MYH9 Binds to Lncrna Gene PTCSC2 and Regulates FOXE1 in the 9Q22 Thyroid Cancer Risk Locus

Total Page:16

File Type:pdf, Size:1020Kb

MYH9 Binds to Lncrna Gene PTCSC2 and Regulates FOXE1 in the 9Q22 Thyroid Cancer Risk Locus MYH9 binds to lncRNA gene PTCSC2 and regulates FOXE1 in the 9q22 thyroid cancer risk locus Yanqiang Wanga,b, Huiling Hea,b, Wei Lia,b, John Phayc, Rulong Shend, Lianbo Yue,f, Baris Hanciogluf, and Albert de la Chapellea,b,1 aHuman Cancer Genetics Program, The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210; bDepartment of Cancer Biology and Genetics, The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210; cDepartment of Surgery, The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210; dDepartment of Pathology, The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210; eCenter for Biostatistics, The Ohio State University, Columbus, OH 43210; and fDepartment of Biomedical Informatics, The Ohio State University, Columbus, OH 43210 Contributed by Albert de la Chapelle, December 7, 2016 (sent for review August 30, 2016; reviewed by Bryan Haugen and Shioko Kimura) A locus on chromosome 9q22 harbors a SNP (rs965513) firmly expression of both PTCSC2 and FOXE1 (15). Three enhancer associated with risk of papillary thyroid carcinoma (PTC). The locus elements are located in a 33-kb linkage disequilibrium (LD) also comprises the forkhead box E1 (FOXE1) gene, which is impli- block within PTCSC2. Previous studies showed that PTCSC2 is cated in thyroid development, and a long noncoding RNA (lncRNA) transcribed in the opposite direction of FOXE1.Exon1andintron1 gene, papillary thyroid cancer susceptibility candidate 2 (PTCSC2). of PTCSC2 isoform C overlap with the FOXE1 promoter region How these might interact is not known. Here we report that PTCSC2 (15). Although the promoter region shared by PTCSC2 and FOXE1 binds myosin-9 (MYH9). In a bidirectional promoter shared by FOXE1 was found to be regulated via long-range looping interactions (18), and PTCSC2, MYH9 inhibits the promoter activity in both directions. the detailed mechanisms and their bearing on thyroid cancer need This inhibition can be reversed by PTCSC2, which acts as a suppres- to be explored. sor. RNA knockdown of FOXE1 in primary thyroid cells profoundly In the present study, we identified myosin-9 (MYH9) as an interferes with the p53 pathway. We propose that the interaction lncRNA binding protein that targets the FOXE1 promoter re- between the lncRNA, its binding protein MYH9, and the coding gene gion through interactions with PTCSC2 and performs its regu- FOXE1 underlies the predisposition to PTC triggered by rs965513. latory function in thyroid cancer via downstream pathways. BIOCHEMISTRY These findings provide a better defined description of the com- lncRNA | MYH9 | transcriptional regulation | bidirectional promoter | plex mechanisms involved in the 9q22 thyroid cancer locus. thyroid cancer Results hyroid cancer is the most common endocrine malignancy. Identification of MYH9 as an LncRNA Binding Protein. To begin to TBased on the data from the National Cancer Institute for unravel the underlying mechanisms by identifying proteins po- 2016, in total 64,300 new patients will be diagnosed with thyroid tentially interacting with PTCSC2, we performed biotin-labeled cancer, accounting for 3.8% of all cancer patients in the United RNA pull-down assays with protein lysate from normal thyroid States (https://seer.cancer.gov/statfacts/html/thyro.html). Thyroid tissue. Two PTCSC2 isoforms—isoform C (1,947 nt) and isoform cancer is classified into four main types: papillary, follicular, D (1,804 nt)—were used as baits by generating both sense and medullary, and anaplastic. Papillary thyroid carcinoma (PTC) ac- antisense (negative control) RNA probes for each of them (Fig. S1). – counts for 85 90% of all thyroid cancers (1). In efforts to explain Asaresult,astrand-specificbinding protein was discovered between the genomic background to PTC, genome-wide association studies (GWASs) have disclosed an SNP marker, rs965513, in 9q22.33 Significance strongly associated with PTC in European populations [odds ratio − (OR) = 1.75; P = 1.7 × 10 27] (2, 3). The association has been observed in other populations as well (4–8). Additionally, rs965513 Papillary thyroid carcinoma (PTC) is the most common endocrine has been reported to be associated with levels of thyroid-related cancer and displays strong heritability. So far, the most significant hormones, hypothyroidism, goiter, and other abnormal thyroid known predisposing variant is rs965513 in 9q22. Although a long functions (2, 3, 9). SNP rs965513 resides in an intron of a recently noncoding RNA, papillary thyroid cancer susceptibility candidate 2 PTCSC2 detected long noncoding RNA (lncRNA), papillary thyroid cancer ( ), has been characterized in this locus, its mode of action in susceptibility candidate 2 (PTCSC2), which is located between two the carcinogenetic process is unknown. Here, we identify myosin- PTCSC2 coding genes, DNA damage recognition and repair factor (XPA) 9 (MYH9) as a binding protein of that regulates the bi- PTCSC2 and forkhead box E1 (FOXE1)(Fig. S1). directional promoter shared by and forkhead box E1 FOXE1 PTCSC2 FOXE1, also known as thyroid transcription factor 2, is a single ( ). can rescue the promoter inhibition caused by exon coding gene belonging to the forkhead/winged helix-domain MYH9. The p53 pathway is profoundly affected by the inhibition FOXE1 PTCSC2 protein family (10). FOXE1 knockout mice were born alive but of . Our study discovers fundamental roles for , FOXE1 exhibited an ectopic or completely absent thyroid gland and severe MYH9, and in thyroid cancer and provides a description of cleft palate, accompanied by up-regulated TSH and down-regulated the regulatory mechanism. free thyroxine hormone levels (11). FOXE1 is essential for thyroid Author contributions: Y.W., H.H., and A.d.l.C. designed research; Y.W. performed research; gland development and the maintenance of thyroid differentiated W.L., J.P., and R.S. contributed new reagents/analytic tools; Y.W., L.Y., and B.H. analyzed status (12, 13). It also plays a role in the predisposition and devel- data; and Y.W., H.H., B.H., and A.d.l.C. wrote the paper. opment of thyroid cancer (14, 15). Moreover, somatic mutational Reviewers: B.H., University of Colorado School of Medicine; and S.K., National Cancer inactivation of FOXE1 has been found in PTC, suggesting FOXE1 Institute/National Institutes of Health. may contribute to tumorigenesis in a subset of thyroid cancers (16). The authors declare no conflict of interest. PTCSC2 was discovered in the region harboring rs965513 on Data deposition: The data reported in this paper have been deposited in the Gene Ex- 9q22 (15). Similar to some 25% of all known lncRNA genes (17), pression Omnibus (GEO) database, www.ncbi.nlm.nih.gov/geo (accession no. GSE83919). PTCSC2 has one unspliced isoform and several spliced isoforms, 1To whom correspondence should be addressed. Email: [email protected]. all of which display thyroid-specific expression. In PTC tumors, This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. the risk allele [A] of rs965513 is significantly associated with low 1073/pnas.1619917114/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1619917114 PNAS Early Edition | 1of6 Downloaded by guest on September 30, 2021 As PTCSC2 and FOXE1 are transcribed in opposite directions (15) and there is only 174 bp between their transcription start sites (TSSs), we postulated that they are transcribed by the same pro- moter with bidirectional activity. To test this hypothesis, a shorter (1,607 bp) and a longer (2,520 bp) fragment cloned from the FOXE1 promoter region were used for luciferase reporter assay in the BCPAP cell line with both forward (Promoter-S and Pro- moter-L) and inverted (Promoter-Inv-S and Promoter-Inv-L) orientation, as indicated (Fig. 3A). Although the inverted promoters showed lower activity compared with their corresponding forward ones, the fragment with both orientations exhibited a significantly higher level of promoter activity in comparison with the no pro- moter control. Moreover, cotransfection with the MYH9 expression vector showed that MYH9 can significantly inhibit the luciferase activity in both the forward and inverted promoter fragments. That MYH9 functions as a suppressor was disclosed in both the shorter and longer promoter fragments (Fig. 3 B and C). Thus, MYH9 had an impact on the FOXE1 gene, but its role in the presence of lncRNA PTCSC2 was not known. We transiently cotransfected the PTCSC2 expression vector in the BCPAP cell line Fig. 1. Identification of MYH9 as a PTCSC2 binding protein. (A) RNA pull- in the presence or absence of the MYH9 expression construct. down experiment with nontumorous thyroid tissue extract. Biotin pull-down Because there was no significant difference in promoter activity assays followed by SDS/PAGE separation were used to isolate the binding protein of PTCSC2 isoform C. Antisense RNA of PTCSC2 isoform C was used as between the two fragments of different sizes, only the longer pro- the negative control. The arrows indicate the binding protein bands ∼226 moter sets were used. Interestingly, an increase in promoter lucif- KD and 42 KD in size, respectively. (B) Information for the identification of erase activity was observed with cotransfection of the PTCSC2 MYH9 by MS. The full length of MYH9 protein is 1,960 amino acids, as shown expression vector, irrespective of MYH9 overexpression. The ef- in the lower diagram. Blue boxes indicate the peptides identified by MS fect was significant on the forward Promoter-L, whereas it was not analysis. (C) qRT-PCR detection of the indicated RNAs retrieved by MYH9 significant on the inverted Promoter-Inv-L, suggesting a rescuing antibody (RIP assay) in the BCPAP cell line with stable PTCSC2 isoform C effect on the inhibition caused by MYH9 (Fig. 4 A and B). overexpression. IgG was used as a negative control. The relative fold change PTCSC2 < ’ To further validate the roles of MYH9 and in regu- was normalized to the IgG control.
Recommended publications
  • Accumulation of Rare Coding Variants in Genes Implicated in Risk of Human Cleft Lip with Or Without Cleft Palate
    HHS Public Access Author manuscript Author ManuscriptAuthor Manuscript Author Am J Med Manuscript Author Genet A. Author Manuscript Author manuscript; available in PMC 2020 July 01. Published in final edited form as: Am J Med Genet A. 2019 July ; 179(7): 1260–1269. doi:10.1002/ajmg.a.61183. Accumulation of Rare Coding Variants in Genes Implicated in Risk of Human Cleft Lip with or without Cleft Palate Nicholas J. Marini1,*, Kripa Asrani1, Wei Yang2, Jasper Rine1, Gary M Shaw2 1California Institute for Quantitative Biosciences, Department of Molecular and Cellular Biology, University of California, Berkeley, CA 94720, USA 2Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA Abstract Cleft lip with/without cleft palate (CLP) is a common craniofacial malformation with complex etiologies, reflecting both genetic and environmental factors. Most of the suspected genetic risk for CLP has yet to be identified. To further classify risk loci and estimate the contribution of rare variants, we sequenced the exons in 49 candidate genes in 323 CLP cases and 211 non-malformed controls. Our findings indicated that rare, protein-altering variants displayed markedly higher burdens in CLP cases at relevant loci. First, putative loss-of-function mutations (nonsense, frameshift) were significantly enriched among cases: 13 of 323 cases (~4%) harbored such alleles within these 49 genes, versus one such change in controls (p = 0.01). Second, in gene-level analyses, the burden of rare alleles showed greater case-association for several genes previously implicated in cleft risk. For example, BHMT displayed a 10-fold increase in protein-altering variants in CLP cases (p = 0.03), including multiple case occurrences of a rare frameshift mutation (K400fs).
    [Show full text]
  • 4-6 Weeks Old Female C57BL/6 Mice Obtained from Jackson Labs Were Used for Cell Isolation
    Methods Mice: 4-6 weeks old female C57BL/6 mice obtained from Jackson labs were used for cell isolation. Female Foxp3-IRES-GFP reporter mice (1), backcrossed to B6/C57 background for 10 generations, were used for the isolation of naïve CD4 and naïve CD8 cells for the RNAseq experiments. The mice were housed in pathogen-free animal facility in the La Jolla Institute for Allergy and Immunology and were used according to protocols approved by the Institutional Animal Care and use Committee. Preparation of cells: Subsets of thymocytes were isolated by cell sorting as previously described (2), after cell surface staining using CD4 (GK1.5), CD8 (53-6.7), CD3ε (145- 2C11), CD24 (M1/69) (all from Biolegend). DP cells: CD4+CD8 int/hi; CD4 SP cells: CD4CD3 hi, CD24 int/lo; CD8 SP cells: CD8 int/hi CD4 CD3 hi, CD24 int/lo (Fig S2). Peripheral subsets were isolated after pooling spleen and lymph nodes. T cells were enriched by negative isolation using Dynabeads (Dynabeads untouched mouse T cells, 11413D, Invitrogen). After surface staining for CD4 (GK1.5), CD8 (53-6.7), CD62L (MEL-14), CD25 (PC61) and CD44 (IM7), naïve CD4+CD62L hiCD25-CD44lo and naïve CD8+CD62L hiCD25-CD44lo were obtained by sorting (BD FACS Aria). Additionally, for the RNAseq experiments, CD4 and CD8 naïve cells were isolated by sorting T cells from the Foxp3- IRES-GFP mice: CD4+CD62LhiCD25–CD44lo GFP(FOXP3)– and CD8+CD62LhiCD25– CD44lo GFP(FOXP3)– (antibodies were from Biolegend). In some cases, naïve CD4 cells were cultured in vitro under Th1 or Th2 polarizing conditions (3, 4).
    [Show full text]
  • FOXE1-Dependent Regulation of Macrophage Chemotaxis by Thyroid Cells in Vitro and in Vivo
    International Journal of Molecular Sciences Article FOXE1-Dependent Regulation of Macrophage Chemotaxis by Thyroid Cells In Vitro and In Vivo Sara C. Credendino 1,2, Marta De Menna 1,3, Irene Cantone 1 , Carmen Moccia 1 , Matteo Esposito 1, Luigi Di Guida 1, Mario De Felice 1,2 and Gabriella De Vita 1,* 1 Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy; [email protected] (S.C.C.); [email protected] (M.D.M.); [email protected] (I.C.); [email protected] (C.M.); [email protected] (M.E.); [email protected] (L.D.G.); [email protected] (M.D.F.) 2 Institute of Experimental Endocrinology and Oncology “G. Salvatore”, National Research Council (CNR), 80131 Naples, Italy 3 DBMR-Department for BioMedical Research, University of Bern, 3012 Bern, Switzerland * Correspondence: [email protected]; Tel.: +39-081-746-3240 Abstract: Forkhead box E1 (FOXE1) is a lineage-restricted transcription factor involved in thyroid cancer susceptibility. Cancer-associated polymorphisms map in regulatory regions, thus affecting the extent of gene expression. We have recently shown that genetic reduction of FOXE1 dosage modifies multiple thyroid cancer phenotypes. To identify relevant effectors playing roles in thyroid cancer development, here we analyse FOXE1-induced transcriptional alterations in thyroid cells that do not express endogenous FOXE1. Expression of FOXE1 elicits cell migration, while transcriptome analysis reveals that several immune cells-related categories are highly enriched in differentially Citation: Credendino, S.C.; expressed genes, including several upregulated chemokines involved in macrophage recruitment. De Menna, M.; Cantone, I.; Moccia, C.; Accordingly, FOXE1-expressing cells induce chemotaxis of co-cultured monocytes.
    [Show full text]
  • Downloaded from Refseq Database ( Duct Development (After E7.5), in Which Both Ducts Re
    Roly et al. BMC Genomics (2020) 21:688 https://doi.org/10.1186/s12864-020-07106-8 RESEARCH ARTICLE Open Access Transcriptional landscape of the embryonic chicken Müllerian duct Zahida Yesmin Roly, Rasoul Godini, Martin A. Estermann, Andrew T. Major, Roger Pocock and Craig A. Smith* Abstract Background: Müllerian ducts are paired embryonic tubes that give rise to the female reproductive tract in vertebrates. Many disorders of female reproduction can be attributed to anomalies of Müllerian duct development. However, the molecular genetics of Müllerian duct formation is poorly understood and most disorders of duct development have unknown etiology. In this study, we describe for the first time the transcriptional landscape of the embryonic Müllerian duct, using the chicken embryo as a model system. RNA sequencing was conducted at 1 day intervals during duct formation to identify developmentally-regulated genes, validated by in situ hybridization. Results: This analysis detected hundreds of genes specifically up-regulated during duct morphogenesis. Gene ontology and pathway analysis revealed enrichment for developmental pathways associated with cell adhesion, cell migration and proliferation, ERK and WNT signaling, and, interestingly, axonal guidance. The latter included factors linked to neuronal cell migration or axonal outgrowth, such as Ephrin B2, netrin receptor, SLIT1 and class A semaphorins. A number of transcriptional modules were identified that centred around key hub genes specifying matrix-associated signaling factors; SPOCK1, HTRA3 and ADGRD1. Several novel regulators of the WNT and TFG-β signaling pathway were identified in Müllerian ducts, including APCDD1 and DKK1, BMP3 and TGFBI.A number of novel transcription factors were also identified, including OSR1, FOXE1, PRICKLE1, TSHZ3 and SMARCA2.
    [Show full text]
  • Potential Targets of FOXL2, a Transcription Factor Involved in Craniofacial and Follicular Development, Identified by Transcriptomics
    Potential targets of FOXL2, a transcription factor involved in craniofacial and follicular development, identified by transcriptomics Frank Batista*, Daniel Vaiman*†, Jean Dausset‡§, Marc Fellous*¶, and Reiner A. Veitia*¶ʈ *De´partement de Ge´ne´ tique et De´veloppement, Institut Cochin, Institut National de la Sante´et de la Recherche Me´dicale U567, Centre National de la Recherche Scientifique Unite´Mixte de Recherche 8104, and Faculte´deMe´ decine Rene´Descartes, Universite´Paris V UM 3, 75014 Paris, France; †De´partement de Ge´ne´ tique Animale, Institut National de la Recherche Agronomique, 75338 Paris Cedex 07, France; ‡Fondation Jean Dausset, Centre d’Etude du Polymorphisme Humain, 75010 Paris, France; and ¶Universite´Denis Diderot/Paris VII, 75005 Paris, France Contributed by Jean Dausset, December 21, 2006 (sent for review November 16, 2006) FOXL2 is a gene encoding a forkhead transcription factor, polyAla has recently been reported in a nonsyndromic (i.e., not whose mutations are responsible for the blepharophimosis-ptosis- BPES-related) case of premature ovarian failure (9). epicanthus inversus syndrome that often involves premature ovar- In humans, FOXL2 is one of the earliest known markers of ian failure. FOXL2 is one of the earliest ovarian markers and it ovarian differentiation (3). Thus, it may play a role at an early stage offers, along with its targets, an excellent model to study ovarian of development of the ovarian somatic compartment. Because development and function in normal and pathological conditions. FOXL2 is still strongly expressed in postnatal and adult follicular We have recently shown that the aromatase gene is a target of cells, it may also play a role throughout female fertile life in FOXL2, and only three other targets have been reported so far.
    [Show full text]
  • Endogenous Rnai Pathways in C. Elegans* Allison C
    Endogenous RNAi pathways in C. elegans* Allison C. Billi1, Sylvia E. J. Fischer2, and John K. Kim1§ 1Life Sciences Institute, Department of Human Genetics, University of Michigan, Ann Arbor MI, USA 2Department of Molecular Biology, Massachusetts General Hospital, Department of Genetics, Harvard Medical School, Boston MA, USA Table of Contents 1. Introduction ............................................................................................................................3 2. Features and targets of 26G RNAs .............................................................................................. 4 3. Biogenesis of 26G RNAs: The ERI complex ................................................................................. 5 3.1. RRF-3 .........................................................................................................................8 3.2. DRH-3 ........................................................................................................................8 3.3. ERI-5 ..........................................................................................................................9 3.4. DCR-1 ........................................................................................................................9 3.5. DCR-1 helicase domain .................................................................................................. 9 3.6. ERI-1b ...................................................................................................................... 10 3.7. ERI-3 .......................................................................................................................
    [Show full text]
  • Pleiotropy Method Identifies Genetic Overlap Between Orofacial Clefts at Multiple Loci from GWAS of Multi-Ethnictrios
    medRxiv preprint doi: https://doi.org/10.1101/2020.11.13.20231654; this version posted November 16, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. It is made available under a CC-BY-ND 4.0 International license . Pleiotropy method identifies genetic overlap between orofacial clefts at multiple loci from GWAS of multi-ethnic trios Debashree Ray1;2;B, Sowmya Venkataraghavan1, Wanying Zhang1, Elizabeth J. Leslie3, Jacqueline B. Hetmanski1, Mary L Marazita4;5;6, Ingo Ruczinski2, Margaret A. Taub2 and Terri H. Beaty1;2;B 1Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA. 2Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA. 3Department of Human Genetics, School of Medicine, Emory University, Atlanta, GA, USA. 4Department of Oral Biology, School of Dental Medicine, University of Pittsburgh, PA, USA. 5Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, PA, USA. 6Center for Craniofacial and Dental Genetics, University of Pittsburgh, PA, USA. BEmail: [email protected]; [email protected] Based on epidemiologic and embryologic patterns, nonsyndromic orofacial clefts are commonly categorized into cleft lip with or without cleft palate (CL/P) and cleft palate alone (CP). While nearly forty risk genes have been identified for CL/P, few risk genes are known for CP. We used a new statistical method, PLACO, to identify genetic variants influencing risk of both CL/P and CP. In a combined multi-ethnic genome-wide study of 2,771 CL/P and 611 CP case-parent trios, we discovered 6 new loci of genetic overlap between CL/P and CP; 3 new loci between pairwise OFC subtypes; and 4 loci not previously implicated in OFCs.
    [Show full text]
  • DISRUPTION of NEURAL CREST ENHANCER LANDSCAPES AS an ETIOLOGICAL MECHANISM for HUMAN NEUROCRISTOPATHIES I Naugural
    DISRUPTION OF NEURAL CREST ENHANCER LANDSCAPES AS AN ETIOLOGICAL MECHANISM FOR HUMAN NEUROCRISTOPATHIES I n a u g u r a l – D i s s e r t a t i o n zur Erlangung des Doktorgrades der Mathematisch-Naturwissenschaftlichen Fakultät der Universität zu Köln vorgelegt von Michaela Bartusel aus Koblenz Köln 2019 Berichterstatter/in: Prof. Dr. Brunhilde Wirth Prof. Dr. Mirka Uhlirova Tag der letzten mündlichen Prüfung: 06.12.2019 Table of Contents TABLE OF CONTENTS Table of Contents .................................................................................................................................................. 3 Kurzzusammenfassung .......................................................................................................................................... 7 Abstract ................................................................................................................................................................ 9 1 Introduction ............................................................................................................................................11 1.1 Transcriptional regulation of the genome .................................................................................................... 11 1.1.1 Enhancers as key cis-regulatory elements .............................................................................................. 12 1.1.2 Enhancers in human disease ..................................................................................................................
    [Show full text]
  • Genetic and Genomics Laboratory Tools and Approaches
    Genetic and Genomics Laboratory Tools and Approaches Meredith Yeager, PhD Cancer Genomics Research Laboratory Division of Cancer Epidemiology and Genetics [email protected] DCEG Radiation Epidemiology and Dosimetry Course 2019 www.dceg.cancer.gov/RadEpiCourse (Recent) history of genetics 2 Sequencing of the Human Genome Science 291, 1304-1351 (2001) 3 The Human Genome – 2019 • ~3.3 billion bases (A, C, G, T) • ~20,000 protein-coding genes, many non-coding RNAs (~2% of the genome) • Annotation ongoing – the initial sequencing in 2001 is still being refined, assembled and annotated, even now – hg38 • Variation (polymorphism) present within humans – Population-specific – Cosmopolitan 4 Types of polymorphisms . Single nucleotide polymorphisms (SNPs) . Common SNPs are defined as > 5% in at least one population . Abundant in genome (~50 million and counting) ATGGAACGA(G/C)AGGATA(T/A)TACGCACTATGAAG(C/A)CGGTGAGAGG . Repeats of DNA (long, short, complex, simple), insertions/deletions . A small fraction of SNPs and other types of variation are very or slightly deleterious and may contribute by themselves or with other genetic or environmental factors to a phenotype or disease 5 Different mutation rates at the nucleotide level Mutation type Mutation rate (per generation) Transition on a CpG 1.6X10-7 Transversion on a CpG 4.4X10-8 Transition: purine to purine Transition out of CpG 1.2X10-8 Transversion: purine to pyrimidine Transversion out of CpG 5.5X10-9 Substitution (average) 2.3X10-8 A and G are purines Insertion/deletion (average) 2.3X10-9 C and T are pyrimidines Mutation rate (average) 2.4X10-8 . Size of haploid genome : 3.3X109 nucleotides .
    [Show full text]
  • Expresión Y Función De Los Factores De Transcripción Foxe1 Y Sox9 En La Célula Folicular Tiroidea
    UNIVERSIDAD AUTÓNOMA DE MADRID DEPARTAMENTO DE BIOQUÍMICA EXPRESIÓN Y FUNCIÓN DE LOS FACTORES DE TRANSCRIPCIÓN FOXE1 Y SOX9 EN LA CÉLULA FOLICULAR TIROIDEA TESIS DOCTORAL ARÍSTIDES LÓPEZ MÁRQUEZ MADRID, 2017 Departamento de Bioquímica Facultad de Medicina Universidad Autónoma de Madrid Expresión y función de los factores de transcripción FoxE1 y Sox9 en la célula folicular tiroidea Memoria de Tesis Doctoral presentada por: Arístides López Márquez Licenciado en Bioquímica Para optar al grado de Doctor por la Universidad Autónoma de Madrid Directora de Tesis: Dra. Pilar Santisteban Sanz Profesora de Investigación del CSIC Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM Madrid, 2017 Pilar Santisteban Sanz, Doctora en Biología y Profesora de Investigación del Instituto de Investigaciones Biomédicas “Alberto Sols” CSIC-UAM, CERTIFICA QUE: Arístides López Márquez, Licenciado en Bioquímica por la Universidad Autónoma de Madrid, ha realizado, bajo mi dirección, el trabajo: “Expresión y función de los factores de transcripción FoxE1 y Sox9 en la célula folicular tiroidea” En esta memoria de tesis doctoral se ha estudiado la regulación de la expresión por TSH y TGFβ de los factores de transcripción FoxE1 y Sox9 en la diferenciación de las células foliculares tiroideas. Mediante análisis in sílico y ensayos funcionales se ha descrito la existencia de una intercomunicación cruzada entre las vías de cAMP/PKA y las proteínas Smads. Asimismo se ha descrito la participación de otros factores de transcripción como Nkx2-1 y Pax8 en ese circuito regulatorio del control de la diferenciación tiroidea. Para llevar a cabo este trabajo se han usado como modelos cultivos celulares, células madre embrionarias (mESC), células foliculares de tiroides de rata y ratón embrionario y adulto.
    [Show full text]
  • Engineered Type 1 Regulatory T Cells Designed for Clinical Use Kill Primary
    ARTICLE Acute Myeloid Leukemia Engineered type 1 regulatory T cells designed Ferrata Storti Foundation for clinical use kill primary pediatric acute myeloid leukemia cells Brandon Cieniewicz,1* Molly Javier Uyeda,1,2* Ping (Pauline) Chen,1 Ece Canan Sayitoglu,1 Jeffrey Mao-Hwa Liu,1 Grazia Andolfi,3 Katharine Greenthal,1 Alice Bertaina,1,4 Silvia Gregori,3 Rosa Bacchetta,1,4 Norman James Lacayo,1 Alma-Martina Cepika1,4# and Maria Grazia Roncarolo1,2,4# Haematologica 2021 Volume 106(10):2588-2597 1Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, USA; 2Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford School of Medicine, Stanford, CA, USA; 3San Raffaele Telethon Institute for Gene Therapy, Milan, Italy and 4Center for Definitive and Curative Medicine, Stanford School of Medicine, Stanford, CA, USA *BC and MJU contributed equally as co-first authors #AMC and MGR contributed equally as co-senior authors ABSTRACT ype 1 regulatory (Tr1) T cells induced by enforced expression of interleukin-10 (LV-10) are being developed as a novel treatment for Tchemotherapy-resistant myeloid leukemias. In vivo, LV-10 cells do not cause graft-versus-host disease while mediating graft-versus-leukemia effect against adult acute myeloid leukemia (AML). Since pediatric AML (pAML) and adult AML are different on a genetic and epigenetic level, we investigate herein whether LV-10 cells also efficiently kill pAML cells. We show that the majority of primary pAML are killed by LV-10 cells, with different levels of sensitivity to killing. Transcriptionally, pAML sensitive to LV-10 killing expressed a myeloid maturation signature.
    [Show full text]
  • FOXE1 Regulates Migration and Invasion in Thyroid Cancer Cells and Targets ZEB1
    27 3 Endocrine-Related J Morillo-Bernal et al. Role of FOXE1 in thyroid 27:3 137–151 Cancer cancer RESEARCH FOXE1 regulates migration and invasion in thyroid cancer cells and targets ZEB1 Jesús Morillo-Bernal1, Lara P Fernández1,2 and Pilar Santisteban1,3 1Instituto de Investigaciones Biomédicas ‘Alberto Sols’, Consejo Superior Investigaciones Científicas, and Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain 2Molecular Oncology Group, IMDEA Food Institute, CEI UAM-CSIC, Madrid, Spain 3Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain Correspondence should be addressed to P Santisteban: [email protected] Abstract FOXE1 is a thyroid-specific transcription factor essential for thyroid gland development Key Words and maintenance of the differentiated state. Interestingly, a strong association has been f thyroid cancer recently described between FOXE1 expression and susceptibility to thyroid cancer, but f FOXE1 little is known about the mechanisms underlying FOXE1-induced thyroid tumorigenesis. f ZEB1 Here, we used a panel of human thyroid cancer-derived cell lines covering the spectrum f SNP of thyroid cancer phenotypes to examine FOXE1 expression and to test for correlations f EMT between FOXE1 expression, the allele frequency of two SNPs and a length polymorphism in or near the FOXE1 locus associated with cancer susceptibility, and the migration ability of thyroid cancer cell lines. Results showed that FOXE1 expression correlated with differentiation status according to histological sub-type, but not with SNP genotype or cell migration ability. However, loss-and-gain-of-function experiments revealed that FOXE1 modulates cell migration, suggesting a role in epithelial-to-mesenchymal transition (EMT).
    [Show full text]