Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 702
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 29: 399-413, 2011.
Article in English | MEDLINE | ID: mdl-21219176

ABSTRACT

The discovery of genetic defects causing congenital neutropenia has illuminated mechanisms controlling differentiation, circulation, and decay of neutrophil granulocytes. Deficiency of the mitochondrial proteins HAX1 and AK2 cause premature apoptosis of myeloid progenitor cells associated with dissipation of the mitochondrial membrane potential, whereas mutations in ELA2/ELANE and G6PC3 are associated with signs of increased endoplasmic reticulum stress. Mutations in the transcriptional repressor GFI1 and the cytoskeletal regulator WASP also lead to defective neutrophil production. This unexpected diversity of factors suggests that multiple pathways are involved in the pathogenesis of congenital neutropenia.


Subject(s)
Neutrophils/cytology , Congenital Bone Marrow Failure Syndromes , Endoplasmic Reticulum , Humans , Mutation , Neutropenia/congenital , Neutropenia/genetics , Neutropenia/pathology , Neutrophils/metabolism
2.
Nature ; 582(7810): 109-114, 2020 06.
Article in English | MEDLINE | ID: mdl-32494068

ABSTRACT

Advances in genetics and sequencing have identified a plethora of disease-associated and disease-causing genetic alterations. To determine causality between genetics and disease, accurate models for molecular dissection are required; however, the rapid expansion of transcriptional populations identified through single-cell analyses presents a major challenge for accurate comparisons between mutant and wild-type cells. Here we generate mouse models of human severe congenital neutropenia (SCN) using patient-derived mutations in the GFI1 transcription factor. To determine the effects of SCN mutations, we generated single-cell references for granulopoietic genomic states with linked epitopes1, aligned mutant cells to their wild-type equivalents and identified differentially expressed genes and epigenetic loci. We find that GFI1-target genes are altered sequentially, as cells go through successive states of differentiation. These insights facilitated the genetic rescue of granulocytic specification but not post-commitment defects in innate immune effector function, and underscore the importance of evaluating the effects of mutations and therapy within each relevant cell state.


Subject(s)
Disease Models, Animal , Granulocyte Precursor Cells/pathology , Mutation , Neutropenia/genetics , Neutropenia/pathology , Neutrophils/pathology , Animals , Candida albicans/immunology , Candida albicans/pathogenicity , Cell Lineage , DNA-Binding Proteins/genetics , Female , Humans , Immunity, Innate , Male , Mice , Mice, Transgenic , Neutropenia/congenital , Neutropenia/immunology , Neutrophils/immunology , Transcription Factors/genetics
3.
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
4.
Semin Immunol ; 54: 101515, 2021 04.
Article in English | MEDLINE | ID: mdl-34772606

ABSTRACT

A considerable amount of continuous proliferation and differentiation is required to produce daily a billion new neutrophils in an adult human. Of the few cytokines and factors known to control neutrophil production, G-CSF is the guardian of granulopoiesis. G-CSF/CSF3R signaling involves the recruitment of non-receptor protein tyrosine kinases and their dependent signaling pathways of serine/threonine kinases, tyrosine phosphatases, and lipid second messengers. These pathways converge to activate the families of STAT and C/EBP transcription factors. CSF3R mutations are associated with human disorders of neutrophil production, including severe congenital neutropenia, neutrophilia, and myeloid malignancies. More than three decades after their identification, cloning, and characterization of G-CSF and G-CSF receptor, fundamental questions remain about their physiology.


Subject(s)
Granulocyte Colony-Stimulating Factor , Neutropenia , Adult , Congenital Bone Marrow Failure Syndromes , Granulocyte Colony-Stimulating Factor/metabolism , Hematopoiesis , Humans , Neutropenia/congenital , Neutropenia/genetics , Neutropenia/pathology , Neutrophils/metabolism , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Receptors, Granulocyte Colony-Stimulating Factor/metabolism
5.
Blood ; 139(5): 779-791, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34115842

ABSTRACT

Severe congenital neutropenia is an inborn disorder of granulopoiesis. Approximately one third of cases do not have a known genetic cause. Exome sequencing of 104 persons with congenital neutropenia identified heterozygous missense variants of CLPB (caseinolytic peptidase B) in 5 severe congenital neutropenia cases, with 5 more cases identified through additional sequencing efforts or clinical sequencing. CLPB encodes an adenosine triphosphatase that is implicated in protein folding and mitochondrial function. Prior studies showed that biallelic mutations of CLPB are associated with a syndrome of 3-methylglutaconic aciduria, cataracts, neurologic disease, and variable neutropenia. However, 3-methylglutaconic aciduria was not observed and, other than neutropenia, these clinical features were uncommon in our series. Moreover, the CLPB variants are distinct, consisting of heterozygous variants that cluster near the adenosine triphosphate-binding pocket. Both genetic loss of CLPB and expression of CLPB variants result in impaired granulocytic differentiation of human hematopoietic progenitor cells and increased apoptosis. These CLPB variants associate with wild-type CLPB and inhibit its adenosine triphosphatase and disaggregase activity in a dominant-negative fashion. Finally, expression of CLPB variants is associated with impaired mitochondrial function but does not render cells more sensitive to endoplasmic reticulum stress. Together, these data show that heterozygous CLPB variants are a new and relatively common cause of congenital neutropenia and should be considered in the evaluation of patients with congenital neutropenia.


Subject(s)
Congenital Bone Marrow Failure Syndromes/genetics , Endopeptidase Clp/genetics , Neutropenia/congenital , Cells, Cultured , Endopeptidase Clp/chemistry , Exome , Female , Genetic Variation , Heterozygote , Humans , Infant , Male , Models, Molecular , Mutation , Neutropenia/genetics
6.
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
7.
J Pediatr Hematol Oncol ; 46(2): e199-e201, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38113221

ABSTRACT

Bloom syndrome (BS) is a rare autosomal recessive inherited disorder. Patients with BS have photosensitivity, telangiectatic facial erythema, and stunted growth. They usually have mild microcephaly, and distinctive facial features such as a narrow, slender face, micrognathism, and a prominent nose. Kostmann disease (KD) is a subgroup of severe congenital neutropenias. The diagnosis of severe congenital neutropenia is based on clinical symptoms, bone marrow findings, and genetic mutation. Here, we report a female patient with a triangular face, nasal prominence, and protruding ears presenting with recurrent infections and severe neutropenia. Molecular genetic testing revealed a compound heterozygous variant in the HCLS-1-associated protein X-1 gene [(c.130_131insA) p.(trp44*), c.430 dup(p.Val144fs)] and a new homozygous variant in Bloom Syndrome RecQ like helicase gene [c.2074+2T>C p.(?)]. She was diagnosed with both BS and KD. To the best of our knowledge, this is the first case of coexisting BS and KD in a patient ever reported.


Subject(s)
Bloom Syndrome , Neutropenia , Neutropenia/congenital , Humans , Female , Bloom Syndrome/complications , Bloom Syndrome/genetics , Bloom Syndrome/diagnosis , Congenital Bone Marrow Failure Syndromes , Neutropenia/complications , Neutropenia/genetics , Mutation
8.
Br J Haematol ; 202(2): 393-411, 2023 07.
Article in English | MEDLINE | ID: mdl-37193639

ABSTRACT

HAX1-related congenital neutropenia (HAX1-CN) is a rare autosomal recessive disorder caused by pathogenic variants in the HAX1 gene. HAX1-CN patients suffer from bone marrow failure as assessed by a maturation arrest of the myelopoiesis revealing persistent severe neutropenia from birth. The disorder is strongly associated with severe bacterial infections and a high risk of developing myelodysplastic syndrome or acute myeloid leukaemia. This study aimed to describe the long-term course of the disease, the treatment, outcome and quality of life in patients with homozygous HAX1 mutations reported to the European branch of the Severe Chronic Neutropenia International Registry. We have analysed a total of 72 patients with different types of homozygous (n = 68), compound heterozygous (n = 3), and digenic (n = 1) HAX1 mutations. The cohort includes 56 paediatric (<18 years) and 16 adult patients. All patients were initially treated with G-CSF with a sufficient increase in absolute neutrophil counts. Twelve patients required haematopoietic stem cell transplantation for leukaemia (n = 8) and non-leukaemic indications (n = 4). While previous genotype-phenotype reports documented a striking correlation between two main transcript variants and clinical neurological phenotypes, our current analysis reveals novel mutation subtypes and clinical overlaps between all genotypes including severe secondary manifestations, e.g., high incidence of secondary ovarian insufficiency.


Subject(s)
Neutropenia , Quality of Life , Humans , Proteins/genetics , Mutation , Neutropenia/congenital , Registries , Adaptor Proteins, Signal Transducing/genetics
9.
Blood ; 137(10): 1340-1352, 2021 03 11.
Article in English | MEDLINE | ID: mdl-33227812

ABSTRACT

Heterozygous de novo missense variants of SRP54 were recently identified in patients with congenital neutropenia (CN) who display symptoms that overlap with Shwachman-Diamond syndrome (SDS). Here, we investigate srp54 knockout zebrafish as the first in vivo model of SRP54 deficiency. srp54-/- zebrafish experience embryonic lethality and display multisystemic developmental defects along with severe neutropenia. In contrast, srp54+/- zebrafish are viable, fertile, and show only mild neutropenia. Interestingly, injection of human SRP54 messenger RNAs (mRNAs) that carry mutations observed in patients (T115A, T117Δ, and G226E) aggravated neutropenia and induced pancreatic defects in srp54+/- fish, mimicking the corresponding human clinical phenotypes. These data suggest that the various phenotypes observed in patients may be a result of mutation-specific dominant-negative effects on the functionality of the residual wild-type SRP54 protein. Overexpression of mutated SRP54 also consistently induced neutropenia in wild-type fish and impaired the granulocytic maturation of human promyelocytic HL-60 cells and healthy cord blood-derived CD34+ hematopoietic stem and progenitor cells. Mechanistically, srp54-mutant fish and human cells show impaired unconventional splicing of the transcription factor X-box binding protein 1 (Xbp1). Moreover, xbp1 morphants recapitulate phenotypes observed in srp54 deficiency and, importantly, injection of spliced, but not unspliced, xbp1 mRNA rescues neutropenia in srp54+/- zebrafish. Together, these data indicate that SRP54 is critical for the development of various tissues, with neutrophils reacting most sensitively to the loss of SRP54. The heterogenic phenotypes observed in patients that range from mild CN to SDS-like disease may be the result of different dominant-negative effects of mutated SRP54 proteins on downstream XBP1 splicing, which represents a potential therapeutic target.


Subject(s)
Congenital Bone Marrow Failure Syndromes/genetics , Neutropenia/congenital , Signal Recognition Particle/genetics , X-Box Binding Protein 1/genetics , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Disease Models, Animal , Gene Deletion , Gene Expression Regulation, Developmental , Gene Knockout Techniques , HL-60 Cells , Humans , Models, Molecular , Mutation , Neutropenia/genetics , RNA Splicing , RNA, Messenger/genetics
10.
J Pediatr Hematol Oncol ; 45(4): e441-e445, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36897312

ABSTRACT

Severe congenital neutropenia is a rare disorder. The survival and quality of life of patients radically improved through infection prevention, use of granulocyte colony-stimulating factor, and the appropriate use of antibiotics during infections. The aim of this study was to evaluate the precautions taken by families to prevent infections, the level of knowledge regarding the disease, and the impact of external factors such as education and economic status on behavior and compliance in patients and caregivers in terms of the following treatment protocols. Questionnaires were designed with the aim of determining how the social, cultural, and economic conditions of the families of children with severe congenital neutropenia affected their behavior and knowledge levels. They were completed using one-on-one video interviews with the caregivers. Thirty-one patients from 25 families were enrolled into the study. No correlations between family disease knowledge, parent education levels, working status of the mother, sibling numbers, economic status, ease of hospital access, and/or residential location were found. An increase in disease knowledge of patients and caregivers, as well as proven approaches to living with the disease, would directly correlate to increased life quality and long-term survival rates of patients.


Subject(s)
Health Knowledge, Attitudes, Practice , Neutropenia , Child , Humans , Quality of Life , Neutropenia/congenital , Congenital Bone Marrow Failure Syndromes , Granulocyte Colony-Stimulating Factor
11.
J Pediatr Hematol Oncol ; 45(5): e643-e649, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37053506

ABSTRACT

INTRODUCTION: Severe congenital neutropenia (SCN) is one of the primary immunodeficiency diseases developed by genetic alterations. Mutations in several genes including HAX-1 , G6PC3 , jagunal , and VPS45 account for autosomal recessive SCN. PATIENTS AND METHODS: Patients with SCN registered in the Iranian Primary Immunodeficiency Registry and referred to our clinic at the Children's Medical Center were reviewed. RESULTS: Thirty-seven eligible patients with a mean age of 28.51 ± 24.38 months at the time of diagnosis were included. Nineteen cases had consanguineous parents and 10 cases had confirmed or unconfirmed positive family history. The most prevalent infectious symptoms were oral infections followed by respiratory infections. We identified HAX-1 mutation in 4, ELANE mutation in 4 cases, G6PC3 mutation in 1, and WHIM syndrome in 1 case. Other patients remained genetically unclassified. After the median follow-up of 36 months from the time of diagnosis, the overall survival was 88.88%. The mean event-free survival was 185.84 months (95% CI: 161.02, 210.66). DISCUSSION: Autosomal recessive SCN is more common in countries with high rates of consanguinity like Iran. The genetic classification was possible only for a few patients in our study. This might suggest that there are other autosomal recessive genes causative of neutropenia that have yet to be described.


Subject(s)
Neutropenia , Child , Humans , Infant , Child, Preschool , Iran/epidemiology , Neutropenia/congenital , Mutation , Adaptor Proteins, Signal Transducing/genetics , Disease Progression
12.
BMC Pediatr ; 23(1): 206, 2023 04 29.
Article in English | MEDLINE | ID: mdl-37120535

ABSTRACT

Jagunal homolog 1 (JAGN1) has been recognized as an essential protein in neutrophil function. The mutated JAGN1 is responsible for immunodeficiency related to innate and humoral defense mechanisms. This deficiency impairs neutrophil development and function, leading to recurrent infections and facial dysmorphism as phenotypic consequences of severe congenital neutropenia (SCN). We report two siblings having the reported JAGN1 mutation with different clinical manifestations. Recurrent abscess formation unresponsive to antibiotic therapy, a history of delayed umbilical separation, frequent bacterial or fungal infection, dysmorphic face, failure to thrive, and other coexisting organ abnormalities should prompt physicians to syndromic immunodeficiencies involving neutrophils. Genetic investigations to elucidate the responsible mutation is critical as clinical management varies. Once the diagnosis is confirmed, a multi-disciplinary team should perform further workups to investigate other coexisting malformations and neurodevelopmental evaluation.


Subject(s)
Neutropenia , Humans , Mutation , Neutropenia/genetics , Neutropenia/congenital , Neutrophils/metabolism , Congenital Bone Marrow Failure Syndromes , Membrane Proteins/genetics
13.
BMC Pediatr ; 23(1): 592, 2023 11 23.
Article in English | MEDLINE | ID: mdl-37993852

ABSTRACT

BACKGROUND: Congenital neutropenia is a rare disease. Recurrent infections since young age are the presentation. The most common mutation causing severe congenital neutropenia (SCN) and cyclic neutropenia (CyN) is the ELANE gene. The objectives of this study were to screen the three common genetic mutations of ELANE, HAX1 and GFI1 in children with chronic neutropenia and to describe the clinical characteristics of children who had the mutations. METHODS: Infants having ANC < 1,000/cu mm or children aged > 1 year having ANC < 1,500/cu mm at least 3 times in 3 months were enrolled in the study. Patients who had acquired neutropenia due to infection, immune deficiency, or drugs were excluded. The ELANE gene was first studied; and if mutations were not identified, the HAX1 and GFI1 genes were further examined. RESULTS: A total of 60 patients were enrolled in the study. The median (range) age, ratio of female to male, ANC, and last follow-up age were 9.2 (0.5-45.2) months, 1:1.2, 248 (0-1,101) /cu mm, and 19.9 (3.5-202.3) months, respectively. Infections were noted in 67.3% of all patients. ELANE gene mutation was found in only four patients (6.7%), and the rest (56 patients) showed no mutations in the HAX1 and GFI1 genes. In patients without mutations, 66.0% had normal ANC during the follow-up, with a median (range) age for normal ANC of 19.8 (4.0-60.0) months. Two novel mutations p. Ala79del (c.234_236del) and p. Val197GlufsTer18 (c.589_590insAGGCCGGC) were identified, and they respectively cause SCN and CyN. Patients with the two novel mutations presented with several episodes of infection, including pneumonia, sepsis, abscess, otitis media, and gum infection. CONCLUSION: The genetic screening for ELANE, HAX1, and GFI1 gene mutations in 60 patients with chronic neutropenia could identify four patients (6.7%) with ELANE gene mutation and two novel mutations, p. Ala79del in exon 3 and p. Val197GlufsTer18 in exon 4 causing SCN; and CyN, respectively.


Subject(s)
Leukocyte Elastase , Neutropenia , Infant , Humans , Male , Child , Female , Leukocyte Elastase/genetics , Neutropenia/genetics , Neutropenia/congenital , Mutation , Adaptor Proteins, Signal Transducing/genetics , DNA-Binding Proteins/genetics , Transcription Factors/genetics
14.
Curr Opin Hematol ; 29(1): 27-33, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34854832

ABSTRACT

PURPOSE OF REVIEW: Myeloid diseases are often characterized by a disturbed regulation of myeloid cell proliferation, survival, and maturation. This may either result in a severe paucity of functional neutrophils (neutropenia), an excess production of mature cells (myeloproliferative disorders) or in clonal expansions of dysplastic or immature myeloid cells (myelodysplasia and acute myeloid leukemia). Although these conditions can be regarded as separate entities, caused by the accumulation of distinct sets of somatic gene mutations, it becomes increasingly clear that they may also evolve as the prime consequence of a congenital defect resulting in severe neutropenia. Prominent examples of such conditions include the genetically heterogeneous forms of severe congenital neutropenia (SCN) and Shwachman-Diamond Syndrome. CSF3 treatment is a successful therapy to alleviate neutropenia in the majority of these patients but does not cure the disease nor does it prevent malignant transformation. Allogeneic stem cell transplantation is currently the only therapeutic option to cure SCN, but is relatively cumbersome, e.g., hampered by treatment-related mortality and donor availability. Hence, there is a need for new therapeutic approaches. RECENT FINDINGS: Developments in disease modeling, amongst others based on induced pluripotent stem cell and CRISPR/Cas9 based gene-editing technologies, have created new insights in disease biology and possibilities for treatment. In addition, they are fueling expectations for advanced disease monitoring to prevent malignant transformation. SUMMARY: This review highlights the recent progress made in SCN disease modeling and discusses the challenges that are still ahead of us to gain a better understanding of the biological heterogeneity of the disease and its consequences for patient care.


Subject(s)
Myelodysplastic Syndromes , Neutropenia , Congenital Bone Marrow Failure Syndromes , Humans , Mutation , Neutropenia/congenital , Neutropenia/genetics , Neutropenia/therapy
15.
Scand J Immunol ; 95(4): e13136, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34964150

ABSTRACT

BACKGROUND AND OBJECTIVES: Glucose-6-phosphate catalytic subunit 3 (G6PC3) deficiency is characterized by severe congenital neutropenia with recurrent pyogenic infections, a prominent superficial venous pattern and cardiovascular and urogenital malformations caused by an alteration of glucose homeostasis, with increased endoplasmic reticulum stress and cell apoptosis. METHODS: We reviewed our patients with G6PC3 deficiency diagnosed along the last decade in Mexico; we also searched the PubMed/Medline database for the terms ('G6PC3 deficiency' OR 'Dursun syndrome' OR 'Severe congenital neutropenia type 4'), and selected articles published in English from 2009 to 2020. RESULTS: We found 89 patients reported from at least 14 countries in 4 continents. We describe five new cases from Mexico. Of the 94 patients, 56% are male, 48% from Middle East countries and none of them had adverse reactions to live vaccines; all presented with at least 1 severe infection prior to age 2. Seventy-five per cent had syndromic features, mainly atrial septal defect in 55% and prominent superficial veins in 62%. CONCLUSIONS: With a total of 94 patients reported in the past decade, we delineate the most frequent laboratory and genetic features, their treatment and outcomes, and to expand the knowledge of syndromic and non-syndromic phenotypes in these patients.


Subject(s)
Glucose-6-Phosphatase , Neutropenia , Catalytic Domain , Congenital Bone Marrow Failure Syndromes , Female , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Humans , Male , Neutropenia/congenital , Neutropenia/genetics
16.
Haematologica ; 107(9): 2081-2095, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35295078

ABSTRACT

Prolonged cytopenias are a non-specific sign with a wide differential diagnosis. Among inherited disorders, cytopenias predisposing to leukemia require a timely and accurate diagnosis to ensure appropriate medical management, including adequate monitoring and stem cell transplantation prior to the development of leukemia. We aimed to define the types and prevalences of the genetic causes leading to persistent cytopenias in children. The study comprises children with persistent cytopenias, myelodysplastic syndrome, aplastic anemia, or suspected inherited bone marrow failure syndromes, who were referred for genetic evaluation from all pediatric hematology centers in Israel during 2016-2019. For variant detection, we used Sanger sequencing of commonly mutated genes and a custom-made targeted next-generation sequencing panel covering 226 genes known to be mutated in inherited cytopenias; the minority subsequently underwent whole exome sequencing. In total, 189 children with persistent cytopenias underwent a genetic evaluation. Pathogenic and likely pathogenic variants were identified in 59 patients (31.2%), including 47 with leukemia predisposing syndromes. Most of the latter (32, 68.1%) had inherited bone marrow failure syndromes, nine (19.1%) had inherited thrombocytopenia predisposing to leukemia, and three each (6.4%) had predisposition to myelodysplastic syndrome or congenital neutropenia. Twelve patients had cytopenias with no known leukemia predisposition, including nine children with inherited thrombocytopenia and three with congenital neutropenia. In summary, almost one third of 189 children referred with persistent cytopenias had an underlying inherited disorder; 79.7% of whom had a germline predisposition to leukemia. Precise diagnosis of children with cytopenias should direct follow-up and management programs and may positively impact disease outcome.


Subject(s)
Anemia, Aplastic , Leukemia , Myelodysplastic Syndromes , Neutropenia , Thrombocytopenia , Anemia, Aplastic/genetics , Child , Congenital Bone Marrow Failure Syndromes , Disease Susceptibility , Humans , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Neutropenia/congenital , Neutropenia/genetics , Thrombocytopenia/diagnosis , Thrombocytopenia/genetics
17.
Am J Hematol ; 97(1): 18-29, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34677878

ABSTRACT

Septins play key roles in mammalian cell division and cytokinesis but have not previously been implicated in a germline human disorder. A male infant with severe neutropenia and progressive dysmyelopoiesis with tetraploid myeloid precursors was identified. No known genetic etiologies for neutropenia or bone marrow failure were found. However, next-generation sequencing of germline samples from the patient revealed a novel, de novo germline stop-loss mutation in the X-linked gene SEPT6 that resulted in reduced SEPT6 staining in bone marrow granulocyte precursors and megakaryocytes. Patient skin fibroblast-derived induced pluripotent stem cells (iPSCs) produced reduced myeloid colonies, particularly of the granulocyte lineage. CRISPR/Cas9 knock-in of the patient's mutation or complete knock-out of SEPT6 was not tolerated in non-patient-derived iPSCs or human myeloid cell lines, but SEPT6 knock-out was successful in an erythroid cell line and resulting clones revealed a propensity to multinucleation. In silico analysis predicts that the mutated protein hinders the dimerization of SEPT6 coiled-coils in both parallel and antiparallel arrangements, which could in turn impair filament formation. These data demonstrate a critical role for SEPT6 in chromosomal segregation in myeloid progenitors that can account for the unusual predisposition to aneuploidy and dysmyelopoiesis.


Subject(s)
Genetic Diseases, X-Linked/genetics , Germ-Line Mutation , Myelodysplastic Syndromes/genetics , Neutropenia/congenital , Septins/genetics , Cell Line , Cells, Cultured , Genetic Diseases, X-Linked/complications , Humans , Infant, Newborn , Male , Myelodysplastic Syndromes/complications , Neutropenia/complications , Neutropenia/genetics , Tetraploidy
18.
J Pediatr Hematol Oncol ; 44(2): e431-e433, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34224517

ABSTRACT

Glucose-6-phosphatase catalytic subunit 3 (G6PC3) deficiency is a recently identified form of congenital neutropenia associated with developmental anomalies. The severity of neutropenia and the clinical spectrum are highly variable. Aside from infectious complications and extrahematologic features, inflammatory bowel disease and autoinflammatory complications are less frequently observed manifestations. However, amyloidosis has never been reported in G6PC3 deficiency. Here, we present a 12-year-old patient with incidentally discovered neutropenia because of the p.E65A (c.194A>C) variant of the G6PC3 gene. He had recurrent aphthae and abdominal pain episodes, and developed nephrotic-range proteinuria, amyloidosis, and end-stage renal failure during follow-up.


Subject(s)
Amyloidosis , Neutropenia , Child , Congenital Bone Marrow Failure Syndromes , Glucose-6-Phosphatase/genetics , Humans , Male , Neutropenia/complications , Neutropenia/congenital , Neutropenia/genetics
19.
J Pediatr Hematol Oncol ; 44(1): e62-e67, 2022 01 01.
Article in English | MEDLINE | ID: mdl-33560082

ABSTRACT

Severe congenital neutropenia (SCN) is a rare disease. Autosomal recessive forms of SCN are more frequent in countries where consanguineous marriages are common. In this report, we describe a 54-day-old female with neutropenia who presented with ecthyma gangrenosum. Clinical exome sequencing was used to identify the mutation. HAX1 messenger RNA and isoforms were examined by real-time quantitative and conventional polymerase chain reaction. Bone marrow aspiration was stained by hematoxylin and eosin. Granulocytes were tested for apoptosis upon H2O2 exposure. T-cell proliferation was tested by flow cytometry. Clinical exome sequencing revealed a novel homozygous acceptor splice site mutation in intron 3 of HAX1 (c.505-1G>C), which reduced both isoforms A and B of HAX1 messenger RNA. The Western blot studies showed a complete absence of HAX1 protein. The purified neutrophils from the patient showed increased apoptosis upon H2O2 exposure, whereas T-cell proliferative responses to various stimuli were intact. The patient was treated with combined antibiotics, filgrastim, and placed on antibiotics prophylaxis. To the best of our knowledge, our data provide the first experimental evidence for HAX1 deficiency because of a splice site mutation. Although 3 other splice site variants have been deposited in databases, functional studies were missing. This novel variant of HAX1 may explain the SCN and secondary infections in our patients.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Congenital Bone Marrow Failure Syndromes/genetics , Introns , Mutation , Neutropenia/congenital , RNA Splice Sites , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Congenital Bone Marrow Failure Syndromes/metabolism , Humans , Hydrogen Peroxide/pharmacology , Infant , Male , Neutropenia/genetics , Neutropenia/metabolism , T-Lymphocytes/metabolism
20.
ScientificWorldJournal ; 2022: 3356835, 2022.
Article in English | MEDLINE | ID: mdl-35571273

ABSTRACT

Single-nucleotide polymorphisms in the ELANE (Elastase, Neutrophil Expressed) gene are associated with severe congenital neutropenia, while the ELANE gene provides instructions for making a protein called neutrophil elastase. We identified disease susceptibility single-nucleotide polymorphisms (SNPs) in the ELANE gene using several computational tools. We used cutting-edge computational techniques to investigate the effects of ELANE mutations on the sequence and structure of the protein. Our study suggested that eight nsSNPs (rs28931611, rs57246956, rs137854448, rs193141883, rs201723157, rs201139487, rs137854451, and rs200384291) are the most deleterious in ELANE gene and disturb protein structure and function. The mutants F218L, R34W, G203S, R193W, and T175M have not yet been identified in patients suffering from SCN and cyclic hematopoiesis, while C71Y, P139R, C151Y, G214R, and G203C reported in our study are already associated with both of the disorders. These mutations are shown to destabilize structure and disrupt ELANE protein activation, splicing, and folding and might diminish trypsin-like serine protease efficiency. Prediction of posttranslation modifications highlighted the significance of deleterious nsSNPs because some of nsSNPs affect potential phosphorylation sites. Gene-gene interactions showed the relation of ELANE with other genes depicting its importance in numerous pathways and coexpressions. We identified the deleterious nsSNPs, constructed mutant protein structures, and evaluated the impact of mutation by employing molecular docking. This research sheds light on how ELANE failure upon mutation results in disease progression, including congenital neutropenia, and validation of these novel predicted nsSNPs is required through the wet lab.


Subject(s)
Polymorphism, Single Nucleotide , Humans , Congenital Bone Marrow Failure Syndromes , Molecular Docking Simulation , Mutation , Neutropenia/congenital , Polymorphism, Single Nucleotide/genetics
SELECTION OF CITATIONS
SEARCH DETAIL