Next Generation Sequencing(Next Gen) Sunquest: NGS

Total Page:16

File Type:pdf, Size:1020Kb

Next Generation Sequencing(Next Gen) Sunquest: NGS UNIVERSITY OF MINNESOTA PHYSICIANS OUTREACH LABS Submit this form along with the appropriate Molecular requisition (Molecular Diagnostics or MOLECULAR DIAGNOSTICS (612) 273-8445 Molecular NGS Oncology). DATE: TIME COLLECTED: PCU/CLINIC: AM PM PATIENT IDENTIFICATION DIAGNOSIS (Dx) / DIAGNOSIS CODES (ICD-9) - OUTPATIENTS ONLY SPECIMEN TYPE: o Blood (1) (2) (3) (4) PLEASE COLLECT 5-10CC IN ACD-A OR EDTA TUBE ORDERING PHYSICIAN NAME AND PHONE NUMBER: Tests can be ordered as a full panel, or by individual gene(s). Please contact the genetic counselor with any questions at 612-624-8948 or by pager at 612-899-3291. _______________________________________________ Test Ordered- EPIC: Next generation sequencing(Next Gen) Sunquest: NGS 17-beta-hydroxysteroid dehydrogenase Diabetes mellitus, transient neonatal, 1 Hyperthyroidism, nonautoimmune X deficiency ZFP57 TSHR HSD17B10 Insulin resistance, severe, digenic Hypogonadotropic hypogonadism Adrenal hyperplasia, congenita PPP1R3A Full panel Full panel PPARG CYP17A1 TAC3 Maturity-onset diabetes of the young CYP11B1 TACR3 Full panel CYP11A1 GNRH1 HNF4A Adrenocorticotropic hormone deficiency KISS1 GCK TBX19 WDR11 HNF1A Apparent mineralcorticoid excess HS6ST1 PDX1 HSD11B2 FGFR1 PAX4 Autoimmune polyendocrinopathy PROKR2 NEUROD1 syndrome PROK2 KLF11 AIRE CHD7 ACTH-independent macronodular CEL FGF8 adrenal hyperplasia INS GNRHR GNAS BLK Congenital hypothyroidism KISS1R Estrogen resistance PAX8 NSMF ESR1 Corticosteroid-binding globulin POLR3B ESR2 deficiency Hypoparathyroidism Glucocorticoid deficiency SERPINA6 Full panel Cortisol resistance MRAP Full panel NR3C1 FGD3 PTH Cortisone reductase deficiency 1 MC2R GCM2 H6PD Glucocorticoid-remediable Hypothryoidism, congenital, Cortisone reductase deficiency 2 aldosteronism nongoitrous HSD11B1 CYP11B2 Diabetes insipidus, neurohypophyseal Growth hormone insensitivity with Full panel AVP immunodeficiency TSHB Diabetes mellitus, insulin-resistant, with STAT5B TSHR acanthosis nigricans Hyperparathyroidism THRA INSR Follicle-stimulating hormone deficiency, Diabetes mellitus, neonatal, with isolated congenital hypothyroidism Full panel CDC73 GLIS3 CASR FSHB 4/1/2014 Version 1 Spermatogenic failure Pigmented nodular adrenocortical disease Full panel Full panel DAZL PRKAR1A AURKC PDE11A SEPT12 PDE8B KLHL10 Pituitary hormone deficiency SYCP3 Full panel SPATA16 PROP1 CATSPER1 LHX3 DPY19L2 LHX4 USP9Y llipodystrophy, congenital and POU1F1 pulmonary hypertension Immunodysregulation, polyendocrinopathy, and enteropathy, CAV1 X-linked Lipodystrophy, congenital generalized FOXP3 Full panel Pseudohypoaldosteronism AGPAT2 Full panel BSCL2 CUL3 CAV1 NR3C2 PTRF WNK4 Lipodystrophy, familial partial WNK1 KLHL3 Full panel STX16 LMNA Short stature, idiopathic familial PPARG SHOX PLIN1 Thyroid dyshormonogenesis CIDEC Full panel Obesity, adrenal insufficiency, and red DUOX2 hair due to POMC deficiency SLC5A5 POMC TPO Obesity with impaired prohormone TG processing IYD DUOXA2 PCSK1 SECISBP2 Ovarian dysgenesis/failure Thyroid hormone resistance Full panel THRB FSHR Thyroxine-binding globulin deficiency PSMC3IP SERPINA7 BMP15 DIAPH2 POF1B FOXL2 NOBOX FIGLA Pancreatic agenesis PDX1 4/1/2014 Version 1 .
Recommended publications
  • Chapter 7: Monogenic Forms of Diabetes
    CHAPTER 7 MONOGENIC FORMS OF DIABETES Mark A. Sperling, MD, and Abhimanyu Garg, MD Dr. Mark A. Sperling is Emeritus Professor and Chair, University of Pittsburgh, Department of Pediatrics, Children’s Hospital of Pittsburgh of UPMC, Pittsburgh, PA. Dr. Abhimanyu Garg is Professor of Internal Medicine and Chief of the Division of Nutrition and Metabolic Diseases at University of Texas Southwestern Medical Center, Dallas, TX. SUMMARY Types 1 and 2 diabetes have multiple and complex genetic influences that interact with environmental triggers, such as viral infections or nutritional excesses, to result in their respective phenotypes: young, lean, and insulin-dependence for type 1 diabetes patients or older, overweight, and often manageable by lifestyle interventions and oral medications for type 2 diabetes patients. A small subset of patients, comprising ~2%–3% of all those diagnosed with diabetes, may have characteristics of either type 1 or type 2 diabetes but have single gene defects that interfere with insulin production, secretion, or action, resulting in clinical diabetes. These types of diabetes are known as MODY, originally defined as maturity-onset diabetes of youth, and severe early-onset forms, such as neonatal diabetes mellitus (NDM). Defects in genes involved in adipocyte development, differentiation, and death pathways cause lipodystrophy syndromes, which are also associated with insulin resistance and diabetes. Although these syndromes are considered rare, more awareness of these disorders and increased availability of genetic testing in clinical and research laboratories, as well as growing use of next generation, whole genome, or exome sequencing for clinically challenging phenotypes, are resulting in increased recognition. A correct diagnosis of MODY, NDM, or lipodystrophy syndromes has profound implications for treatment, genetic counseling, and prognosis.
    [Show full text]
  • Ovarian Gene Expression in the Absence of FIGLA, an Oocyte
    BMC Developmental Biology BioMed Central Research article Open Access Ovarian gene expression in the absence of FIGLA, an oocyte-specific transcription factor Saurabh Joshi*1, Holly Davies1, Lauren Porter Sims2, Shawn E Levy2 and Jurrien Dean1 Address: 1Laboratory of Cellular and Developmental Biology, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA and 2Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, USA Email: Saurabh Joshi* - [email protected]; Holly Davies - [email protected]; Lauren Porter Sims - [email protected]; Shawn E Levy - [email protected]; Jurrien Dean - [email protected] * Corresponding author Published: 13 June 2007 Received: 11 December 2006 Accepted: 13 June 2007 BMC Developmental Biology 2007, 7:67 doi:10.1186/1471-213X-7-67 This article is available from: http://www.biomedcentral.com/1471-213X/7/67 © 2007 Joshi et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Background: Ovarian folliculogenesis in mammals is a complex process involving interactions between germ and somatic cells. Carefully orchestrated expression of transcription factors, cell adhesion molecules and growth factors are required for success. We have identified a germ-cell specific, basic helix-loop-helix transcription factor, FIGLA (Factor In the GermLine, Alpha) and demonstrated its involvement in two independent developmental processes: formation of the primordial follicle and coordinate expression of zona pellucida genes. Results: Taking advantage of Figla null mouse lines, we have used a combined approach of microarray and Serial Analysis of Gene Expression (SAGE) to identify potential downstream target genes.
    [Show full text]
  • Table S1 the Four Gene Sets Derived from Gene Expression Profiles of Escs and Differentiated Cells
    Table S1 The four gene sets derived from gene expression profiles of ESCs and differentiated cells Uniform High Uniform Low ES Up ES Down EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol EntrezID GeneSymbol 269261 Rpl12 11354 Abpa 68239 Krt42 15132 Hbb-bh1 67891 Rpl4 11537 Cfd 26380 Esrrb 15126 Hba-x 55949 Eef1b2 11698 Ambn 73703 Dppa2 15111 Hand2 18148 Npm1 11730 Ang3 67374 Jam2 65255 Asb4 67427 Rps20 11731 Ang2 22702 Zfp42 17292 Mesp1 15481 Hspa8 11807 Apoa2 58865 Tdh 19737 Rgs5 100041686 LOC100041686 11814 Apoc3 26388 Ifi202b 225518 Prdm6 11983 Atpif1 11945 Atp4b 11614 Nr0b1 20378 Frzb 19241 Tmsb4x 12007 Azgp1 76815 Calcoco2 12767 Cxcr4 20116 Rps8 12044 Bcl2a1a 219132 D14Ertd668e 103889 Hoxb2 20103 Rps5 12047 Bcl2a1d 381411 Gm1967 17701 Msx1 14694 Gnb2l1 12049 Bcl2l10 20899 Stra8 23796 Aplnr 19941 Rpl26 12096 Bglap1 78625 1700061G19Rik 12627 Cfc1 12070 Ngfrap1 12097 Bglap2 21816 Tgm1 12622 Cer1 19989 Rpl7 12267 C3ar1 67405 Nts 21385 Tbx2 19896 Rpl10a 12279 C9 435337 EG435337 56720 Tdo2 20044 Rps14 12391 Cav3 545913 Zscan4d 16869 Lhx1 19175 Psmb6 12409 Cbr2 244448 Triml1 22253 Unc5c 22627 Ywhae 12477 Ctla4 69134 2200001I15Rik 14174 Fgf3 19951 Rpl32 12523 Cd84 66065 Hsd17b14 16542 Kdr 66152 1110020P15Rik 12524 Cd86 81879 Tcfcp2l1 15122 Hba-a1 66489 Rpl35 12640 Cga 17907 Mylpf 15414 Hoxb6 15519 Hsp90aa1 12642 Ch25h 26424 Nr5a2 210530 Leprel1 66483 Rpl36al 12655 Chi3l3 83560 Tex14 12338 Capn6 27370 Rps26 12796 Camp 17450 Morc1 20671 Sox17 66576 Uqcrh 12869 Cox8b 79455 Pdcl2 20613 Snai1 22154 Tubb5 12959 Cryba4 231821 Centa1 17897
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • Evidence Revealing Deregulation of the KLF11-Mao a Pathway in Association with Chronic Stress and Depressive Disorders
    Neuropsychopharmacology (2015) 40, 1373–1382 & 2015 American College of Neuropsychopharmacology. All rights reserved 0893-133X/15 www.neuropsychopharmacology.org Evidence Revealing Deregulation of The KLF11-Mao A Pathway in Association with Chronic Stress and Depressive Disorders 1 1 1,2 1 3 Sharonda Harris , Shakevia Johnson , Jeremy W Duncan , Chinelo Udemgba , Jeffrey H Meyer , Paul R Albert4, Gwen Lomberk5, Raul Urrutia5, Xiao-Ming Ou1, Craig A Stockmeier1,6 and Jun Ming Wang*,1,2,7 1 2 3 Department of Psychiatry and Human Behavior, Jackson, MS, USA; Program in Neuroscience, Jackson, MS, USA; Centre for Addiction and Mental Health and Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; 4Ottawa Hospital Research Institute (Neuroscience), Ottawa, Ontario, Canada; 5Epigenetics and Chromatin Dynamics Laboratory, GI Research Unit, Mayo Clinic, Rochester, MN, 6 7 USA; Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA The biochemical pathways underlying major depressive disorder (MDD) and chronic stress are not well understood. However, it has been reported that monoamine oxidase A (MAO A, a major neurotransmitter-degrading enzyme) is significantly increased in the brains of human subjects affected with MDD and rats exposed to chronic social defeat (CSD) stress, which is used to model depression. In the current study, we compared the protein levels of a MAO A-transcriptional activator, Kruppel-like factor 11 (KLF11 , also recognized as transforming growth factor-beta-inducible early gene 2) between the brains of 18 human subjects with MDD and 18 control subjects. We found that, indeed, the expression of KLF11 is increased by 36% (po0.02) in the postmortem prefrontal cortex of human subjects with MDD compared with controls.
    [Show full text]
  • Genome-Wide DNA Methylation Analysis of KRAS Mutant Cell Lines Ben Yi Tew1,5, Joel K
    www.nature.com/scientificreports OPEN Genome-wide DNA methylation analysis of KRAS mutant cell lines Ben Yi Tew1,5, Joel K. Durand2,5, Kirsten L. Bryant2, Tikvah K. Hayes2, Sen Peng3, Nhan L. Tran4, Gerald C. Gooden1, David N. Buckley1, Channing J. Der2, Albert S. Baldwin2 ✉ & Bodour Salhia1 ✉ Oncogenic RAS mutations are associated with DNA methylation changes that alter gene expression to drive cancer. Recent studies suggest that DNA methylation changes may be stochastic in nature, while other groups propose distinct signaling pathways responsible for aberrant methylation. Better understanding of DNA methylation events associated with oncogenic KRAS expression could enhance therapeutic approaches. Here we analyzed the basal CpG methylation of 11 KRAS-mutant and dependent pancreatic cancer cell lines and observed strikingly similar methylation patterns. KRAS knockdown resulted in unique methylation changes with limited overlap between each cell line. In KRAS-mutant Pa16C pancreatic cancer cells, while KRAS knockdown resulted in over 8,000 diferentially methylated (DM) CpGs, treatment with the ERK1/2-selective inhibitor SCH772984 showed less than 40 DM CpGs, suggesting that ERK is not a broadly active driver of KRAS-associated DNA methylation. KRAS G12V overexpression in an isogenic lung model reveals >50,600 DM CpGs compared to non-transformed controls. In lung and pancreatic cells, gene ontology analyses of DM promoters show an enrichment for genes involved in diferentiation and development. Taken all together, KRAS-mediated DNA methylation are stochastic and independent of canonical downstream efector signaling. These epigenetically altered genes associated with KRAS expression could represent potential therapeutic targets in KRAS-driven cancer. Activating KRAS mutations can be found in nearly 25 percent of all cancers1.
    [Show full text]
  • Key Genes Regulating Skeletal Muscle Development and Growth in Farm Animals
    animals Review Key Genes Regulating Skeletal Muscle Development and Growth in Farm Animals Mohammadreza Mohammadabadi 1 , Farhad Bordbar 1,* , Just Jensen 2 , Min Du 3 and Wei Guo 4 1 Department of Animal Science, Faculty of Agriculture, Shahid Bahonar University of Kerman, Kerman 77951, Iran; [email protected] 2 Center for Quantitative Genetics and Genomics, Aarhus University, 8210 Aarhus, Denmark; [email protected] 3 Washington Center for Muscle Biology, Department of Animal Sciences, Washington State University, Pullman, WA 99163, USA; [email protected] 4 Muscle Biology and Animal Biologics, Animal and Dairy Science, University of Wisconsin-Madison, Madison, WI 53558, USA; [email protected] * Correspondence: [email protected] Simple Summary: Skeletal muscle mass is an important economic trait, and muscle development and growth is a crucial factor to supply enough meat for human consumption. Thus, understanding (candidate) genes regulating skeletal muscle development is crucial for understanding molecular genetic regulation of muscle growth and can be benefit the meat industry toward the goal of in- creasing meat yields. During the past years, significant progress has been made for understanding these mechanisms, and thus, we decided to write a comprehensive review covering regulators and (candidate) genes crucial for muscle development and growth in farm animals. Detection of these genes and factors increases our understanding of muscle growth and development and is a great help for breeders to satisfy demands for meat production on a global scale. Citation: Mohammadabadi, M.; Abstract: Farm-animal species play crucial roles in satisfying demands for meat on a global scale, Bordbar, F.; Jensen, J.; Du, M.; Guo, W.
    [Show full text]
  • Supplementary Materials
    Supplementary materials Supplementary Table S1: MGNC compound library Ingredien Molecule Caco- Mol ID MW AlogP OB (%) BBB DL FASA- HL t Name Name 2 shengdi MOL012254 campesterol 400.8 7.63 37.58 1.34 0.98 0.7 0.21 20.2 shengdi MOL000519 coniferin 314.4 3.16 31.11 0.42 -0.2 0.3 0.27 74.6 beta- shengdi MOL000359 414.8 8.08 36.91 1.32 0.99 0.8 0.23 20.2 sitosterol pachymic shengdi MOL000289 528.9 6.54 33.63 0.1 -0.6 0.8 0 9.27 acid Poricoic acid shengdi MOL000291 484.7 5.64 30.52 -0.08 -0.9 0.8 0 8.67 B Chrysanthem shengdi MOL004492 585 8.24 38.72 0.51 -1 0.6 0.3 17.5 axanthin 20- shengdi MOL011455 Hexadecano 418.6 1.91 32.7 -0.24 -0.4 0.7 0.29 104 ylingenol huanglian MOL001454 berberine 336.4 3.45 36.86 1.24 0.57 0.8 0.19 6.57 huanglian MOL013352 Obacunone 454.6 2.68 43.29 0.01 -0.4 0.8 0.31 -13 huanglian MOL002894 berberrubine 322.4 3.2 35.74 1.07 0.17 0.7 0.24 6.46 huanglian MOL002897 epiberberine 336.4 3.45 43.09 1.17 0.4 0.8 0.19 6.1 huanglian MOL002903 (R)-Canadine 339.4 3.4 55.37 1.04 0.57 0.8 0.2 6.41 huanglian MOL002904 Berlambine 351.4 2.49 36.68 0.97 0.17 0.8 0.28 7.33 Corchorosid huanglian MOL002907 404.6 1.34 105 -0.91 -1.3 0.8 0.29 6.68 e A_qt Magnogrand huanglian MOL000622 266.4 1.18 63.71 0.02 -0.2 0.2 0.3 3.17 iolide huanglian MOL000762 Palmidin A 510.5 4.52 35.36 -0.38 -1.5 0.7 0.39 33.2 huanglian MOL000785 palmatine 352.4 3.65 64.6 1.33 0.37 0.7 0.13 2.25 huanglian MOL000098 quercetin 302.3 1.5 46.43 0.05 -0.8 0.3 0.38 14.4 huanglian MOL001458 coptisine 320.3 3.25 30.67 1.21 0.32 0.9 0.26 9.33 huanglian MOL002668 Worenine
    [Show full text]
  • Ahn Supp. Fig. 1 AB 1.5 ARRDC4 1.5 ARRDC4 * * * 1.0 1.0
    Ahn_Supp. Fig. 1 AB 1.5 ARRDC4 1.5 ARRDC4 * * * 1.0 1.0 * * 0.5 * 0.5 * * * Relative mRNA levels mRNA Relative Relative mRNA levels mRNA Relative 0.0 0.0 1.5 MLXIP (MondoA) 1.5 MLXIP (MondoA) 1.0 1.0 0.5 0.5 Relative mRNA levels mRNA Relative Relative mRNA levels mRNA Relative 0.0 0.0 MondoA MondoA 0124824 Starvation (6h) -++++++ Glucose Starvation (h) Refeeding (h) --0.51248 C 1.5 ARRDC4 1.5 MLXIP (MondoA) † Con # KD 1.0 1.0 0.5 0.5 * * * * Relative mRNA levels mRNA Relative Relative mRNA levels mRNA Relative * * 0.0 0.0 BasalStarvation Refeeding BasalStarvation Refeeding MondoA Con + + - - + + - - + + - - KD - - + + - - + + - - + + BasalStarvation Refeeding Supplemental Figure 1. Glucose-mediated regulation of ARRDC4 is dependent on MondoA in human skeletal myotubes. (A) (top) ARRDC4 and MLXIP (MondoA) mRNA levels were determined by qRT-PCR in human skeletal myotubes following deprivation of glucose at the indicated time (n=4). (bottom) Representative Western blot analysis of MondoA demonstrating the effect of glucose deprivation. *p<0.05 vs. 0h. (B) (top) ARRDC4 and MLXIP (MondoA) expression in human myotubes following a 6h glucose removal and refeeding at the times indicated (n=4). (bottom) Corresponding Western blot analysis. *p<0.05 vs Starvation 6h. (C) (top) Expression of ARRDC4 and MLXIP in human myotubes following deprivation and refeeding of glucose in the absence or presence of siRNA-mediated MondoA KD (n=4). (bottom) Corresponding Western blot analysis. *p<0.05 vs siControl. # p<0.05. § p<0.05. The data represents mean ± SD. All statistical significance determined by one-way ANOVA with Tukey multiple comparison post-hoc test.
    [Show full text]
  • Open Chromatin Profiling in Adipose Tissue Marks Genomic Regions with Functional Roles in Cardiometabolic Traits
    INVESTIGATION Open Chromatin Profiling in Adipose Tissue Marks Genomic Regions with Functional Roles in Cardiometabolic Traits Maren E. Cannon,*,1 Kevin W. Currin,*,1 Kristin L. Young,† Hannah J. Perrin,* Swarooparani Vadlamudi,* Alexias Safi,‡ Lingyun Song,‡ Ying Wu,* Martin Wabitsch,§ Markku Laakso,** Gregory E. Crawford,‡ and Karen L. Mohlke*,2 *Department of Genetics, †Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, ‡Department of Pediatrics, Division of Medical Genetics and Center for Genomic and Computational Biology, § Duke University, NC 27710, Department of Pediatrics and Adolescents Medicine, Division of Pediatric Endocrinology and Diabetes, University of Ulm, Germany 89081, and **Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland 70210 ORCID ID: 0000-0001-6721-153X (K.L.M.) ABSTRACT Identifying the regulatory mechanisms of genome-wide association study (GWAS) loci KEYWORDS affecting adipose tissue has been restricted due to limited characterization of adipose transcriptional chromatin regulatory elements. We profiled chromatin accessibility in three frozen human subcutaneous adipose accessibility tissue needle biopsies and preadipocytes and adipocytes from the Simpson Golabi-Behmel Syndrome adipose tissue (SGBS) cell strain using an assay for transposase-accessible chromatin (ATAC-seq). We identified preadipocytes 68,571 representative accessible chromatin regions (peaks) across adipose tissue samples (FDR , 5%). GWAS GWAS loci for eight cardiometabolic traits were enriched in these peaks (P , 0.005), with the strongest cardiometabolic enrichment for waist-hip ratio. Of 110 recently described cardiometabolic GWAS loci colocalized with trait adiposetissueeQTLs,59locihadoneormorevariantsoverlappinganadiposetissuepeak.Annotated variants at the SNX10 waist-hip ratio locus and the ATP2A1-SH2B1 body mass index locus showed allelic differences in regulatory assays.
    [Show full text]
  • Tgfβ-Regulated Gene Expression by Smads and Sp1/KLF-Like Transcription Factors in Cancer VOLKER ELLENRIEDER
    ANTICANCER RESEARCH 28 : 1531-1540 (2008) Review TGFβ-regulated Gene Expression by Smads and Sp1/KLF-like Transcription Factors in Cancer VOLKER ELLENRIEDER Signal Transduction Laboratory, Internal Medicine, Department of Gastroenterology and Endocrinology, University of Marburg, Marburg, Germany Abstract. Transforming growth factor beta (TGF β) controls complex induces the canonical Smad signaling molecules which vital cellular functions through its ability to regulate gene then translocate into the nucleus to regulate transcription (2). The expression. TGFβ binding to its transmembrane receptor cellular response to TGF β can be extremely variable depending kinases initiates distinct intracellular signalling cascades on the cell type and the activation status of a cell at a given time. including the Smad signalling and transcription factors and also For instance, TGF β induces growth arrest and apoptosis in Smad-independent pathways. In normal epithelial cells, TGF β healthy epithelial cells, whereas it can also promote tumor stimulation induces a cytostatic program which includes the progression through stimulation of cell proliferation and the transcriptional repression of the c-Myc oncogene and the later induction of an epithelial-to-mesenchymal transition of tumor induction of the cell cycle inhibitors p15 INK4b and p21 Cip1 . cells (1, 3). In the last decade it has become clear that both the During carcinogenesis, however, many tumor cells lose their tumor suppressing and the tumor promoting functions of TGF β ability to respond to TGF β with growth inhibition, and instead, are primarily regulated on the level of gene expression through activate genes involved in cell proliferation, invasion and Smad-dependent and -independent mechanisms (1, 2, 4).
    [Show full text]
  • TCF3 Antibody Product Type: Primary Antibodies Description: Rabbit Polyclonal to TCF3 Immunogen:3 Synthetic Peptides (Human) Conjugated to KLH
    PRODUCT INFORMATION Product name: TCF3 antibody Product type: Primary antibodies Description: Rabbit polyclonal to TCF3 Immunogen:3 synthetic peptides (human) conjugated to KLH. Reacts with: Hu, Ms Tested applications: ELISA and WB. GENE INFORMATION Gene Symbol: TCF3 Gene Name: transcription factor 3 (E2A immunoglobulin enhancer binding factors E12/E47) Ensembl ID: ENSG00000071564 Entrez GeneID:6929 GenBank Accession number: M65214.1 Omim ID:147141 Swiss­Prot:P15923 Molecular weight of TCF3: 67.6kDa (Isoform E12) and 67.265kDa (Isoform E47) Function: Heterodimers between TCF3 and tissue­specific basic helix­loop­helix (bHLH) proteins play major roles in determining tissue­specific cell fate during embryogenesis, like muscle or early B­cell differentiation. Dimers bind DNA on E­box motifs: 5'­CANNTG­3'. Binds to the kappa­E2 site in the kappa immunoglobulin gene enhancer. Subunit structure: Efficient DNA binding requires dimerization with another bHLH protein. Forms a heterodimer with ASH1 and TWIST2. Isoform E12 interacts with GRIPE and FIGLA By similarity. Interacts with PTF1A and TGFB1I1. Component of a nuclear TAL­1 complex composed at least of CBFA2T3, LDB1, TAL1 and TCF3 By similarity. Interacts with UBE2I. Subcellular localization: Nucleus Involvement in disease:Chromosomal aberrations involving TCF3 are cause of forms of pre­B­cell acute lymphoblastic leukemia (B­ALL). Translocation t(1;19)(q23;p13.3) with PBX1; Translocation t(17;19)(q22;p13.3) with HLF. Inversion inv(19)(p13;q13) with TFPT. APPLICATION NOTE Recommended dilution: • ELISA: Antibody specificity was verified by direct ELISA against the 3 immunogen peptides. A titer of 1/15000 has been determined. Appropriate specificity controls were run.
    [Show full text]