Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 2024 06 21.
Article in English | MEDLINE | ID: mdl-38905593

ABSTRACT

Infant acute lymphoblastic leukemia (ALL) is an aggressive malignancy that has historically been associated with a very poor prognosis. Despite large co-operative international trials and incremental increases in intensity of therapy, there has been no significant improvement in outcome over the last 3 decades. Using representative cases, we highlight the key differences between KMT2A-rearranged and KMT2A-germline infant ALL, and how advances in molecular diagnostics are unpicking KMT2A-germline genetics and guiding treatment reduction. We focus on KM2TA-rearranged infant B-cell ALL where the last few years have seen the emergence of novel therapies which both are more effective and less toxic than conventional chemotherapy. Of these, there is promising early data on the efficacy and tolerability of the bi-specific T-cell engager monoclonal antibody, blinatumomab, as well as the use of autologous and allogeneic chimeric antigen receptor T-cell therapy. We discuss how we can improve risk stratification and incorporate these new agents to replace the most toxic elements of currently deployed intensive chemotherapy schedules with their associated unacceptable toxicity.

2.
J Periodontal Res ; 56(4): 819-827, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33811759

ABSTRACT

OBJECTIVE: This study aimed to assess associations between neutrophil-related primary immunodeficiencies (PIDs) and the presence of periodontal disease and other oral diseases and response to periodontal treatment. BACKGROUND: Presence of neutrophil-related PIDs is thought to be a major risk factor for development of periodontitis. METHODS: This study had both a cross-sectional and cohort design. Twenty-four children (age 4-16) with PIDs and 24 age-matched systemically healthy subjects received a dental clinical examination, including measures of probing pocket depths (PPD), clinical attachment loss (CAL) and bleeding on probing (BOP). Those found to be affected by periodontal disease were offered periodontal treatment and reassessed 6 months later. RESULTS: Diagnosis of PIDs was associated with increased odds of presence of periodontal disease (p = .008 adjusted for age, gender, plaque, OR = 10.0, 95% CI = 1.83-54.38) and with continuous measures of periodontal disease such as number of PPDs >4 mm, mean PPD and mean CAL (all p < .001) and BOP (p = .001). However, only 7 out of 24 children were diagnosed with periodontitis. PIDs were also associated with a history of oral ulcers (p = .001, OR 12.47, 95% CI 2.71-57.29). An improvement in periodontal parameters (PPD and CAL) was detected following oral hygiene instructions and non-surgical periodontal therapy. CONCLUSION: Although children affected by neutrophil-associated PIDs exhibited a higher prevalence of periodontal disease compared with systemically healthy children, severe periodontitis was rarely seen. This suggests that good systemic control of the PIDs may reduce their impact on the periodontium.


Subject(s)
Dental Plaque , Periodontal Diseases , Periodontitis , Adolescent , Child , Child, Preschool , Cross-Sectional Studies , Humans , Periodontal Attachment Loss , Periodontal Diseases/complications , Periodontal Diseases/epidemiology , Periodontal Diseases/therapy , Periodontium
3.
J Pediatr Hematol Oncol ; 43(3): e380-e384, 2021 04 01.
Article in English | MEDLINE | ID: mdl-32218099

ABSTRACT

Although outcomes for children with B-cell non-Hodgkin lymphoma are excellent, between 20% and 40% demonstrate residual radiologic abnormalities at disease assessment during consolidation therapy, the significance of which remains uncertain. The authors report the outcomes for all children treated for B-cell non-Hodgkin lymphoma at our center over an 11-year period. Twenty-four of 64 (38%) children had residual radiologic abnormalities at disease remission assessment. Seven (29%) underwent histologic biopsies that were normal. No children with residual radiologic abnormalities experienced disease relapse or death, suggesting that imaging at this time point creates clinical uncertainty without indicating residual disease or predicting relapse.


Subject(s)
Lymphoma, B-Cell/diagnostic imaging , Adolescent , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Disease Management , Female , Humans , Infant , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/pathology , Magnetic Resonance Imaging , Male , Neoplasm, Residual/diagnostic imaging , Neoplasm, Residual/drug therapy , Neoplasm, Residual/pathology , Prognosis , Retrospective Studies , Tomography, X-Ray Computed , Ultrasonography
5.
Blood ; 126(10): 1214-23, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26162409

ABSTRACT

In-depth molecular investigation of familial leukemia has been limited by the rarity of recognized cases. This study examines the genetic events initiating leukemia and details the clinical progression of disease across multiple families harboring germ-line CEBPA mutations. Clinical data were collected from 10 CEBPA-mutated families, representing 24 members with acute myeloid leukemia (AML). Whole-exome (WES) and deep sequencing were performed to genetically profile tumors and define patterns of clonal evolution. Germline CEBPA mutations clustered within the N-terminal and were highly penetrant, with AML presenting at a median age of 24.5 years (range, 1.75-46 years). In all diagnostic tumors tested (n = 18), double CEBPA mutations (CEBPAdm) were detected, with acquired (somatic) mutations preferentially targeting the C-terminal. Somatic CEBPA mutations were unstable throughout the disease course, with different mutations identified at recurrence. Deep sequencing of diagnostic and relapse paired samples confirmed that relapse-associated CEBPA mutations were absent at diagnosis, suggesting recurrence was triggered by novel, independent clones. Integrated WES and deep sequencing subsequently revealed an entirely new complement of mutations at relapse, verifying the presentation of a de novo leukemic episode. The cumulative incidence of relapse in familial AML was 56% at 10 years (n = 11), and 3 patients experienced ≥3 disease episodes over a period of 17 to 20 years. Durable responses to secondary therapies were observed, with prolonged median survival after relapse (8 years) and long-term overall survival (10-year overall survival, 67%). Our data reveal that familial CEBPA-mutated AML exhibits a unique model of disease progression, associated with favorable long-term outcomes.


Subject(s)
CCAAT-Enhancer-Binding Proteins/genetics , Germ-Line Mutation , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Adolescent , Adult , Child , Child, Preschool , Disease Progression , Female , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Infant , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Pedigree , Young Adult
6.
Nat Genet ; 39(4): 486-95, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17353896

ABSTRACT

The autosomal recessive disorder Shwachman-Diamond syndrome, characterized by bone marrow failure and leukemia predisposition, is caused by deficiency of the highly conserved Shwachman-Bodian-Diamond syndrome (SBDS) protein. Here, we identify the function of the yeast SBDS ortholog Sdo1, showing that it is critical for the release and recycling of the nucleolar shuttling factor Tif6 from pre-60S ribosomes, a key step in 60S maturation and translational activation of ribosomes. Using genome-wide synthetic genetic array mapping, we identified multiple TIF6 gain-of-function alleles that suppressed the pre-60S nuclear export defects and cytoplasmic mislocalization of Tif6 observed in sdo1Delta cells. Sdo1 appears to function within a pathway containing elongation factor-like 1, and together they control translational activation of ribosomes. Thus, our data link defective late 60S ribosomal subunit maturation to an inherited bone marrow failure syndrome associated with leukemia predisposition.


Subject(s)
Protein Biosynthesis/genetics , Ribosomes/metabolism , Saccharomyces cerevisiae Proteins/physiology , Saccharomyces cerevisiae/genetics , Carrier Proteins/genetics , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/physiology , Gene Deletion , Intermediate Filament Proteins/genetics , Models, Biological , Models, Molecular , Mutation , Organisms, Genetically Modified , Peptide Elongation Factors/genetics , Peptide Elongation Factors/physiology , Phosphoproteins/genetics , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Ribosomal Proteins , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics
7.
Br J Haematol ; 171(2): 263-272, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26115422

ABSTRACT

Here, we report a high incidence of PAX5 abnormalities observed in 32/68 (47%) of patients with genetically unclassified childhood precursor B-cell acute lymphoblastic leukaemia (pre-B ALL). Various deletions, gains, mutations and rearrangements of PAX5 comprised 45%, 12%, 29% and 14%, respectively, of the abnormalities found. 28% of patients showed more than one abnormality of the gene, implying bi-allelic impairment of PAX5. Novel PAX5-RHOXF2, PAX5-ELK3 and PAX5-CBFA2T2 rearrangements, which lead to aberrant expression of PAX5, were also identified. PAX5 rearrangements demonstrated a complex mechanism of formation including concurrent duplications/deletions of PAX5 and its partner genes. Finally, the splice variant c.1013-2A>G, seen in two patients with loss of one PAX5 allele, was confirmed to be germ-line in one patient and somatic in the other. PAX5 alterations were also found to be clinically associated with a higher white blood cell count (P = 0·015). These findings contribute to the knowledge of PAX5 alterations and their role in the pathogenesis of pre-B ALL.

10.
Nat Commun ; 14(1): 5092, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37608017

ABSTRACT

Clonal tracking of cells using somatic mutations permits exploration of clonal dynamics in human disease. Here, we perform whole genome sequencing of 323 haematopoietic colonies from 10 individuals with the inherited ribosomopathy Shwachman-Diamond syndrome to reconstruct haematopoietic phylogenies. In ~30% of colonies, we identify mutually exclusive mutations in TP53, EIF6, RPL5, RPL22, PRPF8, plus chromosome 7 and 15 aberrations that increase SBDS and EFL1 gene dosage, respectively. Target gene mutations commence in utero, resulting in a profusion of clonal expansions, with only a few haematopoietic stem cell lineages (mean 8, range 1-24) contributing ~50% of haematopoietic colonies across 8 individuals (range 4-100% clonality) by young adulthood. Rapid clonal expansion during disease transformation is associated with biallelic TP53 mutations and increased mutation burden. Our study highlights how convergent somatic mutation of the p53-dependent nucleolar surveillance pathway offsets the deleterious effects of germline ribosomopathy but increases opportunity for TP53-mutated cancer evolution.


Subject(s)
Chromosomes, Human, Pair 7 , Germ Cells , Humans , Young Adult , Adult , Gene Dosage , Hematopoietic Stem Cells , Mutation
11.
Blood ; 115(17): 3553-8, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20061556

ABSTRACT

Chimeric fusion genes are highly prevalent in childhood acute lymphoblastic leukemia (ALL) and are mostly prenatal, early genetic events in the evolutionary trajectory of this cancer. ETV6-RUNX1-positive ALL also has multiple ( approximately 6 per case) copy number alterations (CNAs) as revealed by genome-wide single-nucleotide polymorphism arrays. Recurrent CNAs are probably "driver" events contributing critically to clonal diversification and selection, but at diagnosis, their developmental timing is "buried" in the leukemia's covert natural history. This conundrum can be resolved with twin pairs. We identified and compared CNAs in 5 pairs of monozygotic twins with concordant ETV6-RUNX1-positive ALL and 1 pair discordant for ETV6-RUNX1 positive ALL. We compared, within each pair, CNAs classified as potential "driver" or "passenger" mutations based upon recurrency and, where known, gene function. An average of 5.1 (range 3-11) CNAs (excluding immunoglobulin/T-cell receptor alterations) were identified per case. All "driver" CNAs (total of 32) were distinct within each of the 5 twin pairs with concordant ALL. "Driver" CNAs in another twin with ALL were all absent in the shared ETV6-RUNX1-positive preleukemic clone of her healthy co-twin. These data place all "driver" CNAs secondary to the prenatal gene fusion event and most probably postnatal in the sequential, molecular pathogenesis of ALL.


Subject(s)
Gene Dosage , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Twins, Monozygotic , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Female , Humans , Male , Mutation , Oligonucleotide Array Sequence Analysis , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
13.
J Clin Invest ; 118(9): 3143-50, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18688286

ABSTRACT

X-linked SCID (SCID-X1) is amenable to correction by gene therapy using conventional gammaretroviral vectors. Here, we describe the occurrence of clonal T cell acute lymphoblastic leukemia (T-ALL) promoted by insertional mutagenesis in a completed gene therapy trial of 10 SCID-X1 patients. Integration of the vector in an antisense orientation 35 kb upstream of the protooncogene LIM domain only 2 (LMO2) caused overexpression of LMO2 in the leukemic clone. However, leukemogenesis was likely precipitated by the acquisition of other genetic abnormalities unrelated to vector insertion, including a gain-of-function mutation in NOTCH1, deletion of the tumor suppressor gene locus cyclin-dependent kinase 2A (CDKN2A), and translocation of the TCR-beta region to the STIL-TAL1 locus. These findings highlight a general toxicity of endogenous gammaretroviral enhancer elements and also identify a combinatorial process during leukemic evolution that will be important for risk stratification and for future protocol design.


Subject(s)
Chromosomes, Human, X , Genetic Therapy/adverse effects , Genetic Therapy/methods , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Severe Combined Immunodeficiency/therapy , Adaptor Proteins, Signal Transducing , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA-Binding Proteins/genetics , Follow-Up Studies , Humans , Infant , LIM Domain Proteins , Male , Metalloproteins/genetics , Models, Biological , Mutagenesis , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/therapy , Proto-Oncogene Proteins , Receptor, Notch1/genetics , Receptors, Interleukin-2/genetics , Severe Combined Immunodeficiency/complications
14.
Lancet ; 376(9757): 2009-17, 2010 Dec 11.
Article in English | MEDLINE | ID: mdl-21131038

ABSTRACT

BACKGROUND: Although survival of children with acute lymphoblastic leukaemia has improved greatly in the past two decades, the outcome of those who relapse has remained static. We investigated the outcome of children with acute lymphoblastic leukaemia who relapsed on present therapeutic regimens. METHODS: This open-label randomised trial was undertaken in 22 centres in the UK and Ireland and nine in Australia and New Zealand. Patients aged 1-18 years with first relapse of acute lymphoblastic leukaemia were stratified into high-risk, intermediate-risk, and standard-risk groups on the basis of duration of first complete remission, site of relapse, and immunophenotype. All patients were allocated to receive either idarubicin or mitoxantrone in induction by stratified concealed randomisation. Neither patients nor those giving interventions were masked. After three blocks of therapy, all high-risk group patients and those from the intermediate group with postinduction high minimal residual disease (≥10(-4) cells) received an allogenic stem-cell transplant. Standard-risk and intermediate-risk patients with postinduction low minimal residual disease (<10(-4) cells) continued chemotherapy. The primary outcome was progression-free survival and the method of analysis was intention-to-treat. Randomisation was stopped in December, 2007 because of differences in progression-free and overall survival between the two groups. This trial is registered, reference number ISCRTN45724312. FINDINGS: Of 239 registered patients, 216 were randomly assigned to either idarubicin (109 analysed) or mitoxantrone (103 analysed). Estimated 3-year progression-free survival was 35·9% (95% CI 25·9-45·9) in the idarubicin group versus 64·6% (54·2-73·2) in the mitoxantrone group (p=0·0004), and 3-year overall survival was 45·2% (34·5-55·3) versus 69·0% (58·5-77·3; p=0·004). Differences in progression-free survival between groups were mainly related to a decrease in disease events (progression, second relapse, disease-related deaths; HR 0·56, 0·34-0·92, p=0·007) rather than an increase in adverse treatment effects (treatment death, second malignancy; HR 0·52, 0·24-1·11, p=0·11). INTERPRETATION: As compared with idarubicin, mitoxantrone conferred a significant benefit in progression-free and overall survival in children with relapsed acute lymphobastic leukaemia, a potentially useful clinical finding that warrants further investigation. FUNDING: Cancer Research UK, Leukaemia and Lymphoma Research, Cancer Council NSW, and Sporting Chance Cancer Foundation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Idarubicin/therapeutic use , Mitoxantrone/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Adolescent , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Asparaginase/administration & dosage , Child , Child, Preschool , Cyclophosphamide/administration & dosage , Cytarabine/administration & dosage , Dexamethasone/administration & dosage , Disease-Free Survival , Etoposide/administration & dosage , Female , Humans , Infant , Kaplan-Meier Estimate , Leukocyte Count , Male , Mercaptopurine/administration & dosage , Methotrexate/administration & dosage , Polyethylene Glycols/administration & dosage , Recurrence , Risk Assessment , Treatment Outcome , United Kingdom , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives , Vincristine/administration & dosage
15.
Leuk Lymphoma ; 62(3): 688-695, 2021 03.
Article in English | MEDLINE | ID: mdl-33225799

ABSTRACT

Congenital Acute Myeloid leukemia (CAML) is a rare leukemia diagnosed within the first 28 days of life. Dismal survival rates of approximately 25% at two years from diagnosis have not improved despite multiple treatment protocols, and there lacks international consensus for optimal management of these vulnerable patients. We report a retrospective analysis of our fifteen-year experience from a large UK tertiary pediatric center, focusing on treatment modalities and outcomes, including late therapeutic toxicities. To our knowledge, this is the first UK series of congenital leukemia patients reported. Twelve patients with a median age of 16.4 days (1-60) were diagnosed with CAML in fifteen years. All patients presented unwell; 92% demonstrating skin involvement. 10 (83%) received chemotherapy; with 1 death at presentation and 1 spontaneous remission. 5 (42%) received subsequent stem cell transplant. Only 4 (33%) remain alive, with 5 (42%) dying in disease remission with treatment-related mortality. Documented cardiotoxicity was observed in 3 (25%) patients, with a further 2 (17%) suspected but not receiving postmortem. Treatment of congenital AML raises challenging diagnostic, therapeutic and ethical questions and requires multi-center, international collaboration to see improvements.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Adolescent , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cardiotoxicity , Child , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/therapy , Remission Induction , Retrospective Studies , United Kingdom/epidemiology
16.
Leukemia ; 35(1): 90-106, 2021 01.
Article in English | MEDLINE | ID: mdl-32242051

ABSTRACT

MLL gene rearrangements (MLLr) are a common cause of aggressive, incurable acute lymphoblastic leukemias (ALL) in infants and children, most of which originate in utero. The most common MLLr produces an MLL-AF4 fusion protein. MLL-AF4 promotes leukemogenesis by activating key target genes, mainly through recruitment of DOT1L and increased histone H3 lysine-79 methylation (H3K79me2/3). One key MLL-AF4 target gene is PROM1, which encodes CD133 (Prominin-1). CD133 is a pentaspan transmembrane glycoprotein that represents a potential pan-cancer target as it is found on multiple cancer stem cells. Here we demonstrate that aberrant PROM1/CD133 expression is essential for leukemic cell growth, mediated by direct binding of MLL-AF4. Activation is controlled by an intragenic H3K79me2/3 enhancer element (KEE) leading to increased enhancer-promoter interactions between PROM1 and the nearby gene TAPT1. This dual locus regulation is reflected in a strong correlation of expression in leukemia. We find that in PROM1/CD133 non-expressing cells, the PROM1 locus is repressed by polycomb repressive complex 2 (PRC2) binding, associated with reduced expression of TAPT1, partially due to loss of interactions with the PROM1 locus. Together, these results provide the first detailed analysis of PROM1/CD133 regulation that explains CD133 expression in MLLr ALL.


Subject(s)
AC133 Antigen/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Leukemic , Histones/metabolism , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplastic Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Promoter Regions, Genetic , Biomarkers, Tumor , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Silencing , Humans , Immunophenotyping , Leukemia/genetics , Leukemia/metabolism , Models, Biological , Protein Binding
17.
Nat Commun ; 12(1): 6905, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824279

ABSTRACT

Although 90% of children with acute lymphoblastic leukemia (ALL) are now cured, the prognosis for infant-ALL remains dismal. Infant-ALL is usually caused by a single genetic hit that arises in utero: an MLL/KMT2A gene rearrangement (MLL-r). This is sufficient to induce a uniquely aggressive and treatment-refractory leukemia compared to older children. The reasons for disparate outcomes in patients of different ages with identical driver mutations are unknown. Using the most common MLL-r in infant-ALL, MLL-AF4, as a disease model, we show that fetal-specific gene expression programs are maintained in MLL-AF4 infant-ALL but not in MLL-AF4 childhood-ALL. We use CRISPR-Cas9 gene editing of primary human fetal liver hematopoietic cells to produce a t(4;11)/MLL-AF4 translocation, which replicates the clinical features of infant-ALL and drives infant-ALL-specific and fetal-specific gene expression programs. These data support the hypothesis that fetal-specific gene expression programs cooperate with MLL-AF4 to initiate and maintain the distinct biology of infant-ALL.


Subject(s)
Fetus , Gene Expression Regulation, Neoplastic , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , CRISPR-Cas Systems , DNA-Binding Proteins , Female , Gene Editing , Histone-Lysine N-Methyltransferase , Humans , Liver , Mice , Myeloid-Lymphoid Leukemia Protein/metabolism , Oncogene Proteins, Fusion/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcriptional Elongation Factors
18.
Haematologica ; 95(2): 333-7, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19679886

ABSTRACT

Hermansky Pudlak syndrome type 2 (HPS2) is a rare disorder associated with mutations in the Adaptor Protein 3 (AP-3) complex, which is involved in sorting transmembrane proteins to lysosomes and related organelles. We now report 2 unrelated subjects with HPS2 who show a characteristic clinical phenotype of oculocutaneous albinism, platelet and T-lymphocyte dysfunction and neutropenia. The subjects were homozygous for different deletions within AP3B1 (g.del180242-180866, c.del153-156), which encodes the AP-3beta3A subunit, resulting in frame shifts and introduction of nonsense substitutions (p.E693fsX13, p.E52fsX11). In the subject with p.E693fsX13, this resulted in expression of a truncated variant beta3A protein. Cytotoxic T-lymphocyte (CTL) clones from both study subjects showed increased cell-surface expression of CD63 and reduced cytotoxicity. Platelets showed impaired aggregation and reduced uptake of (3)H-serotonin. These findings are consistent with CTL granule and platelet dense granule defects, respectively. This report extends the clinical and laboratory description of HPS2.


Subject(s)
Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Hermanski-Pudlak Syndrome/genetics , Mutation , Albinism, Oculocutaneous/genetics , Child , Female , Humans , Infant , Male , Phenotype , Platelet Function Tests , T-Lymphocytes, Cytotoxic/metabolism
20.
J Exp Med ; 216(5): 1050-1060, 2019 05 06.
Article in English | MEDLINE | ID: mdl-30914438

ABSTRACT

Studies of allelic variation underlying genetic blood disorders have provided important insights into human hematopoiesis. Most often, the identified pathogenic mutations result in loss-of-function or missense changes. However, assessing the pathogenicity of noncoding variants can be challenging. Here, we characterize two unrelated patients with a distinct presentation of dyserythropoietic anemia and other impairments in hematopoiesis associated with an intronic mutation in GATA1 that is 24 nucleotides upstream of the canonical splice acceptor site. Functional studies demonstrate that this single-nucleotide alteration leads to reduced canonical splicing and increased use of an alternative splice acceptor site that causes a partial intron retention event. The resultant altered GATA1 contains a five-amino acid insertion at the C-terminus of the C-terminal zinc finger and has no observable activity. Collectively, our results demonstrate how altered splicing of GATA1, which reduces levels of the normal form of this master transcription factor, can result in distinct changes in human hematopoiesis.


Subject(s)
Alternative Splicing/genetics , Anemia, Dyserythropoietic, Congenital/genetics , GATA1 Transcription Factor/genetics , Hematopoiesis/genetics , Introns/genetics , Mutation, Missense , Myelodysplastic Syndromes/genetics , Adult , Child , Exons , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , Male , Myelodysplastic Syndromes/pathology , RNA Splice Sites/genetics , Transcription, Genetic/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL