Promoter Hypermethylation Silences Expression of the Hoxa4 Gene and Correlates with Igvh Mutational Status in CLL

Total Page:16

File Type:pdf, Size:1020Kb

Promoter Hypermethylation Silences Expression of the Hoxa4 Gene and Correlates with Igvh Mutational Status in CLL Letters to the Editor 1326 using antigen-specific, antibody-bound microbeads (Mittenyi It remains unclear as to how a single somatic mutation can Biotech, Survey, UK) on an AutoMacs cell separator (Mittenyi give rise to such a heterogeneous clinical presentation, and it Biotech). In addition, haematopoietic progenitor cell-derived seems likely that additional changes in either the signalling colonies were generated from BM aspirate low-density cells. cascade or other genetic alterations may contribute to the Cultures were supplemented with granulocyte–macrophage disease phenotype. Our report adds to the observation that JAK2 colony-stimulating factor (GM-CSF) alone, GM-CSF with Epo mutation is not an infrequent event in CNL. All patients with and GM-CSF, Epo IL-3 and stem cell factor. Individual colony- newly diagnosed CNL should be screened to determine the true forming units granulocyte/macrophage (CFU-GM)-derived co- incidence of the mutation. Given the poor outcome of this lonies from GM-CSF-supplemented and fully supplemented disease, patients with CNL and JAK2 mutation should be consi- cultures were harvested for JAK2 analysis by polymerase chain dered as potential candidates for molecular-targeted therapy. reaction (PCR). For PCR performed on haematopoietic colonies, colonies were placed directly into the PCR reaction without NC Lea1,4, Z Lim1,2,4, NB Westwood1, MJ Arno3,JGa¨ken1, prior DNA extraction. Cell separation kits were used according A Mohamedali1 and GJ Mufti1,2 to the manufacturer’s instructions. The presence of the mutant 1Department of Haematological Medicine, Kings College JAK2 allele was detected using an allele-specific PCR (AS-PCR).4 London, London, UK; The presence of the mutation and the ratio of mutant to wild- 2Department of Haematological Medicine, Guy’s King’s, type JAK2 allele were confirmed by pyrosequencing. Polymer- Thomas’ School of Medicine, Denmark Hill Campus, Kings College Hospital, London, UK and ase chain reaction products were generated using the AS-PCR 3 primer sequences. Sequences were read from a reverse Genomics Center, School of Biomedical and Health 0 0 Sciences, Kings College London, London, UK sequencing primer 5 -TCTCGTCTCCACAGA-3 . Pyrosequen- E-mail: [email protected] cing reactions were run on a Biotage PSQ HS 96 pyrosequencer. 4These authors contributed equally to this work Allele-specific polymerase chain reaction was used to detect the JAK2 V617F mutation in whole-blood, sub-fractionated BM and CFU-GM-derived haematopoietic colonies. The presence of the JAK2 V617F mutation was detected in whole peripheral blood and BM and was also found in all BM cell lineages with References the exception of the lymphoid compartment (CD3- and CD19- positive cells). Day 14 cultures contained only CFU-GM-derived 1 Imbert M, Bain B, Pierre R. Chronic neutrophilic leukaemia. In: Jaffe colonies. BFU-E-derived and mixed lineage colonies were not ES, Harris NL, Stein H et al. (eds). World Health Organisation detected. All colonies picked and amplified by AS-PCR were Classification of Tumours. Pathology and Genetics of Tumours of positive for the V617F mutation (data not shown). Pyrosequen- and Lymphoid Tissues. IARC Press: Lyon, 2001, pp. 27–28. 2 Elliot MA, Hanson CA, Dewald GW, Smoley SA, Lasho TL, Tefferi cing of subfractionated BM cells confirms the presence of the A. WHO-defined chronic neutrophilia leukaemia: a long-term JAK2 mutation in CD14-, CD15-, CD34-, CD61- and GPA- analysis of 12 cases and a critical review of the literature. positive cells and its absence in CD3- and CD19-positive cells Leukaemia 2005; 19: 313–317. (Figure 1). In addition, homo/heterozygosity calling by the 3 James C, Ugo V, LeCouedic JP, Staerk J, Delhommeau F, Lacout C Biotage software indicates that the mutation is homozygous and et al. A unique clonal JAK2 mutation leading to constitutive that the BM subcellular fractions are almost 100% clonal with signaling causes polycythaemia vera. Nature 2005; 434: 1144– 1148. respect to the JAK2 mutant allele. 4 Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S Two other cases have recently described the concurrent et al. Acquired mutation of the tyrosine kinase JAK2 in human presence of the JAK2 V17F mutation in the context of CNL with myeloproliferative disorders. Lancet 2005; 365: 1054–1061. one case diagnosed with concurrent B-cell non-Hodgkin’s 5 Kralovics R, Pssamonti F, Buser A, Soon-Siong Teo BS, lymphoma.7,8 Both case reports described the homozygous Tiedt R, Passweg JR et al. A gain-of-function mutation of presence of the JAK2 mutation within granulocytes as observed JAK2 in myeloproliferative disorders. N Engl J Med 2005; 352: 1779–1790. in our case. In addition, we have also performed a lineage- 6 Jones AV, Kriel S, Zoi K, Waghorn K, Curtis C, Zhang L et al. specific analysis and our data demonstrate that the mutation is Widespread occurrence of the JAK2 V617F mutation in chronic present in all myeloid and erythroid cells tested, but is absent myeloproliferative disorders. Blood 2005; 106: 2162–2168. from the lymphoid compartment. We have also found homo- 7 Steensma DP, Dewald GW, Lasho TL, Powell HL, McClure RF, zygous-mutated JAK2 present in 100% of CFU-GM-derived Levine RL et al. The JAK2 V617F activating tyrosine kinase mutation haematopoietic colonies. These observations concur with separate is an infrequent event in both ‘atypical’ myeloproliferative disorders and myelodysplastic syndromes. Blood 2005; 106: 1207–1209. studies on patients with MPDs, which have shown that the JAK2 8 McLornan DP, Percy MJ, Jones AV, Cross NCP, McMullin MF. mutation is likely to occur as a myeloid lineage-specific Chronic neutrophilic leukaemia with an associated V617F Jak2 mutation. tyrosine kinase mutation. Haematologica 2005; 90: 1696–1697. Promoter hypermethylation silences expression of the HoxA4 gene and correlates with IgVh mutational status in CLL Leukemia (2006) 20, 1326–1329. doi:10.1038/sj.leu.2404254; HoxD). They each encode homeodomain containing transcrip- published online 11 May 2006 tion factors, which are known to be key regulators of embryonic development.1 The Hox genes are also expressed in adult cells The human Hox genes are a large gene family, consisting of 39 where they play important roles in the control of cellular members, which are all located in four gene clusters (HoxA– differentiation, particularly during haematopoiesis.2 Several Hox Leukemia Letters to the Editor 1327 gene family members (particularly those in the 50 end of clusters, paralogs 9–13) are involved in leukaemia-associated transloca- tions2 and aberrant expression of Hox genes is a common event in leukaemogenesis. Indeed overexpression of multiple Hox genes (particularly within the HoxA cluster) has been identified in both lymphoid and myeloid malignancies.2 Although the potential mechanisms leading to increased Hox gene expression have been widely studied, aberrant suppression of Hox genes (which are expressed in normal haematopoietic progenitors) has received little attention. Alterations in the patterns of DNA methylation are critical in the development of all types of cancer,3 and transcriptional repression owing to hypermethylation of promoter-associated CpG islands has been shown to inactivate many genes whose downregulation is known to be important in tumour development, both in haematological malignancies and in solid tumours.4,5 All but one of the HoxA cluster genes (HoxA3) have CpG islands asso- Figure 1 Examples of HoxA4 COBRA analysis in CLL patient ciated with their transcriptional start site (sequence derived from samples. Examples of COBRA analysis for samples which were clones RP1-170O19 (AC004080), CTD-2536K9 (AC010990), unmutated at IgVh (a) or mutated at IgVh (b), using the upstream (upper panel) and downstream (lower panel) primer sets. Upper RP1-167F23 (AC004079) in GenBank), suggesting that altered (unmethylated) and lower (methylated) bands are indicated. The DNA methylation is a potential candidate mechanism for altered presence ( þ ) or absence (À) of hypermethylation is indicated below Hox gene expression in leukaemia. each sample. IVM, 100% in vitro methylated positive control. B cells, We chose to investigate the methylation status of the HoxA4 purified normal B-lymphocytes. gene, which is the most 30 HoxA gene previously identified as expressed in haematopoietic progenitor cells.6 Although Hox genes at the 50 end of the cluster, particularly HoxA9, have been implicated as oncogenes,7 the role of the more 30 genes is less clear. Initially, a test panel of 21 chronic lymphocytic leukaemia (CLL) samples were analysed for HoxA4 methylation using the COBRA assay, which allows semiquantitative assessment of methylation levels at specific CpG sites (the percentage of COBRA PCR product digested in this assay is equivalent to the percentage of methylated DNA in the sample). Quantitation of methylation levels was carried out by measurement of methylated and unmethylated band intensities using the GeneTools system (Syngene). The values were then corrected for the smaller size (and thus lower ethidium bromide binding capacity) of the methylated band and plotted against those Figure 2 Hypermethylation of the HoxA4 CpG island correlates obtained using samples, derived from in vitro methylated DNA, with loss of gene expression. Real time qRT–PCR analysis (using the of known methylation status (100, 66 and 33% methylated DyNAmo SYBR Green qPCR kit (Finnzymes) and an Opticon 2 DNA controls were used). To account for potential variation in the engine
Recommended publications
  • Prospective Isolation of NKX2-1–Expressing Human Lung Progenitors Derived from Pluripotent Stem Cells
    The Journal of Clinical Investigation RESEARCH ARTICLE Prospective isolation of NKX2-1–expressing human lung progenitors derived from pluripotent stem cells Finn Hawkins,1,2 Philipp Kramer,3 Anjali Jacob,1,2 Ian Driver,4 Dylan C. Thomas,1 Katherine B. McCauley,1,2 Nicholas Skvir,1 Ana M. Crane,3 Anita A. Kurmann,1,5 Anthony N. Hollenberg,5 Sinead Nguyen,1 Brandon G. Wong,6 Ahmad S. Khalil,6,7 Sarah X.L. Huang,3,8 Susan Guttentag,9 Jason R. Rock,4 John M. Shannon,10 Brian R. Davis,3 and Darrell N. Kotton1,2 2 1Center for Regenerative Medicine, and The Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA. 3Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, Texas, USA. 4Department of Anatomy, UCSF, San Francisco, California, USA. 5Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA. 6Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, Massachusetts, USA. 7Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA. 8Columbia Center for Translational Immunology & Columbia Center for Human Development, Columbia University Medical Center, New York, New York, USA. 9Department of Pediatrics, Monroe Carell Jr. Children’s Hospital, Vanderbilt University, Nashville, Tennessee, USA. 10Division of Pulmonary Biology, Cincinnati Children’s Hospital, Cincinnati, Ohio, USA. It has been postulated that during human fetal development, all cells of the lung epithelium derive from embryonic, endodermal, NK2 homeobox 1–expressing (NKX2-1+) precursor cells.
    [Show full text]
  • Integrative Bulk and Single-Cell Profiling of Premanufacture T-Cell Populations Reveals Factors Mediating Long-Term Persistence of CAR T-Cell Therapy
    Published OnlineFirst April 5, 2021; DOI: 10.1158/2159-8290.CD-20-1677 RESEARCH ARTICLE Integrative Bulk and Single-Cell Profiling of Premanufacture T-cell Populations Reveals Factors Mediating Long-Term Persistence of CAR T-cell Therapy Gregory M. Chen1, Changya Chen2,3, Rajat K. Das2, Peng Gao2, Chia-Hui Chen2, Shovik Bandyopadhyay4, Yang-Yang Ding2,5, Yasin Uzun2,3, Wenbao Yu2, Qin Zhu1, Regina M. Myers2, Stephan A. Grupp2,5, David M. Barrett2,5, and Kai Tan2,3,5 Downloaded from cancerdiscovery.aacrjournals.org on October 1, 2021. © 2021 American Association for Cancer Research. Published OnlineFirst April 5, 2021; DOI: 10.1158/2159-8290.CD-20-1677 ABSTRACT The adoptive transfer of chimeric antigen receptor (CAR) T cells represents a breakthrough in clinical oncology, yet both between- and within-patient differences in autologously derived T cells are a major contributor to therapy failure. To interrogate the molecular determinants of clinical CAR T-cell persistence, we extensively characterized the premanufacture T cells of 71 patients with B-cell malignancies on trial to receive anti-CD19 CAR T-cell therapy. We performed RNA-sequencing analysis on sorted T-cell subsets from all 71 patients, followed by paired Cellular Indexing of Transcriptomes and Epitopes (CITE) sequencing and single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) on T cells from six of these patients. We found that chronic IFN signaling regulated by IRF7 was associated with poor CAR T-cell persistence across T-cell subsets, and that the TCF7 regulon not only associates with the favorable naïve T-cell state, but is maintained in effector T cells among patients with long-term CAR T-cell persistence.
    [Show full text]
  • Expression of the Murine Hoxa4 Gene Requires Both Autoregulation and a Conserved Retinoic Acid Response Element
    Development 125, 1991-1998 (1998) 1991 Printed in Great Britain © The Company of Biologists Limited 1998 DEV4970 Expression of the murine Hoxa4 gene requires both autoregulation and a conserved retinoic acid response element Alan I. Packer1,3, David A. Crotty1,3,*, Vivian A. Elwell3 and Debra J. Wolgemuth1-4,† 1Department of Genetics and Development and 2Obstetrics and Gynecology, 3The Center for Reproductive Sciences and the 4Columbia Cancer Center, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA *Present address: Department of Biology, California Institute of Technology, Pasadena, CA 91125, USA †Author for correspondence at address 1 (e-mail: [email protected]) Accepted 30 March; published on WWW 6 May 1998 SUMMARY Analysis of the regulatory regions of the Hox genes has flanking the Hoxd4 gene. Administration of retinoic acid to revealed a complex array of positive and negative cis-acting Hoxa4/lacZ transgenic embryos resulted in stage- elements that control the spatial and temporal pattern of dependent ectopic expression of the reporter gene in the expression of these genes during embryogenesis. In this neural tube and hindbrain. When administered to Hoxa4 study we show that normal expression of the murine Hoxa4 null embryos, however, persistent ectopic expression was gene during development requires both autoregulatory and not observed, suggesting that autoregulation is required for retinoic acid-dependent modes of regulation. When maintenance of the retinoic acid-induced expression. introduced into a Hoxa4 null background, expression of a Finally, mutation of the consensus retinoic acid response lacZ reporter gene driven by the Hoxa4 regulatory region element eliminated the response of the reporter gene to (Hoxa4/lacZ) is either abolished or significantly reduced in exogenous retinoic acid, and abolished all embryonic all tissues at E10.5-E12.5.
    [Show full text]
  • IGF1-Mediated HOXA13 Overexpression Promotes Colorectal
    Qiao et al. Cell Death and Disease (2021) 12:564 https://doi.org/10.1038/s41419-021-03833-2 Cell Death & Disease ARTICLE Open Access IGF1-mediated HOXA13 overexpression promotes colorectal cancer metastasis through upregulating ACLY and IGF1R Chenyang Qiao1, Wenjie Huang2,3,JieChen1,WeiboFeng1, Tongyue Zhang2, Yijun Wang2,DanfeiLiu2,XiaoyuJi2, Meng Xie2, Mengyu Sun2,DaimingFan1,KaichunWu1 and Limin Xia 2 Abstract Metastasis is the major reason for the high mortality of colorectal cancer (CRC) patients and its molecular mechanism remains unclear. Here, we report a novel role of Homeobox A13 (HOXA13), a member of the Homeobox (HOX) family, in promoting CRC metastasis. The elevated expression of HOXA13 was positively correlated with distant metastasis, higher AJCC stage, and poor prognosis in two independent CRC cohorts. Overexpression of HOXA13 promoted CRC metastasis whereas downregulation of HOXA13 suppressed CRC metastasis. Mechanistically, HOXA13 facilitated CRC metastasis by transactivating ATP-citrate lyase (ACLY) and insulin-like growth factor 1 receptor (IGF1R). Knockdown of ACLY and IGFIR inhibited HOXA13-medicated CRC metastasis, whereas ectopic overexpression of ACLY and IGFIR rescued the decreased CRC metastasis induced by HOXA13 knockdown. Furthermore, Insulin-like growth factor 1 (IGF1), the ligand of IGF1R, upregulated HOXA13 expression through the PI3K/AKT/HIF1α pathway. Knockdown of HOXA13 decreased IGF1-mediated CRC metastasis. In addition, the combined treatment of ACLY inhibitor ETC-1002 and IGF1R inhibitor Linsitinib 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; dramatically suppressed HOXA13-mediated CRC metastasis. In conclusion, HOXA13 is a prognostic biomarker in CRC patients. Targeting the IGF1-HOXA13-IGF1R positive feedback loop may provide a potential therapeutic strategy for the treatment of HOXA13-driven CRC metastasis.
    [Show full text]
  • Respiratory Adaptations to Lung Morphological Defects in Adult Mice Lacking Hoxa5 Gene Function
    0031-3998/04/5604-0553 PEDIATRIC RESEARCH Vol. 56, No. 4, 2004 Copyright © 2004 International Pediatric Research Foundation, Inc. Printed in U.S.A. Respiratory Adaptations to Lung Morphological Defects in Adult Mice Lacking Hoxa5 Gene Function RICHARD KINKEAD, MICHELLE LEBLANC, ROUMIANA GULEMETOVA, MÉLANIE LALANCETTE-HÉBERT, MARGOT LEMIEUX, ISABEL MANDEVILLE, AND LUCIE JEANNOTTE Département de Pédiatrie [R.K., R.G.], Centre Hospitalier Universitaire de Québec, Centre de recherche de l’Hôpital St-François d’Assise, Québec, Canada, G1L 3L5, Centre de recherche en cancérologie de l’Université Laval [M.LeB., M.L.-H., M.Lem., I.M., L.J.], Centre Hospitalier Universitaire de Québec, L’Hôtel-Dieu de Québec, Québec, Canada, G1R 2J6 ABSTRACT The Hoxa5 mutation is associated with a high perinatal ventilation increase seen during hypoxia was similar for both mortality rate caused by a severe obstruction of the laryngotra- groups of mice; however, the dynamics of the frequency re- cheal airways, pulmonary dysmorphogenesis, and a decreased sponse was genotype-dependent. The hypercapnic ventilatory production of surfactant proteins. Surviving Hoxa5-/- mutant response did not differ between genotypes. These findings reveal mice also display lung anomalies with deficient alveolar septa- the strategies allowing survival of Hoxa5-/- mice facing morpho- tion and areas of collapsed tissue, thus demonstrating the impor- logic anomalies leading to a significant deficit in gas exchange tance of Hoxa5 throughout lung development and maturation. capacity. (Pediatr Res 56: 553–562, 2004) Here, we address the functional consequences of the Hoxa5 mutation on respiration and chemoreflexes by comparing the Abbreviations -/- breathing pattern of Hoxa5 mice to that of wild-type animals FICO2, fractional concentration of carbon dioxide in the under resting conditions and during exposure to moderate ven- inspired gas tilatory stimuli such as hypoxia and hypercapnia.
    [Show full text]
  • Inactivation of HOXA Genes by Hypermethylation in Myeloid and Lymphoid Malignancy Is Frequent and Associated with Poor Prognosis Gordon Strathdee,1Tessa L
    Imaging, Diagnosis, Prognosis Inactivation of HOXA Genes by Hypermethylation in Myeloid and Lymphoid Malignancy is Frequent and Associated with Poor Prognosis Gordon Strathdee,1Tessa L. Holyoake,2 Alyson Sim,1Anton Parker,4 David G. Oscier,4 Junia V. Melo, 5 Stefan Meyer,6,7 Tim Eden,7 Anne M. Dickinson,8 Joanne C. Mountford,2 Heather G. Jorgensen,2 Richard Soutar, 3 and Robert Brown1 Abstract Purpose: The HOX genes comprise a large family of homeodomain-containing transcription factors, present in four separate clusters, which are key regulators of embryonic development, hematopoietic differentiation, and leukemogenesis. We aimed to study the role of DNA methyl- ation as an inducer of HOX gene silencing in leukemia. Experimental Design: Three hundred and seventy-eight samples of myeloid and lymphoid leukemia were quantitatively analyzed (by COBRA analysis and pyrosequencing of bisulfite- modified DNA) for methylation of eight HOXA and HOXB cluster genes. The biological significance of the methylation identified was studied by expression analysis and through re-expression of HOXA5 in a chronic myeloid leukemia (CML) blast crisis cell line model. Results: Here, we identify frequent hypermethylation and gene inactivation of HOXA and HOXB cluster genes in leukemia. In particular, hypermethylation of HOXA4 and HOXA5 was frequently observed (26-79%) in all types of leukemias studied. HOXA6 hypermethylation was predomi- nantly restricted to lymphoid malignancies, whereas hypermethylation of other HOXA and HOXB genes was only observed in childhood leukemia. HOX gene methylation exhibited clear correla- tions with important clinical variables, most notably in CML, in which hypermethylation of both HOXA5 (P = 0.00002) and HOXA4 (P = 0.006) was strongly correlated with progression to blast crisis.
    [Show full text]
  • Transcriptional Regulation by Brn-3 POU Domain-Containing Transcription Factors
    Transcriptional regulation by Brn-3 POU domain-containing transcription factors Jonathan Hancock Dennis A thesis submitted for the degree of Doctor of Philosophy Medical Molecular Biology Unit Department of Molecular Pathology University College London London January 2001 ProQuest Number: U644107 All rights reserved INFORMATION TO ALL USERS The quality of this reproduction is dependent upon the quality of the copy submitted. In the unlikely event that the author did not send a complete manuscript and there are missing pages, these will be noted. Also, if material had to be removed, a note will indicate the deletion. uest. ProQuest U644107 Published by ProQuest LLC(2016). Copyright of the Dissertation is held by the Author. All rights reserved. This work is protected against unauthorized copying under Title 17, United States Code. Microform Edition © ProQuest LLC. ProQuest LLC 789 East Eisenhower Parkway P.O. Box 1346 Ann Arbor, Ml 48106-1346 ABSTRACT Bm-3a and Bm-3b are closely related members of the POU domain-containing transcription factors. While Bm-3a is associated with neuronal differentiation and Bm- 3b with neuronal proliferation, their expression is not strictly limited to the nervous system. It has been shown that these Bm-3 proteins are expressed in non-neuronal tissues including cervical epithelium, testis and breast. Moreover, these Bm-3 proteins functionally interact with the estrogen receptor and in association with the estrogen receptor have differing transactivation potentials. The p i60 steroid receptor coactivators (Srcs) are also able to interact with the estrogen receptor in a ligand dependent manner and are able to enhance estrogen dependent transcription.
    [Show full text]
  • Gene- and Tissue-Level Interactions in Normal Gastrointestinal Development and Hirschsprung Disease
    Gene- and tissue-level interactions in normal gastrointestinal development and Hirschsprung disease Sumantra Chatterjeea,b,1, Priyanka Nandakumara,1, Dallas R. Auera,b, Stacey B. Gabrielc, and Aravinda Chakravartia,b,2 aCenter for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205; bCenter for Human Genetics and Genomics, New York University School of Medicine, New York, NY 10016; and cGenomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142 Contributed by Aravinda Chakravarti, November 1, 2019 (sent for review May 21, 2019; reviewed by William J. Pavan and Tatjana Sauka-Spengler) The development of the gut from endodermal tissue to an organ between the longitudinal and circular muscles, and the sub- with multiple distinct structures and functions occurs over a mucosal (Meissner’s) plexus, between the circular muscle and prolonged time during embryonic days E10.5–E14.5 in the mouse. the submucosal layer. The myenteric and submucoal plexuses During this process, one major event is innervation of the gut by provide motor innervation to both muscular layers of the gut, enteric neural crest cells (ENCCs) to establish the enteric nervous and secretomotor innervation of the mucosa nearest the lumen system (ENS). To understand the molecular processes underpinning of the gut, respectively (6). gut and ENS development, we generated RNA-sequencing profiles The many stages of gut development require numerous initiat- from wild-type mouse guts at E10.5, E12.5, and E14.5 from both ing signaling events activating transcription factors (TFs) targeting sexes. We also generated these profiles from homozygous Ret null diverse genes and pathways varying across development (7, 8).
    [Show full text]
  • Three-Dimensional Regulation of HOXA Cluster Genes by a Cis-Element in Hematopoietic Stem Cell and Leukemia
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.16.017533; this version posted April 18, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Three-dimensional regulation of HOXA cluster genes by a cis-element in hematopoietic stem cell and leukemia. Xue Qing David Wang1, Haley Gore1, Pamela Himadewi1, Fan Feng2, Lu Yang3, Wanding Zhou1,5, Yushuai Liu1, Xinyu Wang4, Chun-wei Chen3, Jianzhong Su4, Jie Liu2, Gerd Pfeifer1,*, Xiaotian Zhang1,* 1. Center for Epigenetics, Van Andel Research Institute, Grand Rapids, Michigan 2. Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan. 3. Department of System Biology, City of Hope Cancer Center, California 4. Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China 5. Current address: Children Hospital of Philadelphia, University of Pennsylvania *. These authors jointly supervise this project Correspondence should be addressed to Xiaotian Zhang: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/2020.04.16.017533; this version posted April 18, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY 4.0 International license. Abstract: Proper gene regulation is crucial for cellular differentiation, and dysregulation of key genes can lead to diseased states such as cancer. The HOX transcription factors play such a role during hematopoiesis, and aberrant expression of certain HOXA genes is found in certain acute myeloid leukemias (AMLs).
    [Show full text]
  • Supplementary File Table S1
    Supplementary file Table S1 Grouping of Homeobox genes according to their main known function. Anatomical Structure Morphogenesis EN1, HOXC10, HOXC13, HOXD3, LBX1, SIX2, SIX4 Organ Morphogenesis CDX1, CDX2, HOXA11, HOXA13, ISL1, LHX1, PAX3, PDHX, PITX2, PITX3, PROX1, SIX6 Body Pattern Formation ALX3, EMX2, HHEX, HOXA11, HOXA2, HOXA4, HOXA5, HOXA6, HOXB1, HOXB5, HOXB6, HOXC5, HOXD10, HOXD8, LMX1B, PITX2 Ectoderm Development PROX1, VAX2 Endoderm Development HOXC11 Brain & Nervous System Development Brain Development ALX1, DLX2, EMX2 Nervous System Development: ARX, DLX5, DLX6, HOXD10, LBX1, LHX1, OTP, PAX3, PHOX2A, PHOX2B Skeletal Development: ALX3, ALX4, DLX3, DLX5, DLX6, EN1, HOXA11, HOXA13, HOXA2, HOXB6, HOXD10, HOXD13, MSX2 Muscle Development: BARX2, MKX, SIRT1, SIRT2, SIX1 Other Homeobox Genes Involved In BARX1, CDX4, CUX1, DLX1, EMX1, EN2, Multicellular Organismal HOXA1, HOXA7, HOXA9, HOXB13, HOXB2, Development: HOXB3, HOXB4, HOXB7, HOXB8, HOXB9, HOXC12, HOXC8, HOXC9, HOXD1, HOXD11, HOXD12, HOXD9, ISL2, LBX2, LMX1A, MEIS1, NKX3-1, OTX1, TLX1, VAX1, VSX1, VSX2 Homeobox Genes Involved In Cell ARX, EMX2, HHEX, HLX, HOPX, LBX1, LHX1, Differentiation: LMX1B, MIXL1, OTP, PHOX2A, SIRT1, VSX2 Other Genes: PHTF1, SIRT3, SIRT6, SIRT7, ZHX1, ZHX2 Homeobox genes include two subsets of genes coding for transcription factors involved in multiple functions. The clustered HOX genes are indicated in bold. Supplementary file Figure S2 5’ Spatial collinearity 3’ HOXA Chr. 7p15.3 HOXB Chr. 17q21.3 HOXC Chr. 12q13.3 HOXD Chr. 2q31 13 12 11 10 9 8 7 6 5 4 3 2 1 Paralogous HOX groups Distribution of the 39 human HOX genes in four clusters located in different chromosomal regions*. Blue indicates anterior HOX genes. Yellow, paralogy group 3 Hox genes, green and purple indicatete central HOX genes and Red the posterior HOX genes.
    [Show full text]
  • Homeobox Genes and Their Functional Significance in Ovarian Tumorigenesis
    10 Homeobox Genes and Their Functional Significance in Ovarian Tumorigenesis Bon Quy Trinh and Honami Naora University of Texas MD Anderson Cancer Center USA 1. Introduction It is widely recognized that many pathways that control normal embryonic patterning are deregulated in human cancers. Mutations or aberrant expression of components of the Wnt, Hedgehog and Notch signaling pathways have been demonstrated to play pivotal roles in tumorigenesis. Homeobox genes constitute an evolutionarily conserved gene super-family that represents another important class of patterning regulators. These genes encode transcription factors that are essential for controlling cell differentiation and specification of the body plan during embryonic development. Although many homeobox genes have been reported to be aberrantly expressed in ovarian cancer, the functional significance of these genes in ovarian tumorigenesis has only emerged in recent years. This chapter discusses recent research studies that demonstrate that homeobox genes have diverse functions in the biology of ovarian cancer. These functions include specifying patterns of histologic differentiation of ovarian cancers, controlling growth and survival of tumor cells, and promoting tumor angiogenesis, cell-cell interactions and tumor cell invasiveness. This chapter discusses how studies of homeobox genes provide insights into our understanding of the cell-of-origin of ovarian cancers, the striking morphologic heterogeneity of these tumors, and the unique clinical behavior of ovarian cancer. 2. Overview of homeobox genes Homeobox genes were first discovered in Drosophila by their mutations that caused homeotic transformation, a phenomenon in which body segments form in inappropriate locations (Gehring & Hiromi, 1986; McGinnis & Krumlauf, 1992). A classic example of a homeotic transformation in Drosophila is the formation of legs rather than antennae caused by ectopic expression of the Antennapedia gene (Schneuwly et al., 1987).
    [Show full text]
  • Homeobox Genes in the Ribbonworm Lineus Sanguineus: Evolutionary Implications
    Proc. Natl. Acad. Sci. USA Vol. 95, pp. 3030–3035, March 1998 Evolution Homeobox genes in the ribbonworm Lineus sanguineus: Evolutionary implications MARIE KMITA-CUNISSE*†,FELIX LOOSLI‡,JACQUES BIE`RNE*, AND WALTER J. GEHRING†§ †Biozentrum, University of Basel, Klingelbergstr. 70, CH-4056 Basel, Switzerland; and *Laboratoire de Biologie Cellulaire et Mole´culaire,Universite´de Reims Champagne-Ardenne, F-51687 Reims, France Contributed by Walter J. Gehring, December 19, 1997 ABSTRACT From our current understanding of the ge- new body plans have evolved. Until recently, extensive studies netic basis of development and pattern formation in Drosoph- of Hox genes have been performed mainly on flies and ila and vertebrates it is commonly thought that clusters of Hox vertebrates. To enable us to draw a reliable scenario about how genes sculpt the morphology of animals in specific body Hox genes have evolved, studies of Hox genes have to be regions. Based on Hox gene conservation throughout the broadened to include representatives of other phyla, in par- animal kingdom it is proposed that these genes and their role ticular different protostomes. in pattern formation evolved early during the evolution of Based on the original hypothesis of Lewis (11) that the metazoans. Knowledge of the history of Hox genes will lead to Bithorax-Complex of Drosophila arose by unequal crossing- a better understanding of the role of Hox genes in the evolution over, leading to tandem duplications of the Hox genes, we have of animal body plans. To infer Hox gene evolution, reliable proposed that the primordial Hox cluster may have arisen by data on lower chordates and invertebrates are crucial.
    [Show full text]