Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Lancet Haematol ; 11(5): e368-e382, 2024 May.
Article in English | MEDLINE | ID: mdl-38697731

ABSTRACT

Diamond-Blackfan anaemia (DBA), first described over 80 years ago, is a congenital disorder of erythropoiesis with a predilection for birth defects and cancer. Despite scientific advances, this chronic, debilitating, and life-limiting disorder continues to cause a substantial physical, psychological, and financial toll on patients and their families. The highly complex medical needs of affected patients require specialised expertise and multidisciplinary care. However, gaps remain in effectively bridging scientific discoveries to clinical practice and disseminating the latest knowledge and best practices to providers. Following the publication of the first international consensus in 2008, advances in our understanding of the genetics, natural history, and clinical management of DBA have strongly supported the need for new consensus recommendations. In 2014 in Freiburg, Germany, a panel of 53 experts including clinicians, diagnosticians, and researchers from 27 countries convened. With support from patient advocates, the panel met repeatedly over subsequent years, engaging in ongoing discussions. These meetings led to the development of new consensus recommendations in 2024, replacing the previous guidelines. To account for the diverse phenotypes including presentation without anaemia, the panel agreed to adopt the term DBA syndrome. We propose new simplified diagnostic criteria, describe the genetics of DBA syndrome and its phenocopies, and introduce major changes in therapeutic standards. These changes include lowering the prednisone maintenance dose to maximum 0·3 mg/kg per day, raising the pre-transfusion haemoglobin to 9-10 g/dL independent of age, recommending early aggressive chelation, broadening indications for haematopoietic stem-cell transplantation, and recommending systematic clinical surveillance including early colorectal cancer screening. In summary, the current practice guidelines standardise the diagnostics, treatment, and long-term surveillance of patients with DBA syndrome of all ages worldwide.


Subject(s)
Anemia, Diamond-Blackfan , Consensus , Humans , Anemia, Diamond-Blackfan/diagnosis , Anemia, Diamond-Blackfan/therapy , Anemia, Diamond-Blackfan/genetics , Disease Management , Hematopoietic Stem Cell Transplantation
2.
Exp Hematol ; 135: 104235, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38740323

ABSTRACT

The emergence of multiomic single-cell technologies over the last decade has led to improved insights into both normal hematopoiesis and its perturbation in a variety of hematological disorders. Diamond-Blackfan anemia (DBA) syndrome is one such disorder where single-cell assays have helped to delineate the cellular and molecular defects underlying the disease. DBA is caused by heterozygous loss-of-function germline variants in genes encoding ribosomal proteins (RPs). Despite the widespread role of ribosomes in hematopoiesis, the most frequent and severe cytopenia in DBA is anemia. In this review we discussed how single-cell studies, including clonogenic cell culture assays, fluorescence-activated cell sorting (FACS) and single-cell RNA sequencing (scRNA-seq), have led to insights into the pathogenesis of DBA. The main therapies are regular blood transfusions, glucocorticoids, or hematopoietic stem cell transplantation (HSCT) but all are associated with significant morbidity and mortality. We will therefore outline how single-cell studies can inform new therapies for DBA. Furthermore, we discussed how DBA serves as a useful model for understanding normal erythropoiesis in terms of its cellular hierarchy, molecular regulation during homeostasis, and response to "stress."


Subject(s)
Anemia, Diamond-Blackfan , Single-Cell Analysis , Anemia, Diamond-Blackfan/therapy , Anemia, Diamond-Blackfan/genetics , Humans , Erythropoiesis/genetics , Hematopoietic Stem Cell Transplantation , Ribosomal Proteins/genetics
3.
Blood Rev ; 61: 101097, 2023 09.
Article in English | MEDLINE | ID: mdl-37263874

ABSTRACT

Diamond-Blackfan anemia (DBA) is a rare bone marrow failure syndrome, usually caused by loss-of function variants in genes encoding ribosomal proteins. The hallmarks of DBA are anemia, congenital anomalies and cancer predisposition. Although DBA usually presents in childhood, the prevalence in later life is increasing due to an expanding repertoire of implicated genes, improvements in genetic diagnosis and increasing life expectancy. Adult patients uniquely suffer the manifestations of end-organ damage caused by the disease and its treatment, and transition to adulthood poses specific issues in disease management. To standardize and optimize care for this rare disease, in this review we provide updated guidance on the diagnosis and management of DBA, with a specific focus on older adolescents and adults. Recommendations are based upon published literature and our pooled clinical experience from three centres in the United Kingdom (U·K.). Uniquely we have also solicited and incorporated the views of affected families, represented by the independent patient organization, DBA U.K.


Subject(s)
Anemia, Diamond-Blackfan , Neoplasms , Adolescent , Humans , Adult , Anemia, Diamond-Blackfan/diagnosis , Anemia, Diamond-Blackfan/epidemiology , Anemia, Diamond-Blackfan/genetics , Rare Diseases , Ribosomal Proteins/genetics , Disease Susceptibility , Mutation
4.
Blood ; 140(14): 1578-1580, 2022 Oct 06.
Article in English | MEDLINE | ID: mdl-36201331
6.
Haematologica ; 107(3): 721-732, 2022 03 01.
Article in English | MEDLINE | ID: mdl-33596642

ABSTRACT

Multiple myeloma is a malignancy of plasma cells initiated and driven by primary and secondary genetic events. However, myeloma plasma cell survival and proliferation might be sustained by non-genetic drivers. Z-DNA-binding protein 1 (ZBP1; also known as DAI) is an interferon-inducible, Z-nucleic acid sensor that triggers RIPK3-MLKL-mediated necroptosis in mice. ZBP1 also interacts with TBK1 and the transcription factor IRF3 but the function of this interaction is unclear, and the role of the ZBP1-IRF3 axis in cancer is not known. Here we show that ZBP1 is selectively expressed in late B-cell development in both human and murine cells and it is required for optimal T-cell-dependent humoral immune responses. In myeloma plasma cells, the interaction of constitutively expressed ZBP1 with TBK1 and IRF3 results in IRF3 phosphorylation. IRF3 directly binds and activates cell cycle genes, in part through co-operation with the plasma cell lineage-defining transcription factor IRF4, thereby promoting myeloma cell proliferation. This generates a novel, potentially therapeutically targetable and relatively selective myeloma cell addiction to the ZBP1-IRF3 axis. Our data also show a noncanonical function of constitutive ZBP1 in human cells and expand our knowledge of the role of cellular immune sensors in cancer biology.


Subject(s)
Multiple Myeloma , Animals , Cell Proliferation , Humans , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Mice , Multiple Myeloma/genetics , Phosphorylation , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
7.
Cell Rep ; 36(11): 109698, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525349

ABSTRACT

Human hematopoiesis is a dynamic process that starts in utero 18-21 days post-conception. Understanding the site- and stage-specific variation in hematopoiesis is important if we are to understand the origin of hematological disorders, many of which occur at specific points in the human lifespan. To unravel how the hematopoietic stem/progenitor cell (HSPC) compartment changes during human ontogeny and the underlying gene regulatory mechanisms, we compare 57,489 HSPCs from 5 different tissues spanning 4 developmental stages through the human lifetime. Single-cell transcriptomic analysis identifies significant site- and developmental stage-specific transitions in cellular architecture and gene regulatory networks. Hematopoietic stem cells show progression from cycling to quiescence and increased inflammatory signaling during ontogeny. We demonstrate the utility of this dataset for understanding aberrant hematopoiesis through comparison to two cancers that present at distinct time points in postnatal life-juvenile myelomonocytic leukemia, a childhood cancer, and myelofibrosis, which classically presents in older adults.


Subject(s)
Cell Lineage/genetics , Gene Regulatory Networks/genetics , Hematopoietic Stem Cells/metabolism , Cell Differentiation , Hematopoiesis , Hematopoietic Stem Cells/cytology , Humans , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis , Transcriptome
8.
Sci Transl Med ; 13(610): eabf0113, 2021 Sep 08.
Article in English | MEDLINE | ID: mdl-34516827

ABSTRACT

Ribosome dysfunction underlies the pathogenesis of many cancers and heritable ribosomopathies. Here, we investigate how mutations in either ribosomal protein large (RPL) or ribosomal protein small (RPS) subunit genes selectively affect erythroid progenitor development and clinical phenotypes in Diamond-Blackfan anemia (DBA), a rare ribosomopathy with limited therapeutic options. Using single-cell assays of patient-derived bone marrow, we delineated two distinct cellular trajectories segregating with ribosomal protein genotypes. Almost complete loss of erythroid specification was observed in RPS-DBA. In contrast, we observed relative preservation of qualitatively abnormal erythroid progenitors and precursors in RPL-DBA. Although both DBA genotypes exhibited a proinflammatory bone marrow milieu, RPS-DBA was characterized by erythroid differentiation arrest, whereas RPL-DBA was characterized by preserved GATA1 expression and activity. Compensatory stress erythropoiesis in RPL-DBA exhibited disordered differentiation underpinned by an altered glucocorticoid molecular signature, including reduced ZFP36L2 expression, leading to milder anemia and improved corticosteroid response. This integrative analysis approach identified distinct pathways of erythroid failure and defined genotype-phenotype correlations in DBA. These findings may help facilitate therapeutic target discovery.


Subject(s)
Anemia, Diamond-Blackfan , Bone Marrow , Erythropoiesis , Humans , Ribosomal Proteins
9.
J Exp Med ; 218(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33416891

ABSTRACT

Juvenile myelomonocytic leukemia (JMML) is a poor-prognosis childhood leukemia usually caused by RAS-pathway mutations. The cellular hierarchy in JMML is poorly characterized, including the identity of leukemia stem cells (LSCs). FACS and single-cell RNA sequencing reveal marked heterogeneity of JMML hematopoietic stem/progenitor cells (HSPCs), including an aberrant Lin-CD34+CD38-CD90+CD45RA+ population. Single-cell HSPC index-sorting and clonogenic assays show that (1) all somatic mutations can be backtracked to the phenotypic HSC compartment, with RAS-pathway mutations as a "first hit," (2) mutations are acquired with both linear and branching patterns of clonal evolution, and (3) mutant HSPCs are present after allogeneic HSC transplant before molecular/clinical evidence of relapse. Stem cell assays reveal interpatient heterogeneity of JMML LSCs, which are present in, but not confined to, the phenotypic HSC compartment. RNA sequencing of JMML LSC reveals up-regulation of stem cell and fetal genes (HLF, MEIS1, CNN3, VNN2, and HMGA2) and candidate therapeutic targets/biomarkers (MTOR, SLC2A1, and CD96), paving the way for LSC-directed disease monitoring and therapy in this disease.


Subject(s)
Hematopoietic Stem Cells/pathology , Leukemia, Myelomonocytic, Juvenile/pathology , Animals , Biomarkers, Tumor/genetics , Cell Line , Female , Humans , Leukemia, Myelomonocytic, Juvenile/genetics , Male , Mice , Mutation/genetics , Neoplastic Stem Cells/pathology , Signal Transduction/genetics , Up-Regulation/genetics
10.
Mol Cell ; 78(3): 477-492.e8, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32386542

ABSTRACT

Myelofibrosis is a severe myeloproliferative neoplasm characterized by increased numbers of abnormal bone marrow megakaryocytes that induce fibrosis, destroying the hematopoietic microenvironment. To determine the cellular and molecular basis for aberrant megakaryopoiesis in myelofibrosis, we performed single-cell transcriptome profiling of 135,929 CD34+ lineage- hematopoietic stem and progenitor cells (HSPCs), single-cell proteomics, genomics, and functional assays. We identified a bias toward megakaryocyte differentiation apparent from early multipotent stem cells in myelofibrosis and associated aberrant molecular signatures. A sub-fraction of myelofibrosis megakaryocyte progenitors (MkPs) are transcriptionally similar to healthy-donor MkPs, but the majority are disease specific, with distinct populations expressing fibrosis- and proliferation-associated genes. Mutant-clone HSPCs have increased expression of megakaryocyte-associated genes compared to wild-type HSPCs, and we provide early validation of G6B as a potential immunotherapy target. Our study paves the way for selective targeting of the myelofibrosis clone and illustrates the power of single-cell multi-omics to discover tumor-specific therapeutic targets and mediators of tissue fibrosis.


Subject(s)
Hematopoiesis/physiology , Megakaryocytes/pathology , Primary Myelofibrosis/blood , Aged , Aged, 80 and over , Cell Differentiation , Female , Gene Expression Regulation , Hematopoiesis/genetics , Hematopoietic Stem Cells/pathology , High-Throughput Nucleotide Sequencing , Humans , Male , Megakaryocytes/physiology , Middle Aged , Mutation , Receptors, Immunologic/genetics , Single-Cell Analysis/methods
11.
Br J Haematol ; 186(2): 321-326, 2019 07.
Article in English | MEDLINE | ID: mdl-30980390

ABSTRACT

Diamond-Blackfan anaemia (DBA) is a rare bone marrow failure syndrome characterised by anaemia, congenital anomalies and cancer predisposition. Although infections are the second leading cause of mortality in non-transplanted patients, immune function is largely unexplored. We identified quantitative deficits in serum immunoglobulins and/or circulating T, natural killer and B lymphocytes in 59 of 107 unselected patients (55·1%) attending our centre over a 7-year period. Immune abnormalities were independent of ribosomal protein genotype and arose in both steroid-treated and steroid-untreated patients. In summary, these data highlight the high prevalence and spectrum of infections and immune defects in DBA.


Subject(s)
Anemia, Diamond-Blackfan , Genotype , Immunity, Cellular , Immunity, Humoral , Adolescent , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/immunology , Anemia, Diamond-Blackfan/mortality , Anemia, Diamond-Blackfan/pathology , Child , Child, Preschool , Female , Humans , Infant , Male , Ribosomal Proteins/genetics , Ribosomal Proteins/immunology , United Kingdom/epidemiology
12.
Genome Biol ; 17: 83, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27142433

ABSTRACT

BACKGROUND: Recent advances in single-cell techniques have provided the opportunity to finely dissect cellular heterogeneity within populations previously defined by "bulk" assays and to uncover rare cell types. In human hematopoiesis, megakaryocytes and erythroid cells differentiate from a shared precursor, the megakaryocyte-erythroid progenitor (MEP), which remains poorly defined. RESULTS: To clarify the cellular pathway in erythro-megakaryocyte differentiation, we correlate the surface immunophenotype, transcriptional profile, and differentiation potential of individual MEP cells. Highly purified, single MEP cells were analyzed using index fluorescence-activated cell sorting and parallel targeted transcriptional profiling of the same cells was performed using a specifically designed panel of genes. Differentiation potential was tested in novel, single-cell differentiation assays. Our results demonstrate that immunophenotypic MEP comprise three distinct subpopulations: "Pre-MEP," enriched for erythroid/megakaryocyte progenitors but with residual myeloid differentiation capacity; "E-MEP," strongly biased towards erythroid differentiation; and "MK-MEP," a previously undescribed, rare population of cells that are bipotent but primarily generate megakaryocytic progeny. Therefore, conventionally defined MEP are a mixed population, as a minority give rise to mixed-lineage colonies while the majority of cells are transcriptionally primed to generate exclusively single-lineage output. CONCLUSIONS: Our study clarifies the cellular hierarchy in human megakaryocyte/erythroid lineage commitment and highlights the importance of using a combination of single-cell approaches to dissect cellular heterogeneity and identify rare cell types within a population. We present a novel immunophenotyping strategy that enables the prospective identification of specific intermediate progenitor populations in erythro-megakaryopoiesis, allowing for in-depth study of disorders including inherited cytopenias, myeloproliferative disorders, and erythromegakaryocytic leukemias.


Subject(s)
Hematopoiesis , Megakaryocyte-Erythroid Progenitor Cells/cytology , Single-Cell Analysis/methods , Adult , Aged , Cell Lineage , Cell Separation/methods , Erythroid Cells/cytology , Erythroid Cells/metabolism , Female , Humans , Male , Megakaryocyte-Erythroid Progenitor Cells/classification , Megakaryocyte-Erythroid Progenitor Cells/metabolism , Megakaryocytes/cytology , Megakaryocytes/metabolism , Middle Aged
13.
Blood ; 125(16): 2553-7, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25755292

ABSTRACT

Diamond-Blackfan anemia (DBA) is a disorder characterized by a selective defect in erythropoiesis. Delineation of the precise defect is hampered by a lack of markers that define cells giving rise to erythroid burst- and erythroid colony-forming unit (BFU-E and CFU-E) colonies, the clonogenic assays that quantify early and late erythroid progenitor (EEP and LEP) potential, respectively. By combining flow cytometry, cell-sorting, and single-cell clonogenic assays, we identified Lin(-)CD34(+)CD38(+)CD45RA(-)CD123(-)CD71(+)CD41a(-)CD105(-)CD36(-) bone marrow cells as EEP giving rise to BFU-E, and Lin(-)CD34(+/-)CD38(+)CD45RA(-)CD123(-)CD71(+)CD41a(-)CD105(+)CD36(+) cells as LEP giving rise to CFU-E, in a hierarchical fashion. We then applied these definitions to DBA and identified that, compared with controls, frequency, and clonogenicity of DBA, EEP and LEP are significantly decreased in transfusion-dependent but restored in corticosteroid-responsive patients. Thus, both quantitative and qualitative defects in erythroid progenitor (EP) contribute to defective erythropoiesis in DBA. Prospective isolation of defined EPs will facilitate more incisive study of normal and aberrant erythropoiesis.


Subject(s)
Anemia, Diamond-Blackfan/blood , Bone Marrow Cells/metabolism , Erythroid Precursor Cells/metabolism , Erythropoiesis , Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/metabolism , Antigens, CD/metabolism , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cells, Cultured , Colony-Forming Units Assay , Endoglin , Flow Cytometry , GATA1 Transcription Factor/genetics , GATA2 Transcription Factor/genetics , Gene Expression , Humans , Immunophenotyping , Prospective Studies , Receptors, Cell Surface/metabolism
14.
Blood ; 124(1): 134-41, 2014 Jul 03.
Article in English | MEDLINE | ID: mdl-24805191

ABSTRACT

HDAC inhibitors (HDACi) increase transcription of some genes through histone hyperacetylation. To test the hypothesis that HDACi-mediated enhanced transcription might be of therapeutic value for inherited enzyme deficiency disorders, we focused on the glycolytic and pentose phosphate pathways (GPPPs). We show that among the 16 genes of the GPPPs, HDACi selectively enhance transcription of glucose 6-phosphate dehydrogenase (G6PD). This requires enhanced recruitment of the generic transcription factor Sp1, with commensurate recruitment of histone acetyltransferases and deacetylases, increased histone acetylation, and polymerase II recruitment to G6PD. These G6PD-selective transcriptional and epigenetic events result in increased G6PD transcription and ultimately restored enzymatic activity in B cells and erythroid precursor cells from patients with G6PD deficiency, a disorder associated with acute or chronic hemolytic anemia. Therefore, restoration of enzymatic activity in G6PD-deficient nucleated cells is feasible through modulation of G6PD transcription. Our findings also suggest that clinical consequences of pathogenic missense mutations in proteins with enzymatic function can be overcome in some cases by enhancement of the transcriptional output of the affected gene.


Subject(s)
Epigenesis, Genetic/drug effects , Glucosephosphate Dehydrogenase Deficiency/genetics , Glucosephosphate Dehydrogenase/biosynthesis , Glucosephosphate Dehydrogenase/genetics , Histone Deacetylase Inhibitors/pharmacology , Transcription, Genetic/drug effects , Cells, Cultured , Chromatin Immunoprecipitation , Glucosephosphate Dehydrogenase Deficiency/enzymology , Humans , Real-Time Polymerase Chain Reaction
16.
Br J Haematol ; 162(4): 530-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23718193

ABSTRACT

Diamond-Blackfan anaemia (DBA) is caused by inactivating mutations in ribosomal protein (RP) genes, with mutations in 13 of the 80 RP genes accounting for 50-60% of cases. The remaining 40-50% cases may harbour mutations in one of the remaining RP genes, but the very low frequencies render conventional genetic screening as challenging. We, therefore, applied custom enrichment technology combined with high-throughput sequencing to screen all 80 RP genes. Using this approach, we identified and validated inactivating mutations in 15/17 (88%) DBA patients. Target enrichment combined with high-throughput sequencing is a robust and improved methodology for the genetic diagnosis of DBA.


Subject(s)
Anemia, Diamond-Blackfan/genetics , DNA Mutational Analysis , DNA, Ribosomal/genetics , High-Throughput Nucleotide Sequencing , Ribosomal Proteins/genetics , Adolescent , Adult , Child , Child, Preschool , Codon, Nonsense , DNA, Ribosomal/blood , Female , Frameshift Mutation , Gene Library , Humans , Male , Sequence Alignment , Sequence Analysis, DNA , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL
...