Supplementary Table 2. the Major Known Families of Demethylases++

Total Page:16

File Type:pdf, Size:1020Kb

Supplementary Table 2. the Major Known Families of Demethylases++ Supplementary Table 2. The Major Known Families of Demethylases++ and Related Pathology Mark Family Enzyme Synonyms Substrates Associated pathology and references removed KDM1 KDM1A LSD1, AOF2, Kme2/1 Histone (H3K4, H3K9), p53, Cancer development in bladder, lung, BHC110 DNMT1, STAT3, MYPT1, colon1 and breast2 E2F1 KDM1B LSD2, AOF1 Histone (H3K4, H3K9) Deletion leads to depletion of target gene transcription3 KDM2 KDM2A FBXL11, JHDM1A Kme2/1 (H3K36), p65 Maintain heterochromatin state in prostate cancer4 5 KDM2B FBXL10, JHDM1B Histone (H3K36) Regulate pancreatic cancer and senescence6 7 KDM3 KDM3A JMJD1A, JHDM2A, Kme2/1 Histone (H3K9) Co-activator of AR-mediated transcription, TSGA Regulated by HIF1α and enhances tumor growth under hypoxic condition8 KDM3B JMJD1B Frequent deletion of 5q31 (JMJD1B) in myeloid leukemia9 10 KDM4 KDM4A JMJD2A, JHDM3A Kme3/2/1 Histone (H3K9, H3K36, Overexpressed in prostate cancer H1.4K26) KDM4B JMJD2B, JHDM3B Cofactor of ER in breast cancer and mammary gland development11 KDM4C JMJD2C, JHDM3C, Overexpressed in esophageal squamous GASC1 cell carcinoma12 and breast cancer13 14 KDM4D JMJD2D, JHDM3D Histone (H3K9, H1.4K26) Activates AR in prostate cancer 15 KDM5 KDM5A JARID1A, RBP2 Kme3/2/1 Histone (H3K4) Overexpressed in gastric cancer and in the drug-tolerant subpopulation of cancer cells16 KDM5B JARID1B, PLU1 Carcinogenesis in prostate,17 lung,18 breast19 and melanoma20 KDM5C JARID1C, SMCX Regulation of oncogenic HPV expression21 KDM5D JARID1D, SMCY Deleted in prostate cancer22 23 KDM6 KDM6A UTX Kme3/2/1 Histone (H3K27) Mutated in human cancers and enables pRB-mediated cell fate control24 KDM6B JMJD3 Promotes EMT25 and activation of INK4A/ARF locus in response to Onc and senescence26 27 KDM7 KDM7A KIAA1718, Kme2/1 Histone (H3K9, H3K27) Downregulate angiogenesis in cancer cells JHDM1D 28 PHF2 JHDM1E Histone (H3K9), ARID5B Alteration in breast carcinoma PHF8 JHDM1F Histone (H3K9, H4K20) Associated with cleft lip/palate and mental retardation29 KDM8 KDM8* JMJD5 Kme2/1 Histone (H3K36), (NFATc1 Overexpressed in various cancers and hydroxylation proposed) regulate cancer cell proliferation30 JMJD6* PSR, PTDSR Rme2/1 Histone (H4R3, H3R2), Drive cell proliferation and motility in splicing protein breast cancer cells, marker of poor lysyl-hydroxylation prognosis31 32 NO66 Kme3/2 Histone (H3K4, H3K36), Overexpressed in lung carcinomas (hydroxylation proposed) 33 LOXL2 Kme3 Histone (H3K4) Induced by hypoxia and repress E-cadherin PADI PADI4 PAD4 Rme1 Histone (multiple arginines Ectopic expression leads to inhibition of 34 on H3 and H4), p300 tumor growth in a p53 dependent manner PME-1 PPME1, PME1 -CO2me PP2A Overexpressed in malignant glioblastoma (leucyl) and protects ERK from PP2A35 ++ Modified from Curr Opin Chem Biol 2012;16:525-34. 2010;138:981-92. REFERENCES FOR SUPPLEMENTARY 16. Sharma SV, Lee DY, Li B, Quinlan MP, Takahashi F, Maheswaran TABLE 2 S, et al. A chromatin-mediated reversible drug-tolerant state in cancer cell subpopulations. Cell 2010;141:69-80. 17. Xiang Y, Zhu Z, Han G, Ye X, Xu B, Peng Z, et al. JARID1B is a 1. Hayami S, Kelly JD, Cho HS, Yoshimatsu M, Unoki M, Tsunoda histone H3 lysine 4 demethylase up-regulated in prostate cancer. T, et al. Overexpression of LSD1 contributes to human carcino- Proc Natl Acad Sci U S A 2007;104:19226-31. genesis through chromatin regulation in various cancers. Int J 18. Hayami S, Yoshimatsu M, Veerakumarasivam A, Unoki M, Iwai Cancer 2011;128:574-86. Y, Tsunoda T, et al. Overexpression of the JmjC histone demethyl- 2. Lim S, Janzer A, Becker A, Zimmer A, Schüle R, Buettner R, et ase KDM5B in human carcinogenesis: involvement in the prolif- al. Lysine-specific demethylase 1 (LSD1) is highly expressed in eration of cancer cells through the E2F/RB pathway. Mol Cancer ER-negative breast cancers and a biomarker predicting aggressive 2010;9:59. biology. Carcinogenesis 2010;31:512-20. 19. Barrett A, Madsen B, Copier J, Lu PJ, Cooper L, Scibetta AG, et 3. Fang R, Barbera AJ, Xu Y, Rutenberg M, Leonor T, Bi Q, et al. al. PLU-1 nuclear protein, which is upregulated in breast cancer, Human LSD2/KDM1b/AOF1 regulates gene transcription by shows restricted expression in normal human adult tissues: a new modulating intragenic H3K4me2 methylation. Mol Cell 2010; cancer/testis antigen? Int J Cancer 2002;101:581-8. 39:222-33. 20. Roesch A, Fukunaga-Kalabis M, Schmidt EC, Zabierowski SE, 4. Frescas D, Guardavaccaro D, Kuchay SM, Kato H, Poleshko A, Brafford PA, Vultur A, et al. A temporarily distinct subpopulation Basrur V, et al. KDM2A represses transcription of centromeric of slow-cycling melanoma cells is required for continuous tumor satellite repeats and maintains the heterochromatic state. Cell Cy- growth. Cell 2010;141:583-94. cle 2008;7:3539-47. 21. Smith JA, White EA, Sowa ME, Powell ML, Ottinger M, Harper 5. Tzatsos A, Paskaleva P, Ferrari F, Deshpande V, Stoykova S, Con- JW, et al. Genome-wide siRNA screen identifies SMCX, EP400, tino G, et al. KDM2B promotes pancreatic cancer via Polycomb- and Brd4 as E2-dependent regulators of human papillomavirus dependent and -independent transcriptional programs. J Clin In- oncogene expression. Proc Natl Acad Sci U S A 2010;107:3752-7. vest 2013;123:727-39. 22. Perinchery G, Sasaki M, Angan A, Kumar V, Carroll P, Dahiya R. 6. He J, Kallin EM, Tsukada Y, Zhang Y. The H3K36 demethylase Deletion of Y-chromosome specific genes in human prostate can- Jhdm1b/Kdm2b regulates cell proliferation and senescence cer. J Urol 2000;163:1339-42. through p15(Ink4b). Nat Struct Mol Biol 2008;15:1169-75. 23. van Haaften G, Dalgliesh GL, Davies H, Chen L, Bignell G, 7. Yamane K, Toumazou C, Tsukada Y, Erdjument-Bromage H, Greenman C, et al. Somatic mutations of the histone H3K27 de- Tempst P, Wong J, et al. JHDM2A, a JmjC-containing H3K9 de- methylase gene UTX in human cancer. Nat Genet 2009;41:521-3. methylase, facilitates transcription activation by androgen recep- 24. Wang JK, Tsai MC, Poulin G, Adler AS, Chen S, Liu H, et al. The tor. Cell 2006;125:483-95. histone demethylase UTX enables RB-dependent cell fate control. 8. Krieg AJ, Rankin EB, Chan D, Razorenova O, Fernandez S, Giac- Genes Dev 2010;24:327-32. cia AJ. Regulation of the histone demethylase JMJD1A by hypox- 25. Ramadoss S, Chen X, Wang CY. Histone demethylase KDM6B ia-inducible factor 1 alpha enhances hypoxic gene expression and promotes epithelial-mesenchymal transition. J Biol Chem 2012; tumor growth. Mol Cell Biol 2010;30:344-53. 287:44508-17. 9. Hu Z, Gomes I, Horrigan SK, Kravarusic J, Mar B, Arbieva Z, et 26. Agger K, Cloos PA, Rudkjaer L, Williams K, Andersen G, Chris- al. A novel nuclear protein, 5qNCA (LOC51780) is a candidate tensen J, et al. The H3K27me3 demethylase JMJD3 contributes to for the myeloid leukemia tumor suppressor gene on chromosome the activation of the INK4A-ARF locus in response to oncogene- 5 band q31. Oncogene 2001;20:6946-54. and stress-induced senescence. Genes Dev 2009;23:1171-6. 10. Bok RA, Small EJ. Bloodborne biomolecular markers in prostate 27. Osawa T, Muramatsu M, Wang F, Tsuchida R, Kodama T, Minami cancer development and progression. Nat Rev Cancer 2002;2:918- T, et al. Increased expression of histone demethylase JHDM1D 26. under nutrient starvation suppresses tumor growth via down-regu- 11. Kawazu M, Saso K, Tong KI, McQuire T, Goto K, Son DO, et al. lating angiogenesis. Proc Natl Acad Sci U S A 2011;108:20725-9. Histone demethylase JMJD2B functions as a co-factor of estrogen 28. Sinha S, Singh RK, Alam N, Roy A, Roychoudhury S, Panda CK. receptor in breast cancer proliferation and mammary gland devel- Alterations in candidate genes PHF2, FANCC, PTCH1 and XPA opment. PLoS One 2011;6:e17830. at chromosomal 9q22.3 region: pathological significance in early- 12. Cloos PA, Christensen J, Agger K, Maiolica A, Rappsilber J, Antal and late-onset breast carcinoma. Mol Cancer 2008;7:84. T, et al. The putative oncogene GASC1 demethylates tri- and di- 29. Loenarz C, Ge W, Coleman ML, Rose NR, Cooper CD, Klose RJ, methylated lysine 9 on histone H3. Nature 2006;442:307-11. et al. PHF8, a gene associated with cleft lip/palate and mental re- 13. Liu G, Bollig-Fischer A, Kreike B, van de Vijver MJ, Abrams J, tardation, encodes for an Nepsilon-dimethyl lysine demethylase. Ethier SP, et al. Genomic amplification and oncogenic properties Hum Mol Genet 2010;19:217-22. of the GASC1 histone demethylase gene in breast cancer. Onco- 30. Hsia DA, Tepper CG, Pochampalli MR, Hsia EY, Izumiya C, gene 2009;28:4491-500. Huerta SB, et al. KDM8, a H3K36me2 histone demethylase that 14. Shin S, Janknecht R. Activation of androgen receptor by histone acts in the cyclin A1 coding region to regulate cancer cell prolifer- demethylases JMJD2A and JMJD2D. Biochem Biophys Res ation. Proc Natl Acad Sci U S A 2010;107:9671-6. Commun 2007;359:742-6. 31. Lee YF, Miller LD, Chan XB, Black MA, Pang B, Ong CW, et al. 15. Zeng J, Ge Z, Wang L, Li Q, Wang N, Björkholm M, et al. The JMJD6 is a driver of cellular proliferation and motility and a histone demethylase RBP2 Is overexpressed in gastric cancer and marker of poor prognosis in breast cancer. Breast Cancer Res its inhibition triggers senescence of cancer cells. Gastroenterology 2012;14:R85. 32. Suzuki C, Takahashi K, Hayama S, Ishikawa N, Kato T, Ito T, et 34. Tanikawa C, Ueda K, Nakagawa H, Yoshida N, Nakamura Y, al. Identification of Myc-associated protein with JmjC domain as Matsuda K. Regulation of protein Citrullination through p53/ a novel therapeutic target oncogene for lung cancer.
Recommended publications
  • KDM5B Promotes Drug Resistance by Regulating Melanoma-Propagating Cell Subpopulations Xiaoni Liu1,2, Shang-Min Zhang1, Meaghan K
    Published OnlineFirst December 6, 2018; DOI: 10.1158/1535-7163.MCT-18-0395 Cancer Biology and Translational Studies Molecular Cancer Therapeutics KDM5B Promotes Drug Resistance by Regulating Melanoma-Propagating Cell Subpopulations Xiaoni Liu1,2, Shang-Min Zhang1, Meaghan K. McGeary1,2, Irina Krykbaeva1,2, Ling Lai3, Daniel J. Jansen4, Stephen C. Kales4, Anton Simeonov4, Matthew D. Hall4, Daniel P. Kelly3, Marcus W. Bosenberg1,2,5, and Qin Yan1 Abstract Tumor heterogeneity is a major challenge for cancer elevated KDM5B expression, melanoma cells shift toward a À treatment, especially due to the presence of various sub- more drug-tolerant, CD34 state upon exposure to BRAF populations with stem cell or progenitor cell properties. inhibitor or combined BRAF inhibitor and MEK inhibitor þ À þ In mouse melanomas, both CD34 p75 (CD34 )and treatment. KDM5B loss or inhibition shifts melanoma cells À À À þ CD34 p75 (CD34 ) tumor subpopulations were charac- to the more BRAF inhibitor–sensitive CD34 state. These terized as melanoma-propagating cells (MPC) that exhibit results support that KDM5B is a critical epigenetic regulator some of those key features. However, these two subpopula- that governs the transition of key MPC subpopulations tions differ from each other in tumorigenic potential, with distinct drug sensitivity. This study also emphasizes ability to recapitulate heterogeneity, and chemoresistance. the importance of continuing to advance our understand- þ À In this study, we demonstrate that CD34 and CD34 ing of intratumor heterogeneity and ultimately develop V600E subpopulations carrying the BRAF mutation confer novel therapeutics by altering the heterogeneous character- differential sensitivity to targeted BRAF inhibition.
    [Show full text]
  • Crystal Structure of LSD1 in Complex with 4-[5-(Piperidin-4-Ylmethoxy)-2-(P-Tolyl) Pyridin-3-Yl]Benzonitrile
    molecules Article Crystal Structure of LSD1 in Complex with 4-[5-(Piperidin-4-ylmethoxy)-2-(p-tolyl) pyridin-3-yl]benzonitrile Hideaki Niwa 1 ID , Shin Sato 1, Tomoko Hashimoto 2, Kenji Matsuno 2 and Takashi Umehara 1,* ID 1 Laboratory for Epigenetics Drug Discovery, RIKEN Center for Biosystems Dynamics Research (BDR), 1-7-22 Suehiro-cho, Tsurumi, Yokohama 230-0045, Japan; [email protected] (H.N.); [email protected] (S.S.) 2 Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1 Nakano, Hachioji, Tokyo 192-0015, Japan; [email protected] (T.H.); [email protected] (K.M.) * Correspondence: [email protected]; Tel.: +81-45-503-9457 Received: 28 May 2018; Accepted: 22 June 2018; Published: 26 June 2018 Abstract: Because lysine-specific demethylase 1 (LSD1) regulates the maintenance of cancer stem cell properties, small-molecule inhibitors of LSD1 are expected to be useful for the treatment of several cancers. Reversible inhibitors of LSD1 with submicromolar inhibitory potency have recently been reported, but their exact binding modes are poorly understood. In this study, we synthesized a recently reported reversible inhibitor, 4-[5-(piperidin-4-ylmethoxy)-2-(p-tolyl)pyridin-3- yl]benzonitrile, which bears a 4-piperidinylmethoxy group, a 4-methylphenyl group, and a 4-cyanophenyl group on a pyridine ring, and determined the crystal structure of LSD1 in complex with this inhibitor at 2.96 Å. We observed strong electron density for the compound, showing that its cyano group forms a hydrogen bond with Lys661, which is a critical residue in the lysine demethylation reaction located deep in the catalytic center of LSD1.
    [Show full text]
  • Imidazopyridines As Potent KDM5 Demethylase Inhibitors Promoting Reprogramming Efficiency of Human Ipscs
    Article Imidazopyridines as Potent KDM5 Demethylase Inhibitors Promoting Reprogramming Efficiency of Human iPSCs Yasamin Dabiri, Rodrigo A. Gama- Brambila, Katerina Taskova, ..., Jichang Wang, Miguel A. Andrade-Navarro, Xinlai Cheng [email protected] HIGHLIGHTS O4I3 supports the maintenance and generation of human iPSCs O4I3 is a potent H3K4 demethylase KDM5 inhibitor in vitro and in cells KDM5A, but not KDM5B, serves as an epigenetic barrier of reprogramming Chemical or genetic inhibition of KDM5A tends to promote the reprogramming efficiency Dabiri et al., iScience 12,168– 181 February 22, 2019 ª 2019 The Author(s). https://doi.org/10.1016/ j.isci.2019.01.012 Article Imidazopyridines as Potent KDM5 Demethylase Inhibitors Promoting Reprogramming Efficiency of Human iPSCs Yasamin Dabiri,1 Rodrigo A. Gama-Brambila,1 Katerina Taskova,2,3 Kristina Herold,4 Stefanie Reuter,4 James Adjaye,5 Jochen Utikal,6 Ralf Mrowka,4 Jichang Wang,7 Miguel A. Andrade-Navarro,2,3 and Xinlai Cheng1,8,* SUMMARY Pioneering human induced pluripotent stem cell (iPSC)-based pre-clinical studies have raised safety concerns and pinpointed the need for safer and more efficient approaches to generate and maintain patient-specific iPSCs. One approach is searching for compounds that influence pluripotent stem cell reprogramming using functional screens of known drugs. Our high-throughput screening of drug-like hits showed that imidazopyridines—analogs of zolpidem, a sedative-hypnotic drug—are able to improve reprogramming efficiency and facilitate reprogramming of resistant human primary fibro- blasts. The lead compound (O4I3) showed a remarkable OCT4 induction, which at least in part is 1Institute of Pharmacy and due to the inhibition of H3K4 demethylase (KDM5, also known as JARID1).
    [Show full text]
  • High-Throughput Discovery of Novel Developmental Phenotypes
    High-throughput discovery of novel developmental phenotypes The Harvard community has made this article openly available. Please share how this access benefits you. Your story matters Citation Dickinson, M. E., A. M. Flenniken, X. Ji, L. Teboul, M. D. Wong, J. K. White, T. F. Meehan, et al. 2016. “High-throughput discovery of novel developmental phenotypes.” Nature 537 (7621): 508-514. doi:10.1038/nature19356. http://dx.doi.org/10.1038/nature19356. Published Version doi:10.1038/nature19356 Citable link http://nrs.harvard.edu/urn-3:HUL.InstRepos:32071918 Terms of Use This article was downloaded from Harvard University’s DASH repository, and is made available under the terms and conditions applicable to Other Posted Material, as set forth at http:// nrs.harvard.edu/urn-3:HUL.InstRepos:dash.current.terms-of- use#LAA HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Nature. Manuscript Author Author manuscript; Manuscript Author available in PMC 2017 March 14. Published in final edited form as: Nature. 2016 September 22; 537(7621): 508–514. doi:10.1038/nature19356. High-throughput discovery of novel developmental phenotypes A full list of authors and affiliations appears at the end of the article. Abstract Approximately one third of all mammalian genes are essential for life. Phenotypes resulting from mouse knockouts of these genes have provided tremendous insight into gene function and congenital disorders. As part of the International Mouse Phenotyping Consortium effort to generate and phenotypically characterize 5000 knockout mouse lines, we have identified 410 Users may view, print, copy, and download text and data-mine the content in such documents, for the purposes of academic research, subject always to the full Conditions of use:http://www.nature.com/authors/editorial_policies/license.html#terms #Corresponding author: [email protected].
    [Show full text]
  • 82594016.Pdf
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector Structure Article Molecular Mimicry and Ligand Recognition in Binding and Catalysis by the Histone Demethylase LSD1-CoREST Complex Riccardo Baron,1,* Claudia Binda,2 Marcello Tortorici,2 J. Andrew McCammon,1 and Andrea Mattevi2,* 1Department of Chemistry and Biochemistry, Center for Theoretical Biological Physics, and Department of Pharmacology, Howard Hughes Medical Institute, University of California at San Diego, La Jolla, CA 92093-0365, USA 2Department of Genetics and Microbiology, University of Pavia, Via Ferrata 1, 27100, Pavia, Italy *Correspondence: [email protected] (R.B.), [email protected] (A.M.) DOI 10.1016/j.str.2011.01.001 SUMMARY (LSD1 or KDM1A, according to the newly adopted nomencla- ture) (Shi et al., 2004; Forneris et al., 2005). This enzyme acts Histone demethylases LSD1 and LSD2 (KDM1A/B) on mono- and dimethylated Lys4 of histone H3 through a catalyze the oxidative demethylation of Lys4 of FAD-dependent oxidative process (Figure 1A). LSD1 is often histone H3. We used molecular dynamics simula- associated to the histone deacetylases (HDAC) 1 and 2 and to tions to probe the diffusion of the oxygen substrate. the corepressor protein CoREST, which tightly binds to LSD1 Oxygen can reach the catalytic center independently enhancing both its stability and enzymatic activity. A number of from the presence of a bound histone peptide, studies have indicated that LSD1-CoREST interacts with various protein complexes involved in gene regulation and chromatin implying that LSD1 can complete subsequent deme- modification (Forneris et al., 2008; Mosammaparast and Shi, thylation cycles without detaching from the nucleo- 2010).
    [Show full text]
  • Partial Deletion of Chromosome 6P Causing Developmental Delay and Mild Dysmorphisms in a Child
    Vrachnis et al. Mol Cytogenet (2021) 14:39 https://doi.org/10.1186/s13039-021-00557-y CASE REPORT Open Access Partial deletion of chromosome 6p causing developmental delay and mild dysmorphisms in a child: molecular and developmental investigation and literature search Nikolaos Vrachnis1,2,3* , Ioannis Papoulidis4, Dionysios Vrachnis5, Elisavet Siomou4, Nikolaos Antonakopoulos1,2, Stavroula Oikonomou6, Dimitrios Zygouris2, Nikolaos Loukas7, Zoi Iliodromiti8, Efterpi Pavlidou9, Loretta Thomaidis6 and Emmanouil Manolakos4 Abstract Background: The interstitial 6p22.3 deletions concern rare chromosomal events afecting numerous aspects of both physical and mental development. The syndrome is characterized by partial deletion of chromosome 6, which may arise in a number of ways. Case presentation: We report a 2.8-year old boy presenting with developmental delay and mild dysmorphisms. High-resolution oligonucleotide microarray analysis revealed with high precision a 2.5 Mb interstitial 6p deletion in the 6p22.3 region which encompasses 13 genes. Conclusions: Identifcation and in-depth analysis of cases presenting with mild features of the syndrome will sharpen our understanding of the genetic spectrum of the 6p22.3 deletion. Keywords: 6p22.3 deletion, Syndrome, Developmental delay, Intellectual disability, Dysmorphism, Behavioral abnormalities, High-resolution microarray analysis Background dysmorphic features, and structural organ defects, as well Te interstitial deletion of chromosomal region 6p22.3 as intellectual disability. is a rare condition with variable phenotypic expression. We report herein a case of interstitial deletion of chro- To date, more than 30 children and adolescents with this mosome 6p investigated by array-CGH in a 2.8-year old deletion have been reported [1–11].
    [Show full text]
  • The Box C/D Snornp Assembly Factor Bcd1 Interacts with the Histone
    The box C/D snoRNP assembly factor Bcd1 interacts with the histone chaperone Rtt106 and controls its transcription dependent activity Benoît Bragantini, Christophe Charron, Maxime Bourguet, Arnaud Paul, Decebal Tiotiu, Benjamin Rothé, Hélène Marty, Guillaume Terral, Steve Hessmann, Laurence Decourty, et al. To cite this version: Benoît Bragantini, Christophe Charron, Maxime Bourguet, Arnaud Paul, Decebal Tiotiu, et al.. The box C/D snoRNP assembly factor Bcd1 interacts with the histone chaperone Rtt106 and controls its transcription dependent activity. Nature Communications, Nature Publishing Group, 2021, 12 (1), pp.1859. 10.1038/s41467-021-22077-4. hal-03181046 HAL Id: hal-03181046 https://hal.univ-lorraine.fr/hal-03181046 Submitted on 1 Apr 2021 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. Distributed under a Creative Commons Attribution| 4.0 International License ARTICLE https://doi.org/10.1038/s41467-021-22077-4 OPEN The box C/D snoRNP assembly factor Bcd1 interacts with the histone chaperone Rtt106 and controls its transcription dependent
    [Show full text]
  • Investigation of the Underlying Hub Genes and Molexular Pathogensis in Gastric Cancer by Integrated Bioinformatic Analyses
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Investigation of the underlying hub genes and molexular pathogensis in gastric cancer by integrated bioinformatic analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract The high mortality rate of gastric cancer (GC) is in part due to the absence of initial disclosure of its biomarkers. The recognition of important genes associated in GC is therefore recommended to advance clinical prognosis, diagnosis and and treatment outcomes. The current investigation used the microarray dataset GSE113255 RNA seq data from the Gene Expression Omnibus database to diagnose differentially expressed genes (DEGs). Pathway and gene ontology enrichment analyses were performed, and a proteinprotein interaction network, modules, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. Finally, validation of hub genes was performed. The 1008 DEGs identified consisted of 505 up regulated genes and 503 down regulated genes.
    [Show full text]
  • The Complexity of the Epigenome in Cancer and Recent Clinical Advances
    Published OnlineFirst August 17, 2012; DOI: 10.1158/1078-0432.CCR-12-2037 Clinical Cancer Molecular Pathways Research Molecular Pathways: The Complexity of the Epigenome in Cancer and Recent Clinical Advances Mariarosaria Conte1 and Lucia Altucci1,2 Abstract Human cancer is causally linked to genomic and epigenomic deregulations. Epigenetic abnormalities occurring within signaling pathways regulating proliferation, migration, growth, differentiation, tran- scription, and death signals may be critical in the progression of malignancies. Consequently, iden- tification of epigenetic marks and their bioimplications in tumors represents a crucial step toward defining new therapeutic strategies both in cancer treatment and prevention. Alterations of writers, readers, and erasers in cancer may affect, for example, the methylation and acetylation state of huge areas of chromatin, suggesting that epi-based treatments may require "distinct" therapeutic strategies com- pared with "canonical" targeted treatments. Whereas anticancer treatments targeting histone deacetylase and DNA methylation have entered the clinic, additional chromatin modification enzymes have not yet been pharmacologically targeted for clinical use in patients. Thus, a greater insight into alterations occurring on chromatin modifiers and their impact in tumorigenesis represents a crucial advance- ment in exploiting epigenetic targeting in cancer prevention and treatment. Here, the interplay of the best known epi-mutations and how their targeting might be optimized are addressed. Clin Cancer Res; 18 (20); 1–9. Ó2012 AACR. Background strategies both in cancer treatment and prevention (2). A number of epigenetic deregulations, such as DNA Alterations of writers, readers, and erasers in cancer may methylation, histone modifications, and microRNA-based affect the status of chromatin in vast areas of the epigenome, modulation, have been progressively reported as causally thus suggesting that epi-based treatments may require "dis- involved in tumorigenesis and progression.
    [Show full text]
  • RBPJ/CBF1 Interacts with L3MBTL3/MBT1 to Promote Repression of Notch Signaling Via Histone Demethylase KDM1A/LSD1
    Published online: October 13, 2017 Article RBPJ/CBF1 interacts with L3MBTL3/MBT1 to promote repression of Notch signaling via histone demethylase KDM1A/LSD1 Tao Xu1,§, Sung-Soo Park1,§, Benedetto Daniele Giaimo2,§, Daniel Hall3, Francesca Ferrante2, Diana M Ho4, Kazuya Hori4,†, Lucas Anhezini5,‡, Iris Ertl6,¶, Marek Bartkuhn7, Honglai Zhang1, Eléna Milon1, Kimberly Ha1, Kevin P Conlon1, Rork Kuick8, Brandon Govindarajoo9, Yang Zhang9, Yuqing Sun1, Yali Dou1, Venkatesha Basrur1, Kojo SJ Elenitoba-Johnson1, Alexey I Nesvizhskii1,9, Julian Ceron6, Cheng-Yu Lee5, Tilman Borggrefe2, Rhett A Kovall3 & Jean-François Rual1,* Abstract Introduction Notch signaling is an evolutionarily conserved signal transduction The Notch signal transduction pathway is a conserved signaling pathway that is essential for metazoan development. Upon ligand mechanism that is fundamental for morphogenesis in multicellular binding, the Notch intracellular domain (NOTCH ICD) translocates organisms (Bray, 2006; Kopan & Ilagan, 2009; Hori et al, 2013). The into the nucleus and forms a complex with the transcription factor biological action of Notch is highly pleiotropic, and impaired Notch RBPJ (also known as CBF1 or CSL) to activate expression of Notch signaling leads to a broad spectrum of developmental disorders target genes. In the absence of a Notch signal, RBPJ acts as a tran- (Louvi & Artavanis-Tsakonas, 2012) and many types of cancer scriptional repressor. Using a proteomic approach, we identified (Aster et al, 2017). The developmental outcome of Notch signaling L3MBTL3 (also known as MBT1) as a novel RBPJ interactor. L3MBTL3 is strictly dependent on the cell context and can influence cell fate competes with NOTCH ICD for binding to RBPJ. In the absence of in a remarkable number of different ways, for example, differentia- NOTCH ICD, RBPJ recruits L3MBTL3 and the histone demethylase tion, proliferation, and apoptosis (Bray, 2006; Kopan & Ilagan, 2009; KDM1A (also known as LSD1) to the enhancers of Notch target Hori et al, 2013).
    [Show full text]
  • Supplementary Table S4. FGA Co-Expressed Gene List in LUAD
    Supplementary Table S4. FGA co-expressed gene list in LUAD tumors Symbol R Locus Description FGG 0.919 4q28 fibrinogen gamma chain FGL1 0.635 8p22 fibrinogen-like 1 SLC7A2 0.536 8p22 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 DUSP4 0.521 8p12-p11 dual specificity phosphatase 4 HAL 0.51 12q22-q24.1histidine ammonia-lyase PDE4D 0.499 5q12 phosphodiesterase 4D, cAMP-specific FURIN 0.497 15q26.1 furin (paired basic amino acid cleaving enzyme) CPS1 0.49 2q35 carbamoyl-phosphate synthase 1, mitochondrial TESC 0.478 12q24.22 tescalcin INHA 0.465 2q35 inhibin, alpha S100P 0.461 4p16 S100 calcium binding protein P VPS37A 0.447 8p22 vacuolar protein sorting 37 homolog A (S. cerevisiae) SLC16A14 0.447 2q36.3 solute carrier family 16, member 14 PPARGC1A 0.443 4p15.1 peroxisome proliferator-activated receptor gamma, coactivator 1 alpha SIK1 0.435 21q22.3 salt-inducible kinase 1 IRS2 0.434 13q34 insulin receptor substrate 2 RND1 0.433 12q12 Rho family GTPase 1 HGD 0.433 3q13.33 homogentisate 1,2-dioxygenase PTP4A1 0.432 6q12 protein tyrosine phosphatase type IVA, member 1 C8orf4 0.428 8p11.2 chromosome 8 open reading frame 4 DDC 0.427 7p12.2 dopa decarboxylase (aromatic L-amino acid decarboxylase) TACC2 0.427 10q26 transforming, acidic coiled-coil containing protein 2 MUC13 0.422 3q21.2 mucin 13, cell surface associated C5 0.412 9q33-q34 complement component 5 NR4A2 0.412 2q22-q23 nuclear receptor subfamily 4, group A, member 2 EYS 0.411 6q12 eyes shut homolog (Drosophila) GPX2 0.406 14q24.1 glutathione peroxidase
    [Show full text]
  • An RBPJ-Drosophila Model Reveals Dependence of RBPJ Protein Stability on the Formation of Transcription–Regulator Complexes
    cells Article An RBPJ-Drosophila Model Reveals Dependence of RBPJ Protein Stability on the Formation of Transcription–Regulator Complexes Bernd M. Gahr 1,2, Franziska Brändle 1, Mirjam Zimmermann 1 and Anja C. Nagel 1,* 1 Institute of Genetics (240), University of Hohenheim, Garbenstr. 30, 70599 Stuttgart, Germany; [email protected] (B.M.G.); [email protected] (F.B.); [email protected] (M.Z.) 2 Present address: Molecular Cardiology, Department of Internal Medicine II, University of Ulm, Albert-Einstein-Allee 23, 89081 Ulm, Germany * Correspondence: [email protected]; Tel.: +49-711-45922210 Received: 23 August 2019; Accepted: 10 October 2019; Published: 14 October 2019 Abstract: Notch signaling activity governs widespread cellular differentiation in higher animals, including humans, and is involved in several congenital diseases and different forms of cancer. Notch signals are mediated by the transcriptional regulator RBPJ in a complex with activated Notch (NICD). Analysis of Notch pathway regulation in humans is hampered by a partial redundancy of the four Notch receptor copies, yet RBPJ is solitary, allowing its study in model systems. In Drosophila melanogaster, the RBPJ orthologue is encoded by Suppressor of Hairless [Su(H)]. Using genome engineering, we replaced Su(H) by murine RBPJ in order to study its function in the fly. In fact, RBPJ largely substitutes for Su(H)’s function, yet subtle phenotypes reflect increased Notch signaling activity. Accordingly, the binding of RBPJ to Hairless (H) protein, the general Notch antagonist in Drosophila, was considerably reduced compared to that of Su(H). An H-binding defective RBPJLLL mutant matched the respective Su(H)LLL allele: homozygotes were lethal due to extensive Notch hyperactivity.
    [Show full text]