
Collapse
Affiliation(s)
- Dooyoung Lee
- a Department of Agricultural Biotechnology , Seoul National University , Seoul , Republic of Korea
| | - Chanseok Shin
- a Department of Agricultural Biotechnology , Seoul National University , Seoul , Republic of Korea.,b Research Institute of Agriculture and Life Sciences, and Plant Genomics and Breeding Institute , Seoul National University , Seoul , Republic of Korea
| |
Collapse
|
123
|
Heme enables proper positioning of Drosha and DGCR8 on primary microRNAs. Nat Commun 2017; 8:1737. [PMID: 29170488 PMCID: PMC5700927 DOI: 10.1038/s41467-017-01713-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 10/09/2017] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs regulate the expression of many proteins and require specific maturation steps. Primary microRNA transcripts (pri-miRs) are cleaved by Microprocessor, a complex containing the RNase Drosha and its partner protein, DGCR8. Although DGCR8 is known to bind heme, the molecular role of heme in pri-miR processing is unknown. Here we show that heme is critical for Microprocessor to process pri-miRs with high fidelity. Furthermore, the degree of inherent heme dependence varies for different pri-miRs. Heme-dependent pri-miRs fail to properly recruit Drosha, but heme-bound DGCR8 can correct erroneous binding events. Rather than changing the oligomerization state, heme induces a conformational change in DGCR8. Finally, we demonstrate that heme activates DGCR8 to recognize pri-miRs by specifically binding the terminal loop near the 3′ single-stranded segment. Drosha and DGCR8 constitute the core Microprocessor complex, which processes primary microRNAs (pri-miRs) into mature microRNAs. Here the authors show that heme is essential for the proper processing of pri-miRs by Drosha-DGCR8, and the molecular mechanism by which heme enhances processing fidelity.
Collapse
|
124
|
Fehlmann T, Backes C, Kahraman M, Haas J, Ludwig N, Posch AE, Würstle ML, Hübenthal M, Franke A, Meder B, Meese E, Keller A. Web-based NGS data analysis using miRMaster: a large-scale meta-analysis of human miRNAs. Nucleic Acids Res 2017; 45:8731-8744. [PMID: 28911107 PMCID: PMC5587802 DOI: 10.1093/nar/gkx595] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 07/04/2017] [Indexed: 01/21/2023] Open
Abstract
The analysis of small RNA NGS data together with the discovery of new small RNAs is among the foremost challenges in life science. For the analysis of raw high-throughput sequencing data we implemented the fast, accurate and comprehensive web-based tool miRMaster. Our toolbox provides a wide range of modules for quantification of miRNAs and other non-coding RNAs, discovering new miRNAs, isomiRs, mutations, exogenous RNAs and motifs. Use-cases comprising hundreds of samples are processed in less than 5 h with an accuracy of 99.4%. An integrative analysis of small RNAs from 1836 data sets (20 billion reads) indicated that context-specific miRNAs (e.g. miRNAs present only in one or few different tissues / cell types) still remain to be discovered while broadly expressed miRNAs appear to be largely known. In total, our analysis of known and novel miRNAs indicated nearly 22 000 candidates of precursors with one or two mature forms. Based on these, we designed a custom microarray comprising 11 872 potential mature miRNAs to assess the quality of our prediction. MiRMaster is a convenient-to-use tool for the comprehensive and fast analysis of miRNA NGS data. In addition, our predicted miRNA candidates provided as custom array will allow researchers to perform in depth validation of candidates interesting to them.
Collapse
Affiliation(s)
- Tobias Fehlmann
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Christina Backes
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Mustafa Kahraman
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany.,Hummingbird Diagnostics GmbH, Heidelberg, Germany
| | - Jan Haas
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Heidelberg, Germany.,Klaus Tschira Institute for Integrative Computational Cardiology, Heidelberg, Germany
| | - Nicole Ludwig
- Department of Human Genetics, Saarland University, Homburg, Germany
| | | | | | - Matthias Hübenthal
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Benjamin Meder
- Department of Internal Medicine III, University Hospital Heidelberg, Heidelberg, Germany.,German Center for Cardiovascular Research (DZHK), Heidelberg, Germany.,Klaus Tschira Institute for Integrative Computational Cardiology, Heidelberg, Germany
| | - Eckart Meese
- Department of Human Genetics, Saarland University, Homburg, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| |
Collapse
|
125
|
Bouvy-Liivrand M, Hernández de Sande A, Pölönen P, Mehtonen J, Vuorenmaa T, Niskanen H, Sinkkonen L, Kaikkonen MU, Heinäniemi M. Analysis of primary microRNA loci from nascent transcriptomes reveals regulatory domains governed by chromatin architecture. Nucleic Acids Res 2017; 45:9837-9849. [PMID: 28973462 PMCID: PMC5737680 DOI: 10.1093/nar/gkx680] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 07/21/2017] [Indexed: 01/09/2023] Open
Abstract
Changes in mature microRNA (miRNA) levels that occur downstream of signaling cascades play an important role during human development and disease. However, the regulation of primary microRNA (pri-miRNA) genes remains to be dissected in detail. To address this, we followed a data-driven approach and developed a transcript identification, validation and quantification pipeline for characterizing the regulatory domains of pri-miRNAs. Integration of 92 nascent transcriptomes and multilevel data from cells arising from ecto-, endo- and mesoderm lineages reveals cell type-specific expression patterns, allows fine-resolution mapping of transcription start sites (TSS) and identification of candidate regulatory regions. We show that inter- and intragenic pri-miRNA transcripts span vast genomic regions and active TSS locations differ across cell types, exemplified by the mir-29a∼29b-1, mir-100∼let-7a-2∼125b-1 and miR-221∼222 clusters. Considering the presence of multiple TSS as an important regulatory feature at miRNA loci, we developed a strategy to quantify differential TSS usage. We demonstrate that the TSS activities associate with cell type-specific super-enhancers, differential stimulus responsiveness and higher-order chromatin structure. These results pave the way for building detailed regulatory maps of miRNA loci.
Collapse
Affiliation(s)
- Maria Bouvy-Liivrand
- School of Medicine, University of Eastern Finland, Kuopio, North-Savo 70200, Finland
| | | | - Petri Pölönen
- School of Medicine, University of Eastern Finland, Kuopio, North-Savo 70200, Finland
| | - Juha Mehtonen
- School of Medicine, University of Eastern Finland, Kuopio, North-Savo 70200, Finland
| | - Tapio Vuorenmaa
- School of Medicine, University of Eastern Finland, Kuopio, North-Savo 70200, Finland
| | - Henri Niskanen
- A. I. Virtanen Institute,University of Eastern Finland, Kuopio, North-Savo 70200, Finland
| | - Lasse Sinkkonen
- Life Sciences Research Unit, University of Luxembourg, Belvaux L-4367, Luxembourg
| | - Minna Unelma Kaikkonen
- A. I. Virtanen Institute,University of Eastern Finland, Kuopio, North-Savo 70200, Finland
| | - Merja Heinäniemi
- School of Medicine, University of Eastern Finland, Kuopio, North-Savo 70200, Finland
| |
Collapse
|
126
|
Church VA, Pressman S, Isaji M, Truscott M, Cizmecioglu NT, Buratowski S, Frolov MV, Carthew RW. Microprocessor Recruitment to Elongating RNA Polymerase II Is Required for Differential Expression of MicroRNAs. Cell Rep 2017; 20:3123-3134. [PMID: 28954229 PMCID: PMC5639929 DOI: 10.1016/j.celrep.2017.09.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 08/18/2017] [Accepted: 09/04/2017] [Indexed: 01/24/2023] Open
Abstract
The cellular abundance of mature microRNAs (miRNAs) is dictated by the efficiency of nuclear processing of primary miRNA transcripts (pri-miRNAs) into pre-miRNA intermediates. The Microprocessor complex of Drosha and DGCR8 carries this out, but it has been unclear what controls Microprocessor's differential processing of various pri-miRNAs. Here, we show that Drosophila DGCR8 (Pasha) directly associates with the C-terminal domain of the RNA polymerase II elongation complex when it is phosphorylated by the Cdk9 kinase (pTEFb). When association is blocked by loss of Cdk9 activity, a global change in pri-miRNA processing is detected. Processing of pri-miRNAs with a UGU sequence motif in their apical junction domain increases, while processing of pri-miRNAs lacking this motif decreases. Therefore, phosphorylation of RNA polymerase II recruits Microprocessor for co-transcriptional processing of non-UGU pri-miRNAs that would otherwise be poorly processed. In contrast, UGU-positive pri-miRNAs are robustly processed by Microprocessor independent of RNA polymerase association.
Collapse
Affiliation(s)
- Victoria A Church
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Sigal Pressman
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Mamiko Isaji
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Mary Truscott
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL 60607, USA
| | - Nihal Terzi Cizmecioglu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Middle East Technical University, Department of Biological Sciences, 06800, Ankara, Turkey
| | - Stephen Buratowski
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Maxim V Frolov
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL 60607, USA
| | - Richard W Carthew
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA.
| |
Collapse
|
127
|
CRISPR/Cas9 editing reveals novel mechanisms of clustered microRNA regulation and function. Sci Rep 2017; 7:8585. [PMID: 28819307 PMCID: PMC5561095 DOI: 10.1038/s41598-017-09268-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/18/2017] [Indexed: 12/27/2022] Open
Abstract
MicroRNAs (miRNAs) are important regulators of diverse physiological and pathophysiological processes. MiRNA families and clusters are two key features in miRNA biology. Here we explore the use of CRISPR/Cas9 as a powerful tool to delineate the function and regulation of miRNA families and clusters. We focused on four miRNA clusters composed of miRNA members of the same family, homo-clusters or different families, hetero-clusters. Our results highlight different regulatory mechanisms in miRNA cluster expression. In the case of the miR-497~195 cluster, editing of miR-195 led to a significant decrease in the expression of the other miRNA in the cluster, miR-497a. Although no gene editing was detected in the miR-497a genomic locus, computational simulation revealed alteration in the three dimensional structure of the pri-miR-497~195 that may affect its processing. In cluster miR-143~145 our results imply a feed-forward regulation, although structural changes cannot be ruled out. Furthermore, in the miR-17~92 and miR-106~25 clusters no interdependency in miRNA expression was observed. Our findings suggest that CRISPR/Cas9 is a powerful gene editing tool that can uncover novel mechanisms of clustered miRNA regulation and function.
Collapse
|
128
|
Sun G, Riggs AD. A Simple and Cost-Effective Approach for In Vitro Production of Sliced siRNAs as Potent Triggers for RNAi. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 8:345-355. [PMID: 28918034 PMCID: PMC5537206 DOI: 10.1016/j.omtn.2017.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 07/08/2017] [Accepted: 07/09/2017] [Indexed: 12/25/2022]
Abstract
We have studied the molecular properties of in-vitro-transcribed sliced small interfering RNAs (tsli-siRNAs) as an alternative RNAi agent for chemically synthesized siRNA. We describe here a simple and cost-effective procedure for high-purity production of tsli-siRNA using bacteriophage T7 RNA polymerases. tsli-siRNAs exhibit potent gene knockdown effects, with efficacy comparable with that of chemically synthesized sli-siRNAs and classical siRNAs. Furthermore, we found that it is very easy to prepare potent tsli-siRNAs with modified bases, such as 2′-fluorine- or biotin-16-modified tsli-siRNAs. tsli-siRNAs can cause a mild innate immune response, which can be easily eliminated by alkaline phosphatase treatment. On the other hand, this feature, which can be useful as a trigger of the innate immune response, can be enhanced by polynucleotide kinase treatment. Because of the simplicity of preparation and purification, the procedure presented here could be useful for the production of RNAi or immunostimulatory reagents.
Collapse
Affiliation(s)
- Guihua Sun
- Department of Diabetes Complications & Metabolism, Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, 1500 E. Duarte Road, Duarte, CA 91010, USA.
| | - Arthur D Riggs
- Department of Diabetes Complications & Metabolism, Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope, 1500 E. Duarte Road, Duarte, CA 91010, USA.
| |
Collapse
|
129
|
Lee D, Nam JW, Shin C. DROSHA targets its own transcript to modulate alternative splicing. RNA (NEW YORK, N.Y.) 2017; 23:1035-1047. [PMID: 28400409 PMCID: PMC5473138 DOI: 10.1261/rna.059808.116] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 04/06/2017] [Indexed: 05/23/2023]
Abstract
The nuclear RNase III enzyme DROSHA interacts with its cofactor DGCR8 to form the Microprocessor complex, which initiates microRNA (miRNA) maturation by cleaving hairpin structures embedded in primary transcripts. Apart from its central role in the biogenesis of miRNAs, DROSHA is also known to recognize and cleave miRNA-like hairpins in a subset of transcripts without apparent small RNA production. Here, we report that the human DROSHA transcript is one such noncanonical target of DROSHA. Mammalian DROSHA genes have evolved a conserved hairpin structure spanning a specific exon-intron junction, which serves as a substrate for the Microprocessor in human cells but not in murine cells. We show that it is this hairpin element that decides whether the overlapping exon is alternatively or constitutively spliced. We further demonstrate that DROSHA promotes skipping of the overlapping exon in human cells independently of its cleavage function. Our findings add to the expanding list of noncanonical DROSHA functions.
Collapse
Affiliation(s)
- Dooyoung Lee
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
| | - Jin-Wu Nam
- Department of Life Science, Hanyang University, Seoul 04763, Republic of Korea
| | - Chanseok Shin
- Department of Agricultural Biotechnology, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
- Plant Genomics and Breeding Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
130
|
Pfister EL, Chase KO, Sun H, Kennington LA, Conroy F, Johnson E, Miller R, Borel F, Aronin N, Mueller C. Safe and Efficient Silencing with a Pol II, but Not a Pol lII, Promoter Expressing an Artificial miRNA Targeting Human Huntingtin. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 7:324-334. [PMID: 28624208 PMCID: PMC5424568 DOI: 10.1016/j.omtn.2017.04.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 01/08/2023]
Abstract
Huntington's disease is a devastating, incurable neurodegenerative disease affecting up to 12 per 100,000 patients worldwide. The disease is caused by a mutation in the Huntingtin (Htt) gene. There is interest in reducing mutant Huntingtin by targeting it at the mRNA level, but the maximum tolerable dose and long-term effects of such a treatment are unknown. Using a self-complementary AAV9 vector, we delivered a mir-155-based artificial miRNA under the control of the chicken β-actin or human U6 promoter. In mouse brain, the artificial miRNA reduced the human huntingtin mRNA by 50%. The U6, but not the CβA promoter, produced the artificial miRNA at supraphysiologic levels. Embedding the antisense strand in a U6-mir-30 scaffold reduced expression of the antisense strand but increased the sense strand. In mice treated with scAAV9-U6-mir-155-HTT or scAAV9-CβA-mir-155-HTT, activated microglia were present around the injection site 1 month post-injection. Six months post-injection, mice treated with scAAV9-CβA-mir-155-HTT were indistinguishable from controls. Those that received scAAV9-U6-mir-155-HTT showed behavioral abnormalities and striatal damage. In conclusion, miRNA backbone and promoter can be used together to modulate expression levels and strand selection of artificial miRNAs, and in brain, the CβA promoter can provide an effective and safe dose of a human huntingtin miRNA.
Collapse
Affiliation(s)
- Edith L Pfister
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Kathryn O Chase
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Huaming Sun
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Lori A Kennington
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Faith Conroy
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Emily Johnson
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Rachael Miller
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Florie Borel
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Neil Aronin
- Department of Medicine and RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Christian Mueller
- Department of Pediatrics and Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
131
|
Kim B, Jeong K, Kim VN. Genome-wide Mapping of DROSHA Cleavage Sites on Primary MicroRNAs and Noncanonical Substrates. Mol Cell 2017; 66:258-269.e5. [DOI: 10.1016/j.molcel.2017.03.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/24/2017] [Accepted: 03/17/2017] [Indexed: 02/08/2023]
|
132
|
Wang Z, Zong S, Wang Z, Wu L, Chen P, Yun B, Cui Y. Microfluidic chip based micro RNA detection through the combination of fluorescence and surface enhanced Raman scattering techniques. NANOTECHNOLOGY 2017; 28:105501. [PMID: 28139463 DOI: 10.1088/1361-6528/aa527b] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
We present a novel microfluidic chip based method for the detection of micro RNA (miRNA) via the combination of fluorescence and surface enhanced Raman scattering (SERS) spectroscopies. First, silver nanoparticles (Ag NPs) are immobilized onto a glass slide, forming a SERS enhancing substrate. Then a specificially designed molecular beacon (MB) is attached to the SERS substrate. The 3' end of the MB is decorated with a thiol group to facilitate the attachment of the MB, while the 5' end of the MB is labeled with an organic dye 6-FAM, which is used both as the fluorophore and SERS reporter. In the absence of target miRNA, the MB will form a hairpin structure, making 6-FAM close to the Ag NPs. Hence, the fluorescence of 6-FAM will be quenched and the Raman signal of 6-FAM will be enhanced. On the contrary, with target miRNA present, hybridization between the miRNA and MB will unfold the MB and increase the distance between 6-FAM and the Ag NPs. Thus the fluorescence of 6-FAM will recover and the SERS signal of 6-FAM will decrease. So the target miRNA will simultaneously introduce opposite changing trends in the intensities of the fluorescence and SERS signals. By combining the opposite changes in the two optical spectra, an improved sensitivity and linearity toward the target miRNA is achieved as compared with using solely fluorescence or SERS. Moreover, introducing the microfluidic chip can reduce the reaction time, reagent dosage and complexity of detection. With the improved sensitivity and simplicity, we anticipate that the presented method can have great potential in the investigation of miRNA related diseases.
Collapse
|
133
|
The RNA-binding protein QKI5 regulates primary miR-124-1 processing via a distal RNA motif during erythropoiesis. Cell Res 2017; 27:416-439. [PMID: 28244490 DOI: 10.1038/cr.2017.26] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 10/18/2016] [Accepted: 11/11/2016] [Indexed: 02/07/2023] Open
Abstract
MicroRNA (miRNA) biogenesis is finely controlled by complex layers of post-transcriptional regulators, including RNA-binding proteins (RBPs). Here, we show that an RBP, QKI5, activates the processing of primary miR-124-1 (pri-124-1) during erythropoiesis. QKI5 recognizes a distal QKI response element and recruits Microprocessor through interaction with DGCR8. Furthermore, the recruited Microprocessor is brought to pri-124-1 stem loops by a spatial RNA-RNA interaction between two complementary sequences. Thus, mutations disrupting their base-pairing affect the strength of QKI5 activation. When erythropoiesis proceeds, the concomitant decrease of QKI5 releases Microprocessor from pri-124-1 and reduces mature miR-124 levels to facilitate erythrocyte maturation. Mechanistically, miR-124 targets TAL1 and c-MYB, two transcription factors involved in normal erythropoiesis. Importantly, this QKI5-mediated regulation also gives rise to a unique miRNA signature, which is required for erythroid differentiation. Taken together, these results demonstrate the pivotal role of QKI5 in primary miRNA processing during erythropoiesis and provide new insights into how a distal element on primary transcripts affects miRNA biogenesis.
Collapse
|
134
|
Bioinformatics Study of Structural Patterns in Plant MicroRNA Precursors. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6783010. [PMID: 28280737 PMCID: PMC5322449 DOI: 10.1155/2017/6783010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 12/18/2016] [Accepted: 01/12/2017] [Indexed: 01/26/2023]
Abstract
According to the RNA world theory, RNAs which stored genetic information and catalyzed chemical reactions had their contribution in the formation of current living organisms. In recent years, researchers studied this molecule diversity, i.a. focusing on small non-coding regulatory RNAs. Among them, of particular interest is evolutionarily ancient, 19–24 nt molecule of microRNA (miRNA). It has been already recognized as a regulator of gene expression in eukaryotes. In plants, miRNA plays a key role in the response to stress conditions and it participates in the process of growth and development. MicroRNAs originate from primary transcripts (pri-miRNA) encoded in the nuclear genome. They are processed from single-stranded stem-loop RNA precursors containing hairpin structures. While the mechanism of mature miRNA production in animals is better understood, its biogenesis in plants remains less clear. Herein, we present the results of bioinformatics analysis aimed at discovering how plant microRNAs are recognized within their precursors (pre-miRNAs). The study has been focused on sequential and structural motif identification in the neighbourhood of microRNA.
Collapse
|
135
|
Dalgaard LT, Eliasson L. An 'alpha-beta' of pancreatic islet microribonucleotides. Int J Biochem Cell Biol 2017; 88:208-219. [PMID: 28122254 DOI: 10.1016/j.biocel.2017.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 11/17/2022]
Abstract
MicroRNAs (miRNAs) are cellular, short, non-coding ribonucleotides acting as endogenous posttranscriptional repressors following incorporation in the RNA-induced silencing complex. Despite being chemically and mechanistically very similar, miRNAs exert a multitude of different cellular effects by acting on mRNA species, whose gene-products partake in a wide array of processes. Here, the aim was to review the knowledge of miRNA expression and action in the islet of Langerhans. We have focused on: 1) physiological consequences of islet or beta cell specific inhibition of miRNA processing, 2) mechanisms regulating processing of miRNAs in islet cells, 3) presence and function of miRNAs in alpha versus beta cells - the two main cell types of islets, and 4) miRNA mediators of beta cell decompensation. It is clear that miRNAs regulate pancreatic islet development, maturation, and function in vivo. Moreover, processing of miRNAs appears to be altered by obesity, diabetes, and aging. A number of miRNAs (such as miR-7, miR-21, miR-29, miR-34a, miR-212/miR-132, miR-184, miR-200 and miR-375) are involved in mediating beta cell dysfunction and/or compensation induced by hyperglycemia, oxidative stress, cytotoxic cytokines, and in rodent models of fetal metabolic programming prediabetes and overt diabetes. Studies of human type 2 diabetic islets underline that these miRNA families could have important roles also in human type 2 diabetes. Furthermore, there is a genuine gap of knowledge regarding miRNA expression and function in pancreatic alpha cells. Progress in this area would be enhanced by improved in vitro alpha cell models and better tools for islet cell sorting.
Collapse
Affiliation(s)
| | - Lena Eliasson
- Lund University Diabetes Center, Department of Clinical Sciences Malmö, CRC, SUS, Malmö, Sweden.
| |
Collapse
|
136
|
Ramalho-Carvalho J, Fromm B, Henrique R, Jerónimo C. Deciphering the function of non-coding RNAs in prostate cancer. Cancer Metastasis Rev 2017; 35:235-62. [PMID: 27221068 DOI: 10.1007/s10555-016-9628-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The advent of next-generation sequencing methods is fuelling the discovery of multiple non-coding RNA transcripts with direct implication in cell biology and homeostasis. This new layer of biological regulation seems to be of particular importance in human pathogenesis, including cancer. The aberrant expression of ncRNAs is a feature of prostate cancer, as they promote tumor-suppressive or oncogenic activities, controlling multicellular events leading to carcinogenesis and tumor progression. From the small RNAs involved in the RNAi pathway to the long non-coding RNAs controlling chromatin remodeling, alternative splicing, and DNA repair, the non-coding transcriptome represents the significant majority of transcriptional output. As such, ncRNAs appear as exciting new diagnostic, prognostic, and therapeutic tools. However, additional work is required to characterize the RNA species, their functions, and their applicability to clinical practice in oncology. In this review, we summarize the most important features of ncRNA biology, emphasizing its relevance in prostate carcinogenesis and its potential for clinical applications.
Collapse
Affiliation(s)
- João Ramalho-Carvalho
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Porto, Portugal.,Biomedical Sciences Graduate Program, Institute of Biomedical Sciences Abel Salazar-University of Porto (ICBAS-UP), Porto, Portugal
| | - Bastian Fromm
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Nydalen, N-0424, Oslo, Norway
| | - Rui Henrique
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Porto, Portugal.,Departments of Pathology, Portuguese Oncology Institute of Porto, Porto, Portugal.,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology & Epigenetics Group - Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Porto, Portugal. .,Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar, University of Porto (ICBAS-UP), Porto, Portugal. .,Portuguese Oncology Institute of Porto, Research Center-LAB 3, F Bdg, 1st floor, Rua Dr António Bernardino de Almeida, 4200-072, Porto, Portugal.
| |
Collapse
|
137
|
Roden C, Gaillard J, Kanoria S, Rennie W, Barish S, Cheng J, Pan W, Liu J, Cotsapas C, Ding Y, Lu J. Novel determinants of mammalian primary microRNA processing revealed by systematic evaluation of hairpin-containing transcripts and human genetic variation. Genome Res 2017; 27:374-384. [PMID: 28087842 PMCID: PMC5340965 DOI: 10.1101/gr.208900.116] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 01/06/2017] [Indexed: 12/26/2022]
Abstract
Mature microRNAs (miRNAs) are processed from hairpin-containing primary miRNAs (pri-miRNAs). However, rules that distinguish pri-miRNAs from other hairpin-containing transcripts in the genome are incompletely understood. By developing a computational pipeline to systematically evaluate 30 structural and sequence features of mammalian RNA hairpins, we report several new rules that are preferentially utilized in miRNA hairpins and govern efficient pri-miRNA processing. We propose that a hairpin stem length of 36 ± 3 nt is optimal for pri-miRNA processing. We identify two bulge-depleted regions on the miRNA stem, located ∼16-21 nt and ∼28-32 nt from the base of the stem, that are less tolerant of unpaired bases. We further show that the CNNC primary sequence motif selectively enhances the processing of optimal-length hairpins. We predict that a small but significant fraction of human single-nucleotide polymorphisms (SNPs) alter pri-miRNA processing, and confirm several predictions experimentally including a disease-causing mutation. Our study enhances the rules governing mammalian pri-miRNA processing and suggests a diverse impact of human genetic variation on miRNA biogenesis.
Collapse
Affiliation(s)
- Christine Roden
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA.,Yale Stem Cell Center and Yale Cancer Center, Yale University, New Haven, Connecticut 06520, USA.,Yale Center for RNA Science and Medicine, New Haven, Connecticut 06520, USA.,Graduate Program in Biological and Biomedical Sciences, Yale University, New Haven, Connecticut 06510, USA
| | - Jonathan Gaillard
- Yale Stem Cell Center and Yale Cancer Center, Yale University, New Haven, Connecticut 06520, USA.,School of Medicine, Yale University, New Haven, Connecticut 06510, USA
| | - Shaveta Kanoria
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, USA
| | - William Rennie
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, USA
| | - Syndi Barish
- Graduate Program in Biological and Biomedical Sciences, Yale University, New Haven, Connecticut 06510, USA
| | - Jijun Cheng
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA.,Yale Stem Cell Center and Yale Cancer Center, Yale University, New Haven, Connecticut 06520, USA.,Yale Center for RNA Science and Medicine, New Haven, Connecticut 06520, USA
| | - Wen Pan
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA.,Yale Stem Cell Center and Yale Cancer Center, Yale University, New Haven, Connecticut 06520, USA.,Yale Center for RNA Science and Medicine, New Haven, Connecticut 06520, USA
| | - Jun Liu
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA.,Yale Stem Cell Center and Yale Cancer Center, Yale University, New Haven, Connecticut 06520, USA.,Yale Center for RNA Science and Medicine, New Haven, Connecticut 06520, USA
| | - Chris Cotsapas
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA.,Department of Neurology, Yale School of Medicine, New Haven, Connecticut 06511, USA
| | - Ye Ding
- Wadsworth Center, New York State Department of Health, Albany, New York 12208, USA
| | - Jun Lu
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06510, USA.,Yale Stem Cell Center and Yale Cancer Center, Yale University, New Haven, Connecticut 06520, USA.,Yale Center for RNA Science and Medicine, New Haven, Connecticut 06520, USA.,Yale Cooperative Center of Excellence in Hematology, Yale University, New Haven, Connecticut 06520, USA
| |
Collapse
|
138
|
Tian C, Yao S, Liu L, Ding Y, Ye Q, Dong X, Gao Y, Yang N, Li Q. Klf4 inhibits tumor growth and metastasis by targeting microRNA-31 in human hepatocellular carcinoma. Int J Mol Med 2016; 39:47-56. [PMID: 27909734 PMCID: PMC5179175 DOI: 10.3892/ijmm.2016.2812] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 11/18/2016] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs or miRs) are short, endogenous non-coding RNA molecules, demonstrating abnormal expression in cancer initiation and progression. In this study, we profiled 18 differentially regulated miRNAs, including miRNA-31, using miRNA array. Kruppel (or Krüppel)-like factor 4 (Klf4) is a transcription factor and putative tumor suppressor. Both were found to be significantly downregulated in liver cancer tissues and cells. However, little is known about the correlation between Klf4 and miRNA-31 in hepatocellular carcinoma (HCC). The mRNA expression of Klf4 was decreased and inversely associated with the clinical stage, T classification and hepatitis B in patients with HCC, while the expression of miR-31 was lower (r=0.326, P=0.018). Using cell counting kit 8 (CCK8) and Transwell migration assays, we found that Klf4 and miR-31 inhibited the proliferation and metastasis of liver cancer cells. Moreover, we demonstrated that Klf4 directly binds to the promoter of miR-31 and activates its transcription. In vitro experiments confirmed that Klf4 regulated miR-31 and thereby inhibited HCC cell growth and metastasis. Taken together, our findings indicate that Klf4 directly regulates miR-31 in HCC. Thus, miR-31 may serve as a potential diagnostic marker and therapeutic target in HCC.
Collapse
Affiliation(s)
- Chuan Tian
- Department of Oncology, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, P.R. China
| | - Shanshan Yao
- Department of Oncology, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, P.R. China
| | - Li Liu
- Department of Pharmacy, Guiyang Hospital of Guizhou Aviation Industry Group, Guiyang, Guizhou, P.R. China
| | - Youcheng Ding
- Department of General Surgery, Shanghai East Hospital, Tongji University, Shanghai, P.R. China
| | - Qingwang Ye
- Department of Liver Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Xiao Dong
- Department of Oncology, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, P.R. China
| | - Yong Gao
- Department of Oncology, Shanghai East Hospital, Tongji University, Shanghai, P.R. China
| | - Ning Yang
- Department of Liver Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, P.R. China
| | - Qi Li
- Department of Oncology, Shanghai First People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, P.R. China
| |
Collapse
|
139
|
Watanabe C, Cuellar TL, Haley B. Quantitative evaluation of first, second, and third generation hairpin systems reveals the limit of mammalian vector-based RNAi. RNA Biol 2016; 13:25-33. [PMID: 26786363 PMCID: PMC4829305 DOI: 10.1080/15476286.2015.1128062] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Incorporating miRNA-like features into vector-based hairpin scaffolds has been shown to augment small RNA processing and RNAi efficiency. Therefore, defining an optimal, native hairpin context may obviate a need for hairpin-specific targeting design schemes, which confound the movement of functional siRNAs into shRNA/artificial miRNA backbones, or large-scale screens to identify efficacious sequences. Thus, we used quantitative cell-based assays to compare separate third generation artificial miRNA systems, miR-E (based on miR-30a) and miR-3G (based on miR-16-2 and first described in this study) to widely-adopted, first and second generation formats in both Pol-II and Pol-III expression vector contexts. Despite their unique structures and strandedness, and in contrast to first and second-generation RNAi triggers, the third generation formats operated with remarkable similarity to one another, and strong silencing was observed with a significant fraction of the evaluated target sequences within either promoter context. By pairing an established siRNA design algorithm with the third generation vectors we could readily identify targeting sequences that matched or exceeded the potency of those discovered through large-scale sensor-based assays. We find that third generation hairpin systems enable the maximal level of siRNA function, likely through enhanced processing and accumulation of precisely-defined guide RNAs. Therefore, we predict future gains in RNAi potency will come from improved hairpin expression and identification of optimal siRNA-intrinsic silencing properties rather than further modification of these scaffolds. Consequently, third generation systems should be the primary format for vector-based RNAi studies; miR-3G is advantageous due to its small expression cassette and simplified, cost-efficient cloning scheme.
Collapse
Affiliation(s)
- Colin Watanabe
- a Departments of Bioinformatics and Computational Biology , South San Francisco , CA 94080.,c Genentech, Inc . South San Francisco , CA 94080 , USA
| | - Trinna L Cuellar
- b Molecular Biology South San Francisco , CA 94080.,c Genentech, Inc . South San Francisco , CA 94080 , USA
| | - Benjamin Haley
- b Molecular Biology South San Francisco , CA 94080.,c Genentech, Inc . South San Francisco , CA 94080 , USA
| |
Collapse
|
140
|
Mishima T, Sadovsky E, Gegick ME, Sadovsky Y. Determinants of effective lentivirus-driven microRNA expression in vivo. Sci Rep 2016; 6:33345. [PMID: 27627961 PMCID: PMC5024309 DOI: 10.1038/srep33345] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/24/2016] [Indexed: 01/08/2023] Open
Abstract
Manipulation of microRNA (miRNA) levels, including overexpression of mature species, has become an important biological tool, even motivating miRNA-based therapeutics. To assess key determinants of miRNA overexpression in a mammalian system in vivo, we sought to bypass the laborious generation of a transgenic animal by exploiting placental trophoblast-specific gene manipulation using lentiviral vectors, which has been instrumental in elucidating trophoblast biology. We examined the impact of several key components of miRNA stem loops and their flanking sequences on the efficiency of mature miRNA expression in vivo. By combining established and novel approaches for miRNA expression, we engineered lentivirus-driven miRNA expression plasmids, which we tested in the mouse placenta. We found that reverse sense inserts minimized single-strand splicing and degradation, and that maintaining longer, poly-A-containing arms flanking the miRNA stem-loop markedly enhanced transgenic miRNA expression. Additionally, we accomplished overexpression of diverse mammalian, drosophila, or C. elegans miRNAs, either based on native context or using a “cassette” replacement of the mature miRNA sequence. Together, we have identified primary miRNA sequences that are paramount for effective expression of mature miRNAs, and validated their role in mice. Principles established by our findings may guide the design of efficient miRNA vectors for in vivo use.
Collapse
Affiliation(s)
- Takuya Mishima
- Magee-Womens Research Institute, Department of Obstetrics, Gynaecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213 USA
| | - Elena Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynaecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213 USA
| | - Margaret E Gegick
- Magee-Womens Research Institute, Department of Obstetrics, Gynaecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213 USA
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynaecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, 15213 USA
| |
Collapse
|
141
|
Narayanan A, Hill-Teran G, Moro A, Ristori E, Kasper DM, A. Roden C, Lu J, Nicoli S. In vivo mutagenesis of miRNA gene families using a scalable multiplexed CRISPR/Cas9 nuclease system. Sci Rep 2016; 6:32386. [PMID: 27572667 PMCID: PMC5004112 DOI: 10.1038/srep32386] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/08/2016] [Indexed: 01/06/2023] Open
Abstract
A large number of microRNAs (miRNAs) are grouped into families derived from the same phylogenetic ancestors. miRNAs within a family often share the same physiological functions despite differences in their primary sequences, secondary structures, or chromosomal locations. Consequently, the generation of animal models to analyze the activity of miRNA families is extremely challenging. Using zebrafish as a model system, we successfully provide experimental evidence that a large number of miRNAs can be simultaneously mutated to abrogate the activity of an entire miRNA family. We show that injection of the Cas9 nuclease and two, four, ten, and up to twenty-four multiplexed single guide RNAs (sgRNAs) can induce mutations in 90% of the miRNA genomic sequences analyzed. We performed a survey of these 45 mutations in 10 miRNA genes, analyzing the impact of our mutagenesis strategy on the processing of each miRNA both computationally and in vivo. Our results offer an effective approach to mutate and study the activity of miRNA families and pave the way for further analysis on the function of complex miRNA families in higher multicellular organisms.
Collapse
Affiliation(s)
- Anand Narayanan
- Yale Cardiovascular Research Center, Section of Cardiology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Guillermina Hill-Teran
- Yale Cardiovascular Research Center, Section of Cardiology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Albertomaria Moro
- Yale Cardiovascular Research Center, Section of Cardiology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Emma Ristori
- Yale Cardiovascular Research Center, Section of Cardiology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Dionna M. Kasper
- Yale Cardiovascular Research Center, Section of Cardiology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Christine A. Roden
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale University, New Haven, CT, 06520, USA
| | - Jun Lu
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale University, New Haven, CT, 06520, USA
| | - Stefania Nicoli
- Yale Cardiovascular Research Center, Section of Cardiology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
142
|
Bofill-De Ros X, Gu S. Guidelines for the optimal design of miRNA-based shRNAs. Methods 2016; 103:157-66. [PMID: 27083402 PMCID: PMC4921303 DOI: 10.1016/j.ymeth.2016.04.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 03/28/2016] [Accepted: 04/04/2016] [Indexed: 12/21/2022] Open
Abstract
RNA interference (RNAi) is an extremely useful tool for inhibiting gene expression. It can be triggered by transfected synthetic small interfering RNA (siRNA) or by expressed small hairpin RNA (shRNA). The cellular machinery processes the latter into siRNA in vivo. shRNA is preferred or required in genetic screens and specific RNAi approaches in gene therapy settings. Despite its many successes, the field of shRNAs faces many challenges. Insufficient knockdowns and off-target effects become obstacles for shRNA usage in many applications. Numerous failures are triggered by pitfalls in shRNA design that is often associated with impoverished biogenesis. Here, based on current understanding of the miRNA maturation pathway, we discuss the principles of different shRNA design (pre-miRNA-like, pri-miRNA-like and Ago-shRNA) with an emphasis on the RNA structure. We also provide detailed instructions for an optimal design of pre-miRNA-like shRNA.
Collapse
Affiliation(s)
- Xavier Bofill-De Ros
- Gene Regulation and Chromosome Biology Laboratory, Center For Cancer Research, National Cancer Institute, Frederick, MD, United States
| | - Shuo Gu
- Gene Regulation and Chromosome Biology Laboratory, Center For Cancer Research, National Cancer Institute, Frederick, MD, United States.
| |
Collapse
|
143
|
Chen Y, Zubovic L, Yang F, Godin K, Pavelitz T, Castellanos J, Macchi P, Varani G. Rbfox proteins regulate microRNA biogenesis by sequence-specific binding to their precursors and target downstream Dicer. Nucleic Acids Res 2016; 44:4381-95. [PMID: 27001519 PMCID: PMC4872098 DOI: 10.1093/nar/gkw177] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 02/26/2016] [Accepted: 03/07/2016] [Indexed: 12/26/2022] Open
Abstract
Rbfox proteins regulate tissue-specific splicing by targeting a conserved GCAUG sequence within pre-mRNAs. We report here that sequence-specific binding of the conserved Rbfox RRM to miRNA precursors containing the same sequence motif in their terminal loops, including miR-20b and miR-107, suppresses their nuclear processing. The structure of the complex between precursor miR-20b and Rbfox RRM shows the molecular basis for recognition, and reveals changes in the stem-loop upon protein binding. In mammalian cells, Rbfox2 downregulates mature miR-20b and miR-107 levels and increases the expression of their downstream targets PTEN and Dicer, respectively, suggesting that Rbfox2 indirectly regulates many more cellular miRNAs. Thus, some of the widespread cellular functions of Rbfox2 protein are attributable to regulation of miRNA biogenesis, and might include the mis-regulation of miR-20b and miR-107 in cancer and neurodegeneration.
Collapse
Affiliation(s)
- Yu Chen
- Center for Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento (TN), Italy
| | - Lorena Zubovic
- Department of Chemistry, University of Washington, Seattle, WA 98195-1700, USA
| | - Fan Yang
- Center for Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento (TN), Italy
| | - Katherine Godin
- Center for Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento (TN), Italy
| | - Tom Pavelitz
- Center for Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento (TN), Italy
| | - Javier Castellanos
- Center for Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento (TN), Italy
| | - Paolo Macchi
- Department of Chemistry, University of Washington, Seattle, WA 98195-1700, USA
| | - Gabriele Varani
- Center for Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Trento (TN), Italy
| |
Collapse
|
144
|
Abstract
DROSHA and its partner DGCR8 form a heterotrimeric complex named Microprocessor, which is essential for microRNA biogenesis. A recent study by Kwon et al. in Cell reveals the structure of a DROSHA construct in complex with the C-terminal region of DGCR8, thereby unveiling the topology and interactions between components of the Microprocessor and insights into its 'ruler'-based cleavage activity and function.
Collapse
|
145
|
Galka-Marciniak P, Olejniczak M, Starega-Roslan J, Szczesniak MW, Makalowska I, Krzyzosiak WJ. siRNA release from pri-miRNA scaffolds is controlled by the sequence and structure of RNA. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:639-49. [DOI: 10.1016/j.bbagrm.2016.02.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 02/19/2016] [Accepted: 02/23/2016] [Indexed: 01/17/2023]
|
146
|
Zhao Y, Alexandrov PN, Lukiw WJ. Anti-microRNAs as Novel Therapeutic Agents in the Clinical Management of Alzheimer's Disease. Front Neurosci 2016; 10:59. [PMID: 26941600 PMCID: PMC4766517 DOI: 10.3389/fnins.2016.00059] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/08/2016] [Indexed: 12/21/2022] Open
Abstract
Overview- One hundred and ten years since its first description Alzheimer's disease (AD) still retains its prominent status: (i) as the industrialized world's number one cause of age-related intellectual impairment and cognitive decline; (ii) as this country's most rapidly expanding socioeconomic and healthcare concern; and (iii) as an insidious, progressive and lethal neurological disorder of the human central nervous system (CNS) for which there is currently no adequate treatment or cure (Alzheimer, 1991; Alzheimer et al., 1991, 1995) [https://www.alz.org/facts/downloads/facts_figures_2015.pdf (2015)]. The concept of small non-coding RNAs (ncRNAs) as being involved in the etiopathogenesis of AD and age-related human neurodegenerative disease was first proposed about 25 years ago, however it was not until 2007 that specific microRNA (miRNA) abundance, speciation and localization to the hippocampal CA1 region (an anatomical area of the human CNS specifically targeted by the AD process) was shown to strongly associate with AD-type change when compared to age-matched controls (Lukiw et al., 1992; Lukiw, 2007; Schipper et al., 2007; Cogswell et al., 2008; Guerreiro et al., 2012). Currently about 400 reports address the potential link between disruptions in miRNA signaling and the development of various features associated with AD neuropathology (http://www.ncbi.nlm.nih.gov/pubmed/?term=micro+RNA+alzheimer's+disease). In this “Perspectives” paper we will highlight some of the most recent literature on anti-miRNA (AM; antagomir) therapeutic strategies and some very recent technological advances in the analysis and characterization of defective miRNA signaling pathways in AD compared to neurologically normal age-matched controls.
Collapse
Affiliation(s)
- Yuhai Zhao
- LSU Neuroscience Center, Louisiana State University Health Science CenterNew Orleans, LA, USA; Department of Cell Biology and Anatomy, Louisiana State University Health Science CenterNew Orleans, LA, USA
| | | | - Walter J Lukiw
- LSU Neuroscience Center, Louisiana State University Health Science CenterNew Orleans, LA, USA; Department of Ophthalmology, LSU Neuroscience Center, Louisiana State University Health Science CenterNew Orleans, LA, USA; Department Neurology, LSU Neuroscience Center, Louisiana State University Health Science CenterNew Orleans, LA, USA
| |
Collapse
|
147
|
van den Berg FT, Rossi JJ, Arbuthnot P, Weinberg MS. Design of Effective Primary MicroRNA Mimics With Different Basal Stem Conformations. MOLECULAR THERAPY-NUCLEIC ACIDS 2016; 5:e278. [PMID: 26756196 PMCID: PMC5012551 DOI: 10.1038/mtna.2015.53] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 11/30/2015] [Indexed: 12/03/2022]
Abstract
Primary microRNA (pri-miRNA) mimics are important mediators of effective gene silencing and are well suited for sustained therapeutic applications. Pri-miRNA mimics are processed in the endogenous miRNA biogenesis pathway, where elements of the secondary RNA structure are crucial for efficient miRNA production. Cleavage of the pri-miRNA to a precursor miRNA (pre-miRNA) by Drosha-DGCR8 typically occurs adjacent to a basal stem of ~11 bp. However, a number of pri-miRNA structures are expected to contain slightly shorter or longer basal stems, which may be further disrupted in predicted folding of the expressed pri-miRNA sequence. We investigated the function and processing of natural and exogenous RNA guides from pri-miRNAs with various basal stems (9–13 bp), where a canonical hairpin was predicted to be well or poorly maintained in predicted structures of the expressed sequence. We have shown that RNA guides can be effectively derived from pri-miRNAs with various basal stem conformations, while predicted guide region stability can explain the function of pri-miRNA mimics, in agreement with previously proposed design principles. This study provides insight for the design of effective mimics based on naturally occurring pri-miRNAs and has identified several novel scaffolds suitable for use in gene silencing applications.
Collapse
Affiliation(s)
- Fiona T van den Berg
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Department of Molecular Medicine and Haematology, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - John J Rossi
- Department of Molecular and Cellular Biology, Beckman Research Institute of City of Hope, Duarte, California, USA
| | - Patrick Arbuthnot
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Department of Molecular Medicine and Haematology, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Marc S Weinberg
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Department of Molecular Medicine and Haematology, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa.,HIV Pathogenesis Research Unit, Department of Molecular Medicine and Haematology, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa.,Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
148
|
Denzler R, Stoffel M. The Long, the Short, and the Unstructured: A Unifying Model of miRNA Biogenesis. Mol Cell 2016; 60:4-6. [PMID: 26431024 DOI: 10.1016/j.molcel.2015.09.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this issue, Fang and Bartel (2015) report the identification of novel sequence and structural features of human pri-miRNAs, which--together with previously identified sequence motifs--define a unifying model of mammalian pri-miRNAs and advances the de novo design of artificial pri-miRNAs.
Collapse
Affiliation(s)
- Rémy Denzler
- Institute of Molecular Health Sciences, ETH Zurich, Otto-Stern-Weg 7, HPL H36, 8093 Zurich, Switzerland.
| | - Markus Stoffel
- Institute of Molecular Health Sciences, ETH Zurich, Otto-Stern-Weg 7, HPL H36, 8093 Zurich, Switzerland.
| |
Collapse
|
149
|
Kwon SC, Nguyen TA, Choi YG, Jo MH, Hohng S, Kim VN, Woo JS. Structure of Human DROSHA. Cell 2015; 164:81-90. [PMID: 26748718 DOI: 10.1016/j.cell.2015.12.019] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 11/28/2015] [Accepted: 12/07/2015] [Indexed: 01/29/2023]
Abstract
MicroRNA maturation is initiated by RNase III DROSHA that cleaves the stem loop of primary microRNA. DROSHA functions together with its cofactor DGCR8 in a heterotrimeric complex known as Microprocessor. Here, we report the X-ray structure of DROSHA in complex with the C-terminal helix of DGCR8. We find that DROSHA contains two DGCR8-binding sites, one on each RNase III domain (RIIID), which mediate the assembly of Microprocessor. The overall structure of DROSHA is surprisingly similar to that of Dicer despite no sequence homology apart from the C-terminal part, suggesting that DROSHA may have evolved from a Dicer homolog. DROSHA exhibits unique features, including non-canonical zinc-finger motifs, a long insertion in the first RIIID, and the kinked link between Connector helix and RIIID, which explains the 11-bp-measuring "ruler" activity of DROSHA. Our study implicates the evolutionary origin of DROSHA and elucidates the molecular basis of Microprocessor assembly and primary microRNA processing.
Collapse
Affiliation(s)
- S Chul Kwon
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea; School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Tuan Anh Nguyen
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea; School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Yeon-Gil Choi
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea; School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Myung Hyun Jo
- Department of Physics and Astronomy, Seoul National University, Seoul 08826, Korea; National Center for Creative Research Initiatives, Seoul National University, Seoul 08826, Korea; Institute of Applied Physics, Seoul National University, Seoul 08826, Korea
| | - Sungchul Hohng
- Department of Physics and Astronomy, Seoul National University, Seoul 08826, Korea; National Center for Creative Research Initiatives, Seoul National University, Seoul 08826, Korea; Institute of Applied Physics, Seoul National University, Seoul 08826, Korea; Department of Biophysics and Chemical Biology, Seoul National University, Seoul 08826, Korea
| | - V Narry Kim
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea; School of Biological Sciences, Seoul National University, Seoul 08826, Korea.
| | - Jae-Sung Woo
- Center for RNA Research, Institute for Basic Science, Seoul 08826, Korea; School of Biological Sciences, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
150
|
RNA Binding Proteins in the miRNA Pathway. Int J Mol Sci 2015; 17:ijms17010031. [PMID: 26712751 PMCID: PMC4730277 DOI: 10.3390/ijms17010031] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 12/13/2015] [Accepted: 12/23/2015] [Indexed: 12/21/2022] Open
Abstract
microRNAs (miRNAs) are short ~22 nucleotides (nt) ribonucleic acids which post-transcriptionally regulate gene expression. miRNAs are key regulators of all cellular processes, and the correct expression of miRNAs in an organism is crucial for proper development and cellular function. As a result, the miRNA biogenesis pathway is highly regulated. In this review, we outline the basic steps of miRNA biogenesis and miRNA mediated gene regulation focusing on the role of RNA binding proteins (RBPs). We also describe multiple mechanisms that regulate the canonical miRNA pathway, which depends on a wide range of RBPs. Moreover, we hypothesise that the interaction between miRNA regulation and RBPs is potentially more widespread based on the analysis of available high-throughput datasets.
Collapse
|