Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 565
Filter
1.
J Clin Immunol ; 44(8): 183, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39172268

ABSTRACT

Severe congenital neutropenia (SCN) comprises a diverse range of rare hematological disorders characterized by recurrent, often life-threatening infections that manifest within the first months of life. Mutations in the ELANE gene are the most prevalent cause of SCN. While over 230 mutations in ELANE have been documented, including substitutions, frameshifts, nonsense mutations, and splice site alterations, the occurrence of deep intronic mutations has not been previously reported. Herein, we present the case of a young girl who exhibited recurrent fever, respiratory infections, skin abscesses, and gingivitis shortly after birth. Laboratory analysis revealed markedly diminished neutrophil levels alongside elevated monocyte and eosinophil counts. Bone marrow examination disclosed a halt in myelopoiesis maturation. ELANE gene full-length sequencing identified a novel de novo deep intron mutation in ELANE (c.598 + 79G > T), subsequently confirmed by Sanger sequencing. cDNA sequencing of the patient demonstrated aberrant gene splicing. Utilizing a mini-gene splicing assay for ELANE intronic variants, we identified a mutant ELANE allele (c.597 + 1_597 + 83ins) leading to the creation of a premature termination codon (p.Gly200ValfsTer40). Confocal microscopy revealed heightened expression of myeloperoxidase and neutrophil elastase in the patient, suggesting a potential role for the unfolded protein response in the pathogenesis of the deep intron ELANE mutation. In summary, our findings illustrate the first reported instance of de novo deep intron ELANE mutations associated with SCN, underscoring the importance of exploring deep intronic regions in SCN patients lacking identifiable disease-causing gene mutations.


Subject(s)
Congenital Bone Marrow Failure Syndromes , Introns , Leukocyte Elastase , Mutation , Neutropenia , Humans , Female , Neutropenia/genetics , Neutropenia/congenital , Neutropenia/diagnosis , Leukocyte Elastase/genetics , Congenital Bone Marrow Failure Syndromes/genetics , Congenital Bone Marrow Failure Syndromes/diagnosis , Introns/genetics , Mutation/genetics , Genetic Predisposition to Disease , Alleles
3.
Medicine (Baltimore) ; 103(30): e39086, 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39058847

ABSTRACT

RATIONALE: Severe congenital neutropenia (SCN) is a rare and heterogeneous genetic disease. By describing the diagnosis and treatment of a child with SCN and periodontitis, this case provides a reference for the oral health management of a child with SCN and periodontitis. PATIENT CONCERNS: We describe a boy with clinical manifestations of oral bleeding, neutropenia, recurrent fever, and other recurrent infections. The absolute neutrophil count (ANC) was <0.50 × 109/L most of the time. Morphological examination of bone marrow cells showed active granulocyte hyperplasia and dysmaturation. DIAGNOSES: According to the clinical manifestations, hematological examination and gene detection results, the child was diagnosed as SCN with chronic periodontitis. INTERVENTIONS: Periodontal treatment was performed after informed consent was obtained from the child guardian. These included supragingival and subgingival cleaning, hydrogen peroxide and saline irrigation, placement of iodoglycerin in the gingival sulcus, and oral hygiene instruction. Hematopoietic stem cell transplantation (HSCT) was performed later. OUTCOMES: One month after initial periodontal treatment, oral hygiene was well maintained and gingival swelling had subsided. Probing depth (PD) index on periodontal probing and bleeding was significantly reduced. However, there was no significant change in blood routine and other indicators before and after periodontal treatment. CONCLUSION: Once SCN is diagnosed, individualized treatment plans can be developed according to the characteristics of the disease and its impact on oral health, which can effectively control the interaction between SCN and periodontal disease and reduce the occurrence of serious infection.


Subject(s)
Congenital Bone Marrow Failure Syndromes , Neutropenia , Humans , Male , Neutropenia/congenital , Neutropenia/therapy , Neutropenia/diagnosis , Congenital Bone Marrow Failure Syndromes/complications , Congenital Bone Marrow Failure Syndromes/diagnosis , Child , Oral Health , Chronic Periodontitis/therapy , Chronic Periodontitis/complications , Chronic Periodontitis/diagnosis , Oral Hygiene , Hematopoietic Stem Cell Transplantation/methods , Periodontitis/therapy , Periodontitis/complications
4.
J Pediatr Hematol Oncol ; 46(6): e463-e465, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38934594

ABSTRACT

Severe congenital neutropenia is an inherited bone marrow failure disorder characterized by profoundly low neutrophil counts and promyelocytic maturation arrest in bone marrow. Severe congenital neutropenia is most often caused by heterozygous ELANE mutations. In vitro and mouse xenograft studies using CRISPR/Cas9 have shown that introduction of frameshift/nonsense mutations in mutant ELANE may restore neutrophil counts, providing a model for gene therapy. Here, we present 2 children with inherited nonsense mutations in ELANE analogous to those proposed for gene therapy. Their normal peripheral blood neutrophil counts provide support for this approach through human "experiments of nature."


Subject(s)
Codon, Nonsense , Congenital Bone Marrow Failure Syndromes , Genetic Therapy , Leukocyte Elastase , Neutropenia , Humans , Neutropenia/congenital , Neutropenia/genetics , Neutropenia/therapy , Congenital Bone Marrow Failure Syndromes/genetics , Congenital Bone Marrow Failure Syndromes/therapy , Genetic Therapy/methods , Leukocyte Elastase/genetics , Male , Female , Exons/genetics , Infant , Child , Child, Preschool
6.
Front Immunol ; 15: 1349919, 2024.
Article in English | MEDLINE | ID: mdl-38840904

ABSTRACT

Introduction: According to the PRISMA criteria, a systematic review has been conducted to investigate the clinical relevance between patients with severe congenital neutropenia (SCN) and cyclic congenital neutropenia (CyN) induced by ELANE mutations. Methods: We have searched PubMed, EMBASE, Web of Science, Scopus, Cochrane, CNKI, Wanfang Medicine, and VIP for ELANE mutation related literature published from 1997 to 2022. Using Microsoft Excel collect and organize data, SPSS 25, GraphPad Prism 8.0.1, and Omap analyze and plot statistical. Compare the gender, age, geography, mutation sites, infection characteristics, treatment, and other factors of SCN and CyN patients induced by ELANE mutations, with a focus on exploring the relationship between genotype and clinical characteristics, genotype and prognosis. Results: This study has included a total of 467 patients with SCN and 90 patients with CyN. The onset age of SCN and CyN are both less than 1 year old, and the onset and diagnosis age of SCN are both younger than CyN. The mutation of ELANE gene is mainly missense mutation, and hot spot mutations include S126L, P139L, G214R, c.597+1G>A. The high-frequency mutations with severe outcomes are A57V, L121H, L121P, c.597+1G>A, c.597+1G>T, S126L, C151Y, C151S, G214R, C223X. Respiratory tract, skin and mucosa are the most common infection sites, Staphylococcus aureus, Pseudomonas aeruginosa and Escherichia coli are the most common. Discussion: Patients with refractory G-CSF are more likely to develop severe outcomes. The commonly used pre-treatment schemes for transplantation are Bu-Cy-ATG and Flu-Bu-ATG. The prognosis of transplantation is mostly good, but the risk of GVHD is high. Systematic Review Registration: https://www.crd.york.ac.uk/PROSPERO/. PROSPERO, identifier CRD42023434656.


Subject(s)
Mutation , Neutropenia , Humans , Neutropenia/genetics , Neutropenia/congenital , Congenital Bone Marrow Failure Syndromes/genetics , Prognosis , Male , Female , Clinical Relevance
8.
Blood Adv ; 8(15): 4050-4065, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-38739706

ABSTRACT

ABSTRACT: A variety of autosomal recessive mutations in the JAGN1 gene cause severe congenital neutropenia (CN). However, the underlying pathomechanism remains poorly understood, mainly because of the limited availability of primary hematopoietic stem cells from JAGN1-CN patients and the absence of animal models. In this study, we aimed to address these limitations by establishing a zebrafish model of JAGN1-CN. We found 2 paralogs of the human JAGN1 gene, namely jagn1a and jagn1b, which play distinct roles during zebrafish hematopoiesis. Using various approaches such as morpholino-based knockdown, CRISPR/Cas9-based gene editing, and misexpression of a jagn1b harboring a specific human mutation, we successfully developed neutropenia while leaving other hematopoietic lineages unaffected. Further analysis of our model revealed significant upregulation of apoptosis and genes involved in the unfolded protein response (UPR). However, neither UPR nor apoptosis is the primary mechanism that leads to neutropenia in zebrafish. Instead, Jagn1b has a critical role in granulocyte colony-stimulating factor receptor signaling and steady-state granulopoiesis, shedding light on the pathogenesis of neutropenia associated with JAGN1 mutations. The establishment of a zebrafish model for JAGN1-CN represents a significant advancement in understanding the specific pathologic pathways underlying the disease. This model provides a valuable in vivo tool for further investigation and exploration of potential therapeutic strategies.


Subject(s)
Congenital Bone Marrow Failure Syndromes , Neutropenia , Signal Transduction , Unfolded Protein Response , Zebrafish , Animals , Humans , Apoptosis , Congenital Bone Marrow Failure Syndromes/genetics , Disease Models, Animal , Hematopoiesis/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation , Neutropenia/congenital , Neutropenia/genetics , Receptors, Granulocyte Colony-Stimulating Factor/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Mol Ther ; 32(6): 1628-1642, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38556793

ABSTRACT

Severe congenital neutropenia (CN) is an inherited pre-leukemia bone marrow failure syndrome commonly caused by autosomal-dominant ELANE mutations (ELANE-CN). ELANE-CN patients are treated with daily injections of recombinant human granulocyte colony-stimulating factor (rhG-CSF). However, some patients do not respond to rhG-CSF, and approximately 15% of ELANE-CN patients develop myelodysplasia or acute myeloid leukemia. Here, we report the development of a curative therapy for ELANE-CN through inhibition of ELANE mRNA expression by introducing two single-strand DNA breaks at the opposing DNA strands of the ELANE promoter TATA box using CRISPR-Cas9D10A nickases-termed MILESTONE. This editing effectively restored defective neutrophil differentiation of ELANE-CN CD34+ hematopoietic stem and progenitor cells (HSPCs) in vitro and in vivo, without affecting the functions of the edited neutrophils. CRISPResso analysis of the edited ELANE-CN CD34+ HSPCs revealed on-target efficiencies of over 90%. Simultaneously, GUIDE-seq, CAST-Seq, and rhAmpSeq indicated a safe off-target profile with no off-target sites or chromosomal translocations. Taken together, ex vivo gene editing of ELANE-CN HSPCs using MILESTONE in the setting of autologous stem cell transplantation could be a universal, safe, and efficient gene therapy approach for ELANE-CN patients.


Subject(s)
CRISPR-Cas Systems , Congenital Bone Marrow Failure Syndromes , Gene Editing , Genetic Therapy , Leukocyte Elastase , Neutropenia , Promoter Regions, Genetic , Gene Editing/methods , Humans , Neutropenia/congenital , Neutropenia/therapy , Neutropenia/genetics , Genetic Therapy/methods , Congenital Bone Marrow Failure Syndromes/therapy , Congenital Bone Marrow Failure Syndromes/genetics , Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Animals , Mice , Neutrophils/metabolism , Hematopoietic Stem Cells/metabolism , Mutation , Disease Models, Animal , Granulocyte Colony-Stimulating Factor/genetics , Genetic Diseases, X-Linked/therapy , Genetic Diseases, X-Linked/genetics
10.
Eur J Haematol ; 113(2): 146-162, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38600884

ABSTRACT

BACKGROUND: Congenital neutropenias are characterized by severe infections and a high risk of myeloid transformation; the causative genes vary across ethnicities. The Israeli population is characterized by an ethnically diverse population with a high rate of consanguinity. OBJECTIVE: To evaluate the clinical and genetic spectrum of congenital neutropenias in Israel. METHODS: We included individuals with congenital neutropenias listed in the Israeli Inherited Bone Marrow Failure Registry. Sanger sequencing was performed for ELANE or G6PC3, and patients with wild-type ELANE/G6PC3 were referred for next-generation sequencing. RESULTS: Sixty-five patients with neutropenia were included. Of 51 patients with severe congenital neutropenia, 34 were genetically diagnosed, most commonly with variants in ELANE (15 patients). Nine patients had biallelic variants in G6PC3, all of consanguineous Muslim Arab origin. Other genes involved were SRP54, JAGN1, TAZ, and SLC37A4. Seven patients had cyclic neutropenia, all with pathogenic variants in ELANE, and seven had Shwachman-Diamond syndrome caused by biallelic SBDS variants. Eight patients (12%) developed myeloid transformation, including six patients with an unknown underlying genetic cause. Nineteen (29%) patients underwent hematopoietic stem cell transplantation, mostly due to insufficient response to treatment with granulocyte-colony stimulating factor or due to myeloid transformation. CONCLUSIONS: The genetic spectrum of congenital neutropenias in Israel is characterized by a high prevalence of G6PC3 variants and an absence of HAX1 mutations. Similar to other registries, for 26% of the patients, a molecular diagnosis was not achieved. However, myeloid transformation was common in this group, emphasizing the need for close follow-up.


Subject(s)
Congenital Bone Marrow Failure Syndromes , Mutation , Neutropenia , Humans , Neutropenia/genetics , Neutropenia/congenital , Neutropenia/epidemiology , Neutropenia/diagnosis , Male , Israel/epidemiology , Female , Child , Congenital Bone Marrow Failure Syndromes/genetics , Congenital Bone Marrow Failure Syndromes/diagnosis , Child, Preschool , Adolescent , Genetic Predisposition to Disease , Adult , Hematopoietic Stem Cell Transplantation , Infant , Consanguinity , Glucose-6-Phosphatase/genetics , Alleles , Registries , High-Throughput Nucleotide Sequencing , Young Adult , Phenotype , Genetic Association Studies
11.
J Clin Pathol ; 77(9): 586-604, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-38589208

ABSTRACT

The congenital neutropenia syndromes are rare haematological conditions defined by impaired myeloid precursor differentiation or function. Patients are prone to severe infections with high mortality rates in early life. While some patients benefit from granulocyte colony-stimulating factor treatment, they may still face an increased risk of bone marrow failure, myelodysplastic syndrome and acute leukaemia. Accurate diagnosis is crucial for improved outcomes; however, diagnosis depends on familiarity with a heterogeneous group of rare disorders that remain incompletely characterised. The clinical and pathological overlap between reactive conditions, primary and congenital neutropenias, bone marrow failure, and myelodysplastic syndromes further clouds diagnostic clarity.We review the diagnostically useful clinicopathological and morphological features of reactive causes of neutropenia and the most common primary neutropenia disorders: constitutional/benign ethnic neutropenia, chronic idiopathic neutropenia, cyclic neutropenia, severe congenital neutropenia (due to mutations in ELANE, GFI1, HAX1, G6PC3, VPS45, JAGN1, CSF3R, SRP54, CLPB and WAS), GATA2 deficiency, Warts, hypogammaglobulinaemia, infections and myelokathexis syndrome, Shwachman-Diamond Syndrome, the lysosomal storage disorders with neutropenia: Chediak-Higashi, Hermansky-Pudlak, and Griscelli syndromes, Cohen, and Barth syndromes. We also detail characteristic cytogenetic and molecular factors at diagnosis and in progression to myelodysplastic syndrome/leukaemia.


Subject(s)
Congenital Bone Marrow Failure Syndromes , Neutropenia , Humans , Neutropenia/congenital , Neutropenia/diagnosis , Diagnosis, Differential , Congenital Bone Marrow Failure Syndromes/diagnosis , Congenital Bone Marrow Failure Syndromes/pathology , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Mutation
14.
Front Immunol ; 15: 1369243, 2024.
Article in English | MEDLINE | ID: mdl-38469307

ABSTRACT

Severe congenital neutropenia (SCN) is caused by germline mutations, most commonly in ELANE, impacting neutrophil maturation and leading to high risk of life-threatening infections. Most patients with ELANE-mutant SCN can achieve safe neutrophil counts with chronic Granulocyte-Colony Stimulating Factor (G-CSF). However, up to 10% of patients have neutropenia refractory to G-CSF and require allogeneic stem cell transplant. Traditional conditioning for these patients includes busulfan and cyclophosphamide which is associated with significant toxicities. We present five patients with SCN without myeloid malignancy transplanted using a reduced toxicity regimen of busulfan, fludarabine and thymoglobulin. 5 pediatric patients with SCN underwent matched sibling donor bone marrow transplant (MSD-BMT) between 2014-2022 on or per CHP14BT057 (NCT02928991), a prospective, single center trial testing elimination of cyclophosphamide from conditioning in pediatric patients with single lineage inherited BMF syndromes. All patients had MSDs and no evidence of MDS. Conditioning consisted of PK-adjusted busulfan, fludarabine, and thymoglobulin, with calcineurin inhibitor and mycophenolate mofetil GVHD prophylaxis. With median follow-up of 48.4 months, overall and event-free survival were 100%. There was no acute GVHD and one instance of chronic limited GVHD. Patients exhibited >95% donor myeloid chimerism at 5 years post-BMT. Two patients experienced CMV reactivation without end-organ disease, and no other viral reactivation or significant infections occurred. MSD-BMT with reduced toxicity myeloablation for SCN provides excellent outcomes while minimizing toxicity. These data suggest that busulfan, fludarabine, and ATG can be considered an efficacious, low-toxicity standard of care regimen for patients with SCN undergoing MSD-BMT.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Neutropenia , Neutropenia/congenital , Humans , Child , Bone Marrow Transplantation/adverse effects , Congenital Bone Marrow Failure Syndromes , Busulfan/therapeutic use , Busulfan/pharmacology , Hematopoietic Stem Cell Transplantation/methods , Siblings , Prospective Studies , Neutropenia/complications , Cyclophosphamide/therapeutic use , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Granulocyte Colony-Stimulating Factor/therapeutic use
15.
J Pediatr Hematol Oncol ; 46(3): e214-e219, 2024 04 01.
Article in English | MEDLINE | ID: mdl-38408162

ABSTRACT

BACKGROUND: Multisystemic findings of inherited bone marrow failure syndromes may cause difficulty in diagnosis. Exome sequencing (ES) helps to define the etiology of rare diseases and reanalysis offers a valuable new diagnostic approach. Herein, we present the clinical and molecular characteristics of a girl who was referred for cytopenia and frequent infections. CASE REPORT: A 5-year-old girl with cytopenia, dysmorphism, short stature, developmental delay, and myopia was referred for genetic counseling. Reanalysis of the ES data revealed a homozygous splice-site variant in the DNAJC21 (NM_001012339.3:c.983+1G>A), causing Shwachman-Diamond Syndrome (SDS). It was shown by the RNA sequencing that exon 7 was skipped, causing an 88-nucleotide deletion. CONCLUSIONS: Precise genetic diagnosis enables genetic counseling and improves patient management by avoiding inappropriate treatment and unnecessary testing. This report would contribute to the clinical and molecular understanding of this rare type of SDS caused by DNAJC21 variants and expand the phenotypic features of this condition.


Subject(s)
Bone Marrow Diseases , Cytopenia , Female , Humans , Child, Preschool , Congenital Bone Marrow Failure Syndromes/genetics , Exome/genetics , Shwachman-Diamond Syndrome , Homozygote , Bone Marrow Diseases/diagnosis , Bone Marrow Diseases/genetics
16.
FASEB J ; 38(4): e23478, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38372965

ABSTRACT

Carnitine derivatives of disease-specific acyl-CoAs are the diagnostic hallmark for long-chain fatty acid ß-oxidation disorders (lcFAOD), including carnitine shuttle deficiencies, very-long-chain acyl-CoA dehydrogenase deficiency (VLCADD), long-chain 3-hydroxyacyl-CoA dehydrogenase deficiency (LCHADD) and mitochondrial trifunctional protein deficiency (MPTD). The exact consequence of accumulating lcFAO-intermediates and their influence on cellular lipid homeostasis is, however, still unknown. To investigate the fate and cellular effects of the accumulating lcFAO-intermediates and to explore the presence of disease-specific markers, we used tracer-based lipidomics with deuterium-labeled oleic acid (D9-C18:1) in lcFAOD patient-derived fibroblasts. In line with previous studies, we observed a trend towards neutral lipid accumulation in lcFAOD. In addition, we detected a direct connection between the chain length and patterns of (un)saturation of accumulating acylcarnitines and the various enzyme deficiencies. Our results also identified two disease-specific candidate biomarkers. Lysophosphatidylcholine(14:1) (LPC(14:1)) was specifically increased in severe VLCADD compared to mild VLCADD and control samples. This was confirmed in plasma samples showing an inverse correlation with enzyme activity, which was better than the classic diagnostic marker C14:1-carnitine. The second candidate biomarker was an unknown lipid class, which we identified as S-(3-hydroxyacyl)cysteamines. We hypothesized that these were degradation products of the CoA moiety of accumulating 3-hydroxyacyl-CoAs. S-(3-hydroxyacyl)cysteamines were significantly increased in LCHADD compared to controls and other lcFAOD, including MTPD. Our findings suggest extensive alternative lipid metabolism in lcFAOD and confirm that lcFAOD accumulate neutral lipid species. In addition, we present two disease-specific candidate biomarkers for VLCADD and LCHADD, that may have significant relevance for disease diagnosis, prognosis, and monitoring.


Subject(s)
Cardiomyopathies , Congenital Bone Marrow Failure Syndromes , Lipid Metabolism, Inborn Errors , Lipidomics , Mitochondrial Diseases , Mitochondrial Myopathies , Mitochondrial Trifunctional Protein/deficiency , Muscular Diseases , Nervous System Diseases , Rhabdomyolysis , Humans , Mitochondrial Diseases/diagnosis , Carnitine , Cysteamine , Lipids
17.
Eur J Paediatr Neurol ; 49: 60-65, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38377647

ABSTRACT

Fatty acid oxidation (FAO) disorders are autosomal recessive genetic disorders affecting either the transport or the oxidation of fatty acids. Acute symptoms arise during prolonged fasting, intercurrent infections, or intense physical activity. Metabolic crises are characterized by alteration of consciousness, hypoglycemic coma, hepatomegaly, cardiomegaly, arrhythmias, rhabdomyolysis, and can lead to death. In this retrospective and multicentric study, the data of 54 patients with FAO disorders were collected. Overall, 35 patients (64.8%) were diagnosed after newborn screening (NBS), 17 patients on clinical presentation (31.5%), and two patients after family screening (3.7%). Deficiencies identified included medium-chain acyl-CoA dehydrogenase (MCAD) deficiency (75.9%), very long-chain acyl-CoA dehydrogenase (VLCAD) deficiency (11.1%), long-chain hydroxyacyl-CoA dehydrogenase (LCHAD) deficiency (3.7%), mitochondrial trifunctional protein (MTP) deficiency (1.8%), and carnitine palmitoyltransferase 2 (CPT 2) deficiency (7.4%). The NBS results of 25 patients were reviewed and the neurological outcome of this population was compared with that of the patients who were diagnosed on clinical presentation. This article sought to provide a comprehensive overview of how NBS implementation in Southern Belgium has dramatically improved the neurological outcome of patients with FAO disorders by preventing metabolic crises and death. Further investigations are needed to better understand the physiopathology of long-term complications in order to improve the quality of life of patients and to ensure optimal management.


Subject(s)
Acyl-CoA Dehydrogenase/deficiency , Cardiomyopathies , Carnitine O-Palmitoyltransferase/deficiency , Lipid Metabolism, Inborn Errors , Metabolism, Inborn Errors , Mitochondrial Trifunctional Protein/deficiency , Neonatal Screening , Rhabdomyolysis , Humans , Infant, Newborn , Retrospective Studies , Male , Female , Neonatal Screening/methods , Lipid Metabolism, Inborn Errors/diagnosis , Lipid Metabolism, Inborn Errors/complications , Belgium/epidemiology , Infant , Congenital Bone Marrow Failure Syndromes/complications , Congenital Bone Marrow Failure Syndromes/diagnosis , Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Fatty Acids/metabolism , Child, Preschool , Muscular Diseases/diagnosis , Child , Mitochondrial Myopathies/diagnosis , Mitochondrial Myopathies/complications , Mitochondrial Diseases/diagnosis , Mitochondrial Diseases/complications , Nervous System Diseases/etiology , Nervous System Diseases/diagnosis
18.
Leuk Res ; 137: 107441, 2024 02.
Article in English | MEDLINE | ID: mdl-38301422

ABSTRACT

Inherited bone marrow failure syndromes and germline predisposition syndromes (IBMFS/GPS) are associated with increased risk for hematologic malignancies, particularly myeloid neoplasms, such as myelodysplastic neoplasms (MDS) and acute myeloid leukemia (AML). The diagnosis of MDS in these syndromes poses difficulty due to frequent bone marrow hypocellularity and the presence of some degree of dysplastic features related to the underlying germline defect causing abnormal maturation of one or more cell lines. Yet, the diagnosis of MDS is usually associated with a worse outcome in several IBMFS/GPS. Criteria for the diagnosis of MDS in IBMFS/GPS have not been standardized with some authors suggesting a mixture of morphologic, cytogenetic, and genetic criteria. This review highlights these challenges and suggests a more standardized approach to nomenclature and diagnostic criteria.


Subject(s)
Bone Marrow Diseases , Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Humans , Bone Marrow Diseases/genetics , Bone Marrow Diseases/complications , Bone Marrow Diseases/pathology , Congenital Bone Marrow Failure Syndromes/complications , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Leukemia, Myeloid, Acute/genetics , Genetic Predisposition to Disease , Germ Cells/pathology
20.
Blood Adv ; 8(7): 1667-1682, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38286463

ABSTRACT

ABSTRACT: Congenital neutropenia (CN) is a genetic disorder characterized by persistent or intermittent low peripheral neutrophil counts, thus increasing susceptibility to bacterial and fungal infections. Various forms of CN, caused by distinct genetic mutations, exhibit differential responses to granulocyte colony-stimulating factor (G-CSF) therapy, with the underlying mechanisms not fully understood. This study presents an in-depth comparative analysis of clinical and immunological features in 5 CN patient groups (severe congenital neutropenia [SCN]1, SCN3, cyclic neutropenia [CyN], warts, hypogammaglobulinaemia, infections and myelokathexis [WHIM], and Shwachman-Bodian-Diamond Syndrome [SBDS]) associated with mutations in ELANE, HAX1, CXCR4, and SBDS genes. Our analysis led to the identification of 11 novel mutations in ELANE and 1 each in HAX1, CXCR4, and G6PC3 genes. Investigating bone marrow (BM) granulopoiesis and blood absolute neutrophil count after G-CSF treatment, we found that SCN1 and SCN3 presented with severe early-stage disruption between the promyelocyte and myelocyte, leading to a poor response to G-CSF. In contrast, CyN, affected at the late polymorphonuclear stage of neutrophil development, showed a strong G-CSF response. WHIM, displaying normal neutrophil development, responded robustly to G-CSF, whereas SBDS, with moderate disruption from the early myeloblast stage, exhibited a moderate response. Notably, SCN1 uniquely impeded neutrophil development, whereas SCN3, CyN, WHIM, and SBDS also affected eosinophils and basophils. In addition, SCN1, SCN3, and CyN presented with elevated serum immunoglobulins, increased BM plasma cells, and higher A Proliferation-Inducing Ligand levels. Our study reveals a strong correlation between the stage and severity of granulocyte development disruption and the efficacy of G-CSF therapy.


Subject(s)
Congenital Bone Marrow Failure Syndromes , Eosinophils , Granulocyte Colony-Stimulating Factor , Neutropenia/congenital , Humans , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte Colony-Stimulating Factor/therapeutic use , Mutation , Adaptor Proteins, Signal Transducing
SELECTION OF CITATIONS
SEARCH DETAIL