KDM4A) Rescues 3P Haploinsufficiency

Total Page:16

File Type:pdf, Size:1020Kb

KDM4A) Rescues 3P Haploinsufficiency Inhibition of Lysine Demethylase 4A (KDM4A) Rescues 3p Haploinsufficiency Ruhee Dere, Ph.D. 14th International VHL Medical Symposium Oct 29, 2020 Renal Cell Carcinoma – Loss of 3p Tumor Suppressors Co-deletion of multiple tumor suppressors on chromosome 3p contribute to renal cell carcinoma (RCC) 90% of clear cell renal cell carcinoma (ccRCC) have mono-allelic loss of Hsieh J et al., JCO, 2018 the epigenetic regulator SETD2 HistoneHistone Code Code Vs. Vs. Tubulin Tubulin Code Courtesy Dr. Verhey Laurence et al., Trends in Genetics, 2016 SETD2 – A Dual Chromatin & Tubulin Methyltransferase α-tub α-tubK40me3 DAPI Merge Zoom Li F. et al., Cell, 2013 Park IY. et al., Cell, 2016 • SETD2 writes the tri-methyl mark on histone H3 lysine 36 (H3K36me3) SETD2 GST GST GST SETD2 SETD2 • In vitro methylation assays show SETD2 can α -Tubulin methylate α-tubulin • Mass spec identified K40 of α-tubulin as the site for trimethylation Histone • Methyl lysine antibody specific for K40me3 polyMT depolyMT Histones recognized a methyl mark on spindle microtubules Park IY. et al., Cell, 2016 • Loss of Setd2 results in multiple mitotic defects and genomic instability Existence of Dual-Function Epigenetic Regulators Nucleus Cytoplasm Erasers Writers Erasers KDM4A Altered cytoskeleton Writers ?? Cell proliferation Gene Expression SETD2 Genomic stability DNA repair ?? Chromatin structure Migration & metastasis me me me me K40 Readers K40 PBRM1 Readers ?? KDM4A is a Cytoplasmic Protein hTERT RPE-1 HKC MEFs WCE N C CD44 HA-KDM4A WCE N C N C N C KDM4A KDM4A KDM4A Lamin A/C Lamin A/C Lamin A/C LDH LDH LDH • KDM4A is the corresponding demethylase that Li. GM, Cancer Res, 2013 erases the methyl mark laid by SETD2 on chromatin (H3K36me3) • KDM4A is a dioxygenase enzyme requiring Fe, α- ketoglutarate as co-factors for its activity • KDM4A localizes in the cytoplasm KDM4A Binds Methylated Tubulin A RPE1 B C hTERT RPE-1 D HEK293T cells WCE • KDM4A binds α-tubulin • KDM4A binds methylated IP: KDM4A Fold enrichment tubulin over IgG control Mass Spec Analysis • Tubulin 1A (TUB1A) is highly enriched in KDM4A immunoprecipitates KDM4A Demethylates Tubulin A B • Recombinant KDM4A Substrate: Substrate: Recombinant H3K36me3 Purified Tubulin (bovine) demethylates recombinant Recombinant - + Recombinant KDM4A KDM4A - + microtubules H3K36me3 K40me3 • Overexpression of a catalytically H3 (input) Tubulin (input) dead KDM4A (H188A mutant) fails to decrease methylation on C D microtubules • Loss of KDM4A increases K40me3 signal at the mitotic spindle E KDM4A Localizes at the MTOCs • KDM4A localizes at the poles of the mitotic spindle • KDM4A is a centrosome-associated protein that colocalizes with γ- tubulin at the MTOCs during mitosis • Localization of KDM4A at MTOCs is independent of SETD2 methylation on microtubules KDM4A Opposes SETD2- Mediated Methylation on MTs If KDM4A opposes the methyl mark on MTs laid by SETD2, then inhibition of KDM4A should rescue genomic defects associated with SETD2 haploinsufficiency. Chiang et al., Cancer Res, 2018 Loss of KDM4A Rescues Genomic Instability Associated with Mono-allelic Loss of Setd2 Summary • KDM4A is a cytoplasmic protein that binds α-tubulin KDM4A demethylates Erasers • KDM4A microtubules Altered cytoskeleton Writers ?? Cell proliferation SETD2 • KDM4A localizes to the MTOCs Genomic stability ?? Migration & metastasis of the mitotic spindle me me K40 Readers K40 PBRM1 ?? Summary KDM4A activity can be inhibited using the small molecule JIB-04 JIB-04 disrupts O2 binding JIB-04 in the catalytic domain of KDM4A Cascella B., et al., 2017, Chem. Commun. Acknowledgements University of Michigan Kristen Verhey, Ph.D. U.T. Southwestern Elisabeth Martinez, Ph.D. Pratim Chowdhury Dr. Rahul Jangid Research Associate Post-doctoral Fellow Vanderbilt University W. Kimryn Rathmell, M.D., Ph.D. Frank Mason, Ph.D. Baylor College of Medicine Cheryl L. Walker, Ph.D. Nicolas Young, Ph.D. Funding Sung Jung, Ph.D. VHL Alliance Gilson Longenbaugh Foundation.
Recommended publications
  • Interplay Between Epigenetics and Metabolism in Oncogenesis: Mechanisms and Therapeutic Approaches
    OPEN Oncogene (2017) 36, 3359–3374 www.nature.com/onc REVIEW Interplay between epigenetics and metabolism in oncogenesis: mechanisms and therapeutic approaches CC Wong1, Y Qian2,3 and J Yu1 Epigenetic and metabolic alterations in cancer cells are highly intertwined. Oncogene-driven metabolic rewiring modifies the epigenetic landscape via modulating the activities of DNA and histone modification enzymes at the metabolite level. Conversely, epigenetic mechanisms regulate the expression of metabolic genes, thereby altering the metabolome. Epigenetic-metabolomic interplay has a critical role in tumourigenesis by coordinately sustaining cell proliferation, metastasis and pluripotency. Understanding the link between epigenetics and metabolism could unravel novel molecular targets, whose intervention may lead to improvements in cancer treatment. In this review, we summarized the recent discoveries linking epigenetics and metabolism and their underlying roles in tumorigenesis; and highlighted the promising molecular targets, with an update on the development of small molecule or biologic inhibitors against these abnormalities in cancer. Oncogene (2017) 36, 3359–3374; doi:10.1038/onc.2016.485; published online 16 January 2017 INTRODUCTION metabolic genes have also been identified as driver genes It has been appreciated since the early days of cancer research mutated in some cancers, such as isocitrate dehydrogenase 1 16 17 that the metabolic profiles of tumor cells differ significantly from and 2 (IDH1/2) in gliomas and acute myeloid leukemia (AML), 18 normal cells. Cancer cells have high metabolic demands and they succinate dehydrogenase (SDH) in paragangliomas and fuma- utilize nutrients with an altered metabolic program to support rate hydratase (FH) in hereditary leiomyomatosis and renal cell 19 their high proliferative rates and adapt to the hostile tumor cancer (HLRCC).
    [Show full text]
  • Catalysis by the Jmjc Histone Demethylase KDM4A Integrates Substrate Dynamics, Correlated Motions Cite This: Chem
    Chemical Science View Article Online EDGE ARTICLE View Journal | View Issue Catalysis by the JmjC histone demethylase KDM4A integrates substrate dynamics, correlated motions Cite this: Chem. Sci., 2020, 11, 9950 † All publication charges for this article and molecular orbital control have been paid for by the Royal Society a a b of Chemistry Rajeev Ramanan, Shobhit S. Chaturvedi, Nicolai Lehnert, Christopher J. Schofield, c Tatyana G. Karabencheva-Christova *a and Christo Z. Christov *a 3 The N -methyl lysine status of histones is important in the regulation of eukaryotic transcription. The Fe(II) and 2-oxoglutarate (2OG) -dependent JmjC domain enzymes are the largest family of histone N3-methyl lysine demethylases (KDMs). The human KDM4 subfamily of JmjC KDMs is linked with multiple cancers and some of its members are medicinal chemistry targets. We describe the use of combined molecular dynamics (MD) and Quantum Mechanical/Molecular Mechanical (QM/MM) methods to study the mechanism of KDM4A, which catalyzes demethylation of both tri- and di-methylated forms of histone Creative Commons Attribution-NonCommercial 3.0 Unported Licence. H3 at K9 and K36. The results show that the oxygen activation at the active site of KDM4A is optimized towards the generation of the reactive Fe(IV)-oxo intermediate. Factors including the substrate binding mode, correlated motions of the protein and histone substrates, and molecular orbital control synergistically contribute to the reactivity of the Fe(IV)-oxo intermediate. In silico substitutions were performed to investigate the roles of residues (Lys241, Tyr177, and Asn290) in substrate orientation. The Lys241Ala substitution abolishes activity due to altered substrate orientation consistent with reported experimental studies.
    [Show full text]
  • Mutant IDH, (R)-2-Hydroxyglutarate, and Cancer
    Downloaded from genesdev.cshlp.org on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press REVIEW What a difference a hydroxyl makes: mutant IDH, (R)-2-hydroxyglutarate, and cancer Julie-Aurore Losman1 and William G. Kaelin Jr.1,2,3 1Department of Medical Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02215, USA; 2Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA Mutations in metabolic enzymes, including isocitrate whether altered cellular metabolism is a cause of cancer dehydrogenase 1 (IDH1) and IDH2, in cancer strongly or merely an adaptive response of cancer cells in the face implicate altered metabolism in tumorigenesis. IDH1 of accelerated cell proliferation is still a topic of some and IDH2 catalyze the interconversion of isocitrate and debate. 2-oxoglutarate (2OG). 2OG is a TCA cycle intermediate The recent identification of cancer-associated muta- and an essential cofactor for many enzymes, including tions in three metabolic enzymes suggests that altered JmjC domain-containing histone demethylases, TET cellular metabolism can indeed be a cause of some 5-methylcytosine hydroxylases, and EglN prolyl-4-hydrox- cancers (Pollard et al. 2003; King et al. 2006; Raimundo ylases. Cancer-associated IDH mutations alter the enzymes et al. 2011). Two of these enzymes, fumarate hydratase such that they reduce 2OG to the structurally similar (FH) and succinate dehydrogenase (SDH), are bone fide metabolite (R)-2-hydroxyglutarate [(R)-2HG]. Here we tumor suppressors, and loss-of-function mutations in FH review what is known about the molecular mechanisms and SDH have been identified in various cancers, in- of transformation by mutant IDH and discuss their im- cluding renal cell carcinomas and paragangliomas.
    [Show full text]
  • 1714 Gene Comprehensive Cancer Panel Enriched for Clinically Actionable Genes with Additional Biologically Relevant Genes 400-500X Average Coverage on Tumor
    xO GENE PANEL 1714 gene comprehensive cancer panel enriched for clinically actionable genes with additional biologically relevant genes 400-500x average coverage on tumor Genes A-C Genes D-F Genes G-I Genes J-L AATK ATAD2B BTG1 CDH7 CREM DACH1 EPHA1 FES G6PC3 HGF IL18RAP JADE1 LMO1 ABCA1 ATF1 BTG2 CDK1 CRHR1 DACH2 EPHA2 FEV G6PD HIF1A IL1R1 JAK1 LMO2 ABCB1 ATM BTG3 CDK10 CRK DAXX EPHA3 FGF1 GAB1 HIF1AN IL1R2 JAK2 LMO7 ABCB11 ATR BTK CDK11A CRKL DBH EPHA4 FGF10 GAB2 HIST1H1E IL1RAP JAK3 LMTK2 ABCB4 ATRX BTRC CDK11B CRLF2 DCC EPHA5 FGF11 GABPA HIST1H3B IL20RA JARID2 LMTK3 ABCC1 AURKA BUB1 CDK12 CRTC1 DCUN1D1 EPHA6 FGF12 GALNT12 HIST1H4E IL20RB JAZF1 LPHN2 ABCC2 AURKB BUB1B CDK13 CRTC2 DCUN1D2 EPHA7 FGF13 GATA1 HLA-A IL21R JMJD1C LPHN3 ABCG1 AURKC BUB3 CDK14 CRTC3 DDB2 EPHA8 FGF14 GATA2 HLA-B IL22RA1 JMJD4 LPP ABCG2 AXIN1 C11orf30 CDK15 CSF1 DDIT3 EPHB1 FGF16 GATA3 HLF IL22RA2 JMJD6 LRP1B ABI1 AXIN2 CACNA1C CDK16 CSF1R DDR1 EPHB2 FGF17 GATA5 HLTF IL23R JMJD7 LRP5 ABL1 AXL CACNA1S CDK17 CSF2RA DDR2 EPHB3 FGF18 GATA6 HMGA1 IL2RA JMJD8 LRP6 ABL2 B2M CACNB2 CDK18 CSF2RB DDX3X EPHB4 FGF19 GDNF HMGA2 IL2RB JUN LRRK2 ACE BABAM1 CADM2 CDK19 CSF3R DDX5 EPHB6 FGF2 GFI1 HMGCR IL2RG JUNB LSM1 ACSL6 BACH1 CALR CDK2 CSK DDX6 EPOR FGF20 GFI1B HNF1A IL3 JUND LTK ACTA2 BACH2 CAMTA1 CDK20 CSNK1D DEK ERBB2 FGF21 GFRA4 HNF1B IL3RA JUP LYL1 ACTC1 BAG4 CAPRIN2 CDK3 CSNK1E DHFR ERBB3 FGF22 GGCX HNRNPA3 IL4R KAT2A LYN ACVR1 BAI3 CARD10 CDK4 CTCF DHH ERBB4 FGF23 GHR HOXA10 IL5RA KAT2B LZTR1 ACVR1B BAP1 CARD11 CDK5 CTCFL DIAPH1 ERCC1 FGF3 GID4 HOXA11 IL6R KAT5 ACVR2A
    [Show full text]
  • Maternal Expression of the Histone Demethylase Kdm4a Is Crucial For
    © 2017. Published by The Company of Biologists Ltd | Development (2017) 144, 3264-3277 doi:10.1242/dev.155473 RESEARCH ARTICLE Maternal expression of the histone demethylase Kdm4a is crucial for pre-implantation development Aditya Sankar1,2,3,*,§, Susanne Marije Kooistra1,2,‡,§, Javier Martin Gonzalez4, Claes Ohlsson6, Matti Poutanen5,6 and Kristian Helin1,2,3,¶ ABSTRACT residue, as well as type and degree of modification (Greer and Shi, Regulation of chromatin composition through post-translational 2012; Kooistra and Helin, 2012; Dimitrova et al., 2015). modifications of histones contributes to transcriptional regulation Similar to the other members of the JMJD2/KDM4 family of and is essential for many cellular processes, including differentiation histone demethylases, KDM4A is capable of removing di- and tri- and development. KDM4A (JMJD2A) is a lysine demethylase with methylation from lysines 9 and 36 of histone H3 (Cloos et al., 2006; specificity towards di- and tri-methylated lysine 9 and lysine 36 of Klose et al., 2006; Whetstine et al., 2006; Chen et al., 2006, 2007; histone H3 (H3K9me2/me3 and H3K36me2/me3). Here, we report Ng et al., 2007). KDM4A, like KDM4C, binds to tri-methylated that Kdm4a as a maternal factor plays a key role in embryo survival lysine 4 on histone H3 (H3K4me3) through its double Tudor and is vital for female fertility. Kdm4a−/− female mice ovulate normally domain (Huang et al., 2006; Pedersen et al., 2016, 2014). In this with comparable fertilization but poor implantation rates, and cannot way, the KDM4 proteins provide a failsafe mechanism to prevent support healthy transplanted embryos to term. This is due to a role for spurious accumulation of H3K9me3 at active promotors, thereby Kdm4a in uterine function, where its loss causes reduced expression maintaining transcriptional competence (Pedersen et al., 2016).
    [Show full text]
  • Electronic Supplementary Material (ESI) for Metallomics
    Electronic Supplementary Material (ESI) for Metallomics. This journal is © The Royal Society of Chemistry 2018 Uniprot Entry name Gene names Protein names Predicted Pattern Number of Iron role EC number Subcellular Membrane Involvement in disease Gene ontology (biological process) Id iron ions location associated 1 P46952 3HAO_HUMAN HAAO 3-hydroxyanthranilate 3,4- H47-E53-H91 1 Fe cation Catalytic 1.13.11.6 Cytoplasm No NAD biosynthetic process [GO:0009435]; neuron cellular homeostasis dioxygenase (EC 1.13.11.6) (3- [GO:0070050]; quinolinate biosynthetic process [GO:0019805]; response to hydroxyanthranilate oxygenase) cadmium ion [GO:0046686]; response to zinc ion [GO:0010043]; tryptophan (3-HAO) (3-hydroxyanthranilic catabolic process [GO:0006569] acid dioxygenase) (HAD) 2 O00767 ACOD_HUMAN SCD Acyl-CoA desaturase (EC H120-H125-H157-H161; 2 Fe cations Catalytic 1.14.19.1 Endoplasmic Yes long-chain fatty-acyl-CoA biosynthetic process [GO:0035338]; unsaturated fatty 1.14.19.1) (Delta(9)-desaturase) H160-H269-H298-H302 reticulum acid biosynthetic process [GO:0006636] (Delta-9 desaturase) (Fatty acid desaturase) (Stearoyl-CoA desaturase) (hSCD1) 3 Q6ZNF0 ACP7_HUMAN ACP7 PAPL PAPL1 Acid phosphatase type 7 (EC D141-D170-Y173-H335 1 Fe cation Catalytic 3.1.3.2 Extracellular No 3.1.3.2) (Purple acid space phosphatase long form) 4 Q96SZ5 AEDO_HUMAN ADO C10orf22 2-aminoethanethiol dioxygenase H112-H114-H193 1 Fe cation Catalytic 1.13.11.19 Unknown No oxidation-reduction process [GO:0055114]; sulfur amino acid catabolic process (EC 1.13.11.19) (Cysteamine
    [Show full text]
  • Figure S1. Reverse Transcription‑Quantitative PCR Analysis of ETV5 Mrna Expression Levels in Parental and ETV5 Stable Transfectants
    Figure S1. Reverse transcription‑quantitative PCR analysis of ETV5 mRNA expression levels in parental and ETV5 stable transfectants. (A) Hec1a and Hec1a‑ETV5 EC cell lines; (B) Ishikawa and Ishikawa‑ETV5 EC cell lines. **P<0.005, unpaired Student's t‑test. EC, endometrial cancer; ETV5, ETS variant transcription factor 5. Figure S2. Survival analysis of sample clusters 1‑4. Kaplan Meier graphs for (A) recurrence‑free and (B) overall survival. Survival curves were constructed using the Kaplan‑Meier method, and differences between sample cluster curves were analyzed by log‑rank test. Figure S3. ROC analysis of hub genes. For each gene, ROC curve (left) and mRNA expression levels (right) in control (n=35) and tumor (n=545) samples from The Cancer Genome Atlas Uterine Corpus Endometrioid Cancer cohort are shown. mRNA levels are expressed as Log2(x+1), where ‘x’ is the RSEM normalized expression value. ROC, receiver operating characteristic. Table SI. Clinicopathological characteristics of the GSE17025 dataset. Characteristic n % Atrophic endometrium 12 (postmenopausal) (Control group) Tumor stage I 91 100 Histology Endometrioid adenocarcinoma 79 86.81 Papillary serous 12 13.19 Histological grade Grade 1 30 32.97 Grade 2 36 39.56 Grade 3 25 27.47 Myometrial invasiona Superficial (<50%) 67 74.44 Deep (>50%) 23 25.56 aMyometrial invasion information was available for 90 of 91 tumor samples. Table SII. Clinicopathological characteristics of The Cancer Genome Atlas Uterine Corpus Endometrioid Cancer dataset. Characteristic n % Solid tissue normal 16 Tumor samples Stagea I 226 68.278 II 19 5.740 III 70 21.148 IV 16 4.834 Histology Endometrioid 271 81.381 Mixed 10 3.003 Serous 52 15.616 Histological grade Grade 1 78 23.423 Grade 2 91 27.327 Grade 3 164 49.249 Molecular subtypeb POLE 17 7.328 MSI 65 28.017 CN Low 90 38.793 CN High 60 25.862 CN, copy number; MSI, microsatellite instability; POLE, DNA polymerase ε.
    [Show full text]
  • Active Motif Technical Data Sheet (TDS)
    Recombinant JMJD2A / KDM4A protein Catalog No: 31457, 31857 Quantity: 20, 1000 µg Expressed In: Baculovirus Concentration: 0.6 µg/µl Source: Human Buffer Contents: Full length recombinant JMJD2A / KDM4A protein was expressed in Sf9 cells and is supplied in 25 mM Hepes pH 7.5, 300 mM NaCl, 5% Glycerol, 0.04% Triton X-100, 0.2 mM TCEP. Background: KDM4A (lysine (K)-specific demethylase 4A), also known as JMJD2A (Jumonji Domain Containing 2A) is a nuclear protein that functions as a histone demethylase that preferentially demethylates di- and trimethylated lysine 9 (K9me3) and lysine 36 (K36me3) residues of histone H3, converting these trimethylated histone residues to mono- or dimethylated form. JMJD2A / KDM4A has no activity for monomethylated H3K9 and H3K36. KDM4A functions as a transcriptional repressor, participating in transcriptional repression of ASCL2 and E2F-responsive promoters via the recruitment of histone deacetylases and NCOR1, respectively. An additional KDM4A isoform, KDM4A Isoform 2, that lacks the N-terminal demethylase domain is crucial for muscle differentiation in promoting transcriptional activation of the MyoG gene by directing the removal of repressive chromatin marks at its promoter. Protein Details: Recombinant JMJD2A / KDM4A (accession number NP_055478.2) was expressed in Sf9 cells and contains an N-terminal FLAG-Tag with an observed molecular weight of 125.3 kDa. The recombinant protein is >85% pure by SDS-PAGE. Application Notes: Recombinant JMJD2A / KDM4A is suitable for use in the study of enzyme kinetics, inhibitor screening, and selectivity profiling. Histone Demethylase Assay Conditions: 50 mM HEPES pH 7.5, 0.02% Triton X100, 100 μM 2OG, 100 μM Ascorbate, 50 μM (NH4)2Fe(SO4)2·6H2O, 1 mM TCEP, 100 nM Recombinant JMJD2A / KDM4A protein, and 3.3 μM H3K9me3 (aa 1-21) peptide at 2 hours at room temperature.
    [Show full text]
  • NIH Public Access Author Manuscript Curr Cancer Drug Targets
    NIH Public Access Author Manuscript Curr Cancer Drug Targets. Author manuscript; available in PMC 2013 July 06. NIH-PA Author ManuscriptPublished NIH-PA Author Manuscript in final edited NIH-PA Author Manuscript form as: Curr Cancer Drug Targets. 2013 June 10; 13(5): 558–579. Histone Lysine-Specific Methyltransferases and Demethylases in Carcinogenesis: New Targets for Cancer Therapy and Prevention Xuejiao Tian1,2,3, Saiyang Zhang4, Hong-Min Liu4, Yan-Bing Zhang4, Christopher A Blair1,2, Dan Mercola5, Paolo Sassone-Corsi6, and Xiaolin Zi1,2,3,* 1Department of Urology, University of California, Irvine, Orange, CA 92868, USA 2Department of Pharmaceutical Sciences, University of California, Irvine, Orange, CA 92868, USA 3Department of Pharmacology, University of California, Irvine, Orange, CA 92868, USA 4School of Pharmaceutical Sciences, Zhengzhou University, No.100, Science Avenue, Zhengzhou 450001, Henan, China 5Department of Pathology and Laboratory medicine, University of California, Irvine, Orange, CA 92868, USA 6Department of Biological Chemistry, University of California, Irvine, Orange, CA 92868, USA Abstract Aberrant histone lysine methylation that is controlled by histone lysine methyltransferases (KMTs) and demethylases (KDMs) plays significant roles in carcinogenesis. Infections by tumor viruses or parasites and exposures to chemical carcinogens can modify the process of histone lysine methylation. Many KMTs and KDMs contribute to malignant transformation by regulating the expression of human telomerase reverse transcriptase (hTERT), forming a fused gene, interacting with proto-oncogenes or being up-regulated in cancer cells. In addition, histone lysine methylation participates in tumor suppressor gene inactivation during the early stages of carcinogenesis by regulating DNA methylation and/or by other DNA methylation independent mechanisms.
    [Show full text]
  • Epigenetic Changes in Fibroblasts Drive Cancer Metabolism and Differentiation
    26 12 Endocrine-Related R Mishra et al. TME based epigenetic targets 26:12 R673–R688 Cancer in cancer REVIEW Epigenetic changes in fibroblasts drive cancer metabolism and differentiation Rajeev Mishra1, Subhash Haldar2, Surabhi Suchanti1 and Neil A Bhowmick3,4 1Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan, India 2Department of Biotechnology, Brainware University, Kolkata, India 3Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA 4Department of Research, Greater Los Angeles Veterans Administration, Los Angeles, California, USA Correspondence should be addressed to N A Bhowmick: [email protected] Abstract Genomic changes that drive cancer initiation and progression contribute to the Key Words co-evolution of the adjacent stroma. The nature of the stromal reprogramming involves f endocrine therapy differential DNA methylation patterns and levels that change in response to the tumor resistance and systemic therapeutic intervention. Epigenetic reprogramming in carcinoma- f prostate associated fibroblasts are robust biomarkers for cancer progression and have a f neuroendocrine tumors transcriptional impact that support cancer epithelial progression in a paracrine manner. For prostate cancer, promoter hypermethylation and silencing of the RasGAP, RASAL3 that resulted in the activation of Ras signaling in carcinoma-associated fibroblasts. Stromal Ras activity initiated a process of macropinocytosis that provided prostate cancer epithelia with abundant glutamine for metabolic conversion
    [Show full text]
  • The Biological Axis of Protein Arginine Methylation and Asymmetric Dimethylarginine
    International Journal of Molecular Sciences Review The Biological Axis of Protein Arginine Methylation and Asymmetric Dimethylarginine Melody D. Fulton , Tyler Brown and Y. George Zheng * Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA * Correspondence: [email protected]; Tel.: +1-706-542-0277 Received: 17 June 2019; Accepted: 4 July 2019; Published: 6 July 2019 Abstract: Protein post-translational modifications (PTMs) in eukaryotic cells play important roles in the regulation of functionalities of the proteome and in the tempo-spatial control of cellular processes. Most PTMs enact their regulatory functions by affecting the biochemical properties of substrate proteins such as altering structural conformation, protein–protein interaction, and protein–nucleic acid interaction. Amid various PTMs, arginine methylation is widespread in all eukaryotic organisms, from yeasts to humans. Arginine methylation in many situations can drastically or subtly affect the interactions of substrate proteins with their partnering proteins or nucleic acids, thus impacting major cellular programs. Recently, arginine methylation has become an important regulator of the formation of membrane-less organelles inside cells, a phenomenon of liquid–liquid phase separation (LLPS), through altering π-cation interactions. Another unique feature of arginine methylation lies in its impact on cellular physiology through its downstream amino acid product, asymmetric dimethylarginine (ADMA). Accumulation of ADMA in cells and in the circulating bloodstream is connected with endothelial dysfunction and a variety of syndromes of cardiovascular diseases. Herein, we review the current knowledge and understanding of protein arginine methylation in regards to its canonical function in direct protein regulation, as well as the biological axis of protein arginine methylation and ADMA biology.
    [Show full text]
  • Preconceptional Paternal Glycidamide Exposure Affects Embryonic Gene
    Reproductive Toxicology 32 (2011) 463–471 Contents lists available at SciVerse ScienceDirect Reproductive Toxicology jo urnal homepage: www.elsevier.com/locate/reprotox Preconceptional paternal glycidamide exposure affects embryonic gene expression: Single embryo gene expression study following in vitro fertilization a,∗ b a a a Asgeir Brevik , Vendula Rusnakova , Nur Duale , Hege Holte Slagsvold , Ann-Karin Olsen , c d a a Ritsa Storeng , Mikael Kubista , Gunnar Brunborg , Birgitte Lindeman a Norwegian Institute of Public Health, Division of Environmental Medicine, Department of Chemical Toxicology, P.O. Box 4404, Nydalen, N-0403 Oslo, Norway b Dept. of Gene Expression, Institute of Biotechnology AS CR, v.v.i., Videnska 1083, Prague 4, 142 20, Czech Republic c Oslo University Hospital - Rikshospitalet, 0027 Oslo, Norway d Tataa Biocenter, Odinsgatan 28, 411 03 Göteborg, Sweden a r t i c l e i n f o a b s t r a c t Article history: Recognition of early determinants of disease onset has sparked an interest in paternally transmitted Received 2 March 2011 factors and their impact on the developing embryo. Acrylamide (AA), a widely distributed xenobiotic Received in revised form 10 August 2011 compound, is converted to its active metabolite glycidamide (GA) by the CYP2E1 enzyme. Based on its Accepted 17 September 2011 capacity to induce dominant lethal mutations, we hypothesized that paternal GA exposure would have Available online 29 September 2011 a negative impact on embryonic genome activation, via GA-DNA and protamine adducts persisting in the fertilizing sperm. Using a combination of in vitro fertilization (IVF) techniques and RT-qPCR single Keywords: embryo gene expression (SEGE), we studied the expression of key DNA repair genes and genes important Single-cell gene expression Glycidamide for embryo development, at the 1-, 2-, 4- and 8-cell stage of the developing mouse embryo.
    [Show full text]