137 results on '"Gregory, Richard I."'
Search Results
102. Post-transcriptional control of DGCR8 expression by the Microprocessor
- Author
-
Triboulet, Robinson, primary, Chang, Hao-Ming, additional, LaPierre, Robert J., additional, and Gregory, Richard I., additional
- Published
- 2009
- Full Text
- View/download PDF
103. Many roads to maturity: microRNA biogenesis pathways and their regulation
- Author
-
Winter, Julia, primary, Jung, Stephanie, additional, Keller, Sarina, additional, Gregory, Richard I., additional, and Diederichs, Sven, additional
- Published
- 2009
- Full Text
- View/download PDF
104. Podocyte-Specific Loss of Functional MicroRNAs Leads to Rapid Glomerular and Tubular Injury
- Author
-
Ho, Jacqueline, primary, Ng, Kar Hui, additional, Rosen, Seymour, additional, Dostal, Ales, additional, Gregory, Richard I., additional, and Kreidberg, Jordan A., additional
- Published
- 2008
- Full Text
- View/download PDF
105. Determinants of MicroRNA Processing Inhibition by the Developmentally Regulated RNA-binding Protein Lin28
- Author
-
Piskounova, Elena, primary, Viswanathan, Srinivas R., additional, Janas, Maja, additional, LaPierre, Robert J., additional, Daley, George Q., additional, Sliz, Piotr, additional, and Gregory, Richard I., additional
- Published
- 2008
- Full Text
- View/download PDF
106. MicroRNA Regulation of Stem Cell Fate
- Author
-
Li, Qintong, primary and Gregory, Richard I., additional
- Published
- 2008
- Full Text
- View/download PDF
107. MicroRNA Biogenesis and Cancer
- Author
-
Gregory, Richard I., primary and Shiekhattar, Ramin, additional
- Published
- 2005
- Full Text
- View/download PDF
108. Chromatin modifiers and carcinogenesis
- Author
-
Gregory, Richard I., primary and Shiekhattar, Ramin, additional
- Published
- 2004
- Full Text
- View/download PDF
109. Inhibition of Histone Deacetylases Alters Allelic Chromatin Conformation at the Imprinted U2af1-rs1 Locus in Mouse Embryonic Stem Cells
- Author
-
Gregory, Richard I., primary, O'Neill, Laura P., additional, Randall, Tamzin E., additional, Fournier, Cecile, additional, Khosla, Sanjeev, additional, Turner, Bryan M., additional, and Feil, Robert, additional
- Published
- 2002
- Full Text
- View/download PDF
110. MicroRNA Biogenesis.
- Author
-
Walker, John M., Ying, Shao-Yao, Gregory, Richard I., Chendrimada, Thimmaiah P., and Shiekhattar, Ramin
- Abstract
The recently discovered microRNAs (miRNAs) are a large family of small regulatory RNAs that have been implicated in controlling diverse pathways in a variety of organisms (1,2). For posttranscriptional gene silencing, one strand of the miRNA is used to guide components of the RNA interference machinery, including Argonaute 2, to messenger RNAs (mRNAs) with complementary sequences (3,4). Thus, targeted mRNAs are either cleaved by the endonuclease Argonaute 2 (5,6), or protein synthesis is blocked by an as yet uncharacterized mechanism (7,8). Genes encoding miRNAs are transcribed as long primary miRNAs (pri-miRNAs) that are sequentially processed by components of the nucleus and cytoplasm to yield a mature, approx 22-nucleotide (nt)-long miRNA (9). Two members of the ribonuclease (RNase) III endonuclease protein family, Drosha and Dicer, have been implicated in this two-step processing (10-13). To further our understanding of miRNA biogenesis and function it will be essential to identify the protein complexes involved. We were interested in defining the proteins required for the initial nuclear processing of pri-miRNAs to the approx 60- to 70-nt stem-loop intermediates known as precursor miRNAs (pre-miRNAs) (9,10). This led to our identification of a protein complex we termed Microprocessor, which is necessary and sufficient for processing pri-miRNA to premiRNAs (14). The Microprocessor complex comprises Drosha and the double-stranded RNAbinding protein DiGeorge syndrome critical region 8 gene (DGCR8), which is deleted in DiGeorge syndrome (15,16). In this chapter, we detail the methods used for the biochemical isolation and identification of the Microprocessor complex from human cells. We include a protocol for the in vitro analysis of pri-miRNA processing activity of the purified Microprocessor complex. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
111. DNA Methylation Is Linked to Deacetylation of Histone H3, but Not H4, on the Imprinted Genes Snrpnand U2af1-rs1
- Author
-
Gregory, Richard I., primary, Randall, Tamzin E., additional, Johnson, Colin A., additional, Khosla, Sanjeev, additional, Hatada, Izuho, additional, O'Neill, Laura P., additional, Turner, Bryan M., additional, and Feil, Robert, additional
- Published
- 2001
- Full Text
- View/download PDF
112. Environmental effects on genomic imprinting in mammals
- Author
-
Thompson, Samantha L., primary, Konfortova, Galia, additional, Gregory, Richard I., additional, Reik, Wolf, additional, Dean, Wendy, additional, and Feil, Robert, additional
- Published
- 2001
- Full Text
- View/download PDF
113. Probing Chromatin Structure with Nuclease Sensitivity Assays.
- Author
-
Ward, Andrew, Gregory, Richard I., Khosla, Sanjeev, and Feil, Robert
- Abstract
To further our understanding of genomic imprinting it will be essential to identify key control elements, and to investigate their regulation by both epigenetic modifications (such as DNA methylation) and trans-acting factors. So far, sequence elements that regulate parental allele-specific gene expression have been identified in a number of imprinted loci, either because of their differential DNA methylation or through functional studies in transgenic mice (1,2). A systematic search for allele-specific chromatin features constitutes an alternative strategy to identify elements that regulate imprinting. The validity of such an in vivo chromatin approach derives from the fact that in several known imprinting control-elements, a specialized organization of chromatin characterized by nuclease hypersensitivity is present on only one of the two parental chromosome (3). For example, the differentially methylated 5′-portion of the human SNRPN gene-a sequence element that controls imprinting in the Prader-Willi and Angelman syndromes' domain on chromosome 15q11-q13-has strong DNase-I hypersensitive sites on the unmethylated paternal chromosome (4). A differentially methylated region that regulates the imprinting of H19 and that of the neighboring insulin-like growth factor-2 gene on mouse chromosome 7 was also found to have parental chromosomespecific hypersensitive sites (5,6). The precise nature of the allelic nuclease hypersensitivity in these and other imprinted loci remains to be determined in more detail, for example, by applying complementary chromatin methodologies (7,8). However, it is comMonly observed that a nuclease hypersensitive site corresponds to a small region where nucleosomes are absent or partially disrupted. [ABSTRACT FROM AUTHOR]
- Published
- 2002
- Full Text
- View/download PDF
114. PRC2 Is Required to Maintain Expression of the Maternal Gtl2-Rian-MirgLocus by Preventing De Novo DNA Methylation in Mouse Embryonic Stem Cells
- Author
-
Das, Partha Pratim, Hendrix, David A., Apostolou, Effie, Buchner, Alice H., Canver, Matthew C., Beyaz, Semir, Ljuboja, Damir, Kuintzle, Rachael, Kim, Woojin, Karnik, Rahul, Shao, Zhen, Xie, Huafeng, Xu, Jian, De Los Angeles, Alejandro, Zhang, Yingying, Choe, Junho, Jun, Don Leong Jia, Shen, Xiaohua, Gregory, Richard I., Daley, George Q., Meissner, Alexander, Kellis, Manolis, Hochedlinger, Konrad, Kim, Jonghwan, and Orkin, Stuart H.
- Abstract
Polycomb Repressive Complex 2 (PRC2) function and DNA methylation (DNAme) are typically correlated with gene repression. Here, we show that PRC2 is required to maintain expression of maternal microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) from the Gtl2-Rian-Mirglocus, which is essential for full pluripotency of iPSCs. In the absence of PRC2, the entire locus becomes transcriptionally repressed due to gain of DNAme at the intergenic differentially methylated regions (IG-DMRs). Furthermore, we demonstrate that the IG-DMR serves as an enhancer of the maternal Gtl2-Rian-Mirglocus. Further analysis reveals that PRC2 interacts physically with Dnmt3 methyltransferases and reduces recruitment to and subsequent DNAme at the IG-DMR, thereby allowing for proper expression of the maternal Gtl2-Rian-Mirglocus. Our observations are consistent with a mechanism through which PRC2 counteracts the action of Dnmt3 methyltransferases at an imprinted locus required for full pluripotency.
- Published
- 2015
- Full Text
- View/download PDF
115. LIN28 phosphorylation by MAPK/ERK couples signaling to the post-transcriptional control of pluripotency
- Author
-
Tsanov, Kaloyan M., Pearson, Daniel S., Wu, Zhaoting, Han, Areum, Triboulet, Robinson, Seligson, Marc T., Powers, John T., Osborne, Jihan K., Kane, Susan, Gygi, Steven P., Gregory, Richard I., and Daley, George Q.
- Abstract
Signaling and post-transcriptional gene control are both critical for the regulation of pluripotency1,2, yet how they are integrated to influence cell identity remains poorly understood. LIN28 (also known as LIN28A), a highly conserved RNA-binding protein (RBP), has emerged as a central post-transcriptional regulator of cell fate through blockade of let-7 microRNA (miRNA) biogenesis and direct modulation of mRNA translation3. Here we show that LIN28 is phosphorylated by MAPK/ERK in pluripotent stem cells (PSCs), which increases its levels via post-translational stabilization. LIN28 phosphorylation had little impact on let-7 but enhanced LIN28’s effect on its direct mRNA targets, revealing a mechanism that uncouples LIN28’s let-7-dependent and independent activities. We have linked this mechanism to the induction of pluripotency by somatic cell reprogramming and the transition from naïve to primed pluripotency. Collectively, our findings indicate that MAPK/ERK directly impacts LIN28, defining an axis that connects signaling, post-transcriptional gene control, and cell fate regulation.
- Published
- 2016
- Full Text
- View/download PDF
116. Lin28aRegulates Germ Cell Pool Size and Fertility
- Author
-
Shinoda, Gen, De Soysa, T. Yvanka, Seligson, Marc T., Yabuuchi, Akiko, Fujiwara, Yuko, Huang, Pei Yi, Hagan, John P., Gregory, Richard I., Moss, Eric G., and Daley, George Q.
- Abstract
Overexpression of LIN28Ais associated with human germ cell tumors and promotes primordial germ cell (PGC) development from embryonic stem cells in vitro and in chimeric mice. Knockdown of Lin28ainhibits PGC development in vitro, but how constitutional Lin28adeficiency affects the mammalian reproductive system in vivo remains unknown. Here, we generated Lin28aknockout (KO) mice and found that Lin28adeficiency compromises the size of the germ cell pool in both males and females by affecting PGC proliferation during embryogenesis. Interestingly however, in Lin28aKO males, the germ cell pool partially recovers during postnatal expansion, while fertility remains impaired in both males and females mated to wild‐type mice. Embryonic overexpression of let‐7, a microRNA negatively regulated by Lin28a, reduces the germ cell pool, corroborating the role of the Lin28a/let‐7axis in regulating the germ lineage. STEMCELLS2013;31:1001–1009
- Published
- 2013
- Full Text
- View/download PDF
117. Inhibition of Histone Deacetylases Alters Allelic Chromatin Conformation at the Imprinted U2af1-rs1Locus in Mouse Embryonic Stem Cells*
- Author
-
Gregory, Richard I., O'Neill, Laura P., Randall, Tamzin E., Fournier, Cecile, Khosla, Sanjeev, Turner, Bryan M., and Feil, Robert
- Abstract
Most loci that are regulated by genomic imprinting have differentially methylated regions (DMRs). Previously, we showed that the DMRs of the mouse Snrpnand U2af1-rs1genes have paternal allele-specific patterns of acetylation on histones H3 and H4. To investigate the maintenance of acetylation at these DMRs, we performed chromatin immunoprecipitation on trichostatin-A (TSA)-treated and control cells. In embryonic stem (ES) cells and fibroblasts, brief (6-h) TSA treatment induces global hyperacetylation of H3 and H4. In ES cells only, TSA led to a selective increase in maternal acetylation at U2af1-rs1, at lysine 5 of H4 and at lysine 14 of H3. TSA treatment of ES cells did not affect DNA methylation or expression of U2af1-rs1, but was sufficient to increase DNase I sensitivity along the maternal allele to a level comparable with that of the paternal allele. In fibroblasts, TSA did not alter U2af1-rs1acetylation, and the parental alleles retained their differential DNase I sensitivity. At Snrpn, no changes in acetylation were observed in the TSA-treated cells. Our data suggest that the mechanisms regulating histone acetylation at DMRs are locus and developmental stage-specific and are distinct from those effecting global levels of acetylation. Furthermore, it seems that the allelic U2af1-rs1acetylation determines DNase I sensitivity/chromatin conformation.
- Published
- 2002
- Full Text
- View/download PDF
118. Nucleotide resolution profiling of m3C RNA modification by HAC-seq.
- Author
-
Cui, Jia, Liu, Qi, Sendinc, Erdem, Shi, Yang, and Gregory, Richard I
- Published
- 2021
- Full Text
- View/download PDF
119. MicroRNA-induced pluripotent stem cells
- Author
-
Chang, Hao-Ming and Gregory, Richard I.
- Subjects
microRNA (miRNA) ,Reprogramming ,Induced Pluripotent stem (iPS) cells ,Embryonic Stem cells - Published
- 2011
- Full Text
- View/download PDF
120. The mRNA m6A reader YTHDF2 suppresses proinflammatory pathways and sustains hematopoietic stem cell function
- Author
-
Mapperley, Christopher, van de Lagemaat, Louie N., Lawson, Hannah, Tavosanis, Andrea, Paris, Jasmin, Campos, Joana, Wotherspoon, David, Durko, Jozef, Sarapuu, Annika, Choe, Junho, Ivanova, Ivayla, Krause, Daniela S., von Kriegsheim, Alex, Much, Christian, Morgan, Marcos, Gregory, Richard I., Mead, Adam J., O’Carroll, Dónal, and Kranc, Kamil R.
- Abstract
The mRNA N6-methyladenosine (m6A) modification has emerged as an essential regulator of normal and malignant hematopoiesis. Inactivation of the m6A mRNA reader YTHDF2, which recognizes m6A-modified transcripts to promote m6A-mRNA degradation, results in hematopoietic stem cell (HSC) expansion and compromises acute myeloid leukemia. Here we investigate the long-term impact of YTHDF2 deletion on HSC maintenance and multilineage hematopoiesis. We demonstrate that Ythdf2-deficient HSCs from young mice fail upon serial transplantation, display increased abundance of multiple m6A-modified inflammation-related transcripts, and chronically activate proinflammatory pathways. Consistent with the detrimental consequences of chronic activation of inflammatory pathways in HSCs, hematopoiesis-specific Ythdf2 deficiency results in a progressive myeloid bias, loss of lymphoid potential, HSC expansion, and failure of aged Ythdf2-deficient HSCs to reconstitute multilineage hematopoiesis. Experimentally induced inflammation increases YTHDF2 expression, and YTHDF2 is required to protect HSCs from this insult. Thus, our study positions YTHDF2 as a repressor of inflammatory pathways in HSCs and highlights the significance of m6A in long-term HSC maintenance.
- Published
- 2021
- Full Text
- View/download PDF
121. MicroRNAs and reprogramming.
- Author
-
Hao-Ming Chang and Gregory, Richard I.
- Subjects
- *
RNA , *SOMATIC cells , *PLURIPOTENT stem cells , *GENES , *STEM cells - Abstract
The article discusses research studies on the use of micro ribonucleic acids (RNA) as an effective method for reprogramming somatic cells to become pluripotent stem cells. The studies identified the micro RNA target genes that can stimulate the formation of the stem cells as well as dissect the pathways and mechanisms underlying the reprogramming of the cells. According to the author, the findings of these studies can be challenged by ethical and political issues related to human embryonic stem cell and regenerative medicine.
- Published
- 2011
- Full Text
- View/download PDF
122. MicroRNA Regulation of Stem Cell Fate.
- Author
-
Qintong Li and Gregory, Richard I.
- Subjects
STEM cells ,RNA ,CELL determination ,GENETIC regulation ,MESODERM ,MUSCLES ,GENETIC research - Abstract
The article discusses a study that examines the microRNA regulation of stem cell fate. The study shows that two serum response factor (SRF)-dependent muscle-specific microRNAs, miR-1 and miR-133, promote mesoderm formation from embryonic stem (ES) cells but have opposing functions during further differentiation into cardiac muscle progenitors. The results show that muscle-specific microRNAs repress nonmuscle genes to direct embryonic stem cell differentiation to mesodem and muscle.
- Published
- 2008
- Full Text
- View/download PDF
123. Analysis of chromatin in limited numbers of cells: a PCR–SSCP based assay of allele-specific nuclease sensitivity.
- Author
-
Gregory, Richard I. and Feil, Robert
- Published
- 1999
- Full Text
- View/download PDF
124. The m6A Methyltransferase METTL3 Promotes Translation in Human Cancer Cells.
- Author
-
Lin, Shuibin, Choe, Junho, Du, Peng, Triboulet, Robinson, and Gregory, Richard I.
- Subjects
- *
LUNG cancer risk factors , *CANCER cells , *METHYLTRANSFERASES , *GENETIC translation , *EPIDERMAL growth factor receptors , *ADENOCARCINOMA , *ONCOGENES - Abstract
Summary METTL3 is an RNA methyltransferase implicated in mRNA biogenesis, decay, and translation control through N 6 -methyladenosine (m 6 A) modification. Here we find that METTL3 promotes translation of certain mRNAs including epidermal growth factor receptor (EGFR) and the Hippo pathway effector TAZ in human cancer cells. In contrast to current models that invoke m 6 A reader proteins downstream of nuclear METTL3, we find METTL3 associates with ribosomes and promotes translation in the cytoplasm. METTL3 depletion inhibits translation, and both wild-type and catalytically inactive METTL3 promote translation when tethered to a reporter mRNA. Mechanistically, METTL3 enhances mRNA translation through an interaction with the translation initiation machinery. METTL3 expression is elevated in lung adenocarcinoma and using both loss- and gain-of-function studies, we find that METTL3 promotes growth, survival, and invasion of human lung cancer cells. Our results uncover an important role of METTL3 in promoting translation of oncogenes in human lung cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
125. EZH2 Oncogenic Activity in Castration-Resistant Prostate Cancer Cells Is Polycomb-lndependent.
- Author
-
Kexin Xu, Wu, Zhenhua Jeremy, Groner, Anna C., Housheng Hansen He, Changmeng Cai, Lis, Rosina T., Xiaoqiu Wu, Stack, Edward C., Loda, Massimo, Tao Liu, Han Xu, Cato, Laura, Thornton, James E., Gregory, Richard I., Morrissey, Colm, Vessella, Robert L., Montironi, Rodolfo, Magi-Galluzzi, Cristina, Kantoff, Philip W., and Balk, Steven P.
- Subjects
- *
GENE enhancers , *PROSTATE cancer , *POLYCOMB group protein genetics , *ONCOGENES , *CARCINOGENESIS , *HISTONE methyltransferases , *PHOSPHORYLATION - Abstract
Epigenetic regulators represent a promising new class of therapeutic targets for cancer. Enhancer of zeste homolog 2 (EZH2), a subunit of Polycomb repressive complex 2 (PRC2), silences gene expression via its histone methyltransferase activity. We found that the oncogenic function of EZH2 in cells of castration-resistant prostate cancer is independent of its role as a transcriptional repressor. Instead, it involves the ability of EZH2 to act as a coactivator for critical transcription factors including the androgen receptor. This functional switch is dependent on phosphorylation of EZH2 and requires an intact methyltransferase domain. Hence, targeting the non-PRC2 function of EZH2 may have therapeutic efficacy for treating metastatic, hormone-refractory prostate cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
126. Mapping multiple RNA modifications simultaneously by proximity barcode sequencing.
- Author
-
Sendinc E, Yu H, Hwang Fu YH, Santos J, Johnson Z, Kirstein JR, Niu J, Chabot MB, Cantu VA, Džakula Ž, Lam Q, Anmangandla A, Burcham TS, Davis EM, Miles ZD, Price AD, Purse BW, Gregory RI, and Stengel G
- Abstract
RNA is subject to a multitude of different chemical modifications that collectively represent the epitranscriptome. Individual RNA modifications including N6-methyladenosine (m
6 A) on mRNA play essential roles in the posttranscriptional control of gene expression. Recent technological advances have enabled the transcriptome-wide mapping of certain RNA modifications, to reveal their broad relevance and characteristic distribution patterns. However, convenient methods that enable the simultaneous mapping of multiple different RNA marks within the same sample are generally lacking. Here we present EpiPlex RNA modification profiling, a bead-based proximity barcoding assay with sequencing readout that expands the scope of molecular recognition-based RNA modification detection to multiple targets, while providing relative quantification and enabling low RNA input. Measuring signal intensity against spike-in controls provides relative quantification, indicative of the RNA mod abundance at each locus. We report on changes in the modification status of HEK293T cells upon treatment with pharmacological inhibitors separately targeting METTL3, the dominant m6 A writer enzyme, and the EIF4A3 component of the exon junction complex (EJC). The treatments resulted in decreased or increased m6 A levels, respectively, without effect on inosine levels. Inhibiting the helicase activity of EIF4A3 and EIF4A3 knockdown both cause a significant increase of m6 A sites near exon junctions, consistent with the previously reported role of EIF4A3 in shaping the m6 A landscape. Thus, EpiPlex offers a reliable and convenient method for simultaneous mapping of multiple RNA modifications to facilitate epitranscriptome studies.- Published
- 2024
- Full Text
- View/download PDF
127. METTL1-mediated m 7 G modification of Arg-TCT tRNA drives oncogenic transformation.
- Author
-
Orellana EA, Liu Q, Yankova E, Pirouz M, De Braekeleer E, Zhang W, Lim J, Aspris D, Sendinc E, Garyfallos DA, Gu M, Ali R, Gutierrez A, Mikutis S, Bernardes GJL, Fischer ES, Bradley A, Vassiliou GS, Slack FJ, Tzelepis K, and Gregory RI
- Subjects
- Guanosine analogs & derivatives, Guanosine genetics, Humans, Methylation, Neoplasms pathology, Oncogenes genetics, RNA Processing, Post-Transcriptional genetics, RNA, Messenger genetics, RNA, Transfer genetics, Carcinogenesis genetics, Methyltransferases genetics, Neoplasms genetics, tRNA Methyltransferases genetics
- Abstract
The emerging "epitranscriptomics" field is providing insights into the biological and pathological roles of different RNA modifications. The RNA methyltransferase METTL1 catalyzes N7-methylguanosine (m
7 G) modification of tRNAs. Here we find METTL1 is frequently amplified and overexpressed in cancers and is associated with poor patient survival. METTL1 depletion causes decreased abundance of m7 G-modified tRNAs and altered cell cycle and inhibits oncogenicity. Conversely, METTL1 overexpression induces oncogenic cell transformation and cancer. Mechanistically, we find increased abundance of m7 G-modified tRNAs, in particular Arg-TCT-4-1, and increased translation of mRNAs, including cell cycle regulators that are enriched in the corresponding AGA codon. Accordingly, Arg-TCT expression is elevated in many tumor types and is associated with patient survival, and strikingly, overexpression of this individual tRNA induces oncogenic transformation. Thus, METTL1-mediated tRNA modification drives oncogenic transformation through a remodeling of the mRNA "translatome" to increase expression of growth-promoting proteins and represents a promising anti-cancer target., Competing Interests: Declaration of interests R.I.G. and F.J.S. are co-founders and scientific advisory board members of 28-7 Therapeutics. R.I.G. is a co-founder and scientific advisory board member of Theon Therapeutics. E.Y. is co-funded by STORM Therapeutics., (Copyright © 2021 Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
128. Genetic drivers of m 6 A methylation in human brain, lung, heart and muscle.
- Author
-
Xiong X, Hou L, Park YP, Molinie B, Gregory RI, and Kellis M
- Subjects
- Adenosine genetics, Adenosine metabolism, Genome-Wide Association Study, Heart physiology, Humans, Methylation, Organ Specificity, Polymorphism, Single Nucleotide, RNA Processing, Post-Transcriptional, RNA-Binding Proteins genetics, Reproducibility of Results, Adenosine analogs & derivatives, Brain physiology, Lung physiology, Muscle, Skeletal physiology, Quantitative Trait Loci
- Abstract
The most prevalent post-transcriptional mRNA modification, N
6 -methyladenosine (m6 A), plays diverse RNA-regulatory roles, but its genetic control in human tissues remains uncharted. Here we report 129 transcriptome-wide m6 A profiles, covering 91 individuals and 4 tissues (brain, lung, muscle and heart) from GTEx/eGTEx. We integrate these with interindividual genetic and expression variation, revealing 8,843 tissue-specific and 469 tissue-shared m6 A quantitative trait loci (QTLs), which are modestly enriched in, but mostly orthogonal to, expression QTLs. We integrate m6 A QTLs with disease genetics, identifying 184 GWAS-colocalized m6 A QTL, including brain m6 A QTLs underlying neuroticism, depression, schizophrenia and anxiety; lung m6 A QTLs underlying expiratory flow and asthma; and muscle/heart m6 A QTLs underlying coronary artery disease. Last, we predict novel m6 A regulators that show preferential binding in m6 A QTLs, protein interactions with known m6 A regulators and expression correlation with the m6 A levels of their targets. Our results provide important insights and resources for understanding both cis and trans regulation of epitranscriptomic modifications, their interindividual variation and their roles in human disease., (© 2021. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2021
- Full Text
- View/download PDF
129. Nucleotide resolution profiling of m3C RNA modification by HAC-seq.
- Author
-
Cui J, Liu Q, Sendinc E, Shi Y, and Gregory RI
- Subjects
- Aniline Compounds chemistry, Cytidine analysis, Cytidine metabolism, Humans, Hydrazines chemistry, MCF-7 Cells, RNA, Transfer metabolism, Transcriptome, Cytidine analogs & derivatives, RNA, Transfer chemistry, Sequence Analysis, RNA methods
- Abstract
Cellular RNAs are subject to a myriad of different chemical modifications that play important roles in controlling RNA expression and function. Dysregulation of certain RNA modifications, the so-called 'epitranscriptome', contributes to human disease. One limitation in studying the functional, physiological, and pathological roles of the epitranscriptome is the availability of methods for the precise mapping of individual RNA modifications throughout the transcriptome. 3-Methylcytidine (m3C) modification of certain tRNAs is well established and was also recently detected in mRNA. However, methods for the specific mapping of m3C throughout the transcriptome are lacking. Here, we developed a m3C-specific technique, Hydrazine-Aniline Cleavage sequencing (HAC-seq), to profile the m3C methylome at single-nucleotide resolution. We applied HAC-seq to analyze ribosomal RNA (rRNA)-depleted total RNAs in human cells. We found that tRNAs are the predominant m3C-modified RNA species, with 17 m3C modification sites on 11 cytoplasmic and 2 mitochondrial tRNA isoacceptors in MCF7 cells. We found no evidence for m3C-modification of mRNA or other non-coding RNAs at comparable levels to tRNAs in these cells. HAC-seq provides a novel method for the unbiased, transcriptome-wide identification of m3C RNA modification at single-nucleotide resolution, and could be widely applied to reveal the m3C methylome in different cells and tissues., (© The Author(s) 2020. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2021
- Full Text
- View/download PDF
130. An Informatics Pipeline for Profiling and Annotating RNA Modifications.
- Author
-
Liu Q, Lang X, and Gregory RI
- Subjects
- Cell Line, Tumor, Hep G2 Cells, Humans, Sequence Analysis, RNA methods, Informatics methods, RNA genetics, RNA Processing, Post-Transcriptional genetics, Transcriptome genetics
- Abstract
While over 150 distinct types of chemical modifications are known to occur on various cellular RNAs and can be dynamically controlled, the function of most of these modifications remains poorly defined. Collectively, these RNA modifications have been recently termed the "epitranscriptome". Identification and annotation of individual RNA modifications throughout the transcriptome are key for studying the role of the epitranscriptome in the regulation of gene expression and for elucidating the functional relevance of particular RNA modifications in diverse physiological and disease processes. In this protocol, we demonstrate how to identify and annotate RNA modifications based on the informatic analysis of methylated RNA immunoprecipitation and sequencing (MeRIP-seq) data, using RNAmod, a convenient one-stop online interactive platform for the annotation, analysis, and visualization of mRNA modifications.
- Published
- 2021
- Full Text
- View/download PDF
131. Nucleotide resolution profiling of m 7 G tRNA modification by TRAC-Seq.
- Author
-
Lin S, Liu Q, Jiang YZ, and Gregory RI
- Subjects
- Animals, Genomics, Guanosine analysis, Guanosine genetics, High-Throughput Nucleotide Sequencing methods, Methylation, Mice, RNA, Transfer genetics, Software, Transcriptome, Guanosine analogs & derivatives, RNA, Transfer chemistry, Sequence Analysis, RNA methods
- Abstract
Precise identification of sites of RNA modification is key to studying the functional role of such modifications in the regulation of gene expression and for elucidating relevance to diverse physiological processes. tRNA reduction and cleavage sequencing (TRAC-Seq) is a chemically based approach for the unbiased global mapping of 7-methylguansine (m
7 G) modification of tRNAs at single-nucleotide resolution throughout the tRNA transcriptome. m7 G TRAC-Seq involves the treatment of size-selected (<200 nt) RNAs with the demethylase AlkB to remove major tRNA modifications, followed by sodium borohydride (NaBH4 ) reduction of m7 G sites and subsequent aniline-mediated cleavage of the RNA chain at the resulting abasic sites. The cleaved sites are subsequently ligated with adaptors for the construction of libraries for high-throughput sequencing. The m7 G modification sites are identified using a bioinformatic pipeline that calculates the cleavage scores at individual sites on all tRNAs. Unlike antibody-based methods, such as methylated RNA immunoprecipitation and sequencing (meRIP-Seq) for enrichment of methylated RNA sequences, chemically based approaches, including TRAC-Seq, can provide nucleotide-level resolution of modification sites. Compared to the related method AlkAniline-Seq (alkaline hydrolysis and aniline cleavage sequencing), TRAC-Seq incorporates small RNA selection, AlkB demethylation, and sodium borohydride reduction steps to achieve specific and efficient single-nucleotide resolution profiling of m7 G sites in tRNAs. The m7 G TRAC-Seq protocol could be adapted to chemical cleavage-mediated detection of other RNA modifications. The protocol can be completed within ~9 d for four biological replicates of input and treated samples.- Published
- 2019
- Full Text
- View/download PDF
132. Targeting the RNA m 6 A Reader YTHDF2 Selectively Compromises Cancer Stem Cells in Acute Myeloid Leukemia.
- Author
-
Paris J, Morgan M, Campos J, Spencer GJ, Shmakova A, Ivanova I, Mapperley C, Lawson H, Wotherspoon DA, Sepulveda C, Vukovic M, Allen L, Sarapuu A, Tavosanis A, Guitart AV, Villacreces A, Much C, Choe J, Azar A, van de Lagemaat LN, Vernimmen D, Nehme A, Mazurier F, Somervaille TCP, Gregory RI, O'Carroll D, and Kranc KR
- Subjects
- Animals, Cell Self Renewal, Hematopoiesis, Hematopoietic Stem Cells, Humans, Leukemia, Myeloid, Acute genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, RNA, Small Interfering genetics, RNA-Binding Proteins genetics, THP-1 Cells, Leukemia, Myeloid, Acute therapy, Neoplastic Stem Cells physiology, RNA-Binding Proteins metabolism
- Abstract
Acute myeloid leukemia (AML) is an aggressive clonal disorder of hematopoietic stem cells (HSCs) and primitive progenitors that blocks their myeloid differentiation, generating self-renewing leukemic stem cells (LSCs). Here, we show that the mRNA m
6 A reader YTHDF2 is overexpressed in a broad spectrum of human AML and is required for disease initiation as well as propagation in mouse and human AML. YTHDF2 decreases the half-life of diverse m6 A transcripts that contribute to the overall integrity of LSC function, including the tumor necrosis factor receptor Tnfrsf2, whose upregulation in Ythdf2-deficient LSCs primes cells for apoptosis. Intriguingly, YTHDF2 is not essential for normal HSC function, with YTHDF2 deficiency actually enhancing HSC activity. Thus, we identify YTHDF2 as a unique therapeutic target whose inhibition selectively targets LSCs while promoting HSC expansion., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
133. DROSHA Knockout Leads to Enhancement of Viral Titers for Vectors Encoding miRNA-Adapted shRNAs.
- Author
-
Park HH, Triboulet R, Bentler M, Guda S, Du P, Xu H, Gregory RI, Brendel C, and Williams DA
- Abstract
RNAi-based gene therapy using miRNA-adapted short hairpin RNAs (shRNA
miR ) is a powerful approach to modulate gene expression. However, we have observed low viral titers with shRNAmiR -containing recombinant vectors and hypothesized that this could be due to cleavage of viral genomic RNA by the endogenous microprocessor complex during virus assembly. To test this hypothesis, we targeted DROSHA, the core component of the microprocessor complex, and successfully generated monoallelic and biallelic DROSHA knockout (KO) HEK293T cells for vector production. DROSHA KO was verified by polymerase chain reaction (PCR) and western blot analysis. We produced lentiviral vectors containing Venus with or without shRNA hairpins and generated virus supernatants using DROSHA KO packaging cells. We observed an increase in the fluorescence intensity of hairpin-containing Venus transcripts in DROSHA KO producer cells consistent with reduced microprocessor cleavage of encoded mRNA transcripts, and recovery in the viral titer of hairpin-containing vectors compared with non-hairpin-containing constructs. We confirmed the absence of significant shRNAmiR processing by northern blot analysis and showed that this correlated with an increase in the amount of full-length vector genomic RNA. These findings may have important implications in future production of viral shRNAmiR -containing vectors for RNAi-based therapy., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
134. Mettl1/Wdr4-Mediated m 7 G tRNA Methylome Is Required for Normal mRNA Translation and Embryonic Stem Cell Self-Renewal and Differentiation.
- Author
-
Lin S, Liu Q, Lelyveld VS, Choe J, Szostak JW, and Gregory RI
- Subjects
- Animals, Base Sequence, Cell Differentiation genetics, Cell Line, Cell Self Renewal genetics, Embryonic Stem Cells, GTP-Binding Proteins metabolism, Guanosine genetics, Guanosine metabolism, Humans, Methylation, Methyltransferases metabolism, Mice, Mouse Embryonic Stem Cells, RNA Processing, Post-Transcriptional, RNA, Transfer metabolism, GTP-Binding Proteins genetics, Guanosine analogs & derivatives, Methyltransferases genetics, RNA, Transfer genetics
- Abstract
tRNAs are subject to numerous modifications, including methylation. Mutations in the human N
7 -methylguanosine (m7 G) methyltransferase complex METTL1/WDR4 cause primordial dwarfism and brain malformation, yet the molecular and cellular function in mammals is not well understood. We developed m7 G methylated tRNA immunoprecipitation sequencing (MeRIP-seq) and tRNA reduction and cleavage sequencing (TRAC-seq) to reveal the m7 G tRNA methylome in mouse embryonic stem cells (mESCs). A subset of 22 tRNAs is modified at a "RAGGU" motif within the variable loop. We observe increased ribosome occupancy at the corresponding codons in Mettl1 knockout mESCs, implying widespread effects on tRNA function, ribosome pausing, and mRNA translation. Translation of cell cycle genes and those associated with brain abnormalities is particularly affected. Mettl1 or Wdr4 knockout mESCs display defective self-renewal and neural differentiation. Our study uncovers the complexity of the mammalian m7 G tRNA methylome and highlights its essential role in ESCs with links to human disease., (Copyright © 2018 Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
135. Autoregulatory mechanisms controlling the microprocessor.
- Author
-
Triboulet R and Gregory RI
- Subjects
- Humans, MicroRNAs genetics, Microcomputers, RNA, Double-Stranded, RNA Processing, Post-Transcriptional, RNA-Binding Proteins metabolism
- Abstract
The Microprocessor, comprising the ribonuclease Drosha and its essential cofactor, the double-stranded RNA-binding protein, DGCR8, is essential for the first step of the miRNA biogenesis pathway. It specifically cleaves double-stranded RNA within stem-loop structures of primary miRNA transcripts (pri-miRNAs) to generate precursor (pre-miRNA) intermediates. Pre-miRNAs are subsequently processed by Dicer to their mature ∼22 nt form. Thus, Microprocessor is essential for miRNA maturation, and pri-miRNA cleavage by this complex defines one end of the mature miRNA. Moreover, it is emerging that dysregulation of the Microprocessor is associated with various human diseases. It is therefore important to understand the mechanisms by which the expression of the subunits of the Microprocessor is regulated. Recent findings have uncovered a post-transcriptional mechanism that maintains the integrity of the Microprocessor. These studies revealed that the Microprocessor is involved in the processing of the messenger RNA (mRNA) that encodes DGCR8. This regulatory feedback loop, along with the reported role played by DGCR8 in the stabilization of Drosha protein, is part of a newly identified regulatory mechanism controlling Microprocessor activity.
- Published
- 2011
- Full Text
- View/download PDF
136. Autoregulatory mechanisms controlling the Microprocessor.
- Author
-
Triboulet R and Gregory RI
- Subjects
- Animals, Homeostasis, Humans, Proteins physiology, RNA-Binding Proteins, Ribonuclease III physiology, Gene Expression Regulation, MicroRNAs physiology, Proteins genetics, Ribonuclease III genetics
- Abstract
The Microprocessor, comprising the ribonuclease Drosha and its essential cofactor, the double-stranded RNA-binding protein, DGCR8, is essential for the first step of the miRNA biogenesis pathway. It specifically cleaves double-stranded RNA within stem-loop structures of primary miRNA transcripts (pri-miRNAs) to generate precursor (pre-miRNA) intermediates. Pre-miRNAs are subsequently processed by Dicer to their mature 22 nt form. Thus, Microprocessor is essential for miRNA maturation, and pri-miRNA cleavage by this complex defines one end of the mature miRNA. Moreover, it is emerging that dysregulation of the Microprocessor is associated with various human diseases. It is therefore important to understand the mechanisms by which the expression of the subunits of the Microprocessor is regulated. Recent findings have uncovered a post-transcriptional mechanism that maintains the integrity of the Microprocessor. These studies revealed that the Microprocessor is involved in the processing of the messenger RNA (mRNA) that encodes DGCR8. This regulatory feedback loop, along with the reported role played by DGCR8 in the stabilization of Drosha protein, is part ofa newly identified regulatory mechanism controlling Microprocessor activity.
- Published
- 2010
137. MicroRNA biogenesis: isolation and characterization of the microprocessor complex.
- Author
-
Gregory RI, Chendrimada TP, and Shiekhattar R
- Subjects
- Cell Line, Humans, In Vitro Techniques, MicroRNAs genetics, RNA Interference, Ribonuclease III genetics, Ribonuclease III metabolism, MicroRNAs biosynthesis, RNA Processing, Post-Transcriptional
- Abstract
The recently discovered microRNAs (miRNAs) are a large family of small regulatory RNAs that have been implicated in controlling diverse pathways in a variety of organisms (1, 2). For posttranscriptional gene silencing, one strand of the miRNA is used to guide components of the RNA interference machinery, including Argonaute 2, to messenger RNAs (mRNAs) with complementary sequences (3, 4). Thus, targeted mRNAs are either cleaved by the endonuclease Argonaute 2 (5, 6), or protein synthesis is blocked by an as yet uncharacterized mechanism (7, 8). Genes encoding miRNAs are transcribed as long primary miRNAs (pri-miRNAs) that are sequentially processed by components of the nucleus and cytoplasm to yield a mature, approx 22-nucleotide (nt)-long miRNA (9). Two members of the ribonuclease (RNase) III endonuclease protein family, Drosha and Dicer, have been implicated in this two-step processing (10-13). To further our understanding of miRNA biogenesis and function it will be essential to identify the protein complexes involved. We were interested in defining the proteins required for the initial nuclear processing of pri-miRNAs to the approx 60- to 70-nt stem-loop intermediates known as precursor miRNAs (pre-miRNAs) (9, 10). This led to our identification of a protein complex we termed Microprocessor, which is necessary and sufficient for processing pri-miRNA to premiRNAs (14). The Microprocessor complex comprises Drosha and the double-stranded RNAbinding protein DiGeorge syndrome critical region 8 gene (DGCR8), which is deleted in DiGeorge syndrome (15, 16). In this chapter, we detail the methods used for the biochemical isolation and identification of the Microprocessor complex from human cells. We include a protocol for the in vitro analysis of pri-miRNA processing activity of the purified Microprocessor complex.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.