GATA1: Difference between revisions

Jump to navigation Jump to search
m (Bot: HTTP→HTTPS)
 
imported>GreenC bot
(Remove 15 stray access-date. (GreenC bot job #5))
Line 1: Line 1:
{{Infobox_gene}}
{{Infobox_gene}}
'''Erythroid transcription factor''' also known as '''GATA-binding factor 1''' or '''GATA-1''' is a [[protein]] that in humans is encoded by the ''GATA1'' [[gene]].<ref name="pmid1999341">{{cite journal | vauthors = Caiulo A, Nicolis S, Bianchi P, Zuffardi O, Bardoni B, Maraschio P, Ottolenghi S, Camerino G, Giglioni B | title = Mapping the gene encoding the human erythroid transcriptional factor NFE1-GF1 to Xp11.23 | journal = Human Genetics | volume = 86 | issue = 4 | pages = 388–90 | date = Feb 1991 | pmid = 1999341 | doi = 10.1007/bf00201840 }}</ref>
'''GATA-binding factor 1''' or '''GATA-1''' (also termed '''Erythroid transcription factor''') is the founding member of the [[GATA transcription factor|GATA family of transcription factors]]. This [[protein]] is widely expressed throughout vertebrate species. In humans and mice, it is encoded by the ''GATA1'' and ''Gata1'' genes, respectively. These genes are located on the [[X chromosome]] in both species.<ref name="pmid23838521">{{cite journal | vauthors = Katsumura KR, DeVilbiss AW, Pope NJ, Johnson KD, Bresnick EH | title = Transcriptional mechanisms underlying hemoglobin synthesis | journal = Cold Spring Harbor Perspectives in Medicine | volume = 3 | issue = 9 | pages = a015412 | date = September 2013 | pmid = 23838521 | pmc = 3753722 | doi = 10.1101/cshperspect.a015412 }}</ref><ref name="pmid1999341">{{cite journal | vauthors = Caiulo A, Nicolis S, Bianchi P, Zuffardi O, Bardoni B, Maraschio P, Ottolenghi S, Camerino G, Giglioni B | title = Mapping the gene encoding the human erythroid transcriptional factor NFE1-GF1 to Xp11.23 | journal = Human Genetics | volume = 86 | issue = 4 | pages = 388–90 | date = Feb 1991 | pmid = 1999341 | doi = 10.1007/bf00201840 }}</ref>


GATA-1  is a member of the [[GATA transcription factor]] family and is a key mediator of the development of specific types of blood cells from their precursor cells, termed hematopoietic progenitors or precursors.<ref name="pmid22492510">{{cite journal | vauthors = Bresnick EH, Katsumura KR, Lee HY, Johnson KD, Perkins AS | title = Master regulatory GATA transcription factors: mechanistic principles and emerging links to hematologic malignancies | journal = Nucleic Acids Res. | volume = 40 | issue = 13 | pages = 5819–31 | year = 2012 | pmid = 22492510 | pmc = 3401466 | doi = 10.1093/nar/gks281 }}</ref><ref name="pmid20670937">{{cite journal | vauthors = Bresnick EH, Lee HY, Fujiwara T, Johnson KD, Keles S | title = GATA switches as developmental drivers | journal = J. Biol. Chem. | volume = 285 | issue = 41 | pages = 31087–93 | year = 2010 | pmid = 20670937 | pmc = 2951181 | doi = 10.1074/jbc.R110.159079 }}</ref> This protein plays a  role in erythroid development by regulating a large ensemble of genes that mediate both the development and function of red blood cells. Critical functions in the developing red blood cell (erythroblast) include the establishment of the erythroid cytoskeleton, enzymes that mediate heme biosynthesis, and polypeptide chains that constitute the hemoglobin tetramer. Mutations in the gene encoding GATA-1 have been associated with X-linked [[congenital dyserythropoietic anemia|dyserythropoietic anemia]] and [[thrombocytopenia]].<ref>{{cite web | title = Entrez Gene: GATA1 GATA binding protein 1 (globin transcription factor 1)| url = https://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2623| accessdate = }}</ref>
GATA1 regulates the [[Gene expression|expression]] (i.e. formation of the genes' products) of an ensemble of genes that mediate the development of red blood cells and platelets. Its critical roles in red blood cell formation include promoting the [[Erythropoiesis|maturation]] of precursor cells, e.g. [[erythroblast]]s, to red blood cells and stimulating these cells to erect their [[cytoskeleton]] and [[biosynthesis|biosynthesize]] their oxygen-carrying components viz., [[hemoglobin]] and [[heme]]. GATA1 plays a similarly critical role in the maturation of blood [[platelets]] from [[megakaryoblast]]s, [[promegakaryocyte]]s, and [[megakaryocyte]]s; the latter cells then shed membrane-enclosed fragments of their cytoplasm, i.e. platelets, into the blood.<ref name="pmid23838521"/><ref name="pmid26186939">{{cite journal | vauthors = Gruber TA, Downing JR | title = The biology of pediatric acute megakaryoblastic leukemia | journal = Blood | volume = 126 | issue = 8 | pages = 943–9 | date = August 2015 | pmid = 26186939 | pmc = 4551356 | doi = 10.1182/blood-2015-05-567859 }}</ref>  


== Function ==
In consequence of the vital role that GATA1 has in the proper maturation of red blood cells and platelets, [[Mutations#By effect on function|inactivating mutations]] in the ''GATA1'' gene (i.e. mutations that result in the production of no, reduced levels of, or a less active GATA1) cause [[Sex linkage|X chromosome-linked]] [[anemic]] and/or [[bleeding disorders|bleeding diseases]] due to the reduced formation and functionality of red blood cells and/or platelets, respectively, or, under certain circumstances, the pathological proliferation of megakaryoblasts. These diseases include [[Down syndrome#cancer|transient myeloproliferative disorder]] occurring in Down syndrome, [[acute megakaryoblastic leukemia]] occurring in [[Down syndrome#cancer|Down syndrome]], [[Diamond-Blackfan anemia]], and various combined [[anemia]]-[[thrombocytopenia]] syndromes including a [[gray platelet syndrome]]-type disorder.<ref name="pmid28566565">{{cite journal | vauthors = Fujiwara T | title = GATA Transcription Factors: Basic Principles and Related Human Disorders | journal = The Tohoku Journal of Experimental Medicine | volume = 242 | issue = 2 | pages = 83–91 | date = June 2017 | pmid = 28566565 | doi = 10.1620/tjem.242.83 }}</ref><ref>{{cite web | title = Entrez Gene: GATA1 GATA binding protein 1 (globin transcription factor 1)| url = https://www.ncbi.nlm.nih.gov/sites/entrez?Db=gene&Cmd=ShowDetailView&TermToSearch=2623| accessdate = }}</ref><ref name="pmid29081386">{{cite journal | vauthors = Da Costa L, O'Donohue MF, van Dooijeweert B, Albrecht K, Unal S, Ramenghi U, Leblanc T, Dianzani I, Tamary H, Bartels M, Gleizes PE, Wlodarski M, MacInnes AW | title = Molecular approaches to diagnose Diamond-Blackfan anemia: The EuroDBA experience | journal = European Journal of Medical Genetics | volume = | issue = | pages = | date = October 2017 | pmid = 29081386 | doi = 10.1016/j.ejmg.2017.10.017 }}</ref>


GATA1 is required for the maturation of [[red blood cell]]s, [[megakaryocyte]]s, [[mast cell]]s and [[eosinophil]]s.<ref>{{cite journal | vauthors = Crispino JD | title = GATA1 in normal and malignant hematopoiesis | journal = Seminars in Cell & Developmental Biology | volume = 16 | issue = 1 | pages = 137–47 | date = Feb 2005 | pmid = 15659348 | doi = 10.1016/j.semcdb.2004.11.002 }}</ref> GATA1 mutant mice die in early embryonic development due to inability to form mature erythroid cells. GATA1 mutation in humans causes congenital anemias and thrombocytopenias.<ref>{{cite journal | vauthors = Fujiwara Y, Browne CP, Cunniff K, Goff SC, Orkin SH | title = Arrested development of embryonic red cell precursors in mouse embryos lacking transcription factor GATA-1 | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 93 | issue = 22 | pages = 12355–8 | date = Oct 1996 | pmid = 8901585 | pmc = 37995 | doi=10.1073/pnas.93.22.12355}}</ref><ref>{{cite journal | vauthors = Campbell AE, Wilkinson-White L, Mackay JP, Matthews JM, Blobel GA | title = Analysis of disease-causing GATA1 mutations in murine gene complementation systems | journal = Blood | volume = 121 | issue = 26 | pages = 5218–27 | date = Jun 2013 | pmid = 23704091 | pmc = 3695365 | doi = 10.1182/blood-2013-03-488080 }}</ref>
Reduced levels of GATA1 due to reductions in the translation of GATA1 [[mRNA]] into its transcription factor product are associated with promoting the progression of [[myelofibrosis]], i.e. a malignant disease that involves the replacement of bone marrow cells by fibrous tissue and [[extramedullary hematopoiesis]], i.e. the extension of blood cell-forming cells to sites outside of the [[bone marrow]].<ref name="pmid20458749">{{cite journal | vauthors = Verrucci M, Pancrazzi A, Aracil M, Martelli F, Guglielmelli P, Zingariello M, Ghinassi B, D'Amore E, Jimeno J, Vannucchi AM, Migliaccio AR | title = CXCR4-independent rescue of the myeloproliferative defect of the Gata1low myelofibrosis mouse model by Aplidin | journal = Journal of Cellular Physiology | volume = 225 | issue = 2 | pages = 490–9 | date = November 2010 | pmid = 20458749 | pmc = 3780594 | doi = 10.1002/jcp.22228 }}</ref><ref name="pmid29562644">{{cite journal | vauthors = Song MK, Park BB, Uhm JE | title = Understanding Splenomegaly in Myelofibrosis: Association with Molecular Pathogenesis | journal = International Journal of Molecular Sciences | volume = 19 | issue = 3 | pages = | date = March 2018 | pmid = 29562644 | pmc = 5877759 | doi = 10.3390/ijms19030898 }}</ref>


GATA1 was first described as a [[red blood cell]] lineage transcription factor that activates the [[beta-globin]] gene.<ref>{{cite journal | vauthors = Evans T, Reitman M, Felsenfeld G | title = An erythrocyte-specific DNA-binding factor recognizes a regulatory sequence common to all chicken globin genes | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 85 | issue = 16 | pages = 5976–80 | date = Aug 1988 | pmid = 3413070 | pmc = 281888 | doi=10.1073/pnas.85.16.5976}}</ref> During red blood cell maturation, GATA1 activates nearly all erythroid-specific genes while silencing genes associated with the immature proliferative red blood cell precursor cells (erythroblasts).<ref name="ReferenceA">{{cite journal | vauthors = Welch JJ, Watts JA, Vakoc CR, Yao Y, Wang H, Hardison RC, Blobel GA, Chodosh LA, Weiss MJ | title = Global regulation of erythroid gene expression by transcription factor GATA-1 | journal = Blood | volume = 104 | issue = 10 | pages = 3136–47 | date = Nov 2004 | pmid = 15297311 | doi = 10.1182/blood-2004-04-1603 }}</ref><ref name="ReferenceB">{{cite journal | vauthors = Cheng Y, Wu W, Kumar SA, Yu D, Deng W, Tripic T, King DC, Chen KB, Zhang Y, Drautz D, Giardine B, Schuster SC, Miller W, Chiaromonte F, Zhang Y, Blobel GA, Weiss MJ, Hardison RC | title = Erythroid GATA1 function revealed by genome-wide analysis of transcription factor occupancy, histone modifications, and mRNA expression | journal = Genome Research | volume = 19 | issue = 12 | pages = 2172–84 | date = Dec 2009 | pmid = 19887574 | pmc = 2792182 | doi = 10.1101/gr.098921.109 }}</ref> Genome-wide studies have provided evidence that GATA1 activates and represses a large number of genes.<ref name="ReferenceA"/><ref name="ReferenceB"/><ref>{{cite journal | vauthors = Yu M, Riva L, Xie H, Schindler Y, Moran TB, Cheng Y, Yu D, Hardison R, Weiss MJ, Orkin SH, Bernstein BE, Fraenkel E, Cantor AB | title = Insights into GATA-1-mediated gene activation versus repression via genome-wide chromatin occupancy analysis | journal = Molecular Cell | volume = 36 | issue = 4 | pages = 682–95 | date = Nov 2009 | pmid = 19941827 | pmc = 2800995 | doi = 10.1016/j.molcel.2009.11.002 }}</ref><ref name="pmid19941826">{{cite journal | vauthors = Fujiwara T, O'Geen H, Keles S, Blahnik K, Linnemann AK, Kang YA, Choi K, Farnham PJ, Bresnick EH | title = Discovering hematopoietic mechanisms through genome-wide analysis of GATA factor chromatin occupancy | journal = Mol. Cell | volume = 36 | issue = 4 | pages = 667–81 | year = 2009 | pmid = 19941826 | pmc = 2784893 | doi = 10.1016/j.molcel.2009.11.001 }}</ref>  Many questions remain unanswered regarding the function of a large number of genes. By contrast, other GATA-1 target genes have established activities to control fundamental cell biological functions, including machinery that controls the ability of erythroid precursor cells to proliferate and proteins that control the capacity of the erythroid precursor cell to remodel its organelles such as mitochondria,<ref name="pmid22025678">{{cite journal | vauthors = Kang YA, Sanalkumar R, O'Geen H, Linnemann AK, Chang CJ, Bouhassira EE, Farnham PJ, Keles S, Bresnick EH | title = Autophagy driven by a master regulator of hematopoiesis | journal = Mol. Cell. Biol. | volume = 32 | issue = 1 | pages = 226–39 | year = 2012 | pmid = 22025678 | pmc = 3255705 | doi = 10.1128/MCB.06166-11 }}</ref> proteins that control the RNA content of the erythroid precursor cell,<ref name="pmid25115889">{{cite journal | vauthors = McIver SC, Kang YA, DeVilbiss AW, O'Driscoll CA, Ouellette JN, Pope NJ, Camprecios G, Chang CJ, Yang D, Bouhassira EE, Ghaffari S, Bresnick EH | title = The exosome complex establishes a barricade to erythroid maturation | journal = Blood | volume = 124 | issue = 14 | pages = 2285–97 | year = 2014 | pmid = 25115889 | doi = 10.1182/blood-2014-04-571083 | pmc=4183988}}</ref> and proteins that control signal transduction networks that orchestrate the many dynamic transitions of the developing erythroid precursor.<ref name="pmid26073540">{{cite journal | vauthors = Hewitt KJ, Kim DH, Devadas P, Prathibha R, Zuo C, Sanalkumar R, Johnson KD, Kang YA, Kim JS, Dewey CN, Keles S, Bresnick EH | title = Hematopoietic Signaling Mechanism Revealed from a Stem/Progenitor Cell Cistrome | journal = Mol. Cell | volume = 59 | issue = 1 | pages = 62–74 | year = 2015 | pmid = 26073540 | doi = 10.1016/j.molcel.2015.05.020 | pmc=4499333}}</ref>
== Gene ==
The human ''GATA1'' gene is located on the short (i.e. "p") arm of the [[X chromosome]] at position 11.23. It is 7.74 [[Base pair#Length measurements|kilobases]] in length, consists of 6 [[exons]], and codes for a full length protein, GATA1, of 414 [[amino acids]] as well as a shorter one, GATA1-S. GATA1-S lacks the first 83 amino acids of GATA1 and therefore consists of only 331 amino acids.<ref>https://www.ncbi.nlm.nih.gov/gene/2623</ref><ref>http://genatlas.medecine.univ-paris5.fr/fiche.php?onglet=2&n=5186</ref><ref name="pmid27235756">{{cite journal | vauthors = Shimizu R, Yamamoto M | title = GATA-related hematologic disorders | journal = Experimental Hematology | volume = 44 | issue = 8 | pages = 696–705 | date = August 2016 | pmid = 27235756 | doi = 10.1016/j.exphem.2016.05.010 }}</ref> ''GATA1'' codes for two [[zinc finger]] [[structural motif]]s, C-ZnF and N-ZnF, that are present in both GATA1 and GATA1-S proteins. These motifs are critical for both transcription factors' gene-regulating actions. N-ZnF is a frequent site of disease-causing mutations. Lacking the first 83 amino acids and therefore one of the two activation domains of GATA1, GATA1-S has significantly less gene-regulating activity than GATA1.<ref name="pmid28566565"/><ref name="pmid27235756"/>  


== Structure ==
Studies in ''Gata1''-[[knockout mice]], i.e. mice lacking the ''Gata1'' gene, indicate that this gene is essential for the development and maintenance of blood-based and/or tissue-based hematological cells, particularly [[red blood cell]]s and [[platelets]] but also [[eosinophil]]s, [[basophil]]s, [[mast cell]]s, and [[dendritic cell]]s. The knock-out mice die by day 11.5 of their [[embryonic development]] due to severe anemia that is associated with absence of cells of the red blood cell lineage, excessive numbers of malformed platelet-precursor cells, and an absence of [[platelet]]s. These defects reflect the essential role of Gata-1 in stimulating the development, self-renewal, and/or maturation of red blood cell and platelet [[precursor cell]]s. Studies using mice depleted of their ''Gata1'' gene during adulthood show that: '''1)''' Gata1 is required for the stimulation of [[erythropoiesis]] (i.e. increase in red blood cell formation) in response to stress and '''2)''' ''Gata1''-deficient adult mice invariably develop a form of [[myelofibrosis]].<ref name="pmid28179280">{{cite journal | vauthors = Crispino JD, Horwitz MS | title = GATA factor mutations in hematologic disease | journal = Blood | volume = 129 | issue = 15 | pages = 2103–2110 | date = April 2017 | pmid = 28179280 | pmc = 5391620 | doi = 10.1182/blood-2016-09-687889 }}</ref><ref name="pmid28179282">{{cite journal | vauthors = Katsumura KR, Bresnick EH | title = The GATA factor revolution in hematology | journal = Blood | volume = 129 | issue = 15 | pages = 2092–2102 | date = April 2017 | pmid = 28179282 | pmc = 5391619 | doi = 10.1182/blood-2016-09-687871 | url = }}</ref>


The GATA-1 protein contains multiple functional domains including the C-finger, the N-finger, and N-terminal sequences that have been suggested to constitute a transcriptional activation domain. The C-finger, named for being near the [[C-terminal]], mediates [[Zinc finger]] sequence-specific DNA binding. The primary function of the N-finger, named for being near the [[N-terminal]] is binding to a cofactor named [[ZFPM1|FOG1]] (friend of GATA), although it has also been implicated in binding to naked DNA (DNA studied in the test tube that is not assembled into chromatin)The gene for GATA1 is on the X-chromosome.
== GATA1 proteins ==
In both GATA1 and GATA1-S, C-ZnF (i.e. [[C-terminus]] zinc finger) binds to DNA-specific [[nucleic acid sequence]]s sites viz., (T/A(GATA)A/G), on the expression-regulating sites of its target genes and in doing so either stimulates or suppresses the expression of these target genes. Their N-ZnF (i.e. [[N-terminus]] zinc fingers) interacts with an essential transcription factor-regulating nuclear protein, [[ZFPM1|FOG1]]. FOG1 powerfully promotes or suppresses the actions that the two transcription factors have on most of their target genes. Similar to the knockout of ''Gata1'', knockout of the mouse gene for FOG1, ''[[ZFPM1|Zfpm1]]'', causes total failure of red blood cell development and embryonic lethality by day 11.5. Based primarily on mouse studies, it is proposed that the GATA1-FOG1 complex promotes human erythropoiesis by recruiting and binding with at least two gene expression-regulating complexes, [[Mi-2/NuRD complex]] (a [[chromatin remodeling|chromatin remodeler]]) and [[CTBP1]] (a [[histone deacetylase]]) and three gene expression-regulating proteins, [[KMT5A|SET8]] (a GATA1-inhibiting [[histone methyltransferase]]), [[SMARCA4|BRG1]] (a [[transcription activator]]), and [[Mediator (coactivator)|Mediator]] (a [[Coactivator (genetics)|transcription co-activator]]). Other interactions include those with: [[BRD3]] (remodels DNA [[nucleosome]]s),<ref name="Lamonica_2011">{{cite journal | vauthors = Lamonica JM, Deng W, Kadauke S, Campbell AE, Gamsjaeger R, Wang H, Cheng Y, Billin AN, Hardison RC, Mackay JP, Blobel GA | title = Bromodomain protein Brd3 associates with acetylated GATA1 to promote its chromatin occupancy at erythroid target genes | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 108 | issue = 22 | date = May 2011 | pmid = 21536911 | doi = 10.1073/pnas.1102140108 | pages=E159-68 | pmc=3107332}}</ref><ref>{{cite journal | vauthors = Gamsjaeger R, Webb SR, Lamonica JM, Billin A, Blobel GA, Mackay JP | title = Structural basis and specificity of acetylated transcription factor GATA1 recognition by BET family bromodomain protein Brd3 | journal = Molecular and Cellular Biology | volume = 31 | issue = 13 | date = Jul 2011 | pmid = 21555453 | doi = 10.1128/MCB.05413-11 | pages=2632–40 | pmc=3133386}}</ref><ref>{{cite journal | vauthors = Stonestrom AJ, Hsu SC, Jahn KS, Huang P, Keller CA, Giardine BM, Kadauke S, Campbell AE, Evans P, Hardison RC, Blobel GA | title = Functions of BET proteins in erythroid gene expression | journal = Blood | date = Feb 2015 | pmid = 25696920 | doi = 10.1182/blood-2014-10-607309 | volume=125 | pages=2825–34 | pmc=4424630}},</ref> [[BRD4]] (binds acetylated lysine residues in DNA-associated histone to regulate gene accessibility),<ref name="Lamonica_2011" /> [[FLI1]] (a transcription factor that blocks erythroid differentiation),<ref name="pmid2724402">{{cite journal | vauthors = Lahiri K, Dole MG, Vidwans AS, Kamat J, Kandoth P | title = Acute glomerulonephritis | journal = Journal of Tropical Pediatrics | volume = 35 | issue = 2 | pages = 92 | date = Apr 1989 | pmid = 2724402 | doi = 10.1093/tropej/35.2.92 }}</ref><ref name="pmid12556498">{{cite journal | vauthors = Starck J, Cohet N, Gonnet C, Sarrazin S, Doubeikovskaia Z, Doubeikovski A, Verger A, Duterque-Coquillaud M, Morle F | title = Functional cross-antagonism between transcription factors FLI-1 and EKLF | journal = Molecular and Cellular Biology | volume = 23 | issue = 4 | pages = 1390–402 | date = Feb 2003 | pmid = 12556498 | pmc = 141137 | doi = 10.1128/MCB.23.4.1390-1402.2003 }}</ref> [[HDAC1]] (a [[histone deacetylase]]),<ref name="pmid14668799">{{cite journal | vauthors = Watamoto K, Towatari M, Ozawa Y, Miyata Y, Okamoto M, Abe A, Naoe T, Saito H | title = Altered interaction of HDAC5 with GATA-1 during MEL cell differentiation | journal = Oncogene | volume = 22 | issue = 57 | pages = 9176–84 | date = Dec 2003 | pmid = 14668799 | doi = 10.1038/sj.onc.1206902 }}</ref> [[LMO2]] (regulator of erythrocyte development),<ref name="Osada_1995">{{cite journal | vauthors = Osada H, Grutz G, Axelson H, Forster A, Rabbitts TH | title = Association of erythroid transcription factors: complexes involving the LIM protein RBTN2 and the zinc-finger protein GATA1 | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 92 | issue = 21 | pages = 9585–9 | date = Oct 1995 | pmid = 7568177 | pmc = 40846 | doi = 10.1073/pnas.92.21.9585 }}</ref> [[Zinc finger and BTB domain-containing protein 16|ZBTB16]] (transcription factor regulating [[cell cycle]] progression),<ref name="pmid12242665">{{cite journal | vauthors = Labbaye C, Quaranta MT, Pagliuca A, Militi S, Licht JD, Testa U, Peschle C | title = PLZF induces megakaryocytic development, activates Tpo receptor expression and interacts with GATA1 protein | journal = Oncogene | volume = 21 | issue = 43 | pages = 6669–79 | date = Sep 2002 | pmid = 12242665 | doi = 10.1038/sj.onc.1205884 }}</ref> [[TAL1]] (a transcription factor),<ref name="pmid16407974">{{cite journal | vauthors = Goardon N, Lambert JA, Rodriguez P, Nissaire P, Herblot S, Thibault P, Dumenil D, Strouboulis J, Romeo PH, Hoang T | title = ETO2 coordinates cellular proliferation and differentiation during erythropoiesis | journal = The EMBO Journal | volume = 25 | issue = 2 | pages = 357–66 | date = Jan 2006 | pmid = 16407974 | pmc = 1383517 | doi = 10.1038/sj.emboj.7600934 }}</ref> [[ZFPM2|FOG2]] (a transcription factor regulator),<ref name="pmid10438528">{{cite journal | vauthors = Holmes M, Turner J, Fox A, Chisholm O, Crossley M, Chong B | title = hFOG-2, a novel zinc finger protein, binds the co-repressor mCtBP2 and modulates GATA-mediated activation | journal = The Journal of Biological Chemistry | volume = 274 | issue = 33 | pages = 23491–8 | date = Aug 1999 | pmid = 10438528 | doi = 10.1074/jbc.274.33.23491 }}</ref> and [[GATA2]] (Displacement of GATA2 by GATA1, i.e. the "GATA switch", at certain gene-regulating sites is critical for red blood development in mice and, presumably, humans).<ref name="pmid28179282"/><ref>{{cite journal | vauthors = Fujiwara Y, Browne CP, Cunniff K, Goff SC, Orkin SH | title = Arrested development of embryonic red cell precursors in mouse embryos lacking transcription factor GATA-1 | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 93 | issue = 22 | pages = 12355–8 | date = Oct 1996 | pmid = 8901585 | pmc = 37995 | doi=10.1073/pnas.93.22.12355}}</ref><ref>{{cite journal | vauthors = Campbell AE, Wilkinson-White L, Mackay JP, Matthews JM, Blobel GA | title = Analysis of disease-causing GATA1 mutations in murine gene complementation systems | journal = Blood | volume = 121 | issue = 26 | pages = 5218–27 | date = Jun 2013 | pmid = 23704091 | pmc = 3695365 | doi = 10.1182/blood-2013-03-488080 }}</ref> GATA1-FOG1 and GATA2-FOG1 interactions are critical for platelet formation in mice and may similarly be critical for this in humans.<ref name="pmid28179282"/>


== Disease linkage ==
== Physiology and Pathology ==
GATA1 was first described as a transcription factor that activates the [[hemoglobin B]] gene in the red blood cell precursors of chickens.<ref>{{cite journal | vauthors = Evans T, Reitman M, Felsenfeld G | title = An erythrocyte-specific DNA-binding factor recognizes a regulatory sequence common to all chicken globin genes | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 85 | issue = 16 | pages = 5976–80 | date = Aug 1988 | pmid = 3413070 | pmc = 281888 | doi=10.1073/pnas.85.16.5976}}</ref> Subsequent studies in mice and isolated human cells found that GATA1 stimulates the expression of genes that promote the maturation of precursor cells (e.g. [[erythroblast]]s) to red blood cells while silencing genes that cause these precursors to proliferate and thereby to [[Stem cell#self-renewal|self-renew]].<ref name="ReferenceA">{{cite journal | vauthors = Welch JJ, Watts JA, Vakoc CR, Yao Y, Wang H, Hardison RC, Blobel GA, Chodosh LA, Weiss MJ | title = Global regulation of erythroid gene expression by transcription factor GATA-1 | journal = Blood | volume = 104 | issue = 10 | pages = 3136–47 | date = Nov 2004 | pmid = 15297311 | doi = 10.1182/blood-2004-04-1603 }}</ref><ref name="ReferenceB">{{cite journal | vauthors = Cheng Y, Wu W, Kumar SA, Yu D, Deng W, Tripic T, King DC, Chen KB, Zhang Y, Drautz D, Giardine B, Schuster SC, Miller W, Chiaromonte F, Zhang Y, Blobel GA, Weiss MJ, Hardison RC | title = Erythroid GATA1 function revealed by genome-wide analysis of transcription factor occupancy, histone modifications, and mRNA expression | journal = Genome Research | volume = 19 | issue = 12 | pages = 2172–84 | date = Dec 2009 | pmid = 19887574 | pmc = 2792182 | doi = 10.1101/gr.098921.109 }}</ref> GATA1 stimulates this maturation by, for example, inducing the expression of genes in erythroid cells that contribute to the formation of their [[cytoskeleton]] and that make enzymes necessary for the [[biosynthesis]] of [[hemoglobin]]s and [[heme]], the oxygen-carrying components of red blood cells. GATA1-inactivating mutations may thereby result in a failure to produce sufficient numbers of and/or fully functional red blood cells.<ref name="pmid23838521"/> Also based on mouse and isolated human cell studies, GATA1 appears to play a similarly critical role in the maturation of platelets from their precursor cells. This [[Thrombopoiesis|maturation]] involves the stimulation of [[megakaryoblast]]s to mature ultimately to [[megakaryocyte]]s which cells shed membrane-enclosed fragments of their cytoplasm, i.e. platelets, into the blood. GATA1-inactivating mutations may thereby result in reduced levels of and/or dysfunctional blood platelets.<ref name="pmid23838521"/><ref name="pmid26186939"/>


Mutations in GATA1 cause anemias and thrombocytopenia in human patients.<ref>{{cite journal | vauthors = Crispino JD, Weiss MJ | title = Erythro-megakaryocytic transcription factors associated with hereditary anemia | journal = Blood | volume = 123 | issue = 20 | pages = 3080–8 | date = May 2014 | pmid = 24652993 | pmc = 4023417 | doi = 10.1182/blood-2014-01-453167 }}</ref><ref>{{cite journal | vauthors = Nichols KE, Crispino JD, Poncz M, White JG, Orkin SH, Maris JM, Weiss MJ | title = Familial dyserythropoietic anaemia and thrombocytopenia due to an inherited mutation in GATA1 | journal = Nature Genetics | volume = 24 | issue = 3 | pages = 266–70 | date = Mar 2000 | pmid = 10700180 | doi = 10.1038/73480 }}</ref> Disease-causing GATA1 mutations are present in the zinc finger DNA binding domains as well as protein-protein interaction domains of GATA1.<ref>{{cite journal | vauthors = Campbell AE, Wilkinson-White L, Mackay JP, Matthews JM, Blobel GA | title = Analysis of disease-causing GATA1 mutations in murine gene complementation systems | journal = Blood | volume = 121 | issue = 26 | date = Jun 2013 | pmid = 23704091 | doi = 10.1182/blood-2013-03-488080 | pmc=3695365 | pages=5218–27}}</ref>
Reduced levels of GATA1 due to defective [[translation]] of GATA1 [[mRNA]] in human megakaryocytes is associated with [[myelofibrosis]], i.e. the replacement of bone marrow cells by fibrous tissue. Based primarily on mouse and isolated human cell studies, this myelofibrosis is thought to result from the accumulation of platelet precursor cells in the bone marrow and their release of excessive amounts of cytokines that stimulate bone marrow [[stromal cells]] to become fiber-secreting [[fibroblasts]] and [[osteoblasts]]. Based on mouse studies, low GATA1 levels are also thought to promote the development of splenic [[splenomegaly|enlargement]] and [[extramedullary hematopoiesis]] in human myelofibrosis disease. These effects appear to result directly from the over-proliferation of abnormal platelet precursor cells.<ref name="pmid20458749">{{cite journal | vauthors = Verrucci M, Pancrazzi A, Aracil M, Martelli F, Guglielmelli P, Zingariello M, Ghinassi B, D'Amore E, Jimeno J, Vannucchi AM, Migliaccio AR | title = CXCR4-independent rescue of the myeloproliferative defect of the Gata1low myelofibrosis mouse model by Aplidin | journal = Journal of Cellular Physiology | volume = 225 | issue = 2 | pages = 490–9 | date = November 2010 | pmid = 20458749 | pmc = 3780594 | doi = 10.1002/jcp.22228 }}</ref><ref name="pmid29562644">{{cite journal | vauthors = Song MK, Park BB, Uhm JE | title = Understanding Splenomegaly in Myelofibrosis: Association with Molecular Pathogenesis | journal = International Journal of Molecular Sciences | volume = 19 | issue = 3 | pages = | date = March 2018 | pmid = 29562644 | pmc = 5877759 | doi = 10.3390/ijms19030898 }}</ref><ref name="pmid29611379">{{cite journal | vauthors = Yang N, Park S, Cho MS, Lee M, Hong KS, Mun YC, Seong CM, Huh HJ, Huh J | title = GATA1 Expression in BCR/ABL1-negative Myeloproliferative Neoplasms | journal = Annals of Laboratory Medicine | volume = 38 | issue = 4 | pages = 296–305 | date = July 2018 | pmid = 29611379 | doi = 10.3343/alm.2018.38.4.296 }}</ref><ref name="pmid28240607">{{cite journal | vauthors = Gilles L, Arslan AD, Marinaccio C, Wen QJ, Arya P, McNulty M, Yang Q, Zhao JC, Konstantinoff K, Lasho T, Pardanani A, Stein B, Plo I, Sundaravel S, Wickrema A, Migliaccio A, Gurbuxani S, Vainchenker W, Platanias LC, Tefferi A, Crispino JD | title = Downregulation of GATA1 drives impaired hematopoiesis in primary myelofibrosis | journal = The Journal of Clinical Investigation | volume = 127 | issue = 4 | pages = 1316–1320 | date = April 2017 | pmid = 28240607 | pmc = 5373858 | doi = 10.1172/JCI82905 | url =https://www.jci.org/articles/view/82905 | accessdate = 2018-06-26}}</ref>


Mutations in exon 2 of the GATA1 gene are present in almost all cases of [[Down syndrome]] (DS)-associated [[acute megakaryoblastic leukemia]] (AMKL).<ref name="pmid12172547">{{cite journal | vauthors = Wechsler J, Greene M, McDevitt MA, Anastasi J, Karp JE, Le Beau MM, Crispino JD | title = Acquired mutations in GATA1 in the megakaryoblastic leukemia of Down syndrome | journal = Nature Genetics | volume = 32 | issue = 1 | pages = 148–52 | date = Sep 2002 | pmid = 12172547 | doi = 10.1038/ng955 }}</ref><ref name="pmid12649131">{{cite journal | vauthors = Rainis L, Bercovich D, Strehl S, Teigler-Schlegel A, Stark B, Trka J, Amariglio N, Biondi A, Muler I, Rechavi G, Kempski H, Haas OA, Izraeli S | title = Mutations in exon 2 of GATA1 are early events in megakaryocytic malignancies associated with trisomy 21 | journal = Blood | volume = 102 | issue = 3 | pages = 981–6 | date = Aug 2003 | pmid = 12649131 | doi = 10.1182/blood-2002-11-3599 }}</ref> While AMKL is typically associated with the (1;22) translocation and expression of a mutant fusion protein, the genetic alterations that promote individuals with DS-AMKL are related to the GATA1 mutations, and the formation of a truncated transcription factor (GATA1s).
The clinical features associated with inactivating ''GATA1'' mutations or other causes of reduced GATA1 levels vary greatly with respect not only to the types of disease exhibited but also to disease severity.  This variation depends on at least four factors. '''First''', inactivating mutations in ''GATA1'' cause [[Genetic disorder#X-linked recessive|X-linked recessive diseases]]. Males, with only one ''GATA1'' gene, experience the diseases of these mutations while women, with two GATA1 genes, experience no or extremely mild evidence of these diseases unless they have inactivating mutations in both genes or their mutation is [[Mutation#By effect on function|dominant negative]], i.e. inhibiting the good gene's function. '''Second''', the extent to which a mutation reduces the cellular levels of fully functional GATA1 correlates with disease severity. '''Third''', inactivating ''GATA1'' mutations can cause different disease manifestations. For example, mutations in GATA1's N-ZnF that interfere with its interaction with FOG1 result in reduced red blood cell and platelet levels whereas mutations in N-ZnF that reduce its binding affinity to target genes cause a reduction in red blood cells plus [[thalassemia]]-type and [[porphyria]]-type symptoms. '''Fourth''', the genetic background of individuals can impact the type and severity of symptoms. For example, ''GATA1''-inactivating mutations in individuals with the extra [[chromosome 21]] of Down syndrome exhibit a proliferation of megakaryoblasts that infiltrate and consequentially directly damage liver, heart, marrow, pancreas, and skin plus secondarily life-threatening damage to the lungs and kidneys. These same individuals can develop secondary mutations in other genes that results in [[acute megakaryoblastic leukemia]].<ref name="pmid27235756"/><ref name="pmid22966823">{{cite journal | vauthors = Gamis AS, Smith FO | title = Transient myeloproliferative disorder in children with Down syndrome: clarity to this enigmatic disorder | journal = British Journal of Haematology | volume = 159 | issue = 3 | pages = 277–87 | date = November 2012 | pmid = 22966823 | doi = 10.1111/bjh.12041 }}</ref>


The same mutations in exon 2 of GATA1 present in almost all Down Syndrome-associated transient myeloproliferative disorder (TMD) or transient leukemia (TL), a precursor condition that evolves into AMKL in 30% of patients, that as many as 10% of Down Syndrome children may develop.<ref name="pmid14636651">{{cite journal | vauthors = Greene ME, Mundschau G, Wechsler J, McDevitt M, Gamis A, Karp J, Gurbuxani S, Arceci R, Crispino JD | title = Mutations in GATA1 in both transient myeloproliferative disorder and acute megakaryoblastic leukemia of Down syndrome | journal = Blood Cells, Molecules & Diseases | volume = 31 | issue = 3 | pages = 351–6 | year = 2003 | pmid = 14636651 | doi = 10.1016/j.bcmd.2003.08.001 }}</ref>  The incidence for the GATA1 mutation in about 4% of Down Syndrome patients, but less than 10% of those with the mutation developed AMKL.<ref name="pmid17576817">{{cite journal | vauthors = Pine SR, Guo Q, Yin C, Jayabose S, Druschel CM, Sandoval C | title = Incidence and clinical implications of GATA1 mutations in newborns with Down syndrome | journal = Blood | volume = 110 | issue = 6 | pages = 2128–31 | date = Sep 2007 | pmid = 17576817 | doi = 10.1182/blood-2007-01-069542 }}</ref> This mutation is present in the fetus, suggesting an early role in leukemogenesis. In addition to screening for TL, a GATA1 mutation at birth might serve as a bio-marker for an increased risk of DS-related AMKL.<ref name="pmid14684662">{{cite journal | vauthors = Shimada A, Xu G, Toki T, Kimura H, Hayashi Y, Ito E | title = Fetal origin of the GATA1 mutation in identical twins with transient myeloproliferative disorder and acute megakaryoblastic leukemia accompanying Down syndrome | journal = Blood | volume = 103 | issue = 1 | pages = 366 | date = Jan 2004 | pmid = 14684662 | doi = 10.1182/blood-2003-09-3219 }}</ref>
== Genetic disorders ==
''GATA1'' gene [[mutation]]s are associated with the development of various [[genetic disorders]] which may be familial (i.e. inherited) or newly acquired. In consequence of its X chromosome location, GATA1 mutations generally have a far greater physiological and clinical impact in men, who have only one X chromosome along with its ''GATA1'' gene, than woman, who have two of these chromosomes and genes: GATA1 mutations lead to [[Sex linkage|X-linked diseases]] occurring predominantly in males.<ref name="pmid27235756"/> Mutations in the activation domain of GATA1 (GATA1-S lacks this domain) are associated with the transient myeloproliferative disorder and acute megakaryoblastic leukemaia of Down syndrome while mutations in the N-ZnF motif of GATA1 and GATA1-S are associated with diseases similar to congenital dyserythropoietic anemia, congenital thrombocytopenia, and certain features that occur in [[thalassemia]], [[gray platelet syndrome]], [[congenital erythropoietic porphyria]], and [[myelofibrosis]].<ref name="pmid28566565"/>


== Interactions ==
=== Down syndrome-related disorders ===
{{main article|Down syndrome#Cancer}}


GATA1 has been shown to interact with several proteins - either directly by binding the protein or indirectly (functional interaction without direct binding).
==== Transient myeloproliferative disorder ====
{{main article|Transient myeloproliferative disease}}
Acquired inactivating mutations in the activation domain of GATA1 are the apparent cause of the transient myeloproliferative disorder that occurs in individuals with Down syndrome. These mutations are [[frameshift mutation|frameshift]]s in exon 2 that result in the failure to make GATA1 protein, continued formation of GATA1-S, and therefore a greatly reduced ability to regulate GATA1-targeted genes. The presence of these mutaions is restriced to cells bearing the trisomy 21 [[karyotype]] (i.e. extra [[chromosome 21]]) of Down syndrome: GATA1 inactivating mutations and trisomy 21 are necessary and sufficient for development of the disorder.<ref name="pmid22966823"/> Transient myeloproliferative disorder consists of a relatively mild but pathological proliferation of platelet-precursor cells, primarily [[megakaryoblast]]s, which often show an abnormal morphology that resembles immature [[myeloblast]]s (i.e. [[unipotent]] stem cells which differentiate into [[granulocytes]] and are the malignant proliferating cell in [[acute myeloid leukemia]]). [[Phenotype]] analyses indicate that these blasts belong to the megakaryoblast series. Abnormal findings include the frequent presence of excessive [[blast cell]] numbers, reduced platelet and red blood cell levels, increased circulating [[white blood cell]] levels, and infiltration of platelet-precursor cells into the bone marrow, liver, heart, pancreas, and skin.<ref name="pmid22966823"/> The disorder is thought to develop [[in utero]] and is detected at birth in about 10% of individuals with Down syndrome. It resolves totally within ~3 months but in the following 1-3 years progresses to acute megakaryoblastic leukemia in 20% to 30% of these individuals: transient myeloprolierative disorder is a [[Clone (cell biology)|clonal]] (abnormal cells derived from single parent cells), pre-leukemic condition and is classified as a [[myelodysplastic syndrome]] disease.<ref name="pmid26186939"/><ref name="pmid28566565"/><ref name="pmid28179280"/><ref name="pmid22966823"/>


{{Columns-list|2|
==== Acute megakaryoblastic leukemia ====
* [[BRD3]]<ref name="Lamonica_2011">{{cite journal | vauthors = Lamonica JM, Deng W, Kadauke S, Campbell AE, Gamsjaeger R, Wang H, Cheng Y, Billin AN, Hardison RC, Mackay JP, Blobel GA | title = Bromodomain protein Brd3 associates with acetylated GATA1 to promote its chromatin occupancy at erythroid target genes | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 108 | issue = 22 | date = May 2011 | pmid = 21536911 | doi = 10.1073/pnas.1102140108 | pages=E159-68 | pmc=3107332}}</ref><ref>{{cite journal | vauthors = Gamsjaeger R, Webb SR, Lamonica JM, Billin A, Blobel GA, Mackay JP | title = Structural basis and specificity of acetylated transcription factor GATA1 recognition by BET family bromodomain protein Brd3 | journal = Molecular and Cellular Biology | volume = 31 | issue = 13 | date = Jul 2011 | pmid = 21555453 | doi = 10.1128/MCB.05413-11 | pages=2632–40 | pmc=3133386}}</ref><ref>{{cite journal | vauthors = Stonestrom AJ, Hsu SC, Jahn KS, Huang P, Keller CA, Giardine BM, Kadauke S, Campbell AE, Evans P, Hardison RC, Blobel GA | title = Functions of BET proteins in erythroid gene expression | journal = Blood | date = Feb 2015 | pmid = 25696920 | doi = 10.1182/blood-2014-10-607309 | volume=125 | pages=2825–34 | pmc=4424630}}</ref>
{{main article|Acute megakaryoblastic leukemia}}
* [[BRD4]]<ref name="Lamonica_2011" />
Acute megakaryoblastic leukemia is a subtype of acute myeloid leukemia that is extremely rare in adults and, although still rare, more common in children. The childhood disease is classified into two major subgroups based on its occurrence in individuals with or without [[Down syndrome]]. The disease in Down syndrome occurs in 20% to 30% of individuals who previously had transient myeloproliferative disorder.  Their ''GATA1'' mutations are [[frameshift mutation|frameshift]]s in exon 2 that result in the failure to make GATA1 protein, continued formation of GATA1-S, and thus a greatly reduced ability to regulate GATA1-targeted genes. Transient myeloproliferative disorder is detected at or soon after birth and generally resolves during the next months but is followed within 1-3 years by acute megakaryoblastic leukemia.<ref name="pmid26186939"/> During this 1-3 year interval, individuals accumulate multiple [[Mutation#Somatic mutations|somatic mutations]] in cells bearing inactivating GATA1 mutations plus trisomy 21. These mutations are thought to result from the uncontrolled proliferation of blast cells caused by the ''GATAT1'' mutation in the presence of the extra chromosome 21 and to be responsible for progression of the transient disorder to leukemia. The mutations occur in one or, more commonly, multiple genes including: ''[[TP53]], [[RUNX1]], [[FLT3]], [[ERG (gene)|ERG]], [[DYRK1A]], [[CHAF1B]], [[HLCS]], [[CTCF]], [[STAG2]], [[RAD21]], [[SMC3]], [[SMC1A]], [[NIPBL]], [[SUZ12]], [[PRC2]], [[JAK1]], [[JAK2]], [[JAK3]], [[Thrombopoietin receptor|MPL]], [[KRAS]], [[Neuroblastoma RAS viral oncogene homolog|NRAS]], [[SH2B3]]'',  and ''MIR125B2'' which is the gene for [[microRNA]] MiR125B2.<ref name="pmid26186939"/><ref name="pmid22867885">{{cite journal | vauthors = Seewald L, Taub JW, Maloney KW, McCabe ER | title = Acute leukemias in children with Down syndrome | journal = Molecular Genetics and Metabolism | volume = 107 | issue = 1-2 | pages = 25–30 | date = September 2012 | pmid = 22867885 | doi = 10.1016/j.ymgme.2012.07.011 }}</ref>
* [[FLI1]]<ref name="pmid2724402">{{cite journal | vauthors = Lahiri K, Dole MG, Vidwans AS, Kamat J, Kandoth P | title = Acute glomerulonephritis | journal = Journal of Tropical Pediatrics | volume = 35 | issue = 2 | pages = 92 | date = Apr 1989 | pmid = 2724402 | doi = 10.1093/tropej/35.2.92 }}</ref><ref name="pmid12556498">{{cite journal | vauthors = Starck J, Cohet N, Gonnet C, Sarrazin S, Doubeikovskaia Z, Doubeikovski A, Verger A, Duterque-Coquillaud M, Morle F | title = Functional cross-antagonism between transcription factors FLI-1 and EKLF | journal = Molecular and Cellular Biology | volume = 23 | issue = 4 | pages = 1390–402 | date = Feb 2003 | pmid = 12556498 | pmc = 141137 | doi = 10.1128/MCB.23.4.1390-1402.2003 }}</ref>
 
* [[HDAC1]]<ref name="pmid14668799">{{cite journal | vauthors = Watamoto K, Towatari M, Ozawa Y, Miyata Y, Okamoto M, Abe A, Naoe T, Saito H | title = Altered interaction of HDAC5 with GATA-1 during MEL cell differentiation | journal = Oncogene | volume = 22 | issue = 57 | pages = 9176–84 | date = Dec 2003 | pmid = 14668799 | doi = 10.1038/sj.onc.1206902 }}</ref>
=== Diamond–Blackfan anemia ===
* [[LMO2]]<ref name="Osada_1995">{{cite journal | vauthors = Osada H, Grutz G, Axelson H, Forster A, Rabbitts TH | title = Association of erythroid transcription factors: complexes involving the LIM protein RBTN2 and the zinc-finger protein GATA1 | journal = Proceedings of the National Academy of Sciences of the United States of America | volume = 92 | issue = 21 | pages = 9585–9 | date = Oct 1995 | pmid = 7568177 | pmc = 40846 | doi = 10.1073/pnas.92.21.9585 }}</ref>
{{main article|Diamond-Blackfan anemia}}
* [[TAL1]]<ref name="pmid16407974">{{cite journal | vauthors = Goardon N, Lambert JA, Rodriguez P, Nissaire P, Herblot S, Thibault P, Dumenil D, Strouboulis J, Romeo PH, Hoang T | title = ETO2 coordinates cellular proliferation and differentiation during erythropoiesis | journal = The EMBO Journal | volume = 25 | issue = 2 | pages = 357–66 | date = Jan 2006 | pmid = 16407974 | pmc = 1383517 | doi = 10.1038/sj.emboj.7600934 }}</ref>
Diamond–Blackfan anemia is a familial (i.e. inherited) (45% of cases) or acquired (55% of cases) genetic disease that presents in [[infancy]] or, less commonly, later childhood as [[aplastic anemia]] and the circulation of abnormally enlarged [[red blood cell]]s. Other types of blood cell and platelets circulate at normal levels and appear normal in structure. About half of afflicted individuals have various [[birth defects]].<ref name="pmid29081386"/> The disease is regarded as a uniformly genetic disease although the genes causing it have not been identified in ~30% of cases. In virtually all the remaining cases, [[autosomal recessive]] inactivating mutations occur in any one of 20 of the 80 genes encoding [[ribosomal protein]]s. About 90% of the latter mutations occur in 6 ribosomal protein genes viz., ''[[RPS19]], [[RPL5]], [[RPS26]], [[RPL11]], [[RPL35A]]'', and ''[[RPS24]]''.<ref name="pmid28566565"/><ref name="pmid29081386"/> However, several cases of familial Diamond-Blackfan anemia have been associated with ''GATA1'' gene mutations in the apparent absence of a mutation in ribosomal protein genes. These ''GATA1'' mutations occur in an exon 2 splice site or the [[start codon]] of GATA1, cause the production of the GATA1-S in the absence of the GATA1 transcription factor, and therefore are gene-inactivating in nature. It is proposed that these ''GATA1'' mutations are a cause for Diamond Blackfan anemia.<ref name="pmid28566565"/><ref name="pmid27235756"/><ref name="pmid28179280"/>
* [[Zinc finger and BTB domain-containing protein 16|ZBTB16]]<ref name="pmid12242665">{{cite journal | vauthors = Labbaye C, Quaranta MT, Pagliuca A, Militi S, Licht JD, Testa U, Peschle C | title = PLZF induces megakaryocytic development, activates Tpo receptor expression and interacts with GATA1 protein | journal = Oncogene | volume = 21 | issue = 43 | pages = 6669–79 | date = Sep 2002 | pmid = 12242665 | doi = 10.1038/sj.onc.1205884 }}</ref>
 
* [[ZFPM2]]<ref name="pmid10438528">{{cite journal | vauthors = Holmes M, Turner J, Fox A, Chisholm O, Crossley M, Chong B | title = hFOG-2, a novel zinc finger protein, binds the co-repressor mCtBP2 and modulates GATA-mediated activation | journal = The Journal of Biological Chemistry | volume = 274 | issue = 33 | pages = 23491–8 | date = Aug 1999 | pmid = 10438528 | doi = 10.1074/jbc.274.33.23491 }}</ref>
=== Combined anemia-thrombocytopenia syndromes ===
}}
{{main article|congenital dyserythropoietic anemia}}
Certain ''GATA1''-inactivatng mutations are associated with familial or, less commonly, sporadic X-linked disorders that consist of anemia and thrombocytopenia due to a failure in the maturation of red blood cell and platelet precursors plus other hematological abnormalities. These ''GATA1'' mutations are identified by an initial letter identifying the normal amino acid followed by a number giving the position of this amino acid in GATA1, followed by a final letter identifying the amino acid substituted for the normal one. The amino acids are identified as V=[[valine]]; M=[[methionine]]; G=[[glycine]]; S=[[serine]], D=[[aspartic acid]]; Y=[[tyrosine]], R=[[arginine]]; W=[[tryptophan]], Q=[[glutamine]]]). These mutations and some key abnormalities they cause are:<ref name="pmid28566565"/><ref name="pmid28179280"/><ref name="pmid22886561">{{cite journal | vauthors = Balduini CL, Savoia A | title = Genetics of familial forms of thrombocytopenia | journal = Human Genetics | volume = 131 | issue = 12 | pages = 1821–32 | date = December 2012 | pmid = 22886561 | doi = 10.1007/s00439-012-1215-x }}</ref><ref name="pmid28550189">{{cite journal | vauthors = Russo R, Andolfo I, Gambale A, De Rosa G, Manna F, Arillo A, Wandroo F, Bisconte MG, Iolascon A | title = GATA1 erythroid-specific regulation of SEC23B expression and its implication in the pathogenesis of congenital dyserythropoietic anemia type II | journal = Haematologica | volume = 102 | issue = 9 | pages = e371–e374 | date = September 2017 | pmid = 28550189 | pmc = 5685218 | doi = 10.3324/haematol.2016.162966 | url = }}</ref>  
* V205M: familial disease characterized by severe anemia in fetuses and newborns; bone marrow has increased numbers of malformed platelet and red blood cell precursors.
* G208S and D218G: familial disease characterized by severe bleeding, reduced number of circulating platelets which are malformed (i.e. enlarged), and mild anemia.
* D218Y: familial disease similar to but more severe that the disease cause by G209S and D218G mutations.
* R216W: characterized by a [[beta thalassemia]]-type disease, i.e. [[microcytic anemia]], absence of [[hemoglobin B]], and [[hereditary persistence of fetal hemoglobin]]; symptoms of [[congenital erythropoietic porphyria]]; mild to moderately severe thrombocytopenia with features of the gray platelet syndrome.
* R216Q: familial disease characterized by mild anemia with features of heterozygous rather than homozygous (i.e. overt) beta thalassemia; mild thrombocytopenia with features of the gray platelet syndrome.
* G208R: disease characterized by mild anemia and severe thrombocytopenia with malformed erythroblasts and megakaryoblasts in the bone marrow. Structural features of these cells were similar to those observed in congenital dyserythropoietic anemia.
*-183G>A: rare [[Single-nucleotide polymorphism]] (rs113966884<ref>https://www.ncbi.nlm.nih.gov/SNP/snp_ref.cgi?rs=113966884&pt=1rE4MuqHtulrewqkpcs_tGJAQyAyRLUXCOF_U8H-YbhAT2s16HJ</ref>) in which the [[nucleotide]] [[adenine]] replaces [[guanine]] in DNA at the position 183 nucleotides upstream of the [[Start codon|start]] of ''GATA1''; disorder characterized as mild anemia with structural features in bone marrow red cell precursors similar to those observed in congenital dyserythropoietic anemia.
 
The [[Gray platelet syndrome]] is a rare congenital bleeding disorder caused by reductions or absence of [[alpha-granules]] in platelets. Alpha-granules contain various factors which contribute to blood clotting and other functions. In their absence, platelets are defective. The syndrome is commonly considered to result solely from mutations in the ''[[NBEAL2]]'' gene located on human [[chromosome 3]] at position p21. In these cases, the syndrome follows [[autosomal recessive]] inheritance, causes a mild to moderate bleeding tendency, and may be accompanied by a defect in the secretion of the granule contents in [[neutrophils]]. There are other causes for a congenital platelet alpha-granule-deficient bleeding disorder viz., the autosomal recessive disease of [[Arc syndrome]] caused by mutations in either the ''[[VPS33B]]'' (on human chromosome 15 at q26) or ''[[VIPAS39]]'' (on chromosome 14 at q34); the [[autosomal dominant]] disease of GFI1B-related syndrome caused by mutations in ''[[GFI1B]]'' (located on human chromosome 9 at q34); and the disease caused by R216W and R216Q mutations in GATA1. The GATA1 mutation-related disease resembles the one caused by ''NBEAL2'' mutations in that it is associated with the circulation of a reduced number (i.e. [[thrombocytopenia]]) of abnormally enlarged (i.e. macrothrombocytes), alpha-granule deficient platelets. It differs from the ''NBEAL2''-induced disease in that it is X chromosome-linked, accompanied by a moderately severe bleeding tendency, and associated with abnormalities in red blood cells (e.g. anemia, a [[thalassemia]]-like disorder due to unbalanced hemoglobin production, and/or a [[porphyria]]-like disorder.<ref name="pmid26971401">{{cite journal | vauthors = Nurden AT, Nurden P | title = Should any genetic defect affecting α-granules in platelets be classified as gray platelet syndrome? | journal = American Journal of Hematology | volume = 91 | issue = 7 | pages = 714–8 | date = July 2016 | pmid = 26971401 | doi = 10.1002/ajh.24359 }}</ref><ref name="pmid22886561"/> A recent study found that GATA1 is a strong enhancer of ''NBEAL2'' expression and that the R216W and R216Q inactivating mutations in ''GATA1'' may cause the development of alpha granule-deficient platelets by failing to stimulate the expression of NBDAL2 protein.<ref name="pmid28082341">{{cite journal | vauthors = Wijgaerts A, Wittevrongel C, Thys C, Devos T, Peerlinck K, Tijssen MR, Van Geet C, Freson K | title = The transcription factor GATA1 regulates NBEAL2 expression through a long-distance enhancer | journal = Haematologica | volume = 102 | issue = 4 | pages = 695–706 | date = April 2017 | pmid = 28082341 | pmc = 5395110 | doi = 10.3324/haematol.2016.152777 | url = }}</ref> Given these differences, the ''GATA1'' mutation-related disorder appears better classified as clinically and pathologically different than the gray platelet syndrome.<ref name="pmid26971401"/>
 
== GATA1 in myelofibrosis ==
{{main article|Myelofibrosis}}
Myelofibrosis is a rare hematological malignancy characterized by progressive fibrosis of the bone marrow, [[extramedullary hematopoiesis]] (i.e. formation of blood cells outside of their normal site in the bone marrow), variable reductions in the levels of circulating blood cells, increases in the circulating levels of the precursors to the latter cells, abnormalities in platelet precursor cell maturation, and the clustering of grossly malformed [[megakaryocytes]] in the bone marrow. Ultimately, the disease may progress to [[leukemia]]. Recent studies indicate that the megakaryocytes but not other cell types in rare cases of myelofibrosis have greatly reduced levels of GATA1 as a result of a ribosomal deficiency in [[Translation (biology)|translating]] GATA1 [[mRNA]] into GATA1 transcription factor. The studies suggest that these reduced levels of GATA1 contribute to the progression of myelofibrosis by leading to an impairment in platelet precursor cell maturation, by promoting extramedullary hematopoiesis, and, possibly, by contributing to its [[leukemic]] transformation.<ref name="pmid29562644"/><ref name="pmid29611379"/><ref name="pmid28240607"/>


== References ==
== References ==
Line 61: Line 82:


== External links ==
== External links ==
* [http://www.genecards.org/cgi-bin/carddisp.pl?gene=GATA1 Genecards]
* [https://www.genecards.org/cgi-bin/carddisp.pl?gene=GATA1 Genecards]
* [https://www.ncbi.nlm.nih.gov/bookshelf/br.fcgi?book=gene&part=gata1 GeneReviews/NCBI/NIH/UW entry on GATA1-Related X-Linked Cytopenia]
* [https://www.ncbi.nlm.nih.gov/bookshelf/br.fcgi?book=gene&part=gata1 GeneReviews/NCBI/NIH/UW entry on GATA1-Related X-Linked Cytopenia]
* [http://www.dsi.univ-paris5.fr/genatlas/fiche.php?symbol=GATA1 Geneatlas]
* [http://www.dsi.univ-paris5.fr/genatlas/fiche.php?symbol=GATA1 Geneatlas]
* [http://www.infobiogen.fr/services/chromcancer/Genes/GATA1ID40689chXp11.html Infobiogen]
* [http://www.infobiogen.fr/services/chromcancer/Genes/GATA1ID40689chXp11.html Infobiogen]
Line 71: Line 92:
{{Transcription factors|g2}}
{{Transcription factors|g2}}


[[Category:Human genes]]
Other types of ''GATA2'' mutations cause the over-expression of the GATA2 transcription factor. This overexpression is associated with the development of non-familial AML. Apparently, the ''GATA2'' gene's expression level must be delicately balanced between deficiency and excess in order to avoid life-threatening disease.<ref name="pmid25619630">{{cite journal | vauthors = Mir MA, Kochuparambil ST, Abraham RS, Rodriguez V, Howard M, Hsu AP, Jackson AE, Holland SM, Patnaik MM | title = Spectrum of myeloid neoplasms and immune deficiency associated with germline GATA2 mutations | journal = Cancer Medicine | volume = 4 | issue = 4 | pages = 490–9 | date = April 2015 | pmid = 25619630 | pmc = 4402062 | doi = 10.1002/cam4.384 | url = }}</ref><ref name="pmid28179282">{{cite journal | vauthors = Katsumura KR, Bresnick EH | title = The GATA factor revolution in hematology | journal = Blood | volume = 129 | issue = 15 | pages = 2092–2102 | date = April 2017 | pmid = 28179282 | pmc = 5391619 | doi = 10.1182/blood-2016-09-687871 | url = }}</ref>
[[Category:Transcription factors]]

Revision as of 17:53, 5 September 2018

VALUE_ERROR (nil)
Identifiers
Aliases
External IDsGeneCards: [1]
Orthologs
SpeciesHumanMouse
Entrez
Ensembl
UniProt
RefSeq (mRNA)

n/a

n/a

RefSeq (protein)

n/a

n/a

Location (UCSC)n/an/a
PubMed searchn/an/a
Wikidata
View/Edit Human

GATA-binding factor 1 or GATA-1 (also termed Erythroid transcription factor) is the founding member of the GATA family of transcription factors. This protein is widely expressed throughout vertebrate species. In humans and mice, it is encoded by the GATA1 and Gata1 genes, respectively. These genes are located on the X chromosome in both species.[1][2]

GATA1 regulates the expression (i.e. formation of the genes' products) of an ensemble of genes that mediate the development of red blood cells and platelets. Its critical roles in red blood cell formation include promoting the maturation of precursor cells, e.g. erythroblasts, to red blood cells and stimulating these cells to erect their cytoskeleton and biosynthesize their oxygen-carrying components viz., hemoglobin and heme. GATA1 plays a similarly critical role in the maturation of blood platelets from megakaryoblasts, promegakaryocytes, and megakaryocytes; the latter cells then shed membrane-enclosed fragments of their cytoplasm, i.e. platelets, into the blood.[1][3]

In consequence of the vital role that GATA1 has in the proper maturation of red blood cells and platelets, inactivating mutations in the GATA1 gene (i.e. mutations that result in the production of no, reduced levels of, or a less active GATA1) cause X chromosome-linked anemic and/or bleeding diseases due to the reduced formation and functionality of red blood cells and/or platelets, respectively, or, under certain circumstances, the pathological proliferation of megakaryoblasts. These diseases include transient myeloproliferative disorder occurring in Down syndrome, acute megakaryoblastic leukemia occurring in Down syndrome, Diamond-Blackfan anemia, and various combined anemia-thrombocytopenia syndromes including a gray platelet syndrome-type disorder.[4][5][6]

Reduced levels of GATA1 due to reductions in the translation of GATA1 mRNA into its transcription factor product are associated with promoting the progression of myelofibrosis, i.e. a malignant disease that involves the replacement of bone marrow cells by fibrous tissue and extramedullary hematopoiesis, i.e. the extension of blood cell-forming cells to sites outside of the bone marrow.[7][8]

Gene

The human GATA1 gene is located on the short (i.e. "p") arm of the X chromosome at position 11.23. It is 7.74 kilobases in length, consists of 6 exons, and codes for a full length protein, GATA1, of 414 amino acids as well as a shorter one, GATA1-S. GATA1-S lacks the first 83 amino acids of GATA1 and therefore consists of only 331 amino acids.[9][10][11] GATA1 codes for two zinc finger structural motifs, C-ZnF and N-ZnF, that are present in both GATA1 and GATA1-S proteins. These motifs are critical for both transcription factors' gene-regulating actions. N-ZnF is a frequent site of disease-causing mutations. Lacking the first 83 amino acids and therefore one of the two activation domains of GATA1, GATA1-S has significantly less gene-regulating activity than GATA1.[4][11]

Studies in Gata1-knockout mice, i.e. mice lacking the Gata1 gene, indicate that this gene is essential for the development and maintenance of blood-based and/or tissue-based hematological cells, particularly red blood cells and platelets but also eosinophils, basophils, mast cells, and dendritic cells. The knock-out mice die by day 11.5 of their embryonic development due to severe anemia that is associated with absence of cells of the red blood cell lineage, excessive numbers of malformed platelet-precursor cells, and an absence of platelets. These defects reflect the essential role of Gata-1 in stimulating the development, self-renewal, and/or maturation of red blood cell and platelet precursor cells. Studies using mice depleted of their Gata1 gene during adulthood show that: 1) Gata1 is required for the stimulation of erythropoiesis (i.e. increase in red blood cell formation) in response to stress and 2) Gata1-deficient adult mice invariably develop a form of myelofibrosis.[12][13]

GATA1 proteins

In both GATA1 and GATA1-S, C-ZnF (i.e. C-terminus zinc finger) binds to DNA-specific nucleic acid sequences sites viz., (T/A(GATA)A/G), on the expression-regulating sites of its target genes and in doing so either stimulates or suppresses the expression of these target genes. Their N-ZnF (i.e. N-terminus zinc fingers) interacts with an essential transcription factor-regulating nuclear protein, FOG1. FOG1 powerfully promotes or suppresses the actions that the two transcription factors have on most of their target genes. Similar to the knockout of Gata1, knockout of the mouse gene for FOG1, Zfpm1, causes total failure of red blood cell development and embryonic lethality by day 11.5. Based primarily on mouse studies, it is proposed that the GATA1-FOG1 complex promotes human erythropoiesis by recruiting and binding with at least two gene expression-regulating complexes, Mi-2/NuRD complex (a chromatin remodeler) and CTBP1 (a histone deacetylase) and three gene expression-regulating proteins, SET8 (a GATA1-inhibiting histone methyltransferase), BRG1 (a transcription activator), and Mediator (a transcription co-activator). Other interactions include those with: BRD3 (remodels DNA nucleosomes),[14][15][16] BRD4 (binds acetylated lysine residues in DNA-associated histone to regulate gene accessibility),[14] FLI1 (a transcription factor that blocks erythroid differentiation),[17][18] HDAC1 (a histone deacetylase),[19] LMO2 (regulator of erythrocyte development),[20] ZBTB16 (transcription factor regulating cell cycle progression),[21] TAL1 (a transcription factor),[22] FOG2 (a transcription factor regulator),[23] and GATA2 (Displacement of GATA2 by GATA1, i.e. the "GATA switch", at certain gene-regulating sites is critical for red blood development in mice and, presumably, humans).[13][24][25] GATA1-FOG1 and GATA2-FOG1 interactions are critical for platelet formation in mice and may similarly be critical for this in humans.[13]

Physiology and Pathology

GATA1 was first described as a transcription factor that activates the hemoglobin B gene in the red blood cell precursors of chickens.[26] Subsequent studies in mice and isolated human cells found that GATA1 stimulates the expression of genes that promote the maturation of precursor cells (e.g. erythroblasts) to red blood cells while silencing genes that cause these precursors to proliferate and thereby to self-renew.[27][28] GATA1 stimulates this maturation by, for example, inducing the expression of genes in erythroid cells that contribute to the formation of their cytoskeleton and that make enzymes necessary for the biosynthesis of hemoglobins and heme, the oxygen-carrying components of red blood cells. GATA1-inactivating mutations may thereby result in a failure to produce sufficient numbers of and/or fully functional red blood cells.[1] Also based on mouse and isolated human cell studies, GATA1 appears to play a similarly critical role in the maturation of platelets from their precursor cells. This maturation involves the stimulation of megakaryoblasts to mature ultimately to megakaryocytes which cells shed membrane-enclosed fragments of their cytoplasm, i.e. platelets, into the blood. GATA1-inactivating mutations may thereby result in reduced levels of and/or dysfunctional blood platelets.[1][3]

Reduced levels of GATA1 due to defective translation of GATA1 mRNA in human megakaryocytes is associated with myelofibrosis, i.e. the replacement of bone marrow cells by fibrous tissue. Based primarily on mouse and isolated human cell studies, this myelofibrosis is thought to result from the accumulation of platelet precursor cells in the bone marrow and their release of excessive amounts of cytokines that stimulate bone marrow stromal cells to become fiber-secreting fibroblasts and osteoblasts. Based on mouse studies, low GATA1 levels are also thought to promote the development of splenic enlargement and extramedullary hematopoiesis in human myelofibrosis disease. These effects appear to result directly from the over-proliferation of abnormal platelet precursor cells.[7][8][29][30]

The clinical features associated with inactivating GATA1 mutations or other causes of reduced GATA1 levels vary greatly with respect not only to the types of disease exhibited but also to disease severity. This variation depends on at least four factors. First, inactivating mutations in GATA1 cause X-linked recessive diseases. Males, with only one GATA1 gene, experience the diseases of these mutations while women, with two GATA1 genes, experience no or extremely mild evidence of these diseases unless they have inactivating mutations in both genes or their mutation is dominant negative, i.e. inhibiting the good gene's function. Second, the extent to which a mutation reduces the cellular levels of fully functional GATA1 correlates with disease severity. Third, inactivating GATA1 mutations can cause different disease manifestations. For example, mutations in GATA1's N-ZnF that interfere with its interaction with FOG1 result in reduced red blood cell and platelet levels whereas mutations in N-ZnF that reduce its binding affinity to target genes cause a reduction in red blood cells plus thalassemia-type and porphyria-type symptoms. Fourth, the genetic background of individuals can impact the type and severity of symptoms. For example, GATA1-inactivating mutations in individuals with the extra chromosome 21 of Down syndrome exhibit a proliferation of megakaryoblasts that infiltrate and consequentially directly damage liver, heart, marrow, pancreas, and skin plus secondarily life-threatening damage to the lungs and kidneys. These same individuals can develop secondary mutations in other genes that results in acute megakaryoblastic leukemia.[11][31]

Genetic disorders

GATA1 gene mutations are associated with the development of various genetic disorders which may be familial (i.e. inherited) or newly acquired. In consequence of its X chromosome location, GATA1 mutations generally have a far greater physiological and clinical impact in men, who have only one X chromosome along with its GATA1 gene, than woman, who have two of these chromosomes and genes: GATA1 mutations lead to X-linked diseases occurring predominantly in males.[11] Mutations in the activation domain of GATA1 (GATA1-S lacks this domain) are associated with the transient myeloproliferative disorder and acute megakaryoblastic leukemaia of Down syndrome while mutations in the N-ZnF motif of GATA1 and GATA1-S are associated with diseases similar to congenital dyserythropoietic anemia, congenital thrombocytopenia, and certain features that occur in thalassemia, gray platelet syndrome, congenital erythropoietic porphyria, and myelofibrosis.[4]

Down syndrome-related disorders

Transient myeloproliferative disorder

Acquired inactivating mutations in the activation domain of GATA1 are the apparent cause of the transient myeloproliferative disorder that occurs in individuals with Down syndrome. These mutations are frameshifts in exon 2 that result in the failure to make GATA1 protein, continued formation of GATA1-S, and therefore a greatly reduced ability to regulate GATA1-targeted genes. The presence of these mutaions is restriced to cells bearing the trisomy 21 karyotype (i.e. extra chromosome 21) of Down syndrome: GATA1 inactivating mutations and trisomy 21 are necessary and sufficient for development of the disorder.[31] Transient myeloproliferative disorder consists of a relatively mild but pathological proliferation of platelet-precursor cells, primarily megakaryoblasts, which often show an abnormal morphology that resembles immature myeloblasts (i.e. unipotent stem cells which differentiate into granulocytes and are the malignant proliferating cell in acute myeloid leukemia). Phenotype analyses indicate that these blasts belong to the megakaryoblast series. Abnormal findings include the frequent presence of excessive blast cell numbers, reduced platelet and red blood cell levels, increased circulating white blood cell levels, and infiltration of platelet-precursor cells into the bone marrow, liver, heart, pancreas, and skin.[31] The disorder is thought to develop in utero and is detected at birth in about 10% of individuals with Down syndrome. It resolves totally within ~3 months but in the following 1-3 years progresses to acute megakaryoblastic leukemia in 20% to 30% of these individuals: transient myeloprolierative disorder is a clonal (abnormal cells derived from single parent cells), pre-leukemic condition and is classified as a myelodysplastic syndrome disease.[3][4][12][31]

Acute megakaryoblastic leukemia

Acute megakaryoblastic leukemia is a subtype of acute myeloid leukemia that is extremely rare in adults and, although still rare, more common in children. The childhood disease is classified into two major subgroups based on its occurrence in individuals with or without Down syndrome. The disease in Down syndrome occurs in 20% to 30% of individuals who previously had transient myeloproliferative disorder. Their GATA1 mutations are frameshifts in exon 2 that result in the failure to make GATA1 protein, continued formation of GATA1-S, and thus a greatly reduced ability to regulate GATA1-targeted genes. Transient myeloproliferative disorder is detected at or soon after birth and generally resolves during the next months but is followed within 1-3 years by acute megakaryoblastic leukemia.[3] During this 1-3 year interval, individuals accumulate multiple somatic mutations in cells bearing inactivating GATA1 mutations plus trisomy 21. These mutations are thought to result from the uncontrolled proliferation of blast cells caused by the GATAT1 mutation in the presence of the extra chromosome 21 and to be responsible for progression of the transient disorder to leukemia. The mutations occur in one or, more commonly, multiple genes including: TP53, RUNX1, FLT3, ERG, DYRK1A, CHAF1B, HLCS, CTCF, STAG2, RAD21, SMC3, SMC1A, NIPBL, SUZ12, PRC2, JAK1, JAK2, JAK3, MPL, KRAS, NRAS, SH2B3, and MIR125B2 which is the gene for microRNA MiR125B2.[3][32]

Diamond–Blackfan anemia

Diamond–Blackfan anemia is a familial (i.e. inherited) (45% of cases) or acquired (55% of cases) genetic disease that presents in infancy or, less commonly, later childhood as aplastic anemia and the circulation of abnormally enlarged red blood cells. Other types of blood cell and platelets circulate at normal levels and appear normal in structure. About half of afflicted individuals have various birth defects.[6] The disease is regarded as a uniformly genetic disease although the genes causing it have not been identified in ~30% of cases. In virtually all the remaining cases, autosomal recessive inactivating mutations occur in any one of 20 of the 80 genes encoding ribosomal proteins. About 90% of the latter mutations occur in 6 ribosomal protein genes viz., RPS19, RPL5, RPS26, RPL11, RPL35A, and RPS24.[4][6] However, several cases of familial Diamond-Blackfan anemia have been associated with GATA1 gene mutations in the apparent absence of a mutation in ribosomal protein genes. These GATA1 mutations occur in an exon 2 splice site or the start codon of GATA1, cause the production of the GATA1-S in the absence of the GATA1 transcription factor, and therefore are gene-inactivating in nature. It is proposed that these GATA1 mutations are a cause for Diamond Blackfan anemia.[4][11][12]

Combined anemia-thrombocytopenia syndromes

Certain GATA1-inactivatng mutations are associated with familial or, less commonly, sporadic X-linked disorders that consist of anemia and thrombocytopenia due to a failure in the maturation of red blood cell and platelet precursors plus other hematological abnormalities. These GATA1 mutations are identified by an initial letter identifying the normal amino acid followed by a number giving the position of this amino acid in GATA1, followed by a final letter identifying the amino acid substituted for the normal one. The amino acids are identified as V=valine; M=methionine; G=glycine; S=serine, D=aspartic acid; Y=tyrosine, R=arginine; W=tryptophan, Q=glutamine]). These mutations and some key abnormalities they cause are:[4][12][33][34]

  • V205M: familial disease characterized by severe anemia in fetuses and newborns; bone marrow has increased numbers of malformed platelet and red blood cell precursors.
  • G208S and D218G: familial disease characterized by severe bleeding, reduced number of circulating platelets which are malformed (i.e. enlarged), and mild anemia.
  • D218Y: familial disease similar to but more severe that the disease cause by G209S and D218G mutations.
  • R216W: characterized by a beta thalassemia-type disease, i.e. microcytic anemia, absence of hemoglobin B, and hereditary persistence of fetal hemoglobin; symptoms of congenital erythropoietic porphyria; mild to moderately severe thrombocytopenia with features of the gray platelet syndrome.
  • R216Q: familial disease characterized by mild anemia with features of heterozygous rather than homozygous (i.e. overt) beta thalassemia; mild thrombocytopenia with features of the gray platelet syndrome.
  • G208R: disease characterized by mild anemia and severe thrombocytopenia with malformed erythroblasts and megakaryoblasts in the bone marrow. Structural features of these cells were similar to those observed in congenital dyserythropoietic anemia.
  • -183G>A: rare Single-nucleotide polymorphism (rs113966884[35]) in which the nucleotide adenine replaces guanine in DNA at the position 183 nucleotides upstream of the start of GATA1; disorder characterized as mild anemia with structural features in bone marrow red cell precursors similar to those observed in congenital dyserythropoietic anemia.

The Gray platelet syndrome is a rare congenital bleeding disorder caused by reductions or absence of alpha-granules in platelets. Alpha-granules contain various factors which contribute to blood clotting and other functions. In their absence, platelets are defective. The syndrome is commonly considered to result solely from mutations in the NBEAL2 gene located on human chromosome 3 at position p21. In these cases, the syndrome follows autosomal recessive inheritance, causes a mild to moderate bleeding tendency, and may be accompanied by a defect in the secretion of the granule contents in neutrophils. There are other causes for a congenital platelet alpha-granule-deficient bleeding disorder viz., the autosomal recessive disease of Arc syndrome caused by mutations in either the VPS33B (on human chromosome 15 at q26) or VIPAS39 (on chromosome 14 at q34); the autosomal dominant disease of GFI1B-related syndrome caused by mutations in GFI1B (located on human chromosome 9 at q34); and the disease caused by R216W and R216Q mutations in GATA1. The GATA1 mutation-related disease resembles the one caused by NBEAL2 mutations in that it is associated with the circulation of a reduced number (i.e. thrombocytopenia) of abnormally enlarged (i.e. macrothrombocytes), alpha-granule deficient platelets. It differs from the NBEAL2-induced disease in that it is X chromosome-linked, accompanied by a moderately severe bleeding tendency, and associated with abnormalities in red blood cells (e.g. anemia, a thalassemia-like disorder due to unbalanced hemoglobin production, and/or a porphyria-like disorder.[36][33] A recent study found that GATA1 is a strong enhancer of NBEAL2 expression and that the R216W and R216Q inactivating mutations in GATA1 may cause the development of alpha granule-deficient platelets by failing to stimulate the expression of NBDAL2 protein.[37] Given these differences, the GATA1 mutation-related disorder appears better classified as clinically and pathologically different than the gray platelet syndrome.[36]

GATA1 in myelofibrosis

Myelofibrosis is a rare hematological malignancy characterized by progressive fibrosis of the bone marrow, extramedullary hematopoiesis (i.e. formation of blood cells outside of their normal site in the bone marrow), variable reductions in the levels of circulating blood cells, increases in the circulating levels of the precursors to the latter cells, abnormalities in platelet precursor cell maturation, and the clustering of grossly malformed megakaryocytes in the bone marrow. Ultimately, the disease may progress to leukemia. Recent studies indicate that the megakaryocytes but not other cell types in rare cases of myelofibrosis have greatly reduced levels of GATA1 as a result of a ribosomal deficiency in translating GATA1 mRNA into GATA1 transcription factor. The studies suggest that these reduced levels of GATA1 contribute to the progression of myelofibrosis by leading to an impairment in platelet precursor cell maturation, by promoting extramedullary hematopoiesis, and, possibly, by contributing to its leukemic transformation.[8][29][30]

References

  1. 1.0 1.1 1.2 1.3 Katsumura KR, DeVilbiss AW, Pope NJ, Johnson KD, Bresnick EH (September 2013). "Transcriptional mechanisms underlying hemoglobin synthesis". Cold Spring Harbor Perspectives in Medicine. 3 (9): a015412. doi:10.1101/cshperspect.a015412. PMC 3753722. PMID 23838521.
  2. Caiulo A, Nicolis S, Bianchi P, Zuffardi O, Bardoni B, Maraschio P, Ottolenghi S, Camerino G, Giglioni B (Feb 1991). "Mapping the gene encoding the human erythroid transcriptional factor NFE1-GF1 to Xp11.23". Human Genetics. 86 (4): 388–90. doi:10.1007/bf00201840. PMID 1999341.
  3. 3.0 3.1 3.2 3.3 3.4 Gruber TA, Downing JR (August 2015). "The biology of pediatric acute megakaryoblastic leukemia". Blood. 126 (8): 943–9. doi:10.1182/blood-2015-05-567859. PMC 4551356. PMID 26186939.
  4. 4.0 4.1 4.2 4.3 4.4 4.5 4.6 Fujiwara T (June 2017). "GATA Transcription Factors: Basic Principles and Related Human Disorders". The Tohoku Journal of Experimental Medicine. 242 (2): 83–91. doi:10.1620/tjem.242.83. PMID 28566565.
  5. "Entrez Gene: GATA1 GATA binding protein 1 (globin transcription factor 1)".
  6. 6.0 6.1 6.2 Da Costa L, O'Donohue MF, van Dooijeweert B, Albrecht K, Unal S, Ramenghi U, Leblanc T, Dianzani I, Tamary H, Bartels M, Gleizes PE, Wlodarski M, MacInnes AW (October 2017). "Molecular approaches to diagnose Diamond-Blackfan anemia: The EuroDBA experience". European Journal of Medical Genetics. doi:10.1016/j.ejmg.2017.10.017. PMID 29081386.
  7. 7.0 7.1 Verrucci M, Pancrazzi A, Aracil M, Martelli F, Guglielmelli P, Zingariello M, Ghinassi B, D'Amore E, Jimeno J, Vannucchi AM, Migliaccio AR (November 2010). "CXCR4-independent rescue of the myeloproliferative defect of the Gata1low myelofibrosis mouse model by Aplidin". Journal of Cellular Physiology. 225 (2): 490–9. doi:10.1002/jcp.22228. PMC 3780594. PMID 20458749.
  8. 8.0 8.1 8.2 Song MK, Park BB, Uhm JE (March 2018). "Understanding Splenomegaly in Myelofibrosis: Association with Molecular Pathogenesis". International Journal of Molecular Sciences. 19 (3). doi:10.3390/ijms19030898. PMC 5877759. PMID 29562644.
  9. https://www.ncbi.nlm.nih.gov/gene/2623
  10. http://genatlas.medecine.univ-paris5.fr/fiche.php?onglet=2&n=5186
  11. 11.0 11.1 11.2 11.3 11.4 Shimizu R, Yamamoto M (August 2016). "GATA-related hematologic disorders". Experimental Hematology. 44 (8): 696–705. doi:10.1016/j.exphem.2016.05.010. PMID 27235756.
  12. 12.0 12.1 12.2 12.3 Crispino JD, Horwitz MS (April 2017). "GATA factor mutations in hematologic disease". Blood. 129 (15): 2103–2110. doi:10.1182/blood-2016-09-687889. PMC 5391620. PMID 28179280.
  13. 13.0 13.1 13.2 Katsumura KR, Bresnick EH (April 2017). "The GATA factor revolution in hematology". Blood. 129 (15): 2092–2102. doi:10.1182/blood-2016-09-687871. PMC 5391619. PMID 28179282.
  14. 14.0 14.1 Lamonica JM, Deng W, Kadauke S, Campbell AE, Gamsjaeger R, Wang H, Cheng Y, Billin AN, Hardison RC, Mackay JP, Blobel GA (May 2011). "Bromodomain protein Brd3 associates with acetylated GATA1 to promote its chromatin occupancy at erythroid target genes". Proceedings of the National Academy of Sciences of the United States of America. 108 (22): E159–68. doi:10.1073/pnas.1102140108. PMC 3107332. PMID 21536911.
  15. Gamsjaeger R, Webb SR, Lamonica JM, Billin A, Blobel GA, Mackay JP (Jul 2011). "Structural basis and specificity of acetylated transcription factor GATA1 recognition by BET family bromodomain protein Brd3". Molecular and Cellular Biology. 31 (13): 2632–40. doi:10.1128/MCB.05413-11. PMC 3133386. PMID 21555453.
  16. Stonestrom AJ, Hsu SC, Jahn KS, Huang P, Keller CA, Giardine BM, Kadauke S, Campbell AE, Evans P, Hardison RC, Blobel GA (Feb 2015). "Functions of BET proteins in erythroid gene expression". Blood. 125: 2825–34. doi:10.1182/blood-2014-10-607309. PMC 4424630. PMID 25696920.,
  17. Lahiri K, Dole MG, Vidwans AS, Kamat J, Kandoth P (Apr 1989). "Acute glomerulonephritis". Journal of Tropical Pediatrics. 35 (2): 92. doi:10.1093/tropej/35.2.92. PMID 2724402.
  18. Starck J, Cohet N, Gonnet C, Sarrazin S, Doubeikovskaia Z, Doubeikovski A, Verger A, Duterque-Coquillaud M, Morle F (Feb 2003). "Functional cross-antagonism between transcription factors FLI-1 and EKLF". Molecular and Cellular Biology. 23 (4): 1390–402. doi:10.1128/MCB.23.4.1390-1402.2003. PMC 141137. PMID 12556498.
  19. Watamoto K, Towatari M, Ozawa Y, Miyata Y, Okamoto M, Abe A, Naoe T, Saito H (Dec 2003). "Altered interaction of HDAC5 with GATA-1 during MEL cell differentiation". Oncogene. 22 (57): 9176–84. doi:10.1038/sj.onc.1206902. PMID 14668799.
  20. Osada H, Grutz G, Axelson H, Forster A, Rabbitts TH (Oct 1995). "Association of erythroid transcription factors: complexes involving the LIM protein RBTN2 and the zinc-finger protein GATA1". Proceedings of the National Academy of Sciences of the United States of America. 92 (21): 9585–9. doi:10.1073/pnas.92.21.9585. PMC 40846. PMID 7568177.
  21. Labbaye C, Quaranta MT, Pagliuca A, Militi S, Licht JD, Testa U, Peschle C (Sep 2002). "PLZF induces megakaryocytic development, activates Tpo receptor expression and interacts with GATA1 protein". Oncogene. 21 (43): 6669–79. doi:10.1038/sj.onc.1205884. PMID 12242665.
  22. Goardon N, Lambert JA, Rodriguez P, Nissaire P, Herblot S, Thibault P, Dumenil D, Strouboulis J, Romeo PH, Hoang T (Jan 2006). "ETO2 coordinates cellular proliferation and differentiation during erythropoiesis". The EMBO Journal. 25 (2): 357–66. doi:10.1038/sj.emboj.7600934. PMC 1383517. PMID 16407974.
  23. Holmes M, Turner J, Fox A, Chisholm O, Crossley M, Chong B (Aug 1999). "hFOG-2, a novel zinc finger protein, binds the co-repressor mCtBP2 and modulates GATA-mediated activation". The Journal of Biological Chemistry. 274 (33): 23491–8. doi:10.1074/jbc.274.33.23491. PMID 10438528.
  24. Fujiwara Y, Browne CP, Cunniff K, Goff SC, Orkin SH (Oct 1996). "Arrested development of embryonic red cell precursors in mouse embryos lacking transcription factor GATA-1". Proceedings of the National Academy of Sciences of the United States of America. 93 (22): 12355–8. doi:10.1073/pnas.93.22.12355. PMC 37995. PMID 8901585.
  25. Campbell AE, Wilkinson-White L, Mackay JP, Matthews JM, Blobel GA (Jun 2013). "Analysis of disease-causing GATA1 mutations in murine gene complementation systems". Blood. 121 (26): 5218–27. doi:10.1182/blood-2013-03-488080. PMC 3695365. PMID 23704091.
  26. Evans T, Reitman M, Felsenfeld G (Aug 1988). "An erythrocyte-specific DNA-binding factor recognizes a regulatory sequence common to all chicken globin genes". Proceedings of the National Academy of Sciences of the United States of America. 85 (16): 5976–80. doi:10.1073/pnas.85.16.5976. PMC 281888. PMID 3413070.
  27. Welch JJ, Watts JA, Vakoc CR, Yao Y, Wang H, Hardison RC, Blobel GA, Chodosh LA, Weiss MJ (Nov 2004). "Global regulation of erythroid gene expression by transcription factor GATA-1". Blood. 104 (10): 3136–47. doi:10.1182/blood-2004-04-1603. PMID 15297311.
  28. Cheng Y, Wu W, Kumar SA, Yu D, Deng W, Tripic T, King DC, Chen KB, Zhang Y, Drautz D, Giardine B, Schuster SC, Miller W, Chiaromonte F, Zhang Y, Blobel GA, Weiss MJ, Hardison RC (Dec 2009). "Erythroid GATA1 function revealed by genome-wide analysis of transcription factor occupancy, histone modifications, and mRNA expression". Genome Research. 19 (12): 2172–84. doi:10.1101/gr.098921.109. PMC 2792182. PMID 19887574.
  29. 29.0 29.1 Yang N, Park S, Cho MS, Lee M, Hong KS, Mun YC, Seong CM, Huh HJ, Huh J (July 2018). "GATA1 Expression in BCR/ABL1-negative Myeloproliferative Neoplasms". Annals of Laboratory Medicine. 38 (4): 296–305. doi:10.3343/alm.2018.38.4.296. PMID 29611379.
  30. 30.0 30.1 Gilles L, Arslan AD, Marinaccio C, Wen QJ, Arya P, McNulty M, Yang Q, Zhao JC, Konstantinoff K, Lasho T, Pardanani A, Stein B, Plo I, Sundaravel S, Wickrema A, Migliaccio A, Gurbuxani S, Vainchenker W, Platanias LC, Tefferi A, Crispino JD (April 2017). "Downregulation of GATA1 drives impaired hematopoiesis in primary myelofibrosis". The Journal of Clinical Investigation. 127 (4): 1316–1320. doi:10.1172/JCI82905. PMC 5373858. PMID 28240607. Retrieved 2018-06-26.
  31. 31.0 31.1 31.2 31.3 Gamis AS, Smith FO (November 2012). "Transient myeloproliferative disorder in children with Down syndrome: clarity to this enigmatic disorder". British Journal of Haematology. 159 (3): 277–87. doi:10.1111/bjh.12041. PMID 22966823.
  32. Seewald L, Taub JW, Maloney KW, McCabe ER (September 2012). "Acute leukemias in children with Down syndrome". Molecular Genetics and Metabolism. 107 (1–2): 25–30. doi:10.1016/j.ymgme.2012.07.011. PMID 22867885.
  33. 33.0 33.1 Balduini CL, Savoia A (December 2012). "Genetics of familial forms of thrombocytopenia". Human Genetics. 131 (12): 1821–32. doi:10.1007/s00439-012-1215-x. PMID 22886561.
  34. Russo R, Andolfo I, Gambale A, De Rosa G, Manna F, Arillo A, Wandroo F, Bisconte MG, Iolascon A (September 2017). "GATA1 erythroid-specific regulation of SEC23B expression and its implication in the pathogenesis of congenital dyserythropoietic anemia type II". Haematologica. 102 (9): e371–e374. doi:10.3324/haematol.2016.162966. PMC 5685218. PMID 28550189.
  35. https://www.ncbi.nlm.nih.gov/SNP/snp_ref.cgi?rs=113966884&pt=1rE4MuqHtulrewqkpcs_tGJAQyAyRLUXCOF_U8H-YbhAT2s16HJ
  36. 36.0 36.1 Nurden AT, Nurden P (July 2016). "Should any genetic defect affecting α-granules in platelets be classified as gray platelet syndrome?". American Journal of Hematology. 91 (7): 714–8. doi:10.1002/ajh.24359. PMID 26971401.
  37. Wijgaerts A, Wittevrongel C, Thys C, Devos T, Peerlinck K, Tijssen MR, Van Geet C, Freson K (April 2017). "The transcription factor GATA1 regulates NBEAL2 expression through a long-distance enhancer". Haematologica. 102 (4): 695–706. doi:10.3324/haematol.2016.152777. PMC 5395110. PMID 28082341.

Further reading

External links

Other types of GATA2 mutations cause the over-expression of the GATA2 transcription factor. This overexpression is associated with the development of non-familial AML. Apparently, the GATA2 gene's expression level must be delicately balanced between deficiency and excess in order to avoid life-threatening disease.[1][2]

  1. Mir MA, Kochuparambil ST, Abraham RS, Rodriguez V, Howard M, Hsu AP, Jackson AE, Holland SM, Patnaik MM (April 2015). "Spectrum of myeloid neoplasms and immune deficiency associated with germline GATA2 mutations". Cancer Medicine. 4 (4): 490–9. doi:10.1002/cam4.384. PMC 4402062. PMID 25619630.
  2. Katsumura KR, Bresnick EH (April 2017). "The GATA factor revolution in hematology". Blood. 129 (15): 2092–2102. doi:10.1182/blood-2016-09-687871. PMC 5391619. PMID 28179282.