6 results on '"Natalina Elliott"'
Search Results
2. Trisomy 21 driven pro-inflammatory signalling in fetal bone marrow may play a role in perturbed B-lymphopoiesis and acute lymphoblastic leukemia of Down syndrome
- Author
-
Georgina Buck, J Oswald, Irene Roberts, Anindita Roy, David O'Connor, Natalina Elliott, H Fuchs, Nicholas Fordham, Siobhan Rice, Sorcha O’Byrne, and Labbett E-M.
- Subjects
0301 basic medicine ,Stromal cell ,Immunology ,Mesenchymal stem cell ,Cell Biology ,Hematology ,Biology ,Biochemistry ,CD19 ,Cell biology ,03 medical and health sciences ,Haematopoiesis ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,biology.protein ,Lymphopoiesis ,Stem cell ,Progenitor cell ,B cell ,030215 immunology - Abstract
Introduction: Children with Down syndrome (DS) have a markedly increased risk of acute lymphoblastic leukemia (ALL), suggesting that trisomy 21 (T21) has specific effects on hematopoietic stem and progenitor cell (HSPC) biology in early life. Data from human fetal liver (FL) indicates that T21 alters fetal hematopoiesis, causing multiple defects in lympho-myelopoiesis. The impact of T21 on fetal B lymphopoiesis and how this may underpin the increase in ALL is not well known. We have recently found that fetal bone marrow (FBM) rather than FL is the main site of B lymphopoiesis; with a marked enrichment of fetal-specific progenitors (early lymphoid progenitors, ELP and PreProB progenitors) that lie upstream of adult type ProB progenitors (O'Byrne et al, Blood, in press). Previous preliminary data suggested that B progenitors were also reduced in T21 FBM (Roy et al, Blood. 124, 4331). Aim: To dissect putative molecular mechanisms responsible for the defects in T21 FBM B-lymphopoiesis and its association with childhood DS ALL. Methods: Second trimester human FBM and paediatric ALL samples were obtained from the Human Developmental Biology Resource and UK Childhood Leukaemia Cell Bank respectively. Multiparameter flow cytometry/sorting, transcriptome analysis by RNA-sequencing and microarray, and stromal co-culture assays were used to characterize HSPC and mesenchymal stromal cells (MSC) from normal (NM) disomic (n=21-35) and T21 (n=7-12) human FBM; RNASeq was performed on cytogenetically matched non-DS (n=13) and DS ALL (n=7). Results: In contrast to NM FBM, fetal specific progenitors were virtually absent (CD34+CD10-CD19-CD127+ ELP 2.8±0.4% vs. 0.8±0.4% of CD34+ cells) or very severely reduced (CD34+CD10-CD19+ PreProB 12.8±1 vs 2.6±0.7%) in T21 FBM. This was despite a >4-fold increase in the frequency of immunophenotypic HSC (4.2±1.2% vs 0.9±0.2% of CD34+ cells) and similar frequencies of MPP and LMPP in T21 FBM. As in adult BM, the vast majority of B progenitors in T21 FBM were CD34+CD10+CD19+ ProB progenitors with a frequency (28.8±8.3%) similar to NM FBM (30.3±2.3% of CD34+ cells). Thus, T21 causes a severe block in B-progenitor commitment at the LMPP stage, in tandem with a compensatory expansion of ProB progenitors. Consistent with this, T21 FBM HSC, MPP and LMPP had reduced B cell potential in vitro compared to NM FBM in MS5 co-cultures. RNAseq of NM (n=3) and T21 (n=3) FBM HSPC demonstrated global transcriptomic disruption by T21, with increased gene expression in HSC, MPP, LMPP and ProB progenitors. Cell cycle genes were enriched in T21 ProB progenitors. Despite these functional and global gene expression differences, expression of key B-lineage commitment genes was maintained suggesting the defect in B-lymphopoiesis may be secondary to lineage skewing of multipotent progenitors towards a non-B lymphoid fate and/or mediated by extrinsic factors. GSEA pointed to a role for multiple inflammatory pathways in T21 hematopoiesis with dysregulation of IFNα, IL6 and TGFβ signalling pathways in T21 HSC/LMPP. To investigate the role of the T21 microenvironment, we co-cultured NM HSC, MPP and LMPP with T21 or NM primary FBM MSC. T21 FBM MSC (n=3) had reduced capacity to support B cell differentiation in vitro consistent with perturbation of MSC function by T21. Similar to T21 FBM HSPC, transcriptomic analysis of T21 FBM MSC by microarray showed enrichment for IFNα signalling compared to NM; and T21 HSPC and MSC both showed increased gene expression for IFNα receptors IFNAR1 and IFNAR2, which are encoded on chromosome 21. Since IFNα was undetectable by ELISA of conditioned media from NM and T21 MSC, differences in secreted IFNα from MSC are unlikely to fully explain the increased IFN signalling in T21 HSPC and MSC. This suggests that T21 may drive autocrine rather than paracrine IFN signalling in FBM cells. Finally, RNASeq showed perturbed inflammatory signalling in DS ALL compared to non-DS ALL, suggesting a role for T21-driven inflammatory pathways in the biology of DS ALL. Conclusions: These data show that T21 severely impairs B lymphopoiesis in FBM and is associated with expression of proinflammatory gene expression programs in T21 FBM HSPC and MSC and DS ALL. The compensatory expansion of T21 FBM ProB progenitors, through self-renewal or via an alternative differentiation pathway; with concomitant T21-driven proinflammatory signalling may underpin the increased risk of B progenitor ALL in childhood. Disclosures No relevant conflicts of interest to declare.
- Published
- 2020
3. Epigenome-Wide Association Study of Acute Lymphoblastic Leukemia in Children with Down Syndrome
- Author
-
Beth A. Mueller, Austin L. Brown, Priyatama Pandey, Karen R. Rabin, Libby M. Morimoto, Irene Roberts, Shaobo Li, Adam J. de Smith, Helen M. Hansen, Keren Xu, Alice Y. Kang, Ivo S. Muskens, Xiaomei Ma, Pagna Sok, Joseph L. Wiemels, Philip J. Lupo, Swe Swe Myint, Anindita Roy, Natalina Elliott, and Catherine Metayer
- Subjects
Oncology ,medicine.medical_specialty ,Down syndrome ,business.industry ,Lymphoblastic Leukemia ,Immunology ,Cell Biology ,Hematology ,Epigenome ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,medicine.disease ,Biochemistry ,Internal medicine ,Acute Disease ,medicine ,Humans ,Down Syndrome ,Child ,business - Abstract
Background: Down syndrome (DS) is associated with an up to 30-fold increased risk of B-cell acute lymphoblastic leukemia (ALL), and DS-ALL patients have worse overall survival and increased long-term treatment-related health conditions compared with non-DS ALL patients. In a recent genome-wide association study of DS-ALL, established ALL genetic risk loci were associated with DS-ALL, with several single nucleotide polymorphisms (SNPs) conferring a larger effect on ALL risk in the context of DS than in euploidy. We performed an epigenome-wide association study (EWAS) to elucidate whether epigenetic differences at birth are associated with risk of subsequent DS-ALL. Methods: The DS-ALL Discovery Study included 147 DS-ALL cases and 198 DS controls from the International Study of Down Syndrome Acute Leukemia, with newborn dried bloodspots (DBS) obtained from California (n=326) and Washington state (n=19) biobanks. The DS-ALL Replication Study included 24 DS-ALL cases and 24 DS controls with newborn DBS from the Michigan Neonatal Biobank. DNA was isolated from DBS, bisulfite converted, and assayed using Illumina Infinium MethylationEPIC Beadchip genome-wide DNA methylation arrays. Raw data were processed using "minfi" and "noob" packages in R. Reference-based deconvolution of blood cell proportions was performed using the Identifying Optimal DNA methylation Libraries (IDOL) algorithm, using DNA methylation data from cord blood reference samples, to estimate proportions of B cells, T cells (CD4+ and CD8+), monocytes, granulocytes, natural killer cells, and nucleated red blood cells. We compared each cell type proportion between DS-ALL cases and DS controls using linear regression adjusting for sex, plate, and principal components (PCs) to account for genetic ancestry. To identify single CpG probes associated with DS-ALL risk, we performed a multiethnic EWAS of DS-ALL in each study using linear regression adjusting for sex, plate, and PCs related to: 1) cell-type proportions and 2) genetic ancestry. Differentially methylated regions (DMRs) were identified using DMRcate and comb-p methods. In the Discovery Study, genome-wide SNP array data were available for 131 cases and 130 controls, and data from targeted sequencing of somatic mutations in exons 2/3 of GATA1 were available for 184/198 DS controls. Results: Deconvolution of blood cell proportions in the DS-ALL Discovery Study showed significantly higher B cell proportions in newborns with DS who later developed ALL (mean=0.0128, sd=0.0151) compared with DS controls (mean=0.00826, sd=0.0115) (P=6.4x10 -4, coefficient=0.0052). A significantly higher B cell proportion at birth was also found in DS-ALL cases in the independent Replication Study (cases mean=0.048, sd=0.024; controls mean=0.039, sd=0.028; P=0.03, coefficient=0.015). In the Discovery Study, the B cell difference remained significant (P=5.8x10 -3) with a similar effect size (coefficient=0.0045) after removal of GATA1 mutation-positive DS controls (n=30). We also investigated whether DS-ALL risk SNPs at ARID5B, IKZF1, GATA3, and CDKN2A may confound the association, but the increased B cell proportions in DS-ALL remained significant and effect estimates slightly increased in SNP genotype-adjusted models (coefficient range:0.0055-0.0059). In the EWAS of DS-ALL, 9 CpGs reached epigenome-wide significance (P Conclusions: Increased B cell proportions in newborns with DS may be a risk factor for development of DS-ALL in childhood. This finding, based on DNA methylation data, requires confirmation using conventional cell count measures, and should be explored as a novel biomarker for ALL risk in the non-DS population. Single CpGs and DMRs associated with DS-ALL risk in our Discovery Study require further investigation, including in additional ALL case-control studies in DS and non-DS populations. Disclosures Ma: Celgene/Bristol Myers Squibb: Consultancy, Research Funding.
- Published
- 2021
- Full Text
- View/download PDF
4. Discovery of a CD10-negative B-progenitor in human fetal life identifies unique ontogeny-related developmental programs
- Author
-
Natalina Elliott, Sarah Inglott, Phillip R. Bennett, Hashem Koohy, Clara Bueno, Antonio Agraz-Doblas, Thomas A. Milne, P Ancliff, Irene Roberts, Catherine Garnett, Bethan Psaila, Anastasios Karadimitris, David J. H. F. Knapp, Siobhan Rice, Pablo Menendez, Suzanne M. Watt, Anindita Roy, Adam J. Mead, Laura Godfrey, Nicholas T. Crump, Gemma Buck, Gary Wright, Ignacio Varela, Paresh Vyas, Peng Hua, Nicholas Fordham, Benjamin J. Povinelli, and Sorcha O’Byrne
- Subjects
EXPRESSION ,Immunology ,HUMAN HEMATOPOIESIS ,DISTINCT ,Context (language use) ,Growth ,Biology ,Biochemistry ,Andrology ,03 medical and health sciences ,0302 clinical medicine ,Fetus ,b-lymphocytes ,medicine ,TRANSLOCATIONS ,Lymphopoiesis ,1102 Cardiorespiratory Medicine and Haematology ,ACUTE LYMPHOBLASTIC-LEUKEMIA ,030304 developmental biology ,Progenitor ,CELL DEVELOPMENT ,ARCHITECTURE ,0303 health sciences ,Science & Technology ,REARRANGEMENT ,Hematopoietic stem cell ,1103 Clinical Sciences ,LINEAGE ,Cell Biology ,Hematology ,STEM ,medicine.disease ,Embryonic stem cell ,Leukemia ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,cd19 antigens ,1114 Paediatrics and Reproductive Medicine ,Neprilysin ,Bone marrow ,Life Sciences & Biomedicine - Abstract
Human lymphopoiesis is a dynamic lifelong process that starts in utero 6 weeks post-conception. Although fetal B-lymphopoiesis remains poorly defined, it is key to understanding leukemia initiation in early life. Here, we provide a comprehensive analysis of the human fetal B-cell developmental hierarchy. We report the presence in fetal tissues of 2 distinct CD191 B-progenitors, an adult-type CD10+ve ProB-progenitor and a new CD10-ve PreProB-progenitor, and describe their molecular and functional characteristics. PreProB-progenitors and ProB-progenitors appear early in the first trimester in embryonic liver, followed by a sustained second wave of B-progenitor development in fetal bone marrow (BM), where together they form > 40% of the total hematopoietic stem cell/progenitor pool. Almost one-third of fetal B-progenitors are CD10-ve PreProB-progenitors, whereas, by contrast, PreProB-progenitors are almost undetectable (0.53% +/- 0.24%) in adult BM. Single-cell transcriptomics and functional assays place fetal PreProB-progenitors upstream of ProB-progenitors, identifying them as the first B-lymphoid-restricted progenitor in human fetal life. Although fetalBMPreProB-progenitors and ProB-progenitors both give rise solely to B-lineage cells, they are transcriptionally distinct. As with their fetal counterparts, adult BM PreProB-progenitors give rise only to B-lineage cells in vitro and express the expected B-lineage gene expression program. However, fetal PreProB-progenitors display a distinct, ontogeny-related gene expression pattern that is not seen in adult PreProB-progenitors, and they share transcriptomic signatures with CD10-ve B-progenitor infant acute lymphoblastic leukemia blast cells. These data identify PreProB-progenitors as the earliest B-lymphoid-restricted progenitor in human fetal life and suggest that this fetal-restricted committed B-progenitor might provide a permissive cellular context for prenatal B-progenitor leukemia initiation.
- Published
- 2019
5. FERM domain mutations induce gain of function in JAK3 in adult T-cell leukemia/lymphoma
- Author
-
Susan M. Cleveland, Natalina Elliott, John Janik, Victor R. Grann, Thomas A. Waldmann, and Utpal P. Davé
- Subjects
Models, Molecular ,Moesin ,DNA Mutational Analysis ,Molecular Sequence Data ,Immunology ,Mutation, Missense ,Biology ,medicine.disease_cause ,Biochemistry ,Adult T-cell leukemia/lymphoma ,Cell Line ,Ezrin ,immune system diseases ,Radixin ,hemic and lymphatic diseases ,STAT5 Transcription Factor ,medicine ,Animals ,Humans ,Leukemia-Lymphoma, Adult T-Cell ,Amino Acid Sequence ,Protein Kinase Inhibitors ,Mutation ,Lymphoid Neoplasia ,FERM domain ,Janus kinase 3 ,Janus Kinase 3 ,Cell Biology ,Hematology ,medicine.disease ,Protein Structure, Tertiary ,Leukemia ,Cancer research ,Sequence Alignment - Abstract
Adult T-cell leukemia/lymphoma (ATLL) is an incurable disease where most patients succumb within the first year of diagnosis. Both standard chemotherapy regimens and mAbs directed against ATLL tumor markers do not alter this aggressive clinical course. Therapeutic development would be facilitated by the discovery of genes and pathways that drive or initiate ATLL, but so far amenable drug targets have not been forthcoming. Because the IL-2 signaling pathway plays a prominent role in ATLL pathogenesis, mutational analysis of pathway components should yield interesting results. In this study, we focused on JAK3, the nonreceptor tyrosine kinase that signals from the IL-2R, where activating mutations have been found in diverse neoplasms. We screened 36 ATLL patients and 24 ethnically matched controls and found 4 patients with mutations in JAK3. These somatic, missense mutations occurred in the N-terminal FERM (founding members: band 4.1, ezrin, radixin, and moesin) domain and induced gain of function in JAK3. Importantly, we show that these mutant JAK3s are inhibited with a specific kinase inhibitor already in human clinical testing. Our findings underscore the importance of this pathway in ATLL development and offer a therapeutic handle for this incurable cancer.
- Published
- 2011
- Full Text
- View/download PDF
6. Clinical and Hematologic Impact of Fetal and Perinatal Variables on Mutant GATA1 Clone Size in Neonates with Down Syndrome
- Author
-
Natalina Elliott, Gaëtan Juban, Joanna Bonnici, Georgina W. Hall, Irene Roberts, Alice Norton, Paresh Vyas, Sarah Filippi, Helen Richmond, Anindita Roy, Michael P. H. Stumpf, Kate A. Alford, Neha Bhatnagar, Kelly J. Perkins, and Simon J. McGowan
- Subjects
Fetus ,Immunology ,Mutant ,Myeloid leukemia ,GATA1 ,Cell Biology ,Hematology ,Intrauterine hypoxia ,Biology ,medicine.disease ,Biochemistry ,Andrology ,Haematopoiesis ,medicine.anatomical_structure ,Megakaryocyte ,medicine ,Trisomy - Abstract
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; p150x109/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. Disclosures No relevant conflicts of interest to declare.
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.