author_facet Bhatnagar, Neha
Perkins, Kelly
Filippi, Sarah
Richmond, Helen
Bonnici, Joanna
Alford, Kate
Hall, Georgina
Juban, Gaëtan
McGowan, Simon
Roy, Anindita
Elliott, Natalina
Stumpf, Michael
Norton, Alice
Vyas, Paresh
Roberts, Irene
Bhatnagar, Neha
Perkins, Kelly
Filippi, Sarah
Richmond, Helen
Bonnici, Joanna
Alford, Kate
Hall, Georgina
Juban, Gaëtan
McGowan, Simon
Roy, Anindita
Elliott, Natalina
Stumpf, Michael
Norton, Alice
Vyas, Paresh
Roberts, Irene
author Bhatnagar, Neha
Perkins, Kelly
Filippi, Sarah
Richmond, Helen
Bonnici, Joanna
Alford, Kate
Hall, Georgina
Juban, Gaëtan
McGowan, Simon
Roy, Anindita
Elliott, Natalina
Stumpf, Michael
Norton, Alice
Vyas, Paresh
Roberts, Irene
spellingShingle Bhatnagar, Neha
Perkins, Kelly
Filippi, Sarah
Richmond, Helen
Bonnici, Joanna
Alford, Kate
Hall, Georgina
Juban, Gaëtan
McGowan, Simon
Roy, Anindita
Elliott, Natalina
Stumpf, Michael
Norton, Alice
Vyas, Paresh
Roberts, Irene
Blood
Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
Cell Biology
Hematology
Immunology
Biochemistry
author_sort bhatnagar, neha
spelling Bhatnagar, Neha Perkins, Kelly Filippi, Sarah Richmond, Helen Bonnici, Joanna Alford, Kate Hall, Georgina Juban, Gaëtan McGowan, Simon Roy, Anindita Elliott, Natalina Stumpf, Michael Norton, Alice Vyas, Paresh Roberts, Irene 0006-4971 1528-0020 American Society of Hematology Cell Biology Hematology Immunology Biochemistry http://dx.doi.org/10.1182/blood.v124.21.2349.2349 <jats:title>Abstract</jats:title> <jats:p>Children with Down syndrome (DS; trisomy 21) have an increased risk of acute myeloid leukemia (ML-DS) in the first 5 years of life. In most cases ML-DS is preceded by Transient Abnormal Myelopoiesis (TAM), a fetal/neonatal pre-leukemic disorder unique to DS which regresses after birth. Both TAM and ML-DS harbor acquired N-terminal mutations in the hematopoietic transcription factor gene GATA1. In a prospective study of 200 DS neonates, we recently showed that 29% had acquired GATA1 mutations including 17/200 (8.5%) with clinical or hematologic evidence of TAM; the remaining 20.5% were clinically and hematologically 'silent', with smaller mutant GATA1 clones and lower blast frequency compared to overt TAM. The reasons why some DS neonates develop overt TAM and the factors which determine mutant GATA1 clone size are unknown. To address this, we analysed data from neonates in the prospective Oxford-Imperial DS Cohort Study and investigated the impact of 30 clinical and hematologic factors on clone size using statistical and mathematical modelling. Mutant GATA1 clones were determined in 54 neonates by targeted next generation sequencing of GATA1 exon 2 (mutation detection limit 0.3%). Clone size was determined by analysing original unprocessed reads using less stringent filtering parameters and counting reads containing mutated v total sequence. Correlation analysis identified 4 hematologic variables correlated with mutant GATA1 clone size: circulating nucleated red cells (r=+0.5003; p=0.0001), platelets (r=+0.436; p=0.001), total leukocytes (r=+0.7094; p&lt;0.001) and % blasts (r=+0.7292; p&lt;0.001); clone size was higher in neonates with megakaryocyte fragments (p=0.0024) and platelets &gt;150x109/L (p=0.019). Numbers of neutrophils, monocytes, basophils, eosinophils and lymphocytes did not correlate with GATA1 clone size. Clinical variables significantly correlated with clone size were hepatomegaly (p=0.0016), splenomegaly (p=0.0001) and rash (0.0174). The only pregnancy-related variables affecting mutant GATA1 clone size were intrauterine growth restriction and maternal diabetes (p=0.0156). Linear regression to determine the joint impact of all 30 variables on clone size (r2=0.88) followed by Lasso penalization identified the same 4 hematologic variables (nucleated red cells, platelets, total leukocytes and % blasts); Lasso penalized regression with these 4 variables gave a coefficient of determination of 0.63. Together these data suggest that chronic intrauterine hypoxia may affect expansion/differentiation of mutant GATA1 clones in DS. Consistent with this, nucleated red cells from 3 neonates with TAM all harbored GATA1 mutations identical to those in total circulating nucleated cells. Since neither perinatal infection nor gestational age at birth correlated with mutant GATA1 clone size, infection-related cytokines and the timing of acquisition of a mutant GATA1 clone during fetal development may not play a major role in determining clone size. Finally, a hierarchical model to investigate the impact of GATA1 mutation on hematopoietic stem and progenitor (HSPC) differentiation in DS neonates using a Bayesian approach also predicted increased erythroid cell output from GATA1 mutated HSPC v HSPC without a GATA1 mutation. In conclusion, in neonates with DS the size of the mutant GATA1 clone correlates with the presence of clinical signs of hepatomegaly, splenomegaly and skin rash; mutant GATA1 clone size correlates with the numbers of circulating nucleated red cells, platelets and blast cells suggesting that GATA1 mutant HSPC retain the ability to differentiate down the erythroid and megakaryocyte lineage; intrauterine hypoxia may be one of the factors driving expansion and/or maturation of the GATA1 mutant clone during fetal life in DS.</jats:p> <jats:sec> <jats:title>Disclosures</jats:title> <jats:p>No relevant conflicts of interest to declare.</jats:p> </jats:sec> Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome Blood
doi_str_mv 10.1182/blood.v124.21.2349.2349
facet_avail Online
Free
finc_class_facet Biologie
Medizin
Chemie und Pharmazie
format ElectronicArticle
fullrecord blob:ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE4Mi9ibG9vZC52MTI0LjIxLjIzNDkuMjM0OQ
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE4Mi9ibG9vZC52MTI0LjIxLjIzNDkuMjM0OQ
institution DE-Gla1
DE-Zi4
DE-15
DE-Pl11
DE-Rs1
DE-105
DE-14
DE-Ch1
DE-L229
DE-D275
DE-Bn3
DE-Brt1
DE-Zwi2
DE-D161
imprint American Society of Hematology, 2014
imprint_str_mv American Society of Hematology, 2014
issn 0006-4971
1528-0020
issn_str_mv 0006-4971
1528-0020
language English
mega_collection American Society of Hematology (CrossRef)
match_str bhatnagar2014clinicalandhematologicimpactoffetalandperinatalvariablesonmutantgata1clonesizeinneonateswithdownsyndrome
publishDateSort 2014
publisher American Society of Hematology
recordtype ai
record_format ai
series Blood
source_id 49
title Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
title_unstemmed Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
title_full Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
title_fullStr Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
title_full_unstemmed Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
title_short Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
title_sort clinical and hematologic impact of fetal and perinatal variables on mutant gata1 clone size in neonates with down syndrome
topic Cell Biology
Hematology
Immunology
Biochemistry
url http://dx.doi.org/10.1182/blood.v124.21.2349.2349
publishDate 2014
physical 2349-2349
description <jats:title>Abstract</jats:title> <jats:p>Children with Down syndrome (DS; trisomy 21) have an increased risk of acute myeloid leukemia (ML-DS) in the first 5 years of life. In most cases ML-DS is preceded by Transient Abnormal Myelopoiesis (TAM), a fetal/neonatal pre-leukemic disorder unique to DS which regresses after birth. Both TAM and ML-DS harbor acquired N-terminal mutations in the hematopoietic transcription factor gene GATA1. In a prospective study of 200 DS neonates, we recently showed that 29% had acquired GATA1 mutations including 17/200 (8.5%) with clinical or hematologic evidence of TAM; the remaining 20.5% were clinically and hematologically 'silent', with smaller mutant GATA1 clones and lower blast frequency compared to overt TAM. The reasons why some DS neonates develop overt TAM and the factors which determine mutant GATA1 clone size are unknown. To address this, we analysed data from neonates in the prospective Oxford-Imperial DS Cohort Study and investigated the impact of 30 clinical and hematologic factors on clone size using statistical and mathematical modelling. Mutant GATA1 clones were determined in 54 neonates by targeted next generation sequencing of GATA1 exon 2 (mutation detection limit 0.3%). Clone size was determined by analysing original unprocessed reads using less stringent filtering parameters and counting reads containing mutated v total sequence. Correlation analysis identified 4 hematologic variables correlated with mutant GATA1 clone size: circulating nucleated red cells (r=+0.5003; p=0.0001), platelets (r=+0.436; p=0.001), total leukocytes (r=+0.7094; p&lt;0.001) and % blasts (r=+0.7292; p&lt;0.001); clone size was higher in neonates with megakaryocyte fragments (p=0.0024) and platelets &gt;150x109/L (p=0.019). Numbers of neutrophils, monocytes, basophils, eosinophils and lymphocytes did not correlate with GATA1 clone size. Clinical variables significantly correlated with clone size were hepatomegaly (p=0.0016), splenomegaly (p=0.0001) and rash (0.0174). The only pregnancy-related variables affecting mutant GATA1 clone size were intrauterine growth restriction and maternal diabetes (p=0.0156). Linear regression to determine the joint impact of all 30 variables on clone size (r2=0.88) followed by Lasso penalization identified the same 4 hematologic variables (nucleated red cells, platelets, total leukocytes and % blasts); Lasso penalized regression with these 4 variables gave a coefficient of determination of 0.63. Together these data suggest that chronic intrauterine hypoxia may affect expansion/differentiation of mutant GATA1 clones in DS. Consistent with this, nucleated red cells from 3 neonates with TAM all harbored GATA1 mutations identical to those in total circulating nucleated cells. Since neither perinatal infection nor gestational age at birth correlated with mutant GATA1 clone size, infection-related cytokines and the timing of acquisition of a mutant GATA1 clone during fetal development may not play a major role in determining clone size. Finally, a hierarchical model to investigate the impact of GATA1 mutation on hematopoietic stem and progenitor (HSPC) differentiation in DS neonates using a Bayesian approach also predicted increased erythroid cell output from GATA1 mutated HSPC v HSPC without a GATA1 mutation. In conclusion, in neonates with DS the size of the mutant GATA1 clone correlates with the presence of clinical signs of hepatomegaly, splenomegaly and skin rash; mutant GATA1 clone size correlates with the numbers of circulating nucleated red cells, platelets and blast cells suggesting that GATA1 mutant HSPC retain the ability to differentiate down the erythroid and megakaryocyte lineage; intrauterine hypoxia may be one of the factors driving expansion and/or maturation of the GATA1 mutant clone during fetal life in DS.</jats:p> <jats:sec> <jats:title>Disclosures</jats:title> <jats:p>No relevant conflicts of interest to declare.</jats:p> </jats:sec>
container_issue 21
container_start_page 2349
container_title Blood
container_volume 124
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
_version_ 1792322736177020937
geogr_code not assigned
last_indexed 2024-03-01T11:22:32.429Z
geogr_code_person not assigned
openURL url_ver=Z39.88-2004&ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fvufind.svn.sourceforge.net%3Agenerator&rft.title=Clinical+and+Hematologic+Impact+of+Fetal+and+Perinatal+Variables+on+Mutant+GATA1+Clone+Size+in+Neonates+with+Down+Syndrome&rft.date=2014-12-06&genre=article&issn=1528-0020&volume=124&issue=21&spage=2349&epage=2349&pages=2349-2349&jtitle=Blood&atitle=Clinical+and+Hematologic+Impact+of+Fetal+and+Perinatal+Variables+on+Mutant+GATA1+Clone+Size+in+Neonates+with+Down+Syndrome&aulast=Roberts&aufirst=Irene&rft_id=info%3Adoi%2F10.1182%2Fblood.v124.21.2349.2349&rft.language%5B0%5D=eng
SOLR
_version_ 1792322736177020937
author Bhatnagar, Neha, Perkins, Kelly, Filippi, Sarah, Richmond, Helen, Bonnici, Joanna, Alford, Kate, Hall, Georgina, Juban, Gaëtan, McGowan, Simon, Roy, Anindita, Elliott, Natalina, Stumpf, Michael, Norton, Alice, Vyas, Paresh, Roberts, Irene
author_facet Bhatnagar, Neha, Perkins, Kelly, Filippi, Sarah, Richmond, Helen, Bonnici, Joanna, Alford, Kate, Hall, Georgina, Juban, Gaëtan, McGowan, Simon, Roy, Anindita, Elliott, Natalina, Stumpf, Michael, Norton, Alice, Vyas, Paresh, Roberts, Irene, Bhatnagar, Neha, Perkins, Kelly, Filippi, Sarah, Richmond, Helen, Bonnici, Joanna, Alford, Kate, Hall, Georgina, Juban, Gaëtan, McGowan, Simon, Roy, Anindita, Elliott, Natalina, Stumpf, Michael, Norton, Alice, Vyas, Paresh, Roberts, Irene
author_sort bhatnagar, neha
container_issue 21
container_start_page 2349
container_title Blood
container_volume 124
description <jats:title>Abstract</jats:title> <jats:p>Children with Down syndrome (DS; trisomy 21) have an increased risk of acute myeloid leukemia (ML-DS) in the first 5 years of life. In most cases ML-DS is preceded by Transient Abnormal Myelopoiesis (TAM), a fetal/neonatal pre-leukemic disorder unique to DS which regresses after birth. Both TAM and ML-DS harbor acquired N-terminal mutations in the hematopoietic transcription factor gene GATA1. In a prospective study of 200 DS neonates, we recently showed that 29% had acquired GATA1 mutations including 17/200 (8.5%) with clinical or hematologic evidence of TAM; the remaining 20.5% were clinically and hematologically 'silent', with smaller mutant GATA1 clones and lower blast frequency compared to overt TAM. The reasons why some DS neonates develop overt TAM and the factors which determine mutant GATA1 clone size are unknown. To address this, we analysed data from neonates in the prospective Oxford-Imperial DS Cohort Study and investigated the impact of 30 clinical and hematologic factors on clone size using statistical and mathematical modelling. Mutant GATA1 clones were determined in 54 neonates by targeted next generation sequencing of GATA1 exon 2 (mutation detection limit 0.3%). Clone size was determined by analysing original unprocessed reads using less stringent filtering parameters and counting reads containing mutated v total sequence. Correlation analysis identified 4 hematologic variables correlated with mutant GATA1 clone size: circulating nucleated red cells (r=+0.5003; p=0.0001), platelets (r=+0.436; p=0.001), total leukocytes (r=+0.7094; p&lt;0.001) and % blasts (r=+0.7292; p&lt;0.001); clone size was higher in neonates with megakaryocyte fragments (p=0.0024) and platelets &gt;150x109/L (p=0.019). Numbers of neutrophils, monocytes, basophils, eosinophils and lymphocytes did not correlate with GATA1 clone size. Clinical variables significantly correlated with clone size were hepatomegaly (p=0.0016), splenomegaly (p=0.0001) and rash (0.0174). The only pregnancy-related variables affecting mutant GATA1 clone size were intrauterine growth restriction and maternal diabetes (p=0.0156). Linear regression to determine the joint impact of all 30 variables on clone size (r2=0.88) followed by Lasso penalization identified the same 4 hematologic variables (nucleated red cells, platelets, total leukocytes and % blasts); Lasso penalized regression with these 4 variables gave a coefficient of determination of 0.63. Together these data suggest that chronic intrauterine hypoxia may affect expansion/differentiation of mutant GATA1 clones in DS. Consistent with this, nucleated red cells from 3 neonates with TAM all harbored GATA1 mutations identical to those in total circulating nucleated cells. Since neither perinatal infection nor gestational age at birth correlated with mutant GATA1 clone size, infection-related cytokines and the timing of acquisition of a mutant GATA1 clone during fetal development may not play a major role in determining clone size. Finally, a hierarchical model to investigate the impact of GATA1 mutation on hematopoietic stem and progenitor (HSPC) differentiation in DS neonates using a Bayesian approach also predicted increased erythroid cell output from GATA1 mutated HSPC v HSPC without a GATA1 mutation. In conclusion, in neonates with DS the size of the mutant GATA1 clone correlates with the presence of clinical signs of hepatomegaly, splenomegaly and skin rash; mutant GATA1 clone size correlates with the numbers of circulating nucleated red cells, platelets and blast cells suggesting that GATA1 mutant HSPC retain the ability to differentiate down the erythroid and megakaryocyte lineage; intrauterine hypoxia may be one of the factors driving expansion and/or maturation of the GATA1 mutant clone during fetal life in DS.</jats:p> <jats:sec> <jats:title>Disclosures</jats:title> <jats:p>No relevant conflicts of interest to declare.</jats:p> </jats:sec>
doi_str_mv 10.1182/blood.v124.21.2349.2349
facet_avail Online, Free
finc_class_facet Biologie, Medizin, Chemie und Pharmazie
format ElectronicArticle
format_de105 Article, E-Article
format_de14 Article, E-Article
format_de15 Article, E-Article
format_de520 Article, E-Article
format_de540 Article, E-Article
format_dech1 Article, E-Article
format_ded117 Article, E-Article
format_degla1 E-Article
format_del152 Buch
format_del189 Article, E-Article
format_dezi4 Article
format_dezwi2 Article, E-Article
format_finc Article, E-Article
format_nrw Article, E-Article
geogr_code not assigned
geogr_code_person not assigned
id ai-49-aHR0cDovL2R4LmRvaS5vcmcvMTAuMTE4Mi9ibG9vZC52MTI0LjIxLjIzNDkuMjM0OQ
imprint American Society of Hematology, 2014
imprint_str_mv American Society of Hematology, 2014
institution DE-Gla1, DE-Zi4, DE-15, DE-Pl11, DE-Rs1, DE-105, DE-14, DE-Ch1, DE-L229, DE-D275, DE-Bn3, DE-Brt1, DE-Zwi2, DE-D161
issn 0006-4971, 1528-0020
issn_str_mv 0006-4971, 1528-0020
language English
last_indexed 2024-03-01T11:22:32.429Z
match_str bhatnagar2014clinicalandhematologicimpactoffetalandperinatalvariablesonmutantgata1clonesizeinneonateswithdownsyndrome
mega_collection American Society of Hematology (CrossRef)
physical 2349-2349
publishDate 2014
publishDateSort 2014
publisher American Society of Hematology
record_format ai
recordtype ai
series Blood
source_id 49
spelling Bhatnagar, Neha Perkins, Kelly Filippi, Sarah Richmond, Helen Bonnici, Joanna Alford, Kate Hall, Georgina Juban, Gaëtan McGowan, Simon Roy, Anindita Elliott, Natalina Stumpf, Michael Norton, Alice Vyas, Paresh Roberts, Irene 0006-4971 1528-0020 American Society of Hematology Cell Biology Hematology Immunology Biochemistry http://dx.doi.org/10.1182/blood.v124.21.2349.2349 <jats:title>Abstract</jats:title> <jats:p>Children with Down syndrome (DS; trisomy 21) have an increased risk of acute myeloid leukemia (ML-DS) in the first 5 years of life. In most cases ML-DS is preceded by Transient Abnormal Myelopoiesis (TAM), a fetal/neonatal pre-leukemic disorder unique to DS which regresses after birth. Both TAM and ML-DS harbor acquired N-terminal mutations in the hematopoietic transcription factor gene GATA1. In a prospective study of 200 DS neonates, we recently showed that 29% had acquired GATA1 mutations including 17/200 (8.5%) with clinical or hematologic evidence of TAM; the remaining 20.5% were clinically and hematologically 'silent', with smaller mutant GATA1 clones and lower blast frequency compared to overt TAM. The reasons why some DS neonates develop overt TAM and the factors which determine mutant GATA1 clone size are unknown. To address this, we analysed data from neonates in the prospective Oxford-Imperial DS Cohort Study and investigated the impact of 30 clinical and hematologic factors on clone size using statistical and mathematical modelling. Mutant GATA1 clones were determined in 54 neonates by targeted next generation sequencing of GATA1 exon 2 (mutation detection limit 0.3%). Clone size was determined by analysing original unprocessed reads using less stringent filtering parameters and counting reads containing mutated v total sequence. Correlation analysis identified 4 hematologic variables correlated with mutant GATA1 clone size: circulating nucleated red cells (r=+0.5003; p=0.0001), platelets (r=+0.436; p=0.001), total leukocytes (r=+0.7094; p&lt;0.001) and % blasts (r=+0.7292; p&lt;0.001); clone size was higher in neonates with megakaryocyte fragments (p=0.0024) and platelets &gt;150x109/L (p=0.019). Numbers of neutrophils, monocytes, basophils, eosinophils and lymphocytes did not correlate with GATA1 clone size. Clinical variables significantly correlated with clone size were hepatomegaly (p=0.0016), splenomegaly (p=0.0001) and rash (0.0174). The only pregnancy-related variables affecting mutant GATA1 clone size were intrauterine growth restriction and maternal diabetes (p=0.0156). Linear regression to determine the joint impact of all 30 variables on clone size (r2=0.88) followed by Lasso penalization identified the same 4 hematologic variables (nucleated red cells, platelets, total leukocytes and % blasts); Lasso penalized regression with these 4 variables gave a coefficient of determination of 0.63. Together these data suggest that chronic intrauterine hypoxia may affect expansion/differentiation of mutant GATA1 clones in DS. Consistent with this, nucleated red cells from 3 neonates with TAM all harbored GATA1 mutations identical to those in total circulating nucleated cells. Since neither perinatal infection nor gestational age at birth correlated with mutant GATA1 clone size, infection-related cytokines and the timing of acquisition of a mutant GATA1 clone during fetal development may not play a major role in determining clone size. Finally, a hierarchical model to investigate the impact of GATA1 mutation on hematopoietic stem and progenitor (HSPC) differentiation in DS neonates using a Bayesian approach also predicted increased erythroid cell output from GATA1 mutated HSPC v HSPC without a GATA1 mutation. In conclusion, in neonates with DS the size of the mutant GATA1 clone correlates with the presence of clinical signs of hepatomegaly, splenomegaly and skin rash; mutant GATA1 clone size correlates with the numbers of circulating nucleated red cells, platelets and blast cells suggesting that GATA1 mutant HSPC retain the ability to differentiate down the erythroid and megakaryocyte lineage; intrauterine hypoxia may be one of the factors driving expansion and/or maturation of the GATA1 mutant clone during fetal life in DS.</jats:p> <jats:sec> <jats:title>Disclosures</jats:title> <jats:p>No relevant conflicts of interest to declare.</jats:p> </jats:sec> Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome Blood
spellingShingle Bhatnagar, Neha, Perkins, Kelly, Filippi, Sarah, Richmond, Helen, Bonnici, Joanna, Alford, Kate, Hall, Georgina, Juban, Gaëtan, McGowan, Simon, Roy, Anindita, Elliott, Natalina, Stumpf, Michael, Norton, Alice, Vyas, Paresh, Roberts, Irene, Blood, Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome, Cell Biology, Hematology, Immunology, Biochemistry
title Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
title_full Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
title_fullStr Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
title_full_unstemmed Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
title_short Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
title_sort clinical and hematologic impact of fetal and perinatal variables on mutant gata1 clone size in neonates with down syndrome
title_unstemmed Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
topic Cell Biology, Hematology, Immunology, Biochemistry
url http://dx.doi.org/10.1182/blood.v124.21.2349.2349