Network


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

Hotspot


Dive into the research topics where Catherine Rhee is active.

Publication


Featured researches published by Catherine Rhee.


G3: Genes, Genomes, Genetics | 2013

Functional Analyses of Trichoderma reesei LAE1 Reveal Conserved and Contrasting Roles of This Regulator

Razieh Karimi-Aghcheh; Jin Woo Bok; Pallavi A. Phatale; Kristina M. Smith; Scott E. Baker; Alexander Lichius; Markus Omann; Susanne Zeilinger; Bernhard Seiboth; Catherine Rhee; Nancy P. Keller; Michael Freitag; Christian P. Kubicek

The putative methyltransferase LaeA is a global regulator that affects the expression of multiple secondary metabolite gene clusters in several fungi, and it can modify heterochromatin structure in Aspergillus nidulans. We have recently shown that the LaeA ortholog of Trichoderma reesei (LAE1), a fungus that is an industrial producer of cellulase and hemicellulase enzymes, regulates the expression of cellulases and polysaccharide hydrolases. To learn more about the function of LAE1 in T. reesei, we assessed the effect of deletion and overexpression of lae1 on genome-wide gene expression. We found that in addition to positively regulating 7 of 17 polyketide or nonribosomal peptide synthases, genes encoding ankyrin-proteins, iron uptake, heterokaryon incompatibility proteins, PTH11-receptors, and oxidases/monoxygenases are major gene categories also regulated by LAE1. chromatin immunoprecipitation sequencing with antibodies against histone modifications known to be associated with transcriptionally active (H3K4me2 and -me3) or silent (H3K9me3) chromatin detected 4089 genes bearing one or more of these methylation marks, of which 75 exhibited a correlation between either H3K4me2 or H3K4me3 and regulation by LAE1. Transformation of a laeA-null mutant of A. nidulans with the T. reesei lae1 gene did not rescue sterigmatocystin formation and further impaired sexual development. LAE1 did not interact with A. nidulans VeA in yeast two-hybrid assays, whereas it interacted with the T. reesei VeA ortholog, VEL1. LAE1 was shown to be required for the expression of vel1, whereas the orthologs of velB and VosA are unaffected by lae1 deletion. Our data show that the biological roles of A. nidulans LaeA and T. reesei LAE1 are much less conserved than hitherto thought. In T. reesei, LAE1 appears predominantly to regulate genes increasing relative fitness in its environment.


Stem cell reports | 2014

Bright/Arid3A Acts as a Barrier to Somatic Cell Reprogramming through Direct Regulation of Oct4, Sox2, and Nanog

Melissa Popowski; Troy D. Templeton; Bum Kyu Lee; Catherine Rhee; He Li; Cathrine Miner; Joseph D. Dekker; Shari Orlanski; Yehudit Bergman; Vishwanath R. Iyer; Carol F. Webb; Haley Tucker

Summary We show here that singular loss of the Bright/Arid3A transcription factor leads to reprograming of mouse embryonic fibroblasts (MEFs) and enhancement of standard four-factor (4F) reprogramming. Bright-deficient MEFs bypass senescence and, under standard embryonic stem cell (ESC) culture conditions, spontaneously form clones that in vitro express pluripotency markers, differentiate to all germ lineages, and in vivo form teratomas and chimeric mice. We demonstrate that BRIGHT binds directly to the promoter/enhancer regions of Oct4, Sox2, and Nanog to contribute to their repression in both MEFs and ESCs. Thus, elimination of the BRIGHT barrier may provide an approach for somatic cell reprogramming.


Genes & Development | 2014

Arid3a is essential to execution of the first cell fate decision via direct embryonic and extraembryonic transcriptional regulation

Catherine Rhee; Bum Kyu Lee; Samuel Beck; Azeen Anjum; Kendra R. Cook; Melissa Popowski; Haley O. Tucker; Jonghwan Kim

Despite their origin from the inner cell mass, embryonic stem (ES) cells undergo differentiation to the trophectoderm (TE) lineage by repression of the ES cell master regulator Oct4 or activation of the TE master regulator Caudal-type homeobox 2 (Cdx2). In contrast to the in-depth studies of ES cell self-renewal and pluripotency, few TE-specific regulators have been identified, thereby limiting our understanding of mechanisms underlying the first cell fate decision. Here we show that up-regulation and nuclear entry of AT-rich interactive domain 3a (Arid3a) drives TE-like transcriptional programs in ES cells, maintains trophoblast stem (TS) cell self-renewal, and promotes further trophoblastic differentiation both upstream and independent of Cdx2. Accordingly, Arid3a(-/-) mouse post-implantation placental development is severely impaired, resulting in early embryonic death. We provide evidence that Arid3a directly activates TE-specific and trophoblast lineage-specific genes while directly repressing pluripotency genes via differential regulation of epigenetic acetylation or deacetylation. Our results identify Arid3a as a critical regulator of TE and placental development through execution of the commitment and differentiation phases of the first cell fate decision.


Nature Communications | 2014

CpG island-mediated global gene regulatory modes in mouse embryonic stem cells

Samuel Beck; Bum Kyu Lee; Catherine Rhee; Jawon Song; Andrew J. Woo; Jonghwan Kim

Both transcriptional and epigenetic regulations are fundamental for the control of eukaryotic gene expression. Here we perform a compendium analysis of >200 large sequencing data sets to elucidate the regulatory logic of global gene expression programs in mouse embryonic stem (ES) cells. We define four major classes of DNA-binding proteins (Core, PRC, MYC and CTCF) based on their target co-occupancy, and discover reciprocal regulation between the MYC and PRC classes for the activity of nearly all genes under the control of the CpG island (CGI)-containing promoters. This CGI-dependent regulatory mode explains the functional segregation between CGI-containing and CGI-less genes during early development. By defining active enhancers based on the co-occupancy of the Core class, we further demonstrate their additive roles in CGI-containing gene expression and cell type-specific roles in CGI-less gene expression. Altogether, our analyses provide novel insights into previously unknown CGI-dependent global gene regulatory modes.


Blood | 2016

Interferon-α signaling promotes embryonic HSC maturation.

Peter Geon Kim; Matthew C. Canver; Catherine Rhee; Samantha J. Ross; June V. Harriss; Ho-Chou Tu; Stuart H. Orkin; Haley O. Tucker; George Q. Daley

In the developing mouse embryo, the first hematopoietic stem cells (HSCs) arise in the aorta-gonad-mesonephros (AGM) and mature as they transit through the fetal liver (FL). Compared with FL and adult HSCs, AGM HSCs have reduced repopulation potential in irradiated adult transplant recipients but mechanisms underlying this deficiency in AGM HSCs are poorly understood. By co-expression gene network analysis, we deduced that AGM HSCs show lower levels of interferon-α (IFN-α)/Jak-Stat1-associated gene expression than FL HSCs. Treatment of AGM HSCs with IFN-α enhanced long-term hematopoietic engraftment and donor chimerism. Conversely, IFN-α receptor-deficient AGMs (Ifnαr1(-/-)), had significantly reduced donor chimerism. We identify adenine-thymine-rich interactive domain-3a (Arid3a), a factor essential for FL and B lymphopoiesis, as a key transcriptional co-regulator of IFN-α/Stat1 signaling. Arid3a occupies the genomic loci of Stat1 as well as several IFN-α effector genes, acting to regulate their expression. Accordingly, Arid3a(-/-) AGM HSCs had significantly reduced transplant potential, which was rescued by IFN-α treatment. Our results implicate the inflammatory IFN-α/Jak-Stat pathway in the developmental maturation of embryonic HSCs, whose manipulation may lead to increased potency of reprogrammed HSCs for transplantation.


Developmental Biology | 2017

ARID3A is required for mammalian placenta development

Catherine Rhee; Melissa Edwards; Christine Dang; June Harris; Mark A. Brown; Jonghwan Kim; Haley O. Tucker

Previous studies in the mouse indicated that ARID3A plays a critical role in the first cell fate decision required for generation of trophectoderm (TE). Here, we demonstrate that ARID3A is widely expressed during mouse and human placentation and essential for early embryonic viability. ARID3A localizes to trophoblast giant cells and other trophoblast-derived cell subtypes in the junctional and labyrinth zones of the placenta. Conventional Arid3a knockout embryos suffer restricted intrauterine growth with severe defects in placental structural organization. Arid3a null placentas show aberrant expression of subtype-specific markers as well as significant alteration in cytokines, chemokines and inflammatory response-related genes, including previously established markers of human placentation disorders. BMP4-mediated induction of trophoblast stem (TS)-like cells from human induced pluripotent stem cells results in ARID3A up-regulation and cytoplasmic to nuclear translocation. Overexpression of ARID3A in BMP4-mediated TS-like cells up-regulates TE markers, whereas pluripotency markers are down-regulated. Our results reveal an essential, conserved function for ARID3A in mammalian placental development through regulation of both intrinsic and extrinsic developmental programs.


Nucleic Acids Research | 2018

Implications of CpG islands on chromosomal architectures and modes of global gene regulation

Samuel Beck; Catherine Rhee; Jawon Song; Bum Kyu Lee; Lucy LeBlanc; Laurie Cannon; Jonghwan Kim

Abstract CpG islands (CGIs) have long been implicated in the regulation of vertebrate gene expression. However, the involvement of CGIs in chromosomal architectures and associated gene expression regulations has not yet been thoroughly explored. By combining large-scale integrative data analyses and experimental validations, we show that CGIs clearly reconcile two competing models explaining nuclear gene localizations. We first identify CGI-containing (CGI+) and CGI-less (CGI−) genes are non-randomly clustered within the genome, which reflects CGI-dependent spatial gene segregation in the nucleus and corresponding gene regulatory modes. Regardless of their transcriptional activities, CGI+ genes are mainly located at the nuclear center and encounter frequent long-range chromosomal interactions. Meanwhile, nuclear peripheral CGI− genes forming heterochromatin are activated and internalized into the nuclear center by local enhancer–promoter interactions. Our findings demonstrate the crucial implications of CGIs on chromosomal architectures and gene positioning, linking the critical importance of CGIs in determining distinct mechanisms of global gene regulation in three-dimensional space in the nucleus.


Nucleic Acids Research | 2017

Fbxl19 recruitment to CpG islands is required for Rnf20-mediated H2B mono-ubiquitination

Bum Kyu Lee; Jiwoon Lee; Wenwen Shen; Catherine Rhee; Haewon Chung; Jonghwan Kim

Abstract Histone H2B lysine 120 mono-ubiquitination (H2Bub1) catalyzed by Rnf20 has been implicated in normal differentiation of embryonic stem (ES) and adult stem cells. However, it remains unknown how Rnf20 is recruited to its specific target chromosomal loci for the establishment of H2Bub1. Here, we reveal that Fbxl19, a CxxC domain-containing protein, promotes H2Bub1 at the promoters of CpG island-containing genes by interacting with Rnf20. We show that up-regulation of Fbxl19 increases the level of global H2Bub1 in mouse ES cells, while down-regulation of Fbxl19 reduces the level of H2Bub1. Our genome-wide target mapping unveils the preferential occupancy of Fbxl19 on CpG island-containing promoters, and we further discover that chromosomal binding of Fbxl19 is required for H2Bub1 of its targets. Moreover, we reveal that Fbxl19 is critical for proper differentiation of ES cells in collaboration with Rnf20. Altogether, our results demonstrate that Fbxl19 recruitment to CpG islands is required for Rnf20-mediated H2B mono-ubiquitination.


Nucleic Acids Research | 2017

Mechanisms of transcription factor-mediated direct reprogramming of mouse embryonic stem cells to trophoblast stem-like cells

Catherine Rhee; Bum Kyu Lee; Samuel Beck; Lucy LeBlanc; Haley O. Tucker; Jonghwan Kim

Abstract Direct reprogramming can be achieved by forced expression of master transcription factors. Yet how such factors mediate repression of initial cell-type-specific genes while activating target cell-type-specific genes is unclear. Through embryonic stem (ES) to trophoblast stem (TS)-like cell reprogramming by introducing individual TS cell-specific ‘CAG’ factors (Cdx2, Arid3a and Gata3), we interrogate their chromosomal target occupancies, modulation of global transcription and chromatin accessibility at the initial stage of reprogramming. From the studies, we uncover a sequential, two-step mechanism of cellular reprogramming in which repression of pre-existing ES cell-associated gene expression program is followed by activation of TS cell-specific genes by CAG factors. Therefore, we reveal that CAG factors function as both decommission and pioneer factors during ES to TS-like cell fate conversion.


bioRxiv | 2018

Lymphoid origin of a lineage of intrinsically activated plasmacytoid dendritic cell in mice and humans

Joseph D. Dekker; Catherine Rhee; Zicheng Hu; Bum Kyu Lee; Jiwon Lee; Vishwanath R. Iyer; Lauren I. R. Ehrlich; George Georgiou; Haley O. Tucker; Gregory C. Ippolito

We identified a mouse pDC lineage derived exclusively from the common lymphoid progenitor (CLP) that is dependent on expression of Bcl1a. CLP-derived pDC have a unique gene expression profile that includes hallmark B cell genes not normally expressed in pDCs and therefore we refer to this lineage as “B-pDCs.” Unlike classical pDC, B-pDC express an inherent activation signature, localize preferentially to secondary lymphoid organs and expand more robustly and also induce increased T cell proliferation relative to classical pDCs following Toll-like receptor 9 (TLR9) engagement. B-pDCs lack IFN-α secretion but instead express a distinct cytokine profile and display high levels of the cell-surface receptor tyrosine kinase Axl. Murine B-pDCs represent a CLP-derived DC lineage that is genetically, phenotypically, and functionally homologous to an AXL+ DC subtype recently discovered in human blood1–4.

Collaboration


Dive into the Catherine Rhee's collaboration.

Top Co-Authors

Avatar

Bum Kyu Lee

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Jonghwan Kim

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Haley O. Tucker

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Samuel Beck

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Haewon Chung

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Jiwoon Lee

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Lucy LeBlanc

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Wenwen Shen

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge