Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Meg Byron is active.

Publication


Featured researches published by Meg Byron.


Science | 2013

A Long Noncoding RNA Mediates Both Activation and Repression of Immune Response Genes

Susan Carpenter; Daniel Aiello; Maninjay K. Atianand; Emiliano P. Ricci; Pallavi Gandhi; Lisa L. Hall; Meg Byron; Brian G. Monks; Meabh Henry-Bezy; Jeanne B. Lawrence; Luke A. J. O'Neill; Melissa J. Moore; Daniel R. Caffrey; Katherine A. Fitzgerald

A New Linc in Innate Immunity Long noncoding RNAs (lncRNAs) have recently emerged as important regulators of gene expression in a wide variety of biological processes, although specific roles for these molecules in the immune system have not been described. Carpenter et al. (p. 789, published online 1 August) now define the function of one such lncRNA in the immune system, lincRNA-Cox2. Whole-transcriptome profiling revealed that lincRNA-Cox2 was induced in mouse macrophages in response to activation of Toll-like receptors—molecules that detect microbes and alert the immune system to respond. LincRNA-Cox2 both positively and negatively regulated the expression of distinct groups of inflammatory genes. Negative regulation of gene expression was mediated by lincRNA-Cox interaction with heterogeneous nuclear ribonucleoprotein A/B and A2/B1. In mice, a broadly acting RNA, lincRNA-Cox2, regulates the circuit that controls the inflammatory response. An inducible program of inflammatory gene expression is central to antimicrobial defenses. This response is controlled by a collaboration involving signal-dependent activation of transcription factors, transcriptional co-regulators, and chromatin-modifying factors. We have identified a long noncoding RNA (lncRNA) that acts as a key regulator of this inflammatory response. Pattern recognition receptors such as the Toll-like receptors induce the expression of numerous lncRNAs. One of these, lincRNA-Cox2, mediates both the activation and repression of distinct classes of immune genes. Transcriptional repression of target genes is dependent on interactions of lincRNA-Cox2 with heterogeneous nuclear ribonucleoprotein A/B and A2/B1. Collectively, these studies unveil a central role of lincRNA-Cox2 as a broad-acting regulatory component of the circuit that controls the inflammatory response.


Nature Structural & Molecular Biology | 2011

The three-dimensional folding of the α-globin gene domain reveals formation of chromatin globules

Davide Baù; Amartya Sanyal; Bryan R. Lajoie; Emidio Capriotti; Meg Byron; Jeanne B. Lawrence; Job Dekker; Marc A. Marti-Renom

We developed a general approach that combines chromosome conformation capture carbon copy (5C) with the Integrated Modeling Platform (IMP) to generate high-resolution three-dimensional models of chromatin at the megabase scale. We applied this approach to the ENm008 domain on human chromosome 16, containing the α-globin locus, which is expressed in K562 cells and silenced in lymphoblastoid cells (GM12878). The models accurately reproduce the known looping interactions between the α-globin genes and their distal regulatory elements. Further, we find using our approach that the domain folds into a single globular conformation in GM12878 cells, whereas two globules are formed in K562 cells. The central cores of these globules are enriched for transcribed genes, whereas nontranscribed chromatin is more peripheral. We propose that globule formation represents a higher-order folding state related to clustering of transcribed genes around shared transcription machineries, as previously observed by microscopy.


Proceedings of the National Academy of Sciences of the United States of America | 2006

The X chromosome is organized into a gene-rich outer rim and an internal core containing silenced nongenic sequences

Christine Moulton Clemson; Lisa L. Hall; Meg Byron; John Mcneil; Jeanne B. Lawrence

We investigated whether genes escape X chromosome inactivation by positioning outside of the territory defined by XIST RNA. Results reveal an unanticipated higher order organization of genes and noncoding sequences. All 15 X-linked genes, regardless of activity, position on the border of the XIST RNA territory, which resides outside of the DAPI-dense Barr body. Although more strictly delineated on the inactive X chromosome (Xi), all genes localized predominantly to the outer rim of the Xi and active X chromosome. This outer rim is decorated only by X chromosome DNA paints and is excluded from both the XIST RNA and dense DAPI staining. The only DNA found well within the Barr body and XIST RNA territory was centromeric and Cot-1 DNA; hence, the core of the X chromosome essentially excludes genes and is composed primarily of noncoding repeat-rich DNA. Moreover, we show that this core of repetitive sequences is expressed throughout the nucleus yet is silenced throughout Xi, providing direct evidence for chromosome-wide regulation of “junk” DNA transcription. Collective results suggest that the Barr body, long presumed to be the physical manifestation of silenced genes, is in fact composed of a core of silenced noncoding DNA. Instead of acting at a local gene level, XIST RNA appears to interact with and silence core architectural elements to effectively condense and shut down the Xi.


Nature | 2013

Translating dosage compensation to trisomy 21

Jun Jiang; Yuanchun Jing; Gregory J. Cost; Jen-Chieh Chiang; Heather J. Kolpa; Allison M. Cotton; Dawn M. Carone; Benjamin R. Carone; David A. Shivak; Dmitry Guschin; Jocelynn R. Pearl; Edward J. Rebar; Meg Byron; Philip D. Gregory; Carolyn J. Brown; Fyodor D. Urnov; Lisa L. Hall; Jeanne B. Lawrence

Down’s syndrome is a common disorder with enormous medical and social costs, caused by trisomy for chromosome 21. We tested the concept that gene imbalance across an extra chromosome can be de facto corrected by manipulating a single gene, XIST (the X-inactivation gene). Using genome editing with zinc finger nucleases, we inserted a large, inducible XIST transgene into the DYRK1A locus on chromosome 21, in Down’s syndrome pluripotent stem cells. The XIST non-coding RNA coats chromosome 21 and triggers stable heterochromatin modifications, chromosome-wide transcriptional silencing and DNA methylation to form a ‘chromosome 21 Barr body’. This provides a model to study human chromosome inactivation and creates a system to investigate genomic expression changes and cellular pathologies of trisomy 21, free from genetic and epigenetic noise. Notably, deficits in proliferation and neural rosette formation are rapidly reversed upon silencing one chromosome 21. Successful trisomy silencing in vitro also surmounts the major first step towards potential development of ‘chromosome therapy’.


Journal of Cell Biology | 2003

Clustering of multiple specific genes and gene-rich R-bands around SC-35 domains evidence for local euchromatic neighborhoods

Lindsay S. Shopland; Meg Byron; John Mcneil; Jeanne B. Lawrence

Typically, eukaryotic nuclei contain 10–30 prominent domains (referred to here as SC-35 domains) that are concentrated in mRNA metabolic factors. Here, we show that multiple specific genes cluster around a common SC-35 domain, which contains multiple mRNAs. Nonsyntenic genes are capable of associating with a common domain, but domain “choice” appears random, even for two coordinately expressed genes. Active genes widely separated on different chromosome arms associate with the same domain frequently, assorting randomly into the 3–4 subregions of the chromosome periphery that contact a domain. Most importantly, visualization of six individual chromosome bands showed that large genomic segments (∼5 Mb) have striking differences in organization relative to domains. Certain bands showed extensive contact, often aligning with or encircling an SC-35 domain, whereas others did not. All three gene-rich reverse bands showed this more than the gene-poor Giemsa dark bands, and morphometric analyses demonstrated statistically significant differences. Similarly, late-replicating DNA generally avoids SC-35 domains. These findings suggest a functional rationale for gene clustering in chromosomal bands, which relates to nuclear clustering of genes with SC-35 domains. Rather than random reservoirs of splicing factors, or factors accumulated on an individual highly active gene, we propose a model of SC-35 domains as functional centers for a multitude of clustered genes, forming local euchromatic “neighborhoods.”


Proceedings of the National Academy of Sciences of the United States of America | 2002

An ectopic human XIST gene can induce chromosome inactivation in postdifferentiation human HT-1080 cells

Lisa L. Hall; Meg Byron; Kosuke Sakai; Laura Carrel; Huntington F. Willard; Jeanne B. Lawrence

It has been believed that XIST RNA requires a discrete window in early development to initiate the series of chromatin-remodeling events that form the heterochromatic inactive X chromosome. Here we investigate four adult male HT-1080 fibrosarcoma cell lines expressing ectopic human XIST and demonstrate that these postdifferentiation cells can undergo chromosomal inactivation outside of any normal developmental context. All four clonal lines inactivated the transgene-containing autosome to varying degrees and with variable stability. One clone in particular consistently localized the ectopic XIST RNA to a discrete chromosome territory that exhibited striking hallmarks of inactivation, including long-range transcriptional inactivation. Results suggest that some postdifferentiation cell lines are capable of de novo chromosomal inactivation; however, long-term retention of autosomal inactivation was less common, which suggests that autosomal inactivation may confer a selective disadvantage. These results have fundamental significance for understanding genomic programming in early development.


Nature | 2010

Maternal Rnf12/RLIM is required for imprinted X-chromosome inactivation in mice

JongDae Shin; Michael Bossenz; Young Chung; Hong Ma; Meg Byron; Naoko Taniguchi-Ishigaki; Xiaochun Zhu; Baowei Jiao; Lisa L. Hall; Michael R. Green; Stephen N. Jones; Irm Hermans-Borgmeyer; Jeanne B. Lawrence; Ingolf Bach

Two forms of X-chromosome inactivation (XCI) ensure the selective silencing of female sex chromosomes during mouse embryogenesis. Imprinted XCI begins with the detection of Xist RNA expression on the paternal X chromosome (Xp) at about the four-cell stage of embryonic development. In the embryonic tissues of the inner cell mass, a random form of XCI occurs in blastocysts that inactivates either Xp or the maternal X chromosome (Xm). Both forms of XCI require the non-coding Xist RNA that coats the inactive X chromosome from which it is expressed. Xist has crucial functions in the silencing of X-linked genes, including Rnf12 (refs 3, 4) encoding the ubiquitin ligase RLIM (RING finger LIM-domain-interacting protein). Here we show, by targeting a conditional knockout of Rnf12 to oocytes where RLIM accumulates to high levels, that the maternal transmission of the mutant X chromosome (Δm) leads to lethality in female embryos as a result of defective imprinted XCI. We provide evidence that in Δm female embryos the initial formation of Xist clouds and Xp silencing are inhibited. In contrast, embryonic stem cells lacking RLIM are able to form Xist clouds and silence at least some X-linked genes during random XCI. These results assign crucial functions to the maternal deposit of Rnf12/RLIM for the initiation of imprinted XCI.


Journal of Cellular Physiology | 2008

X‐inactivation reveals epigenetic anomalies in most hESC but identifies sublines that initiate as expected

Lisa L. Hall; Meg Byron; John T. Butler; Klaus A. Becker; Angel Nelson; Michal Amit; Joseph Itskovitz-Eldor; Janet L. Stein; Gary S. Stein; Carol B. Ware; Jeanne B. Lawrence

The clinical and research value of human embryonic stem cells (hESC) depends upon maintaining their epigenetically naïve, fully undifferentiated state. Inactivation of one X chromosome in each cell of mammalian female embryos is a paradigm for one of the earliest steps in cell specialization through formation of facultative heterochromatin. Mouse ES cells are derived from the inner cell mass (ICM) of blastocyst stage embryos prior to X‐inactivation, and cultured murine ES cells initiate this process only upon differentiation. Less is known about human X‐inactivation during early development. To identify a human ES cell model for X‐inactivation and study differences in the epigenetic state of hESC lines, we investigated X‐inactivation in all growth competent, karyotypically normal, NIH approved, female hESC lines and several sublines. In the vast majority of undifferentiated cultures of nine lines examined, essentially all cells exhibit hallmarks of X‐inactivation. However, subcultures of any hESC line can vary in X‐inactivation status, comprising distinct sublines. Importantly, we identified rare sublines that have not yet inactivated Xi and retain competence to undergo X‐inactivation upon differentiation. Other sublines exhibit defects in counting or maintenance of XIST expression on Xi. The few hESC sublines identified that have not yet inactivated Xi may reflect the earlier epigenetic state of the human ICM and represent the most promising source of NIH hESC for study of human X‐inactivation. The many epigenetic anomalies seen indicate that maintenance of fully unspecialized cells, which have not formed Xi facultative heterochromatin, is a delicate epigenetic balance difficult to maintain in culture. J. Cell. Physiol. 216: 445–452, 2008.


Chromosoma | 2004

Ubiquitinated proteins including uH2A on the human and mouse inactive X chromosome: enrichment in gene rich bands

Kelly P. Smith; Meg Byron; Christine Moulton Clemson; Jeanne B. Lawrence

The inactive X chromosome (Xi) forms a heterochromatic structure in the nucleus that is known to have several modifications to specific histones involving acetylation or methylation. Using three different antibodies in four different cell lines, we demonstrate that the Xi in human and mouse cells is highly enriched in ubiquitinated protein(s), much of which is polyubiquitinated. This ubiquitination appears specific for the Xi as it was not observed for centromeres or other regions of heterochromatin. Results using an antibody specific to ubiquitinated H2A provide a clear link between H2A ubiquitination and gene repression, as visualized across an entire inactive chromosome. Interestingly, the ubiquitination of the chromosome persists into mitosis and can be seen in a reproducible banded pattern. This pattern matches that of Xist RNA which forms bands as it detaches from the mitotic X chromosome. Both ubiquitination and Xist RNA appear enriched in gene dense regions and depleted in gene poor bands, but do not correlate with L1 LINE elements which have been suggested as key to X-inactivation. These results provide evidence that ubiquitination along with Xist RNA plays an important role in the formation of facultative heterochromatin during X-inactivation.


Journal of Cell Biology | 2009

AURKB-mediated effects on chromatin regulate binding versus release of XIST RNA to the inactive chromosome

Lisa L. Hall; Meg Byron; Gayle Jeannette Pageau; Jeanne B. Lawrence

How XIST RNA strictly localizes across the inactive X chromosome is unknown; however, prophase release of human XIST RNA provides a clue. Tests of inhibitors that mimic mitotic chromatin modifications implicated an indirect role of PP1 (protein phosphatase 1), potentially via its interphase repression of Aurora B kinase (AURKB), which phosphorylates H3 and chromosomal proteins at prophase. RNA interference to AURKB causes mitotic retention of XIST RNA, unlike other mitotic or broad kinase inhibitors. Thus, AURKB plays an unexpected role in regulating RNA binding to heterochromatin, independent of mechanics of mitosis. H3 phosphorylation (H3ph) was shown to precede XIST RNA release, whereas results exclude H1ph involvement. Of numerous Xi chromatin (chromosomal protein) hallmarks, ubiquitination closely follows XIST RNA retention or release. Surprisingly, H3S10ph staining (but not H3S28ph) is excluded from Xi and is potentially linked to ubiquitination. Results suggest a model of multiple distinct anchor points for XIST RNA. This study advances understanding of RNA chromosome binding and the roles of AURKB and demonstrates a novel approach to manipulate and study XIST RNA.

Collaboration


Dive into the Meg Byron's collaboration.

Top Co-Authors

Avatar

Jeanne B. Lawrence

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Lisa L. Hall

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Ingolf Bach

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

JongDae Shin

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Dawn M. Carone

University of Connecticut

View shared research outputs
Top Co-Authors

Avatar

Kelly P. Smith

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Christine Moulton Clemson

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Lindsay S. Shopland

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Stephen N. Jones

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Xiaochun Zhu

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge