Network


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

Hotspot


Dive into the research topics where Jayantha Gunaratne is active.

Publication


Featured researches published by Jayantha Gunaratne.


Nature Structural & Molecular Biology | 2012

Symmetric dimethylation of H3R2 is a newly identified histone mark that supports euchromatin maintenance.

Valentina Migliori; Julius Muller; Sameer Phalke; Diana Low; Marco Bezzi; Wei Chuen Mok; Sanjeeb Kumar Sahu; Jayantha Gunaratne; Paola Capasso; Christian Bassi; Valentina Cecatiello; Ario de Marco; Walter Blackstock; Vladimir A. Kuznetsov; Bruno Amati; Marina Mapelli; Ernesto Guccione

The asymmetric dimethylation of histone H3 arginine 2 (H3R2me2a) acts as a repressive mark that antagonizes trimethylation of H3 lysine 4. Here we report that H3R2 is also symmetrically dimethylated (H3R2me2s) by PRMT5 and PRMT7 and present in euchromatic regions. Profiling of H3-tail interactors by SILAC MS revealed that H3R2me2s excludes binding of RBBP7, a central component of co-repressor complexes Sin3a, NURD and PRC2. Conversely H3R2me2s enhances binding of WDR5, a common component of the coactivator complexes MLL, SET1A, SET1B, NLS1 and ATAC. The interaction of histone H3 with WDR5 distinguishes H3R2me2s from H3R2me2a, which impedes the recruitment of WDR5 to chromatin. The crystallographic structure of WDR5 and the H3R2me2s peptide elucidates the molecular determinants of this high affinity interaction. Our findings identify H3R2me2s as a previously unknown mark that keeps genes poised in euchromatin for transcriptional activation upon cell-cycle withdrawal and differentiation in human cells.


RNA Biology | 2011

Quantitative mass spectrometry of DENV-2 RNA-interacting proteins reveals that the DEAD-box RNA helicase DDX6 binds the DB1 and DB2 3’ UTR structures

Alex M. Ward; Katell Bidet; Ang Yinglin; Siok Ghee Ler; Kelly Hogue; Walter Blackstock; Jayantha Gunaratne; Mariano A. Garcia-Blanco

Dengue virus (DENV) is a rapidly re-emerging flavivirus that causes dengue fever (DF), dengue hemorrhagic fever (DHF) and dengue shock syndrome (DSS), diseases for which there are no available therapies or vaccines. The DENV-2 positive-strand RNA genome contains 5’ and 3’ untranslated regions (UTRs) that have been shown to form secondary structures required for virus replication and interaction with host cell proteins. In order to comprehensively identify host cell factors that bind the DENV-2 UTRs, we performed RNA chromatography, using the DENV-2 5’ and 3’ UTRs as “bait”, combined with quantitative mass spectrometry. We identified several proteins, including DDX6, G3BP1, G3BP2, Caprin1, and USP10, implicated in P body (PB) and stress granule (SG) function, and not previously known to bind DENV RNAs. Indirect immunofluorescence microscopy showed these proteins to colocalize with the DENV replication complex. Moreover, DDX6 knockdown resulted in reduced amounts of infectious particles and viral RNA in tissue culture supernatants following DENV infection. DDX6 interacted with DENV RNA in vivo during infection and in vitro this interaction was mediated by the DB1 and DB2 structures in the 3’ UTR, possibly by formation of a pseudoknot structure. Additional experiments demonstrate that, in contrast to DDX6, the SG proteins G3BP1, G3BP2, Caprin1 and USP10 bind to the variable region (VR) in the 3’ UTR. These results suggest that the DENV-2 3’ UTR is a site for assembly of PB and SG proteins and, for DDX6, assembly on the 3’ UTR is required for DENV replication.


Science | 2015

Dengue subgenomic RNA binds TRIM25 to inhibit interferon expression for epidemiological fitness

Gayathri Manokaran; Esteban Finol; Chunling Wang; Jayantha Gunaratne; Justin Bahl; Eugenia Z. Ong; Hwee Cheng Tan; October M. Sessions; Alex M. Ward; Duane J. Gubler; Eva Harris; Mariano A. Garcia-Blanco; Eng Eong Ooi

Orchestrating a viral takeover For some pathogenic viruses, outbreaks occur when a new viral strain emerges and displaces the endemic strain. How such a takeover occurs at a molecular level, however, remains an open question. Manokaran et al. examined one example, the emergence of a new clade of dengue virus (DENV) that caused an outbreak in Puerto Rico in 1994. The epidemic strain produced elevated amounts of subgenomic flavivirus RNA (sfRNA), a viral noncoding RNA, relative to amounts of genomic viral RNA. sfRNA bound to and inhibited TRIM25, a protein important for activating the hosts antiviral response, and so by reducing host immunity was able to increase its own fitness. Science, this issue p. 217 Elevated amounts of a viral noncoding RNA that suppresses host immunity likely led to an outbreak of dengue virus. The global spread of dengue virus (DENV) infections has increased viral genetic diversity, some of which appears associated with greater epidemic potential. The mechanisms governing viral fitness in epidemiological settings, however, remain poorly defined. We identified a determinant of fitness in a foreign dominant (PR-2B) DENV serotype 2 (DENV-2) clade, which emerged during the 1994 epidemic in Puerto Rico and replaced an endemic (PR-1) DENV-2 clade. The PR-2B DENV-2 produced increased levels of subgenomic flavivirus RNA (sfRNA) relative to genomic RNA during replication. PR-2B sfRNA showed sequence-dependent binding to and prevention of tripartite motif 25 (TRIM25) deubiquitylation, which is critical for sustained and amplified retinoic acid–inducible gene 1 (RIG-I)–induced type I interferon expression. Our findings demonstrate a distinctive viral RNA–host protein interaction to evade the innate immune response for increased epidemiological fitness.


Genes & Development | 2013

ER stress potentiates insulin resistance through PERK-mediated FOXO phosphorylation

Wei Zhang; Ville Hietakangas; Sheena Wee; Siew Choo Lim; Jayantha Gunaratne; Stephen M. Cohen

Endoplasmic reticulum (ER) stress is emerging as a potential contributor to the onset of type 2 diabetes by making cells insulin-resistant. However, our understanding of the mechanisms by which ER stress affects insulin response remains fragmentary. Here we present evidence that the ER stress pathway acts via a conserved signaling mechanism involving the protein kinase PERK to modulate cellular insulin responsiveness. Insulin signaling via AKT reduces activity of FOXO transcription factors. In some cells, PERK can promote insulin responsiveness. However, we found that PERK also acts oppositely via phosphorylation of FOXO to promote FOXO activity. Inhibition of PERK improves cellular insulin responsiveness at the level of FOXO activity. We suggest that the protein kinase PERK may be a promising pharmacological target for ameliorating insulin resistance.


Proceedings of the National Academy of Sciences of the United States of America | 2012

Ecotopic viral integration site 1 (EVI1) regulates multiple cellular processes important for cancer and is a synergistic partner for FOS protein in invasive tumors

Emilie A. Bard-Chapeau; Justin Jeyakani; Chung H. Kok; Julius Muller; Belinda Q. Chua; Jayantha Gunaratne; Arsen O Batagov; Piroon Jenjaroenpun; Vladimir A. Kuznetsov; Chia Lin Wei; Richard J. D'Andrea; Guillaume Bourque; Nancy A. Jenkins; Neal G. Copeland

Ecotropic viral integration site 1 (EVI1) is an oncogenic dual domain zinc finger transcription factor that plays an essential role in the regulation of hematopoietic stem cell renewal, and its overexpression in myeloid leukemia and epithelial cancers is associated with poor patient survival. Despite the discovery of EVI1 in 1988 and its emerging role as a dominant oncogene in various types of cancer, few EVI1 target genes are known. This lack of knowledge has precluded a clear understanding of exactly how EVI1 contributes to cancer. Using a combination of ChIP-Seq and microarray studies in human ovarian carcinoma cells, we show that the two zinc finger domains of EVI1 bind to DNA independently and regulate different sets of target genes. Strikingly, an enriched fraction of EVI1 target genes are cancer genes or genes associated with cancer. We also show that more than 25% of EVI1-occupied genes contain linked EVI1 and activator protein (AP)1 DNA binding sites, and this finding provides evidence for a synergistic cooperative interaction between EVI1 and the AP1 family member FOS in the regulation of cell adhesion, proliferation, and colony formation. An increased number of dual EVI1/AP1 target genes are also differentially regulated in late-stage ovarian carcinomas, further confirming the importance of the functional cooperation between EVI1 and FOS. Collectively, our data indicate that EVI1 is a multipurpose transcription factor that synergizes with FOS in invasive tumors.


Genes & Development | 2015

Mutant p53 cooperates with the SWI/SNF chromatin remodeling complex to regulate VEGFR2 in breast cancer cells

Neil T. Pfister; Vitalay Fomin; Kausik Regunath; Jeffrey Y. Zhou; Wen Zhou; Laxmi Silwal-Pandit; William A. Freed-Pastor; Oleg Laptenko; Suat Peng Neo; Jill Bargonetti; Mainul Hoque; Bin Tian; Jayantha Gunaratne; Olav Engebraaten; James L. Manley; Anne Lise Børresen-Dale; Paul M. Neilsen; Carol Prives

Mutant p53 impacts the expression of numerous genes at the level of transcription to mediate oncogenesis. We identified vascular endothelial growth factor receptor 2 (VEGFR2), the primary functional VEGF receptor that mediates endothelial cell vascularization, as a mutant p53 transcriptional target in multiple breast cancer cell lines. Up-regulation of VEGFR2 mediates the role of mutant p53 in increasing cellular growth in two-dimensional (2D) and three-dimensional (3D) culture conditions. Mutant p53 binds near the VEGFR2 promoter transcriptional start site and plays a role in maintaining an open conformation at that location. Relatedly, mutant p53 interacts with the SWI/SNF complex, which is required for remodeling the VEGFR2 promoter. By both querying individual genes regulated by mutant p53 and performing RNA sequencing, the results indicate that >40% of all mutant p53-regulated gene expression is mediated by SWI/SNF. We surmise that mutant p53 impacts transcription of VEGFR2 as well as myriad other genes by promoter remodeling through interaction with and likely regulation of the SWI/SNF chromatin remodeling complex. Therefore, not only might mutant p53-expressing tumors be susceptible to anti VEGF therapies, impacting SWI/SNF tumor suppressor function in mutant p53 tumors may also have therapeutic potential.


EMBO Reports | 2012

Mutant p53 interactome identifies nardilysin as a p53R273H-specific binding partner that promotes invasion.

Cynthia R. Coffill; Patricia A. J. Muller; Hue Kian Oh; Suat Peng Neo; Kelly Hogue; Chit Fang Cheok; Karen H. Vousden; David P. Lane; Walter Blackstock; Jayantha Gunaratne

The invasiveness of tumour cells depends on changes in cell shape, polarity and migration. Mutant p53 induces enhanced tumour metastasis in mice, and human cells overexpressing p53R273H have aberrant polarity and increased invasiveness, demonstrating the ‘gain of function’ of mutant p53 in carcinogenesis. We hypothesize that p53R273H interacts with mutant p53‐specific binding partners that control polarity, migration or invasion. Here we analyze the p53R273H interactome using stable isotope labelling by amino acids in cell culture and quantitative mass spectrometry, and identify at least 15 new potential mutant p53‐specific binding partners. The interaction of p53R273H with one of them—nardilysin (NRD1)—promotes an invasive response to heparin binding–epidermal growth factor‐like growth factor that is p53R273H‐dependant but does not require Rab coupling protein or p63. Advanced proteomics has thus allowed the detection of a new mechanism of p53‐driven invasion.


Cell Stem Cell | 2015

ELABELA Is an Endogenous Growth Factor that Sustains hESC Self-Renewal via the PI3K/AKT Pathway

Lena Ho; Shawn Y.X. Tan; Sheena Wee; Yixuan Wu; Sam J.C. Tan; Navin B. Ramakrishna; Serene C. Chng; Srikanth Nama; Iwona Szczerbinska; Yun-Shen Chan; Stuart Avery; Norihiro Tsuneyoshi; Huck-Hui Ng; Jayantha Gunaratne; N. Ray Dunn; Bruno Reversade

ELABELA (ELA) is a peptide hormone required for heart development that signals via the Apelin Receptor (APLNR, APJ). ELA is also abundantly secreted by human embryonic stem cells (hESCs), which do not express APLNR. Here we show that ELA signals in a paracrine fashion in hESCs to maintain self-renewal. ELA inhibition by CRISPR/Cas9-mediated deletion, shRNA, or neutralizing antibodies causes reduced hESC growth, cell death, and loss of pluripotency. Global phosphoproteomic and transcriptomic analyses of ELA-pulsed hESCs show that it activates PI3K/AKT/mTORC1 signaling required for cell survival. ELA promotes hESC cell-cycle progression and protein translation and blocks stress-induced apoptosis. INSULIN and ELA have partially overlapping functions in hESC medium, but only ELA can potentiate the TGFβ pathway to prime hESCs toward the endoderm lineage. We propose that ELA, acting through an alternate cell-surface receptor, is an endogenous secreted growth factor in human embryos and hESCs that promotes growth and pluripotency.


Nature Communications | 2015

An oncogenic role of Agrin in regulating focal adhesion integrity in hepatocellular carcinoma

Sayan Chakraborty; Manikandan Lakshmanan; Hannah Lee-Foon Swa; Jianxiang Chen; Xiaoqian Zhang; Yan Shan Ong; Li Shen Loo; Semih Can Akıncılar; Jayantha Gunaratne; Vinay Tergaonkar; Kam M. Hui; Wanjin Hong

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths globally. The identity and role of cell surface molecules driving complex biological events leading to HCC progression are poorly understood, hence representing major lacunae in HCC therapies. Here, combining SILAC quantitative proteomics and biochemical approaches, we uncover a critical oncogenic role of Agrin, which is overexpressed and secreted in HCC. Agrin enhances cellular proliferation, migration and oncogenic signalling. Mechanistically, Agrin’s extracellular matrix sensor activity provides oncogenic cues to regulate Arp2/3-dependent ruffling, invadopodia formation and epithelial–mesenchymal transition through sustained focal adhesion integrity that drives liver tumorigenesis. Furthermore, Agrin signalling through Lrp4-muscle-specific tyrosine kinase (MuSK) forms a critical oncogenic axis. Importantly, antibodies targeting Agrin reduced oncogenic signalling and tumour growth in vivo. Together, we demonstrate that Agrin is frequently upregulated and important for oncogenic property of HCC, and is an attractive target for antibody therapy.


Proceedings of the National Academy of Sciences of the United States of America | 2013

EVI1 oncoprotein interacts with a large and complex network of proteins and integrates signals through protein phosphorylation

Emilie A. Bard-Chapeau; Jayantha Gunaratne; Pankaj Kumar; Belinda Q. Chua; Julius Muller; Frederic Bard; Walter Blackstock; Neal G. Copeland; Nancy A. Jenkins

Significance Although ecotropic viral integration site 1 (EVI1) oncogenic transcription factor was discovered in 1988, its molecular functions and regulations are still underexplored. Through characterization of few EVI1-interacting proteins, EVI1 was identified as dynamic modulator of transcription and chromatin remodeling. We used proteomics approaches to define the EVI1 interactome. We found associations of EVI1 with not only transcriptional regulators, but also components of signaling pathways, DNA repair, DNA recombination, and mitosis complexes. We also identified functional EVI1 phosphorylation sites modified by casein-kinase II and protein phosphatase-1α that impact EVI1 activity. Thus, our study provides critical molecular insights on EVI1 action and regulation. Ecotropic viral integration site-1 (EVI1) is an oncogenic zinc finger transcription factor whose expression is frequently up-regulated in myeloid leukemia and epithelial cancers. To better understand the mechanisms underlying EVI1-associated disease, we sought to define the EVI1 interactome in cancer cells. By using stable isotope labeling by amino acids in cell culture (SILAC)-based quantitative proteomics, we could confidently assign 78 proteins as EVI1-interacting partners for FLAG-tagged EVI1. Subsequently, we showed that 22 of 27 tested interacting proteins could coimmunoprecipitate with endogenous EVI1 protein, which represented an 81.5% validation rate. Additionally, by comparing the stable isotope labeling by amino acids in cell culture (SILAC) data with high-throughput yeast two hybrid results, we showed that five of these proteins interacted directly with EVI1. Functional classification of EVI1-interacting proteins revealed associations with cellular transcription machinery; modulators of transcription; components of WNT, TGF-β, and RAS pathways; and proteins regulating DNA repair, recombination, and mitosis. We also identified EVI1 phosphorylation sites by MS analysis and showed that Ser538 and Ser858 can be phosphorylated and dephosphorylated by two EVI1 interactome proteins, casein kinase II and protein phosphatase-1α. Finally, mutations that impair EVI1 phosphorylation at these sites reduced EVI1 DNA binding through its C-terminal zinc finger domain and induced cancer cell proliferation. Collectively, these combinatorial proteomic approaches demonstrate that EVI1 interacts with large and complex networks of proteins, which integrate signals from various different signaling pathways important for oncogenesis. Comprehensive analysis of the EVI1 interactome has thus provided an important resource for dissecting the molecular mechanisms of EVI1-associated disease.

Collaboration


Dive into the Jayantha Gunaratne's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wee Joo Chng

National University of Singapore

View shared research outputs
Top Co-Authors

Avatar

Mariano A. Garcia-Blanco

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Alex M. Ward

National University of Singapore

View shared research outputs
Researchain Logo
Decentralizing Knowledge