Network


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

Hotspot


Dive into the research topics where Rachel Munk is active.

Publication


Featured researches published by Rachel Munk.


RNA Biology | 2017

Identification of HuR target circular RNAs uncovers suppression of PABPN1 translation by CircPABPN1

Kotb Abdelmohsen; Amaresh C. Panda; Rachel Munk; Ioannis Grammatikakis; Dawood B. Dudekula; Supriyo De; Jiyoung Kim; Ji Heon Noh; Kyoung Mi Kim; Jennifer L. Martindale; Myriam Gorospe

ABSTRACT HuR influences gene expression programs and hence cellular phenotypes by binding to hundreds of coding and noncoding linear RNAs. However, whether HuR binds to circular RNAs (circRNAs) and impacts on their function is unknown. Here, we have identified en masse circRNAs binding HuR in human cervical carcinoma HeLa cells. One of the most prominent HuR target circRNAs was hsa_circ_0031288, renamed CircPABPN1 as it arises from the PABPN1 pre-mRNA. Further analysis revealed that HuR did not influence CircPABPN1 abundance; interestingly, however, high levels of CircPABPN1 suppressed HuR binding to PABPN1 mRNA. Evaluation of PABPN1 mRNA polysomes indicated that PABPN1 translation was modulated positively by HuR and hence negatively by CircPABPN1. We propose that the extensive binding of CircPABPN1 to HuR prevents HuR binding to PABPN1 mRNA and lowers PABPN1 translation, providing the first example of competition between a circRNA and its cognate mRNA for an RBP that affects translation.


Nucleic Acids Research | 2017

Identification of senescence-associated circular RNAs (SAC-RNAs) reveals senescence suppressor CircPVT1

Amaresh C. Panda; Ioannis Grammatikakis; Kyoung Mi Kim; Supriyo De; Jennifer L. Martindale; Rachel Munk; Xiaoling Yang; Kotb Abdelmohsen; Myriam Gorospe

Abstract Using RNA sequencing (RNA-Seq), we compared the expression patterns of circular RNAs in proliferating (early-passage) and senescent (late-passage) human diploid WI-38 fibroblasts. Among the differentially expressed senescence-associated circRNAs (which we termed ‘SAC-RNAs’), we identified CircPVT1, generated by circularization of an exon of the PVT1 gene, as a circular RNA showing markedly reduced levels in senescent fibroblasts. Reducing CircPVT1 levels in proliferating fibroblasts triggered senescence, as determined by a rise in senescence-associated β-galactosidase activity, higher abundance of CDKN1A/P21 and TP53, and reduced cell proliferation. Although several microRNAs were predicted to bind CircPVT1, only let-7 was found enriched after pulldown of endogenous CircPVT1, suggesting that CircPVT1 might selectively modulate let-7 activity and hence expression of let-7-regulated mRNAs. Reporter analysis revealed that CircPVT1 decreased the cellular pool of available let-7, and antagonizing endogenous let-7 triggered cell proliferation. Importantly, silencing CircPVT1 promoted cell senescence and reversed the proliferative phenotype observed after let-7 function was impaired. Consequently, the levels of several proliferative proteins that prevent senescence, such as IGF2BP1, KRAS and HMGA2, encoded by let-7 target mRNAs, were reduced by silencing CircPVT1. Our findings indicate that the SAC-RNA CircPVT1, elevated in dividing cells and reduced in senescent cells, sequesters let-7 to enable a proliferative phenotype.


Genes & Development | 2016

HuR and GRSF1 modulate the nuclear export and mitochondrial localization of the lncRNA RMRP

Ji Heon Noh; Kyoung Mi Kim; Kotb Abdelmohsen; Je-Hyun Yoon; Amaresh C. Panda; Rachel Munk; Jin Kim; Jessica Curtis; Moad Ca; Wohler Cm; Fred E. Indig; de Paula W; Dawood B. Dudekula; Supriyo De; Yulan Piao; Xiaoling Yang; Jennifer L. Martindale; de Cabo R; Myriam Gorospe

Some mitochondrial long noncoding RNAs (lncRNAs) are encoded by nuclear DNA, but the mechanisms that mediate their transport to mitochondria are poorly characterized. Using affinity RNA pull-down followed by mass spectrometry analysis, we found two RNA-binding proteins (RBPs), HuR (human antigen R) and GRSF1 (G-rich RNA sequence-binding factor 1), that associated with the nuclear DNA-encoded lncRNA RMRP and mobilized it to mitochondria. In cultured human cells, HuR bound RMRP in the nucleus and mediated its CRM1 (chromosome region maintenance 1)-dependent export to the cytosol. After RMRP was imported into mitochondria, GRSF1 bound RMRP and increased its abundance in the matrix. Loss of GRSF1 lowered the mitochondrial levels of RMRP, in turn suppressing oxygen consumption rates and modestly reducing mitochondrial DNA replication priming. Our findings indicate that RBPs HuR and GRSF1 govern the cytoplasmic and mitochondrial localization of the lncRNA RMRP, which is encoded by nuclear DNA but has key functions in mitochondria.


Wiley Interdisciplinary Reviews - Rna | 2017

Emerging roles and context of circular RNAs

Amaresh C. Panda; Ioannis Grammatikakis; Rachel Munk; Myriam Gorospe; Kotb Abdelmohsen

Circular RNAs (circRNAs) represent a large class of noncoding RNAs (ncRNAs) that have recently emerged as regulators of gene expression. They have been shown to suppress microRNAs, thereby increasing the translation and stability of the targets of such microRNAs. In this review, we discuss the emerging functions of circRNAs, including RNA transcription, splicing, turnover, and translation. We also discuss other possible facets of circRNAs that can influence their function depending on the cell context, such as circRNA abundance, subcellular localization, interacting partners (RNA, DNA, and proteins), dynamic changes in interactions following stimulation, and potential circRNA translation. The ensuing changes in gene expression patterns elicited by circRNAs are proposed to drive key cellular processes, such as cell proliferation, differentiation, and survival, that govern health and disease. WIREs RNA 2017, 8:e1386. doi: 10.1002/wrna.1386


Nucleic Acids Research | 2017

High-purity circular RNA isolation method (RPAD) reveals vast collection of intronic circRNAs.

Amaresh C. Panda; Supriyo De; Ioannis Grammatikakis; Rachel Munk; Xiaoling Yang; Yulan Piao; Dawood B. Dudekula; Kotb Abdelmohsen; Myriam Gorospe

Abstract High-throughput RNA sequencing methods coupled with specialized bioinformatic analyses have recently uncovered tens of thousands of unique circular (circ)RNAs, but their complete sequences, genes of origin and functions are largely unknown. Given that circRNAs lack free ends and are thus relatively stable, their association with microRNAs (miRNAs) and RNA-binding proteins (RBPs) can influence gene expression programs. While exoribonuclease treatment is widely used to degrade linear RNAs and enrich circRNAs in RNA samples, it does not efficiently eliminate all linear RNAs. Here, we describe a novel method for the isolation of highly pure circRNA populations involving RNase R treatment followed by Polyadenylation and poly(A)+ RNA Depletion (RPAD), which removes linear RNA to near completion. High-throughput sequencing of RNA prepared using RPAD from human cervical carcinoma HeLa cells and mouse C2C12 myoblasts led to two surprising discoveries: (i) many exonic circRNA (EcircRNA) isoforms share an identical backsplice sequence but have different body sizes and sequences, and (ii) thousands of novel intronic circular RNAs (IcircRNAs) are expressed in cells. In sum, isolating high-purity circRNAs using the RPAD method can enable quantitative and qualitative analyses of circRNA types and sequence composition, paving the way for the elucidation of circRNA functions.


PLOS ONE | 2011

Involvement of mTOR in CXCL12 Mediated T Cell Signaling and Migration

Rachel Munk; Paritosh Ghosh; Manik C. Ghosh; Takeshi Saito; Mai Xu; Arnell Carter; Fred E. Indig; Dennis D. Taub; Dan L. Longo

Background CXCL12 is a pleiotropic chemokine involved in multiple different processes such as immune regulation, inflammatory responses, and cancer development. CXCL12 is also a potent chemokine involved in chemoattraction of T cells to the site of infection or inflammation. Mammalian target of rapamycin (mTOR) is a serine-threonine kinase that modulates different cellular processes, such as metabolism, nutrient sensing, protein translation, and cell growth. The role of mTOR in CXCL12-mediated resting T cell migration has yet to be elucidated. Methodology/Principal Findings Rapamycin, an inhibitor of mTOR, significantly inhibits CXCL12 mediated migration of both primary human resting T cells and human T cell leukemia cell line CEM. p70S6K1, an effector molecule of mTOR signaling pathway, was knocked down by shRNA in CEM cells using a lentiviral gene transfer system. Using p70S6K1 knock down cells, we demonstrate the role of mTOR signaling in T cell migration both in vitro and in vivo. Conclusions Our data demonstrate a new role for mTOR in CXCL12-induced T cell migration, and enrich the current knowledge regarding the clinical use of rapamycin.


European Journal of Immunology | 2016

p(⁷⁰S⁶K¹) in the TORC1 pathway is essential for the differentiation of Th17 Cells, but not Th1, Th2, or Treg cells in mice.

Carl Y. Sasaki; Gang Chen; Rachel Munk; Erez Eitan; Jennifer L. Martindale; Dan L. Longo; Paritosh Ghosh

The TORC1 pathway is necessary for ribosomal biogenesis and initiation of protein translation. Furthermore, the differentiation of Th1 and Th17 cells requires TORC1 activity. To investigate the role of the TORC1 pathway in the differentiation of Th1 and/or Th17 cells in more detail, we compared the differentiation capacity of naïve T cells from wild type and p70S6K1 knockout mice. Expression of many of the genes associated with Th17‐cell differentiation, such as IL17a, IL17f, and IL‐23R, were reduced in p70S6K1 knockout mice. In contrast, the development of Th1, Th2, and Treg cells was unaffected in the absence of p70S6K1. Furthermore, expression of the major transcription factor in Th17‐cell differentiation, retinoic acid receptor‐related orphan receptor gamma T, remained unchanged. However, the acetylation of histone 3 at the promoters of IL17a and IL17f was reduced in the absence of p70S6K1. In accordance with the in vitro data, the kinetics, but not the development, of EAE was affected with the loss of p70S6K1 expression. Collectively, our findings suggested that both in vitro and in vivo differentiation of Th17 cells were positively regulated by p70S6K1.


Wiley Interdisciplinary Reviews - Rna | 2018

Noncoding RNAs in Alzheimer's disease

M. Laura Idda; Rachel Munk; Kotb Abdelmohsen; Myriam Gorospe

Alzheimers disease (AD) is a progressive neurodegenerative disorder and the main cause of dementia among the elderly worldwide. Despite intense efforts to develop drugs for preventing and treating AD, no effective therapies are available as yet, posing a growing burden at the personal, medical, and socioeconomic levels. AD is characterized by the production and aggregation of amyloid β (Aβ) peptides derived from amyloid precursor protein (APP), the presence of hyperphosphorylated microtubule‐associated protein Tau (MAPT), and chronic inflammation leading to neuronal loss. Aβ accumulation and hyperphosphorylated Tau are responsible for the main histopathological features of AD, Aβ plaques, and neurofibrillary tangles (NFTs), respectively. However, the full spectrum of molecular factors that contribute to AD pathogenesis is not known. Noncoding (nc)RNAs, including microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs), regulate gene expression at the transcriptional and posttranscriptional levels in various diseases, serving as biomarkers and potential therapeutic targets. There is rising recognition that ncRNAs have been implicated in both the onset and pathogenesis of AD. Here, we review the ncRNAs implicated posttranscriptionally in the main AD pathways and discuss the growing interest in targeting regulatory ncRNAs therapeutically to combat AD pathology. WIREs RNA 2018, 9:e1463. doi: 10.1002/wrna.1463


Oncotarget | 2017

LncRNA OIP5-AS1/cyrano suppresses GAK expression to control mitosis

Jiyoung Kim; Ji Heon Noh; Seung-Kyu Lee; Rachel Munk; Alexei A. Sharov; Elin Lehrmann; Yongqing Zhang; Weidong Wang; Kotb Abdelmohsen; Myriam Gorospe

Some long noncoding RNAs (lncRNAs) can regulate gene expression programs, in turn affecting specific cellular processes. We sought to identify the mechanism through which the lncRNA OIP5-AS1, which is abundant in the cytoplasm, suppressed cell proliferation. Silencing of OIP5-AS1 in human cervical carcinoma HeLa cells triggered the appearance of many aberrant (monopolar, multipolar, misaligned) mitotic spindles. Through a combination of approaches to pull down mRNAs bound to OIP5-AS1 and identify proteins differentially expressed when OIP5-AS1 was silenced, we identified a subset of human cell cycle regulatory proteins encoded by mRNAs that interacted with OIP5-AS1 in HeLa cells. Further analysis revealed that GAK mRNA, which encodes a cyclin G-associated kinase important for mitotic progression, associated prominently with OIP5-AS1. The interaction between these two transcripts led to a reduction in GAK mRNA stability and GAK protein abundance, as determined in cells in which OIP5-AS1 levels were increased or decreased. Importantly, the aberrant mitotic cell division seen after silencing OIP5-AS1 was partly rescued if GAK was simultaneously silenced. These findings indicate that the abnormal mitoses seen after silencing OIP5-AS1 were caused by an untimely rise in GAK levels and suggest that OIP5-AS1 suppresses cell proliferation at least in part by reducing GAK levels.


International Review of Cell and Molecular Biology | 2017

Senescence-Associated MicroRNAs

Rachel Munk; Amaresh C. Panda; Ioannis Grammatikakis; Myriam Gorospe; Kotb Abdelmohsen

Senescent cells arise as a consequence of cellular damage and can have either a detrimental or advantageous impact on tissues and organs depending on the specific cell type and metabolic state. As senescent cells accumulate in tissues with advancing age, they have been implicated in many age-related declines and diseases. The major facets of senescence include two pathways responsible for establishing and maintaining a senescence program, p53/CDKN1A(p21) and CDKN2A(p16)/RB, as well as the senescence-associated secretory phenotype. Numerous MicroRNAs influence senescence by modulating the abundance of key senescence regulatory proteins, generally by lowering the stability and/or translation of mRNAs that encode such factors. Accordingly, understanding the molecular mechanisms by which MicroRNAs influence senescence will enable diagnostic and therapeutic opportunities directed at senescent cells. Here, we review senescence-associated (SA)-MicroRNAs and discuss their implications in senescence-relevant pathologies.

Collaboration


Dive into the Rachel Munk's collaboration.

Top Co-Authors

Avatar

Kotb Abdelmohsen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Myriam Gorospe

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Amaresh C. Panda

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dan L. Longo

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Ioannis Grammatikakis

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paritosh Ghosh

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Supriyo De

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Xiaoling Yang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Carl Y. Sasaki

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge