Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Eur J Haematol ; 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38967591

ABSTRACT

Shwachman-Diamond syndrome (SDS) is an inherited bone marrow failure disorder that often presents at infancy. Progress has been made in revealing causal mutated genes (SBDS and others), ribosome defects, and hematopoietic aberrations in SDS. However, the mechanism underlying the hematopoietic failure remained unknown, and treatment options are limited. Herein, we investigated the onset of SDS embryonic hematopoietic impairments. We generated SDS and control human-derived induced pluripotent stem cells (iPSCs). SDS iPSCs recapitulated the SDS hematological phenotype. Detailed stepwise evaluation of definitive hematopoiesis revealed defects that started at the early emerging hematopoietic progenitor (EHP) stage after mesoderm and hemogenic endothelium were normally induced. Hematopoietic potential of EHPs was markedly reduced, and the introduction of SBDS in SDS iPSCs improved colony formation. Transcriptome analysis revealed reduced expression of ribosome and oxidative phosphorylation-related genes in undifferentiated and differentiated iPSCs. However, certain pathways (e.g., DNA replication) and genes (e.g., CHCHD2) were exclusively or more severely dysregulated in EHPs compared with earlier and later stages. To our knowledge, this study offers for the first time an insight into the embryonic onset of human hematopoietic defects in an inherited bone marrow failure syndrome and reveals cellular and molecular aberrations at critical stages of hematopoietic development toward EHPs.

2.
Pediatr Blood Cancer ; 71(8): e31070, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38757488

ABSTRACT

Severe aplastic anemia (SAA) is a rare potentially fatal hematologic disorder. Although overall outcomes with treatment are excellent, there are variations in management approach, including differences in treatment between adult and pediatric patients. Certain aspects of treatment are under active investigation in clinical trials. Because of the rarity of the disease, some pediatric hematologists may have relatively limited experience with the complex management of SAA. The following recommendations reflect an up-to-date evidence-based approach to the treatment of children with newly diagnosed SAA.


Subject(s)
Anemia, Aplastic , Humans , Anemia, Aplastic/therapy , Anemia, Aplastic/diagnosis , Child , Evidence-Based Medicine , Practice Guidelines as Topic/standards
3.
Pediatr Blood Cancer ; 71(8): e31075, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38764170

ABSTRACT

Severe aplastic anemia (SAA) is a rare potentially fatal hematologic disorder. Although overall outcomes with treatment are excellent, there are variations in management approach, including differences in treatment between adult and pediatric patients. Certain aspects of treatment are under active investigation in clinical trials. Because of the rarity of the disease, some pediatric hematologists may have relatively limited experience with the complex management of SAA. The following recommendations reflect an up-to-date evidence-based approach to the treatment of children with relapsed or refractory SAA.


Subject(s)
Anemia, Aplastic , Humans , Anemia, Aplastic/therapy , Child , Recurrence , Evidence-Based Medicine , Hematopoietic Stem Cell Transplantation
4.
Haematologica ; 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38618686

ABSTRACT

Not available.

8.
Eur J Haematol ; 108(4): 278-287, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34897809

ABSTRACT

Classification of inherited bone marrow failure syndromes (IBMFSs) according to clinical and genetic diagnoses enables proper adjustment of treatment. Unfortunately, 30% of patients enrolled in the Canadian Inherited Marrow Failure Registry (CIMFR) with features suggesting hereditability could not be classified with a specific syndromic diagnosis. We analyzed the outcome of hematopoietic stem cell transplantation (HSCT) in unclassified IBMFSs (uIBMFSs) and the factors associated with outcome. Twenty-two patients with uIBMFSs and 70 patients with classified IBMFSs underwent HSCT. Five-year overall survival of uIBMFS patients after HSCT was inferior to that of patients with classified IBMFSs (56% vs 76.5%). The outcome of patients with uIBMFS who received cord blood was significantly lower than that of patients who received other stem cell sources (14.8% vs 90.9%). Engraftment failure was higher among patients with uIBMFS who received cord blood than those who received bone marrow. None of the following factors were significantly associated with poor survival: transfusion load, transplant indication, the intensity of conditioning regimen, human leukocyte antigen-identical sibling/alternative donor. We suggest that identifying the genetic diagnosis is essential to modulate the transplant procedure including conditioning agents and stem cell sources for better outcome and the standard cord blood transplantation (CBT) should be avoided in uIBMFS.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Bone Marrow Transplantation , Canada/epidemiology , Congenital Bone Marrow Failure Syndromes , Graft vs Host Disease/diagnosis , Graft vs Host Disease/etiology , HLA Antigens , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Retrospective Studies , Transplantation Conditioning/methods
9.
Front Immunol ; 12: 748519, 2021.
Article in English | MEDLINE | ID: mdl-34777360

ABSTRACT

Inherited defects that abrogate the function of the adenosine deaminase (ADA) enzyme and consequently lead to the accumulation of toxic purine metabolites cause profound lymphopenia and severe combined immune deficiency. Additionally, neutropenia and impaired neutrophil function have been reported among ADA-deficient patients. However, due to the rarity of the disorder, the neutrophil developmental abnormalities and the mechanisms contributing to them have not been characterized. Induced pluripotent stem cells (iPSC) generated from two unrelated ADA-deficient patients and from healthy controls were differentiated through embryoid bodies into neutrophils. ADA deficiency led to a significant reduction in the number of all early multipotent hematopoietic progenitors. At later stages of differentiation, ADA deficiency impeded the formation of granulocyte colonies in methylcellulose cultures, leading to a significant decrease in the number of neutrophils generated from ADA-deficient iPSCs. The viability and apoptosis of ADA-deficient neutrophils isolated from methylcellulose cultures were unaffected, suggesting that the abnormal purine homeostasis in this condition interferes with differentiation or proliferation. Additionally, there was a significant increase in the percentage of hyperlobular ADA-deficient neutrophils, and these neutrophils demonstrated significantly reduced ability to phagocytize fluorescent microspheres. Supplementing iPSCs and methylcellulose cultures with exogenous ADA, which can correct adenosine metabolism, reversed all abnormalities, cementing the critical role of ADA in neutrophil development. Moreover, chemical inhibition of the ribonucleotide reductase (RNR) enzyme, using hydroxyurea or a combination of nicotinamide and trichostatin A in iPSCs from healthy controls, led to abnormal neutrophil differentiation similar to that observed in ADA deficiency, implicating RNR inhibition as a potential mechanism for the neutrophil abnormalities. In conclusion, the findings presented here demonstrate the important role of ADA in the development and function of neutrophils while clarifying the mechanisms responsible for the neutrophil abnormalities in ADA-deficient patients.


Subject(s)
Adenosine Deaminase/physiology , Agammaglobulinemia/immunology , Induced Pluripotent Stem Cells/cytology , Neutrophils/cytology , Severe Combined Immunodeficiency/immunology , Adenosine Deaminase/genetics , Cells, Cultured , Embryoid Bodies/cytology , Fibroblasts/enzymology , Granulocytes/cytology , Humans , Hydroxamic Acids/pharmacology , Hydroxyurea/pharmacology , Infant , Male , Mutation, Missense , Myelopoiesis , Niacinamide/pharmacology , Point Mutation , Ribonucleotide Reductases/antagonists & inhibitors
10.
Am J Hematol ; 96(11): 1491-1504, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34342889

ABSTRACT

The North American Pediatric Aplastic Anemia Consortium (NAPAAC) is a group of pediatric hematologist-oncologists, hematopathologists, and bone marrow transplant physicians from 46 institutions in North America with interest and expertise in aplastic anemia, inherited bone marrow failure syndromes, and myelodysplastic syndromes. The NAPAAC Bone Marrow Failure Diagnosis and Care Guidelines Working Group was established with the charge of harmonizing the approach to the diagnostic workup of aplastic anemia in an effort to standardize best practices in the field. This document outlines the rationale for initial evaluations in pediatric patients presenting with signs and symptoms concerning for severe aplastic anemia.


Subject(s)
Anemia, Aplastic/diagnosis , Anemia, Aplastic/pathology , Bone Marrow/pathology , Child , Diagnosis, Differential , Fetal Hemoglobin/analysis , HLA Antigens/analysis , Humans , North America , Severity of Illness Index
11.
Pediatr Blood Cancer ; 67(10): e28652, 2020 10.
Article in English | MEDLINE | ID: mdl-32779892

ABSTRACT

BACKGROUND: Myelodysplastic syndromes (MDS) represent a group of clonal hematopoietic stem cell disorders that commonly progress to acute myeloid leukemia (AML). The diagnostics, prognostics, and treatment of adult MDS are established but do not directly translate to children and adolescents. Pediatric MDS is a rare disease, characterized by unique cytogenetics and histology compared with adult MDS, and often arises secondary to germline predisposition or cytotoxic exposures. Our objective was to highlight aspects of diagnosis/management that would benefit from further systematic review toward the development of clinical practice guidelines for pediatric MDS. PROCEDURE: The North American Pediatric Aplastic Anemia Consortium (NAPAAC) is composed of collaborative institutions with a strong interest in pediatric bone marrow failure syndromes and hematologic malignancies. The NAPAAC MDS working group developed a national survey distributed to 35 NAPAAC institutions to assess data on (1) clinical presentation of pediatric MDS, (2) diagnostic evaluation, (3) criteria for diagnosis, (4) supportive care and treatment decisions, and (5) role of hematopoietic stem cell transplantation (HSCT). RESULTS: Twenty-eight of 35 institutions returned the survey. Most centers agreed on a common diagnostic workup, though there was considerable variation regarding the criteria for diagnosis. Although there was consensus on supportive care, treatment strategies, including the role of cytoreduction and HSCT, varied across centers surveyed. CONCLUSIONS: There is lack of national consensus on diagnosis and treatment of pediatric MDS. This survey identified key aspects of MDS management that will warrant systematic review toward the goal of developing national clinical practice guidelines for pediatric MDS.


Subject(s)
Decision Making , Hematopoietic Stem Cell Transplantation/methods , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/therapy , Practice Patterns, Physicians'/statistics & numerical data , Anemia, Aplastic/diagnosis , Anemia, Aplastic/therapy , Child , Humans , Prognosis , Retrospective Studies , Surveys and Questionnaires , Survival Rate
12.
Br J Haematol ; 189(5): 976-981, 2020 06.
Article in English | MEDLINE | ID: mdl-32128787

ABSTRACT

Progressive cytopenia is a serious complication among paediatric patients with inherited bone marrow failure syndromes (IBMFS). Androgens have been used to improve blood counts in different bone marrow failure conditions. Little is known about efficacy and toxicity with new androgens (i.e., danazol) in different types of IBMFS. We identified 29 patients from the Canadian Inherited Marrow Failure Registry, who received oxymetholone or danazol. Sixteen (55%) had haematological response including patients with unclassified IBMFS (45%). Danazol showed a better toxicity profile and similar efficacy compared to oxymetholone. Androgens are an effective and safe option to ameliorate bone marrow failure in IBMFS.


Subject(s)
Androgens/therapeutic use , Bone Marrow Failure Disorders/drug therapy , Adolescent , Adult , Androgens/adverse effects , Bone Marrow Failure Disorders/blood , Bone Marrow Failure Disorders/genetics , Bone Marrow Failure Disorders/therapy , Canada/epidemiology , Cell Lineage , Child , Child, Preschool , Combined Modality Therapy , Danazol/adverse effects , Danazol/therapeutic use , Disease Progression , Drug Substitution , Female , Hematopoietic Stem Cell Transplantation , Humans , Infant , Male , Middle Aged , Oxymetholone/adverse effects , Oxymetholone/therapeutic use , Pancytopenia/drug therapy , Pancytopenia/etiology , Registries , Thrombocytopenia/drug therapy , Thrombocytopenia/etiology , Treatment Outcome , Virilism/chemically induced
13.
J Pediatr Hematol Oncol ; 42(3): 170-174, 2020 04.
Article in English | MEDLINE | ID: mdl-32134844

ABSTRACT

The distinction between myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) often relies on an arbitrary marrow blast cutoff of 30% in pediatrics and 20% in adults. There is little data about the treatment of children with extramedullary myeloid malignancy that has features of both, MDS and AML. Herein, we report for the first time 2 patients MDS/AML (1 with Shwachman-Diamond syndrome and 1 with idiopathic MDS and monosomy 7) who presented with extramedullary complications, received treatment with azacitidine, achieved complete remission and subsequently underwent hematopoietic stem cell transplantation.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Adolescent , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 7 , Humans , Leukemia, Myeloid, Acute/genetics , Male , Myelodysplastic Syndromes/genetics , Shwachman-Diamond Syndrome/complications
14.
JCI Insight ; 5(4)2020 02 27.
Article in English | MEDLINE | ID: mdl-31990679

ABSTRACT

Inherited bone marrow failure syndromes, such as Fanconi anemia (FA) and Shwachman-Diamond syndrome (SDS), feature progressive cytopenia and a risk of acute myeloid leukemia (AML). Using deep phenotypic analysis of early progenitors in FA/SDS bone marrow samples, we revealed selective survival of progenitors that phenotypically resembled granulocyte-monocyte progenitors (GMP). Whole-exome and targeted sequencing of GMP-like cells in leukemia-free patients revealed a higher mutation load than in healthy controls and molecular changes that are characteristic of AML: increased G>A/C>T variants, decreased A>G/T>C variants, increased trinucleotide mutations at Xp(C>T)pT, and decreased mutation rates at Xp(C>T)pG sites compared with other Xp(C>T)pX sites and enrichment for Cancer Signature 1 (X indicates any nucleotide). Potential preleukemic targets in the GMP-like cells from patients with FA/SDS included SYNE1, DST, HUWE1, LRP2, NOTCH2, and TP53. Serial analysis of GMPs from an SDS patient who progressed to leukemia revealed a gradual increase in mutational burden, enrichment of G>A/C>T signature, and emergence of new clones. Interestingly, the molecular signature of marrow cells from 2 FA/SDS patients with leukemia was similar to that of FA/SDS patients without transformation. The predicted founding clones in SDS-derived AML harbored mutations in several genes, including TP53, while in FA-derived AML the mutated genes included ARID1B and SFPQ. We describe an architectural change in the hematopoietic hierarchy of FA/SDS with remarkable preservation of GMP-like populations harboring unique mutation signatures. GMP-like cells might represent a cellular reservoir for clonal evolution.


Subject(s)
Bone Marrow Failure Disorders/pathology , Hematopoietic Stem Cells/pathology , Models, Genetic , Bone Marrow Failure Disorders/genetics , Clonal Evolution , Humans , Leukemia, Myeloid, Acute/genetics , Mutation , Myelodysplastic Syndromes/genetics
15.
NPJ Genom Med ; 4: 30, 2019.
Article in English | MEDLINE | ID: mdl-31839986

ABSTRACT

Inherited bone marrow failure syndromes (IBMFSs) are genetically heterogeneous disorders with cytopenia. Many IBMFSs also feature physical malformations and an increased risk of cancer. Point mutations can be identified in about half of patients. Copy number variation (CNVs) have been reported; however, the frequency and spectrum of CNVs are unknown. Unfortunately, current genome-wide methods have major limitations since they may miss small CNVs or may have low sensitivity due to low read depths. Herein, we aimed to determine whether reanalysis of NGS panel data by normalized coverage value could identify CNVs and characterize them. To address this aim, DNA from IBMFS patients was analyzed by a NGS panel assay of known IBMFS genes. After analysis for point mutations, heterozygous and homozygous CNVs were searched by normalized read coverage ratios and specific thresholds. Of the 258 tested patients, 91 were found to have pathogenic point variants. NGS sample data from 165 patients without pathogenic point mutations were re-analyzed for CNVs; 10 patients were found to have deletions. Diamond Blackfan anemia genes most commonly exhibited heterozygous deletions, and included RPS19, RPL11, and RPL5. A diagnosis of GATA2-related disorder was made in a patient with myelodysplastic syndrome who was found to have a heterozygous GATA2 deletion. Importantly, homozygous FANCA deletion were detected in a patient who could not be previously assigned a specific syndromic diagnosis. Lastly, we identified compound heterozygousity for deletions and pathogenic point variants in RBM8A and PARN genes. All deletions were validated by orthogonal methods. We conclude that careful analysis of normalized coverage values can detect CNVs in NGS panels and should be considered as a standard practice prior to do further investigations.

16.
Blood Adv ; 3(10): 1622-1637, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138591

ABSTRACT

Polymorphonuclear neutrophils (PMNs) are the most abundant circulating leukocytes, and the first cells recruited to sites of tissue inflammation. Using a fixation method to preserve native CD marker expression prior to immunophenotyping, we identified a distinct population of "primed for recruitment" PMNs in healthy mouse and human blood that has high expression of adhesion and activation markers compared with the bulk resting-state PMNs. In response to acute tissue inflammation, primed PMNs (pPMNs) were rapidly depleted from the circulation and recruited to the tissue. One hour after acute peritoneal insult, pPMNs became the dominant PMN population in bone marrow (BM) and blood, returning to baseline levels with resolution of inflammation. PMN priming was induced by the granulopoietic factors granulocyte-macrophage-colony-stimulating factor (GM-CSF) and granulocyte-colony-stimulating factor (G-CSF). High levels of pPMNs were observed in neutropenic mice and in pediatric neutropenic patients who were resistant to infection, highlighting an important role of this population in innate immune function.


Subject(s)
Neutrophils/metabolism , Adolescent , Adult , Animals , Healthy Volunteers , Humans , Mice , Young Adult
18.
Angiogenesis ; 22(1): 95-102, 2019 02.
Article in English | MEDLINE | ID: mdl-30168024

ABSTRACT

Vascular complications such as bleeding due to gastrointestinal telangiectatic anomalies, pulmonary arteriovenous malformations, hepatopulmonary syndrome, and retinal vessel abnormalities are being reported in patients with telomere biology disorders (TBDs) more frequently than previously described. The international clinical care consortium of telomere-associated ailments and family support group Dyskeratosis Congenita Outreach, Inc. held a workshop on vascular abnormalities in the TBDs at the National Cancer Institute in October 2017. Clinicians and basic scientists reviewed current data on vascular complications, hypotheses for the underlying biology and developed new collaborations to address the etiology and clinical management of vascular complications in TBDs.


Subject(s)
Arteriovenous Fistula , Pulmonary Artery/abnormalities , Pulmonary Veins/abnormalities , Telangiectasis , Telomere , Animals , Arteriovenous Fistula/genetics , Arteriovenous Fistula/metabolism , Arteriovenous Fistula/pathology , Education , Humans , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pulmonary Veins/metabolism , Pulmonary Veins/pathology , Telangiectasis/genetics , Telangiectasis/metabolism , Telangiectasis/pathology , Telomere/genetics , Telomere/metabolism , Telomere/pathology
19.
J Med Genet ; 56(5): 340-346, 2019 05.
Article in English | MEDLINE | ID: mdl-30327448

ABSTRACT

BACKGROUND: Dandy-Walker malformation features agenesis/hypoplasia of the cerebellar vermis, cystic dilatation of the fourth ventricle and enlargement of posterior fossa. Although Dandy-Walker malformation is relatively common and several genes were linked to the syndrome, the genetic cause in the majority of cases is unknown. OBJECTIVE: To identify the mutated gene responsible for Dandy-Walker malformation, kidney disease and bone marrow failure in four patients from two unrelated families. METHODS: Medical assessment, sonographic, MRI and pathological studies were used to define phenotype. Chromosomal microarray analysis and whole-exome sequence were performed to unravel the genotype. RESULTS: We report four subjects from two unrelated families with homozygous mutations in the Exocyst Complex Component 3-Like-2 gene (EXOC3L2).EXOC3L2 functions in trafficking of post-Golgi vesicles to the plasma membrane. In the first family a missense mutation in a highly conserved amino acid, p.Leu41Gln, was found in three fetuses; all had severe forms of Dandy-Walker malformation that was detectable by prenatal ultrasonography and confirmed by autopsy. In the second family, the affected child carried a nonsense mutation, p.Arg72*, and no detected protein. He had peritrigonal and cerebellar white matter abnormalities with enlargement of the ventricular trigones, developmental delay, pituitary hypoplasia, severe renal dysplasia and bone marrow failure. CONCLUSION: We propose that biallelic EXOC3L2 mutations lead to a novel syndrome that affects hindbrain development, kidney and possibly the bone marrow.


Subject(s)
Alleles , Dandy-Walker Syndrome/diagnosis , Dandy-Walker Syndrome/genetics , Mutation , Phenotype , Vesicular Transport Proteins/genetics , Biopsy , Brain/pathology , DNA Copy Number Variations , Homozygote , Humans , Kidney/metabolism , Magnetic Resonance Imaging , Symptom Assessment , Syndrome , Ultrasonography , Vesicular Transport Proteins/metabolism , Exome Sequencing
20.
Mol Genet Genomic Med ; 6(3): 463-468, 2018 05.
Article in English | MEDLINE | ID: mdl-29633571

ABSTRACT

BACKGROUND: ERCC6L2-associated disorder has recently been described and only five patients were reported so far. The described phenotype included bone marrow, cerebral, and craniofacial abnormalities. The aim of this study was to further define the genetic and phenotypic spectrum of the disorder by summarizing the five published cases and an additional case that we identified through whole-exome sequencing performed at the University of Toronto. METHODS: Clinical data was extracted from the Canadian Inherited Marrow Failure Registry. Whole exome sequencing was performed to identify causative mutations. RESULTS: All six cases had homozygous truncating mutations either at or upstream of the helicase domain of ERCC6L2. All patients displayed bone marrow failure, learning or developmental delay and microcephaly. Our patient was unique in displaying features of cerebellar disease, including ataxia and dysmetria as well as an interval deterioration of the corpus callosum and generalized volume loss on MRI. Another unique feature of our patient was retinal dystrophy with macular involvement. Along with one other patient, our patient displayed craniofacial abnormalities by presenting with low-set prominent ears, a pointed prominent chin, and deep-set eyes. Leukemia is common among patients with inherited bone marrow failure, but thus far, none of the patients have developed this complication. CONCLUSIONS: ERCC6L2-associated disorder is a multisystem disorder. The phenotype spectrum includes bone marrow failure, cerebral, and craniofacial abnormalities, as well as cerebellar and retinal abnormalities.


Subject(s)
Anemia, Aplastic/genetics , Bone Marrow Diseases/genetics , DNA Helicases/genetics , Hemoglobinuria, Paroxysmal/genetics , Adolescent , Adult , Anemia, Aplastic/physiopathology , Bone Marrow Diseases/physiopathology , Bone Marrow Failure Disorders , Canada , Child , Child, Preschool , Craniofacial Abnormalities/genetics , DNA Helicases/physiology , Developmental Disabilities/genetics , Exome , Female , Hemoglobinuria, Paroxysmal/physiopathology , Homozygote , Humans , Infant , Intellectual Disability/genetics , Male , Microcephaly/genetics , Mutation , Nervous System Malformations/genetics , Pedigree , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...