(12) Patent Application Publication (10) Pub. No.: US 2015/0315643 A1 O'garra Et Al

Total Page:16

File Type:pdf, Size:1020Kb

(12) Patent Application Publication (10) Pub. No.: US 2015/0315643 A1 O'garra Et Al US 201503 15643A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2015/0315643 A1 O'Garra et al. (43) Pub. Date: Nov. 5, 2015 (54) BLOODTRANSCRIPTIONAL SIGNATURES Publication Classification OF ACTIVE PULMONARY TUBERCULOSS AND SARCODOSIS (51) Int. C. CI2O I/68 (2006.01) (71) Applicants: BAYLOR RESEARCH INSTITUTE, (52) U.S. C. Dallas, TX (US); MEDICAL CPC ............ CI2O I/6883 (2013.01): CI2O I/6886 RESEARCH COUNCIL, Swindon (2013.01); C12O 2600/158 (2013.01); C12O (GB); IMPERIAL COLLEGE 2600/16 (2013.01); C12O 2600/1 18 (2013.01); HEALTHCARE NHS TRUST, London C12O 2600/1 12 (2013.01); C12O 2600/106 (GB) (2013.01) (72) Inventors: Anne O'Garra, London (GB); Chloe (57) ABSTRACT Bloom, London (GB); Matthew Paul The present invention includes a method of determining a Reddoch Berry, London (GB); Jacques lung disease from a patient Suspected of sarcoidosis, tuber F. Banchereau, Montclair, NJ (US); culosis, lung cancer or pneumonia comprising: obtaining a Damien Chaussabel, Bainbridge Island, sample from whole blood of the patient suspected of sarcoi WA (US); Viginia Maria Pascual, dosis, tuberculosis, lung cancer or pneumonia; detecting Dallas, TX (US) expression of (although not exclusive) six or more disease (21) Appl. No.: 14/651,989 genes, markers, or probes selected from SEQID NOS.: 1 to 1446, wherein increased expression of mRNA of upregulated (22) PCT Fled: Dec. 13, 2013 sarcoidosis, tuberculosis, lung cancer and pneumonia mark ers of SEQID NOS.: 1 to 1446 and/or decreased expression (86) PCT NO.: PCT/US2013/075097 of mRNA of downregulated Sarcoidosis, tuberculosis, lung cancer or pneumonia markers of SEQ ID NOS.: 1 to 1446 S371 (c)(1), relative to the expression of the mRNAs from a normal (2) Date: Jun. 12, 2015 sample; and determining the lung disease based on the expression level of the six or more disease markers of SEQID Related U.S. Application Data NOS.: 1 to 1446 based on a comparison of the expression (60) Provisional application No. 61/736,908, filed on Dec. level of sarcoidosis, tuberculosis, lung cancer, and pneumo 13, 2012. 18. Patent Application Publication Nov. 5, 2015 Sheet 1 of 21 US 2015/0315643 A1 SETTWACJEZIT\/WHON STOHINOO| WINOWñENd£ 8|| SISOGIOOHWS© NEONVOONQTNo.. Patent Application Publication Nov. 5, 2015 Sheet 2 of 21 US 2015/0315643 A1 Patent Application Publication US 2015/0315643 A1 STOHINOO||. NEONVOONQTNo.. WINOWIEN?? SISOGIOOHWS© : S. S S & &S Patent Application Publication Nov. 5, 2015 Sheet 4 of 21 US 2015/0315643 A1 SISOGIOOHWSENILOW? SISOO||OOHWSHALLOW-NON }|E|ONVO9NITT Patent Application Publication Nov. 5, 2015 Sheet 5 of 21 US 2015/0315643 A1 p40.001 p<0.001 p<0.001 p<0.01 p<0.001 p40.05 1000 p40.05 p<0.001 800 WEIGHTED MOLECULAR 600 DISTANCE 400 TO HEALTH 200 CONTROL ACTIVE PNEUMONIA SARCOD TB NON-ACTIVE CANCER SARCOD FIG. 3B FIG. 4A-1 FIG. 4A-2 FIG. 6A-1 FIG. 6A-2 FIG. 4 FIG. 6 Patent Application Publication Nov. 5, 2015 Sheet 6 of 21 US 2015/0315643 A1 FIG. 4A-1 O&O G (900 G | O Q O OG G eos ooooooooo doooooo oo Ooooooel (33 () (366) (3 OOC) OG E)(3G) () G | O (3G) (C) () (3G) (3)O 3 (3 e G () () O (GSO) () ()(36 () (36) () (OOG) (3G) oooooood (c)(3) () () (336:33Ooooooooooooooo (333 (36)(3)(363.36 (3 (33)(3)(33G)3&63()eeeeoo pool (33333opio &o OOOO OO o OOOoel O OOO OO OO koo o (3G) () ()() () ()() () O (E) () () ()() () Q oo o ode (E) () O OO e Oc (33 CE) 2 (3 (3 (3) O o d (3G) H O3 (33 (3)(3 (3G) () ()(3) (3) ()() " () () ()() () (3) () eO & Ge e o Q G GQ e o oo oo od 363 () (3) GE) () () () () () O GE) o oooooo() ()() (3) odo oo o o () GE) (SG) () ()() & () () soo() o() ()()so e() o lo o o do() NE TB ACTIVE SARCODOSIS NON-ACTIVE SARCOD Patent Application Publication Nov. 5, 2015 Sheet 7 of 21 US 2015/0315643 A1 FIG. 4A-2 3. Q() () M4.10 B-CELLS ke oooo eke oooooo M41M9.2 TCELLsB-CELLS 3O6) M5.15 NEUTROPHILS M4.11 PLASMA CELLS M4.14 MONOCYTES pooe GE M11 PLATELETS 6 M2.3 ERYTHROCYTES OG 6.) M3.1 ERYTHROCYTES 38G)6(3)() (33 QQ M36 CYTOTOXCINK M7.21 CYTOTOXC O M12 F INTERFERON M34 IFN MODULES M5.12 LIFN (3) Q M7.18 IMMUNOSUPRESSION G) M8.61 IMMUNE RESPONSES eO M8.89 IMMUNE RESPONSES b M934 MMUNOSUPRESSION 3.33(3) (3)(33 (33 M3.2 |NFLAMMATION NFLAMMATION M42 INFLAMMATION 4 MODULES a M4.13 NFLAMMATION E M5.1 INFLAMMATION M722 NFLAMMATION () () ( M7.24 LYMPHOCYTEACTIVATION M2.1 CELL CYCLE M22 CELL CYCLE M3.3 CELL CYCLE 6: OO M3.5 CELL CYCLE M6.11 CELL CYCLE M6.13 CELL DEATH M6.16 CELL CYCLE () d M76 PROLIFERATION O M77 PROLIFERATION M78 CELL CYCLE (3)(E)(3G)3 M7.16 DCIAPOPTOSIS 6 O M5.10 MIT RESPIRATION M6.2 MITOCHONDRIAL RESPIRATION k d O M6.12 MITOCHONDRIAL STRESS ====/vvy u/ O OVEREXPRESSED PNEUMONIA LUNG CANCER (3 & 8 (3) Q) O UNDEREXPRESSED 100 908070 60504030 20 PERCENTAGE PROBE (P<0.05) Patent Application Publication Nov. 5, 2015 Sheet 8 of 21 US 2015/0315643 A1 p<0.001 p40.001 p<0.001 p<0.001 p<0.05 p<0.001 p40.001 pCO.001 p40.001 pCO.001 cuo p40.001 p40.001 ly 50 4 s: 40 HS i? 3: 3 O cod 30 E R 5 2 e c 20 e f : 10 f : 1 5 O O NON-ACTIVE CANCER NON-ACTIVE CANCER SARCOD SARCOID ACTIVE PNEUMONIA ACTIVE PNEUMONIA SARCOD SARCOD FIG. 4B FIG. 4C p<0.001 p40.001 p<0.001 p<0.05 P-0001 p<0.001 P-0001 p<0.001 if 30 p-0001p-0001 if 25 p-0001p-0001 1 D S 5 SB 2.0 5 20 e Z C2 E 235 s. 1.5 a PS 10 3. H sas Z. ise S 0.5 32 s O 0.0 4 n. NON-ACTIVE CANCER 4. NON-ACTIVE CANCER SARCOD SARCOD ACTIVE PNEUMONIA ACTIVE PNEUMONIA SARCOD SARCOD FIG. 4D FIG. 4E Patent Application Publication Nov. 5, 2015 Sheet 9 of 21 US 2015/0315643 A1 FIG. 5A PERCENTAGE OF GENES IN PATHWAY O 20 40 60 TB ACTIVE SARCOD EIF2 SIGNALING PNEUMONIA (UNDER-ABUNDANT) CANCER O 5 10 15 -log (B-H p-value) PERCENTAGE OF GENES IN PATHWAY O 20 40 60 TB -NN ACTIVE SARCOID - IFNSIGNALING PNEUMONIA (OVER-ABUNDANT) CANCER O 5 10 15 -log (B-H p-value) PERCENTAGE OF GENES IN PATHWAY O 20 40 60 TB-NN ROLE OF PRRS IN ACTIVE SARCOD RECOGNITION OF PNEUMONIA BACTERIA AND VIRUSES (OVER-ABUNDANT) CANCER O 5 10 15 -log (B-H p-value) PERCENTAGE OF GENES IN PATHWAY O 20 40 60 TB -NN ANTIGEN ACTIVE SARCOID - PRESENTATION PNEUMONIA PATHWAY (OVER-ABUNDANT) CANCER O 5 10 15 -log (B-H p-value) Patent Application Publication Nov. 5, 2015 Sheet 10 of 21 US 2015/0315643 A1 GIOOLIHOOOOn10)--/ @@º@z STIETOTIN Patent Application Publication Nov. 5, 2015 Sheet 11 of 21 US 2015/0315643 A1 GIOOLIHOOOOnT9X–—/ STIETOTIN Patent Application Publication Nov. 5, 2015 Sheet 12 of 21 US 2015/0315643 A1 "OIH[?NET)|×]VINOWITEINdCIS Patent Application Publication Nov. 5, 2015 Sheet 13 of 21 US 2015/0315643 A1 GIOOLHOOOOn10)--/ X @ "OIHEFNEI)|×]}|E|ONY/OSONTOTEIS INVOIN?n8W-HECINQÑ„No.,/*%\€)sz Patent Application Publication Nov. 5, 2015 Sheet 14 of 21 US 2015/0315643 A1 FIG. 6A-1 O O o O Ol 6 (3. O Ole o e o e g g o el o e o O & ele o og o O O 2 o ogo O O o O O e e o so o ol go o o O O O Q Q O 99 () O O e e () e o O ele o (3) () 2 O O ( 0 () () ( e. lo is o 6 o 6 6 o O 6 lo 35CD () () e Q goQ oo H5 Q Q Q lo (3) o (E) (3 e o O o O O o o o A B C D E PRE-TREATMENT POST-TREATMENT PRE-TREATMENT v=v-7 PNEUMONIA Patent Application Publication Nov. 5, 2015 Sheet 15 of 21 US 2015/0315643 A1 FIG. 6A-2 O O Go O G M410 BCELLS M9.2 B-CELLS e Q O Ole e M4.1 T-CELLS O O Ole O M5.15 NEUTROPHILS M4.11 PLASMA CELLS M4.14 MONOCYTES O9 O M1.1 PLATELETS O M2.3 ERYTHROCYTES M3.1 ERYTHROCYTES () elo M3.6 CYTOTOXCINK M7.21 CYTOTOXC O () (3 G M12 IN INTERFERON 999 Q M3.4 IFN r- MODULES M5.12 IFN (D seesO () () () M7.18 IMMUNOSUPRESSION () o () M8.61 IMMUNE RESPONSES () M8.89 IMMUNE RESPONSES G3 G3 (3 & O M32MSYNSESSION NFLAMMATION INFLAMMATION M42 NFLAMMATION 1. MODULES O ()o ()ee () (3e O9 M4.13 NFLAMMATION Ole O e M5.1 NFLAMMATION M7.22 NFLAMMATION olo de :: M7.24 LYMPHOCYTEACTIVATION M2.1 CELL CYCLE |o o e M2.2 CELL CYCLE OO M3.3 CELL CYCLE M3.5 CELL CYCLE o o?e o o M6.11 CELL CYCLE 6 M6.13 CELL DEATH oo o o M6.16 CELL CYCLE O M7.6 PROLIFERATION () o|o o o M7.7 PROLIFERATION M7.8 CELL CYCLE M7.16 DCIAPOPTOSIS olo e as M5.10 MIT RESPIRATION () () M6.2 MITOCHONDRIAL RESPRATION (3 l (; 6 (3) : M6.12 MITOCHONDRIAL STRESS 32 52 62 7212 23 33 4243 PATIENTID INADEQUATE GOOD TREATMENT TREATMENT O OVEREXPRESSED RESPONSE RESPONSE 6 (8 & 3) (3) () (O UNDEREXPRESSED =w=/ 100908070 60504030 20 SA RCODOSIS PERCENTAGE PROBE (P<0.05) Patent Application Publication Nov.
Recommended publications
  • The Cross-Talk Between Methylation and Phosphorylation in Lymphoid-Specific Helicase Drives Cancer Stem-Like Properties
    Signal Transduction and Targeted Therapy www.nature.com/sigtrans ARTICLE OPEN The cross-talk between methylation and phosphorylation in lymphoid-specific helicase drives cancer stem-like properties Na Liu1,2,3, Rui Yang1,2, Ying Shi1,2, Ling Chen1,2, Yating Liu1,2, Zuli Wang1,2, Shouping Liu1,2, Lianlian Ouyang4, Haiyan Wang1,2, Weiwei Lai1,2, Chao Mao1,2, Min Wang1,2, Yan Cheng5, Shuang Liu4, Xiang Wang6, Hu Zhou7, Ya Cao1,2, Desheng Xiao1 and Yongguang Tao1,2,6 Posttranslational modifications (PTMs) of proteins, including chromatin modifiers, play crucial roles in the dynamic alteration of various protein properties and functions including stem-cell properties. However, the roles of Lymphoid-specific helicase (LSH), a DNA methylation modifier, in modulating stem-like properties in cancer are still not clearly clarified. Therefore, exploring PTMs modulation of LSH activity will be of great significance to further understand the function and activity of LSH. Here, we demonstrate that LSH is capable to undergo PTMs, including methylation and phosphorylation. The arginine methyltransferase PRMT5 can methylate LSH at R309 residue, meanwhile, LSH could as well be phosphorylated by MAPK1 kinase at S503 residue. We further show that the accumulation of phosphorylation of LSH at S503 site exhibits downregulation of LSH methylation at R309 residue, which eventually promoting stem-like properties in lung cancer. Whereas, phosphorylation-deficient LSH S503A mutant promotes the accumulation of LSH methylation at R309 residue and attenuates stem-like properties, indicating the critical roles of LSH PTMs in modulating stem-like properties. Thus, our study highlights the importance of the crosstalk between LSH PTMs in determining its activity and function in lung cancer stem-cell maintenance.
    [Show full text]
  • Hypoxia and Oxygen-Sensing Signaling in Gene Regulation and Cancer Progression
    International Journal of Molecular Sciences Review Hypoxia and Oxygen-Sensing Signaling in Gene Regulation and Cancer Progression Guang Yang, Rachel Shi and Qing Zhang * Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; [email protected] (G.Y.); [email protected] (R.S.) * Correspondence: [email protected]; Tel.: +1-214-645-4671 Received: 6 October 2020; Accepted: 29 October 2020; Published: 31 October 2020 Abstract: Oxygen homeostasis regulation is the most fundamental cellular process for adjusting physiological oxygen variations, and its irregularity leads to various human diseases, including cancer. Hypoxia is closely associated with cancer development, and hypoxia/oxygen-sensing signaling plays critical roles in the modulation of cancer progression. The key molecules of the hypoxia/oxygen-sensing signaling include the transcriptional regulator hypoxia-inducible factor (HIF) which widely controls oxygen responsive genes, the central members of the 2-oxoglutarate (2-OG)-dependent dioxygenases, such as prolyl hydroxylase (PHD or EglN), and an E3 ubiquitin ligase component for HIF degeneration called von Hippel–Lindau (encoding protein pVHL). In this review, we summarize the current knowledge about the canonical hypoxia signaling, HIF transcription factors, and pVHL. In addition, the role of 2-OG-dependent enzymes, such as DNA/RNA-modifying enzymes, JmjC domain-containing enzymes, and prolyl hydroxylases, in gene regulation of cancer progression, is specifically reviewed. We also discuss the therapeutic advancement of targeting hypoxia and oxygen sensing pathways in cancer. Keywords: hypoxia; PHDs; TETs; JmjCs; HIFs 1. Introduction Molecular oxygen serves as a co-factor in many biochemical processes and is fundamental for aerobic organisms to maintain intracellular ATP levels [1,2].
    [Show full text]
  • Epigenetics of Estrogen Receptor Signaling: Role in Hormonal Cancer Progression and Therapy
    Cancers 2011, 3, 1691-1707; doi:10.3390/cancers3021691 OPEN ACCESS cancers ISSN 2072-6694 www.mdpi.com/journal/cancers Review Epigenetics of Estrogen Receptor Signaling: Role in Hormonal Cancer Progression and Therapy Monica Mann 1, Valerie Cortez 1 and Ratna K. Vadlamudi 2,* 1 Department of Cellular and Structural Biology, UTHSCSA, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA; E-Mails: [email protected] (M.M.); [email protected] (V.C.) 2 Department of Obstetrics and Gynecology, UTHSCSA, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA * Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +1-210-567-4930; Fax: +1-210-567-4958. Received: 4 January 2011; in revised form: 11 March 2011 / Accepted: 25 March 2011 / Published: 29 March 2011 Abstract: Estrogen receptor (ER) signaling plays a key role in hormonal cancer progression. ER is a ligand-dependent transcription factor that modulates gene transcription via recruitment to the target gene chromatin. Emerging evidence suggests that ER signaling has the potential to contribute to epigenetic changes. Estrogen stimulation is shown to induce several histone modifications at the ER target gene promoters including acetylation, phosphorylation and methylation via dynamic interactions with histone modifying enzymes. Deregulation of enzymes involved in the ER-mediated epigenetic pathway could play a vital role in ER driven neoplastic processes. Unlike genetic alterations, epigenetic changes are reversible, and hence offer novel therapeutic opportunities to reverse ERdriven epigenetic changes In this review, we summarize current knowledge on mechanisms by which ER signaling potentiates epigenetic changes in cancer cells via histone modifications.
    [Show full text]
  • Molecular Mechanisms Involved Involved in the Interaction Effects of HCV and Ethanol on Liver Cirrhosis
    Virginia Commonwealth University VCU Scholars Compass Theses and Dissertations Graduate School 2010 Molecular Mechanisms Involved Involved in the Interaction Effects of HCV and Ethanol on Liver Cirrhosis Ryan Fassnacht Virginia Commonwealth University Follow this and additional works at: https://scholarscompass.vcu.edu/etd Part of the Physiology Commons © The Author Downloaded from https://scholarscompass.vcu.edu/etd/2246 This Thesis is brought to you for free and open access by the Graduate School at VCU Scholars Compass. It has been accepted for inclusion in Theses and Dissertations by an authorized administrator of VCU Scholars Compass. For more information, please contact [email protected]. Ryan C. Fassnacht 2010 All Rights Reserved Molecular Mechanisms Involved in the Interaction Effects of HCV and Ethanol on Liver Cirrhosis A thesis submitted in partial fulfillment of the requirements for the degree of Master of Science at Virginia Commonwealth University. by Ryan Christopher Fassnacht, B.S. Hampden Sydney University, 2005 M.S. Virginia Commonwealth University, 2010 Director: Valeria Mas, Ph.D., Associate Professor of Surgery and Pathology Division of Transplant Department of Surgery Virginia Commonwealth University Richmond, Virginia July 9, 2010 Acknowledgement The Author wishes to thank his family and close friends for their support. He would also like to thank the members of the molecular transplant team for their help and advice. This project would not have been possible with out the help of Dr. Valeria Mas and her endearing
    [Show full text]
  • Histone Demethylase JMJD6 Regulates Cellular Migration And
    Shen et al. Stem Cell Research & Therapy (2018) 9:212 https://doi.org/10.1186/s13287-018-0949-3 RESEARCH Open Access Histone demethylase JMJD6 regulates cellular migration and proliferation in adipose-derived mesenchymal stem cells Chongyang Shen1,2, Qingli Quan3*, Chuan Yang1, Yueqiang Wen2 and Hong Li1* Abstract Background: Adipose-derived mesenchymal stem cells (ADSCs) have been extensively explored as a promising therapeutic agent due to their differentiation, proliferation and migration abilities. The epigenetic mechanisms that regulate the fate of mesenchymal stem cells (MSCs) have been described in detail. However, the epigenetic modulation of ADSCs proliferation and migration is poorly understood. Methods: The present study examined histone demethylases roles and expression by RT-PCR, as well as through siRNA screening and ChIP-qPCR assay. Cellular proliferation and migration assays were employed in shRNA- mediated JMJD6 knockdown and control ADSCs. PDE1C inhibition studies were conducted to confirm its role in JMJD6-mediated epigenetic regulation of ADSCs. Results: The data demonstrate that the histone demethylase JMJD6 plays a critical role in regulating the proliferation and migration of ADSCs by removing H4R3me2a at the promoter regions of PDEC1 and suppressing PDEC1 expression. Importantly, the depletion of JMJD6 in ADSCs significantly increased cellular proliferation and motility, which was associated with increases in PDE1C expression and decreases in the levels of both cAMP and cGMP. The increase in proliferation and migration was reversed by treatment with a PDE1C inhibitor, suggesting that JMJD6 attenuates the proliferation and migration of ADSCs as an epigenetic regulator and PDE1C partially contributes to the JMJD6-mediated regulation. Conclusions: Taken together, our results indicate for the first time that JMJD6 plays an important role in the regulation of ADSCs proliferation and migration through the modulation of PDE1C expression.
    [Show full text]
  • Preclinical Evaluation of Protein Disulfide Isomerase Inhibitors for the Treatment of Glioblastoma by Andrea Shergalis
    Preclinical Evaluation of Protein Disulfide Isomerase Inhibitors for the Treatment of Glioblastoma By Andrea Shergalis A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy (Medicinal Chemistry) in the University of Michigan 2020 Doctoral Committee: Professor Nouri Neamati, Chair Professor George A. Garcia Professor Peter J. H. Scott Professor Shaomeng Wang Andrea G. Shergalis [email protected] ORCID 0000-0002-1155-1583 © Andrea Shergalis 2020 All Rights Reserved ACKNOWLEDGEMENTS So many people have been involved in bringing this project to life and making this dissertation possible. First, I want to thank my advisor, Prof. Nouri Neamati, for his guidance, encouragement, and patience. Prof. Neamati instilled an enthusiasm in me for science and drug discovery, while allowing me the space to independently explore complex biochemical problems, and I am grateful for his kind and patient mentorship. I also thank my committee members, Profs. George Garcia, Peter Scott, and Shaomeng Wang, for their patience, guidance, and support throughout my graduate career. I am thankful to them for taking time to meet with me and have thoughtful conversations about medicinal chemistry and science in general. From the Neamati lab, I would like to thank so many. First and foremost, I have to thank Shuzo Tamara for being an incredible, kind, and patient teacher and mentor. Shuzo is one of the hardest workers I know. In addition to a strong work ethic, he taught me pretty much everything I know and laid the foundation for the article published as Chapter 3 of this dissertation. The work published in this dissertation really began with the initial identification of PDI as a target by Shili Xu, and I am grateful for his advice and guidance (from afar!).
    [Show full text]
  • The Dynamic Role of Jumonji C Domain Containing Protein 6 in Placental Development and Disease
    The Dynamic Role of Jumonji C Domain Containing Protein 6 in Placental Development and Disease by Sruthi Alahari A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Physiology University of Toronto © Copyright by Sruthi Alahari 2017 i The Dynamic Role of Jumonji C Domain Containing Protein 6 in Placental Development and Disease Sruthi Alahari Doctor of Philosophy Department of Physiology University of Toronto 2017 Abstract Perturbations in oxygen sensing are a defining feature of placental-associated pathologies such as preeclampsia, a serious disorder of pregnancy. Preeclamptic placentae have markedly elevated levels of Hypoxia Inducible Factor 1α (HIF1A), a master regulator of oxygen homeostasis. Mounting evidence implicates a family of Fe2+ and oxygen-dependent Jumonji C domain containing enzymes (JMJDs) as mediators of the epigenetic code and hypoxic gene expression. While several JMJDs are induced in hypoxia, their role in pregnancy remains unclear. The goal of this study was to characterize JMJD6 function in the placenta in physiological and pathological conditions, and unravel its regulatory relationship with von Hippel Lindau tumour suppressor (VHL), a key executor of the cellular hypoxic response. JMJD6 expression inversely correlated with changes in oxygen tension during placental development, while JMJD6 protein and mRNA were significantly elevated in low oxygen and in early-onset preeclamptic (E-PE) placentae. In vitro demethylation assays revealed that optimal JMJD6-dependent demethylation of its histone targets, H3R2me2s and H4R3me2s, occurred in normoxia, and this was impaired in E-PE placentae due to a hypoxia-iron imbalance. In cytotrophoblast cells, JMJD6 is a positive ii regulator of VHL gene expression in normoxia.
    [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]
  • 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]
  • Genome-Wide Analysis of Thyroid Hormone Receptors Shared and Specific Functions in Neural Cells
    Genome-wide analysis of thyroid hormone receptors shared and specific functions in neural cells Fabrice Chatonneta, Romain Guyota, Gérard Benoîtb, and Frederic Flamanta,1 aInstitut de Génomique Fonctionnelle de Lyon, Université de Lyon, Centre National de la Recherche Scientifique (CNRS), Institut National de la Recherche Agronomique, École Normale Supérieure de Lyon, 69364 Lyon Cedex 07, France; and bCentre de Génétique et de Physiologie Moléculaire et Cellulaire, CNRS, Université Lyon 1, Unité Mixte de Recherche 5334, Villeurbanne F-69622, France Edited by Pierre Chambon, Institut de Génétique et de Biologie Moléculaire et Cellulaire (Centre National de la Recherche Scientifique UMR7104, Institut National de la Santé et de la Recherche Médicale U596, Université de Strasbourg, College de France), Illkirch-Cedex, France, and approved January 4, 2013 (received for review June 25, 2012) TRα1 and TRβ1, the two main thyroid hormone receptors in mam- issue. It would help to develop new selective ligands (6). It might mals, are transcription factors that share similar properties. How- also help to predict the possible detrimental side effects of these ever, their respective functions are very different. This functional synthetic ligands in heart and brain (7) and the toxicity of some divergence might be explained in two ways: it can reflect different environmental contaminants supposed to interfere with TR expression patterns or result from different intrinsic properties of functions (8). The primary sequence and 3D structure of TRα1 β the receptors. We tested this second hypothesis by comparing the and TR 1 are very similar, although differences are observed for – repertoires of 3,3′,5-triiodo-L-thyronine (T3)-responsive genes of key amino acids in the DNA-binding domain (9 12).
    [Show full text]
  • HDAC5-Mediated Deacetylation and Nuclear Localisation of SOX9 Is Critical for Tamoxifen Resistance in Breast Cancer
    www.nature.com/bjc ARTICLE Translational Therapeutics HDAC5-mediated deacetylation and nuclear localisation of SOX9 is critical for tamoxifen resistance in breast cancer Yue Xue1, Wenwen Lian1, Jiaqi Zhi1, Wenjuan Yang1, Qianjin Li1, Xingyi Guo2, Jiahao Gao1, Hao Qu3, Weiqiang Lin4, Zhongqi Li5, Lihua Lai1,6 and Qingqing Wang1 BACKGROUND: Tamoxifen resistance remains a significant clinical challenge for the therapy of ER-positive breast cancer. It has been reported that the upregulation of transcription factor SOX9 in ER+ recurrent cancer is sufficient for tamoxifen resistance. However, the mechanisms underlying the regulation of SOX9 remain largely unknown. METHODS: The acetylation level of SOX9 was detected by immunoprecipitation and western blotting. The expressions of HDACs and SIRTs were evaluated by qRT-PCR. Cell growth was measured by performing MTT assay. ALDH-positive breast cancer stem cells were evaluated by flow cytometry. Interaction between HDAC5 and SOX9 was determined by immunoprecipitation assay. RESULTS: Deacetylation is required for SOX9 nuclear translocation in tamoxifen-resistant breast cancer cells. Furthermore, HDAC5 is the key deacetylase responsible for SOX9 deacetylation and subsequent nuclear translocation. In addition, the transcription factor C-MYC directly promotes the expression of HDAC5 in tamoxifen resistant breast cancer cells. For clinical relevance, high SOX9 and HDAC5 expression are associated with lower survival rates in breast cancer patients treated with tamoxifen. CONCLUSIONS: This study reveals that
    [Show full text]
  • The Critical Role of Histone Methylation in Tumour Progression and As Anti-Cancer Targets in Neuroblastoma
    The critical role of histone methylation in tumour progression and as anti-cancer targets in neuroblastoma Matthew Wong A thesis in fulfilment of the requirements for the degree of Doctor of Philosophy Supervisor: Dr Tao Liu Co-supervisor: A. Prof Patsie Polly School of Women’s and Children’s Health Faculty of Medicine February 2016 PLEASE TYPE THE UNIVERSITY OF NEW SOUTH WALES Thesis/Dissertation Sheet Surname or Family name: First name: Matthew Other name/s: Kwok Kei Wong Abbreviation for degree as given in the University calendar: PhD School: School of Women’s and Children’s Heath Faculty: Faculty of Medicine Title: The critical role of histone methylation in tumour progression and as anti-cancer targets in neuroblastoma N-Myc induces neuroblastoma by regulating the expression of target oncogenes. Histone H3 lysine 79 (H3K79) methylation at Myc-responsive elements of target gene promoters is a strict prerequisites for Myc-induced transcriptional activation. DOT1L is the only known histone methyltransferase that catalyses mono-methylation (me), di- methylation (me2) and tri-methylation (me3) at the histone H3K79 position, which have been linked to gene transcriptional activation. JMJD6 is a bi-functional arginine demethylase and lysyl-hydroxylase. The JMJD6 gene is located on the chromosome 17q25 position. 17q21-gter gain has been identified as the most frequent chromosome alternation in neuroblastoma and an indicator of poor patient prognostic. Here, I investigated the roles of DOT1L and JMJD6 in N-Myc over-expressing neuroblastoma. I found that N-Myc up-regulated DOT1L mRNA and protein expression, by binding to an E-box at the DOT1L gene promoter.
    [Show full text]