Network


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

Hotspot


Dive into the research topics where Kotb Abdelmohsen is active.

Publication


Featured researches published by Kotb Abdelmohsen.


Molecular Cell | 2012

LincRNA-p21 Suppresses Target mRNA Translation

Je-Hyun Yoon; Kotb Abdelmohsen; Subramanya Srikantan; Xiaoling Yang; Jennifer L. Martindale; Supriyo De; Maite Huarte; Ming Zhan; Kevin G. Becker; Myriam Gorospe

Mammalian long intergenic noncoding RNAs (lincRNAs) are best known for modulating transcription. Here we report a posttranscriptional function for lincRNA-p21 as a modulator of translation. Association of the RNA-binding protein HuR with lincRNA-p21 favored the recruitment of let-7/Ago2 to lincRNA-p21, leading to lower lincRNA-p21 stability. Under reduced HuR levels, lincRNA-p21 accumulated in human cervical carcinoma HeLa cells, increasing its association with JUNB and CTNNB1 mRNAs and selectively lowering their translation. With elevated HuR, lincRNA-p21 levels declined, which in turn derepressed JunB and β-catenin translation and increased the levels of these proteins. We propose that HuR controls translation of a subset of target mRNAs by influencing lincRNA-p21 levels. Our findings uncover a role for lincRNA as a posttranscriptional inhibitor of translation.


Journal of Molecular Biology | 2013

Posttranscriptional Gene Regulation by Long Noncoding RNA

Je-Hyun Yoon; Kotb Abdelmohsen; Myriam Gorospe

Eukaryotic cells transcribe a vast number of noncoding RNA species. Among them, long noncoding RNAs (lncRNAs) have been widely implicated in the regulation of gene transcription. However, examples of posttranscriptional gene regulation by lncRNAs are emerging. Through extended base-pairing, lncRNAs can stabilize or promote the translation of target mRNAs, while partial base-pairing facilitates mRNA decay or inhibits target mRNA translation. In the absence of complementarity, lncRNAs can suppress precursor mRNA splicing and translation by acting as decoys of RNA-binding proteins or microRNAs and can compete for microRNA-mediated inhibition leading to increased expression of the mRNA. Through these regulatory mechanisms, lncRNAs can elicit differentiation, proliferation, and cytoprotective programs, underscoring the rising recognition of lncRNA roles in human disease. In this review, we summarize the mechanisms of posttranscriptional gene regulation by lncRNAs identified until now.


PLOS ONE | 2010

microRNA Expression Patterns Reveal Differential Expression of Target Genes with Age

Nicole Noren Hooten; Kotb Abdelmohsen; Myriam Gorospe; Ngozi Ejiogu; Alan B. Zonderman; Michele K. Evans

Recent evidence supports a role for microRNAs (miRNAs) in regulating the life span of model organisms. However, little is known about how these small RNAs contribute to human aging. Here, we profiled the expression of over 800 miRNAs in peripheral blood mononuclear cells from young and old individuals by real-time RT-PCR analysis. This genome-wide assessment of miRNA expression revealed that the majority of miRNAs studied decreased in abundance with age. We identified nine miRNAs (miR-103, miR-107, miR-128, miR-130a, miR-155, miR-24, miR-221, miR-496, miR-1538) that were significantly lower in older individuals. Among them, five have been implicated in cancer pathogenesis. Predicted targets of several of these miRNAs, including PI3 kinase (PI3K), c-Kit and H2AX, were found to be elevated with advancing age, supporting a possible role for them in the aging process. Furthermore, we found that decreasing the levels of miR-221 was sufficient to cause a corresponding increase in the expression of the predicted target, PI3K. Taken together, these findings demonstrate that changes in miRNA expression occur with human aging and suggest that miRNAs and their predicted targets have the potential to be diagnostic indicators of age or age-related diseases.


Wiley Interdisciplinary Reviews - Rna | 2010

Posttranscriptional regulation of cancer traits by HuR

Kotb Abdelmohsen; Myriam Gorospe

Cancer‐related gene expression programs are strongly influenced by posttranscriptional mechanisms. The RNA‐binding protein HuR is highly abundant in many cancers. Numerous HuR‐regulated mRNAs encode proteins implicated in carcinogenesis. Here, we review the collections of HuR target mRNAs that encode proteins responsible for implementing five major cancer traits. By interacting with specific mRNA subsets, HuR enhances the levels of proteins that (1) promote cell proliferation, (2) increase cell survival, (3) elevate local angiogenesis, (4) help the cancer cell evade immune recognition, and (5) facilitate cancer cell invasion and metastasis. We propose that HuR exerts a tumorigenic function by enabling these cancer phenotypes. We discuss evidence that links HuR to several specific cancers and suggests its potential usefulness in cancer diagnosis, prognosis, and therapy. Copyright


PLOS ONE | 2008

p16INK4a translation suppressed by miR-24

Ashish Lal; Hyeon-Ho Kim; Kotb Abdelmohsen; Yuki Kuwano; Rudolf Pullmann; Subramanya Srikantan; Ramesh Subrahmanyam; Jennifer L. Martindale; Xiaoling Yang; Fariyal Ahmed; Francisco Navarro; Derek M. Dykxhoorn; Judy Lieberman; Myriam Gorospe

Background Expression of the tumor suppressor p16INK4a increases during aging and replicative senescence. Methodology/Principal Findings Here, we report that the microRNA miR-24 suppresses p16 expression in human diploid fibroblasts and cervical carcinoma cells. Increased p16 expression with replicative senescence was associated with decreased levels of miR-24, a microRNA that was predicted to associate with the p16 mRNA coding and 3′-untranslated regions. Ectopic miR-24 overexpression reduced p16 protein but not p16 mRNA levels. Conversely, introduction of antisense (AS)-miR-24 blocked miR-24 expression and markedly enhanced p16 protein levels, p16 translation, and the production of EGFP-p16 reporter bearing the miR-24 target recognition sites. Conclusions/Significance Together, our results suggest that miR-24 represses the initiation and elongation phases of p16 translation.


Molecular and Cellular Biology | 2011

miR-130 Suppresses Adipogenesis by Inhibiting Peroxisome Proliferator-Activated Receptor Expression

Eun Kyung Lee; Mi-Jeong Lee; Kotb Abdelmohsen; Wook Kim; Mihee M. Kim; Subramanya Srikantan; Jennifer L. Martindale; Emmette R. Hutchison; Hyeon Ho Kim; Bernard S. Marasa; Roza Selimyan; Josephine M. Egan; Steven R. Smith; Susan K. Fried; Myriam Gorospe

ABSTRACT Adipose tissue development is tightly regulated by altering gene expression. MicroRNAs are strong posttranscriptional regulators of mammalian differentiation. We hypothesized that microRNAs might influence human adipogenesis by targeting specific adipogenic factors. We identified microRNAs that showed varying abundance during the differentiation of human preadipocytes into adipocytes. Among them, miR-130 strongly affected adipocyte differentiation, as overexpressing miR-130 impaired adipogenesis and reducing miR-130 enhanced adipogenesis. A key effector of miR-130 actions was the protein peroxisome proliferator-activated receptor γ (PPARγ), a major regulator of adipogenesis. Interestingly, miR-130 potently repressed PPARγ expression by targeting both the PPARγ mRNA coding and 3′ untranslated regions. Adipose tissue from obese women contained significantly lower miR-130 and higher PPARγ mRNA levels than that from nonobese women. Our findings reveal that miR-130 reduces adipogenesis by repressing PPARγ biosynthesis and suggest that perturbations in this regulation is linked to human obesity.


Molecular and Cellular Biology | 2008

RNA-Binding Proteins HuR and PTB Promote the Translation of Hypoxia-Inducible Factor 1α

Stefanie Galban; Yuki Kuwano; Rudolf Pullmann; Jennifer L. Martindale; Hyeon Ho Kim; Ashish Lal; Kotb Abdelmohsen; Xiaoling Yang; Youngjun Dang; Jun O. Liu; Stephen M. Lewis; Martin Holcik; Myriam Gorospe

ABSTRACT The levels of hypoxia-inducible factor 1α (HIF-1α) are tightly controlled. Here, we investigated the posttranscriptional regulation of HIF-1α expression in human cervical carcinoma HeLa cells responding to the hypoxia mimetic CoCl2. Undetectable in untreated cells, HIF-1α levels increased dramatically in CoCl2-treated cells, while HIF-1α mRNA levels were unchanged. HIF-1α translation was potently elevated by CoCl2 treatment, as determined by de novo translation analysis and by monitoring the polysomal association of HIF-1α mRNA. An internal ribosome entry site in the HIF-1α 5′ untranslated region (UTR) was found to enhance translation constitutively, but it did not further induce translation in response to CoCl2 treatment. Instead, we postulated that RNA-binding proteins HuR and PTB, previously shown to bind HIF-1α mRNA, participated in its translational upregulation after CoCl2 treatment. Indeed, both RNA-binding proteins were found to bind HIF-1α mRNA in a CoCl2-inducible manner as assessed by immunoprecipitation of endogenous ribonucleoprotein complexes. Using a chimeric reporter, polypyrimidine tract-binding protein (PTB) was found to bind the HIF-1α 3′UTR, while HuR associated principally with the 5′UTR. Lowering PTB expression or HuR expression using RNA interference reduced HIF-1α translation and expression levels but not HIF-1α mRNA abundance. Conversely, HIF-1α expression and translation in response to CoCl2 were markedly elevated after HuR overexpression. We propose that HuR and PTB jointly upregulate HIF-1α translation in response to CoCl2.


Cell Cycle | 2007

Posttranscriptional Orchestration of an Anti-Apoptotic Program by HuR

Kotb Abdelmohsen; Ashish Lal; Hyeon Ho Kim; Myriam Gorospe

The RNA-binding protein HuR can stabilize and/or regulate the translation of target mRNAs, thereby affecting the cellular responses to immune, proliferative, and damaging agents. Here, we discuss emerging evidence that HuR elicits a broad anti-apoptotic function through its influence on the expression of multiple target mRNAs. HuR was previously shown to bind to the mRNA encoding the apoptosome inhibitor prothymosin α (ProTα) and enhanced its translation and cytoplasmic abundance. More recently, HuR was shown to increase the stability of a target mRNA encoding the pro-survival deacetylase SIRT1. The discovery that HuR likewise binds to and promotes the expression of mRNAs encoding Bcl-2 and Mcl-1, two major anti-apoptotic effectors, strongly supports HuR’s role as a key upstream coordinator of a constitutive pro-survival program.


Molecular and Cellular Biology | 2008

MKP-1 mRNA Stabilization and Translational Control by RNA-Binding Proteins HuR and NF90†

Yuki Kuwano; Hyeon Ho Kim; Kotb Abdelmohsen; Rudolf Pullmann; Jennifer L. Martindale; Xiaoling Yang; Myriam Gorospe

ABSTRACT The mitogen-activated protein (MAP) kinase phosphatase 1 (MKP-1) plays a major role in dephosphorylating and thereby inactivating the MAP kinases extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38. Here, we examine the posttranscriptional events underlying the robust MKP-1 induction by oxidants in HeLa cells. H2O2 treatment potently stabilized the MKP-1 mRNA and increased the association of MKP-1 mRNA with the translation machinery. Four RNA-binding proteins (RNA-BPs) that influence mRNA turnover and/or translation (HuR, NF90, TIAR, and TIA-1) were found to bind to biotinylated transcripts spanning the MKP-1 AU-rich 3′ untranslated region. By using ribonucleoprotein immunoprecipitation analysis, we showed that H2O2 treatment increased the association of MKP-1 mRNA with HuR and NF90 and decreased its association with the translational repressors TIAR and TIA-1. HuR or NF90 silencing significantly diminished the H2O2-stimulated MKP-1 mRNA stability; HuR silencing also markedly decreased MKP-1 translation. In turn, lowering MKP-1 expression in HuR-silenced cultures resulted in substantially elevated phosphorylation of JNK and p38 after H2O2 treatment. Collectively, MKP-1 upregulation by oxidative stress is potently influenced by increased mRNA stability and translation, mediated at least in part by the RNA-BPs HuR and NF90.


Biological Chemistry | 2008

Posttranscriptional gene regulation by RNA-binding proteins during oxidative stress: implications for cellular senescence.

Kotb Abdelmohsen; Yuki Kuwano; Hyeon Ho Kim; Myriam Gorospe

Abstract To respond adequately to oxidative stress, mammalian cells elicit rapid and tightly controlled changes in gene expression patterns. Besides alterations in the subsets of transcribed genes, two posttranscriptional processes prominently influence the oxidant-triggered gene expression programs: mRNA turnover and translation. Here, we review recent progress in our knowledge of the turnover and translation regulatory (TTR) mRNA-binding proteins (RBPs) that influence gene expression in response to oxidative damage. Specifically, we identify oxidant damage-regulated mRNAs that are targets of TTR-RBPs, we review the oxidant-triggered signaling pathways that govern TTR-RBP function, and we examine emerging evidence that TTR-RBP activity is altered with senescence and aging. Given the potent influence of TTR-RBPs upon oxidant-regulated gene expression profiles, we propose that the senescence-associated changes in TTR-RBPs directly contribute to the impaired responses to oxidant damage that characterize cellular senescence and advancing age.

Collaboration


Dive into the Kotb Abdelmohsen's collaboration.

Top Co-Authors

Avatar

Myriam Gorospe

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiaoling Yang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Subramanya Srikantan

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

Ioannis Grammatikakis

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kevin G. Becker

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ji Heon Noh

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Je-Hyun Yoon

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ashish Lal

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge