Skip to main content
Erschienen in: BMC Pediatrics 1/2024

Open Access 01.12.2024 | Case Report

A novel missense mutation in the MECOM gene in a Chinese boy with radioulnar synostosis with amegakaryocytic thrombocytopenia

verfasst von: Duowen Huang, Mingyan Jiang, Yiping Zhu, Dongjun Li, Xiaoxi Lu, Ju Gao

Erschienen in: BMC Pediatrics | Ausgabe 1/2024

Abstract

Radioulnar synostosis with amegakaryocytic thrombocytopenia (RUSAT) type 2, caused by MDS1 and EVI1 complex locus (MECOM) gene mutations, is a rare inherited bone marrow failure syndrome (IBMFS) with skeletal anomalies, characterized by varying presentation of congenital thrombocytopenia (progressing to pancytopenia), bilateral proximal radioulnar synostosis, and other skeletal abnormalities. Due to limited knowledge and heterogenous manifestations, clinical diagnosis of the disease is challenging. Here we reported a novel MECOM mutation in a Chinese boy with typical clinical features for RUSAT-2. Trio-based whole exome sequencing of buccal swab revealed a novel heterozygous missense mutation in exon 11 of the MECOM gene (chr3:168818673; NM_001105078.3:c.2285G > A). The results strongly suggest that the variant was a germline mutation and disease-causing mutation. The patient received matched unrelated donor hematopoetic stem cell transplantation (HSCT). This finding was not only expanded the pathogenic mutation spectrum of MECOM gene, but also provided key information for clinical diagnosis and treatment of RUSAT-2.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12887-024-04552-1.
Duowen Huang and Mingyan Jiang contirbuted equally.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Radioulnar synostosis with amegakaryocytic thrombocytopenia (RUSAT [MIM: 605,432]), first described by Nielsen and colleagues in 2012 [1], is a rare inherited bone marrow failure syndrome (IBMFS) with a continuous spectrum of clinical phenotype from isolated proximal radioulnar synostosis to severe bone marrow failure without skeletal abnormalities in the patients and affected relatives [2]. Some of the patients were complicated with renal and cardiac malformations, and hearing loss [3, 4].
So far, about 64 variants have been reported, with 5 reported Chinese individuals and family members in the literature reviewed by Voit RA, et al. [5]. Type 2 of RUSAT (RUSAT-2; MIM: 616,738) is caused by a mutation in the MECOM gene (MIM: 165,215) on chromosome 3q26. MECOM is an abbreviation of MDS1 (myelodysplasia syndrome 1) and EVI1 (ecotropic viral integration 1 site) complex locus, which encodes a 1051 amino acid protein and contains 10 zinc finger motifs and its isoforms act as transcriptional factors [2, 6, 7]. RUSAT-2 is not an easily recognizable syndrome due to the limited knowledge of reported cases and the heterozygous clinical manifestations, especially in the cases without bone abnormalities. To our knowledge, all reported RUSAT-2 cases were diagnosed through next generation sequencing and most of them received HSCT.
In the present study, we describe a 1-year-3-month-old Chinese boy with a novel heterozygous missense variant in the MECOM gene identified by next generation sequencing and treated by HSCT. In addition, the genetic and phenotypic spectrum and the outcome of reported RUSAT-2 cases are reviewed and summarized.

Case report

The reported patient was a 1-year-3-month-old Chinese boy. He was referred to our hospital due to petechiae, pallor, and refractory fever for one year. He was the first born of a non-consanguineous Chinese couple, and there was no family history of hematological disorders. The pregnancy and delivery at 39 wk gestation were uneventful. At birth, the weight was 3200 g, the length was 49 cm, and the occipitofrontal circumference (OFC) was 32.5 cm. His hearing and psychomotor development were normal. He did not have facial dysmorphism, microcephaly or organomegaly, but had bilateral clinodactyly of the digits without abnormalities of bones. Bilateral forearms were limited to abduction and internal rotation (Fig. 1).
For the proband, regular laboratory tests were performed. Complete blood count revealed pancytopenia (minimum value of neutrophil count was 0.07 × 109/L (normal reference range 0.8 × 109/L-5.8 × 109/L), minimum value of hemoglobin was 17 g/L (normal reference range 110 g/L-160 g/L), minimum value of platelet count was 1 × 109/L (normal reference range 100 × 109/L-300 × 109/L), reticulocyte count of 1.9%). His liver and kidney function, electrolytes, myocardial enzymes, thyroid function, erythrocyte sedimentation rate (ESR), hemoglobin electrophoresis, and coagulation profile were normal. The virus infectious screening, Coombs’ test, and autoimmune antibody were negative. Bone marrow smear revealed hypocellular, megacaryophthisis. Chromosomal breakage study was normal. Single cell gel electrophoresis (SCGE) revealed damage of lymphocytes in peripheral blood, and the rate of cometal cells was 22% (normal range < 21%). His CD19 positive B-cell count was 0.09 × 109/L (normal range 0.20–2.10 × 109/L). The karyotype was 46, XY.
The X-ray of both forearms showed superior radioulnar synostosis. The X-ray of both hands and feet showed clinodactyly and brachydactyly (Fig. 1). Computerized tomography (CT) of brain revealed subarachnoid hemorrhage. There was no abnormality in his CT of chest and abdomen, echocardiography, ultrasonic test of urinary system, or hearing screening.
We performed trio-based whole-exome sequencing (WES) on the buccal swab and peripheral blood of the patient and peripheral blood of his unaffected parents. Genomic DNA was extracted from buccal swab and peripheral blood. The captured DNA fragments were then sequenced at the Chigene Translational Medical Research Center (Beijing, China). The sequence variants were functionally annotated and filtered using known populations and databases, including 1000 genomes, Signle Nucleotide Polymorphism Database, Genome Aggregation Database, Clin Var, Human Gene Mutation Database, and Online Menedelian Inheritance in Man. Candidate variants were then evaluated in the context of clinical presentation and inheritance mode. The trio-based WES revealed a heterozygous missense variant c.2285G > A in exon 11 of the MECOM gene (NM_001105078.3) in the proband, predicted to result in the amino acid substitution of arginine for lysine at codon 762 (p.Arg762Lys). The variation was not reported previously. No variation was detected at this site in his parents, which was subsequently validated by Sanger sequencing (Fig. 2). The de novo variant was not observed in public variant databases. This novel de novo variant was classified as “likely pathogenic” according to the American College of Medical Genetics and Genomics (ACMG) standards and guidelines [8].
He was diagnosed as RUSAT-2, and treated with intravenous immunoglobulin, methylprednisolone, antibiotics, platelet and packed red cells products. The petechiae and intracranial bleeding disappeared, but he still had pancytopenia. Follow up at 1 year revealed persisting anemia with severe neutropenia and thrombocytopenia. He had been treated with androgen for one month and intermittently supported with blood products. But he was dependent on transfusion, so he received matched unrelated donor hematopoetic stem cell transplantation (HSCT) at the age of 1 year 4 months. After transplantation, the rate of transfusion was decreased with less frequent infection. The last follow-up was at 4 years old. The complete blood count after HSCT was normal (neutrophil count 3.82 × 109/L, hemoglobin 122 g/L, platelet was 317 × 109/L).

Discussion

RUSAT-2 is a rare autosomal dominant bone marrow failure syndrome (IBMFS) caused by mutations in the MECOM gene. As reported, the clinical manifestations of the cases with MECOM mutations presented a continuous spectrum from isolated radioulnar synostosis to severe bone marrow failure without skeletal abnormalities, which are named as MECOM-associated syndromes [2, 9]. Radioulnar synostosis was observed in 45.3% of patients with MECOM mutations reported as RUSAT-2. The other relatively prevalent features, as observed in 42.2% of reported RUSAT-2 cases, were other skeletal malformation, including hypoplasia of middle and end phalanx D5, Toe malposition D2, Thumb under D2. Pancytopenia was the dominant hematologic abnormality (36/64, 56.2%), though 8/64 (12.5%) patients had no cytopenia, and 16/64 (25.0%) patients had thrombocytopenia. Additionally, other malformations such as cardiac malformations (27/64, 26.6%), hearing impairment (9/64, 14.1%), nail, or facial abnormalities (15/64, 23.4%), renal (6/64, 9.4%) and neurological disorder (11/64, 17.2%), and precocious puberty (4/64, 6.3%) have been reported in patients with MECOM mutations, but were not seen in our patient (Supplementary Table 1) [14, 7, 923]. Notably, our patient had consulted an orthopedist for bilateral clinodactyly of the digits. It has been implied that some patients with RUSAT-2 may be initially misdiagnosed as isolated orthopedic disorders. Although the CT brain revealed subarachnoid hemorrhage, the patient had no neurological abnormalities. We speculated that the hemorrhage was caused by severe thrombocytopenia. Interestingly, immune dysfunction, such as decreased B cell and hypogammaglobulinemia had been observed in some patients (10/64, 15.6%) (Supplementary Table 1) [14, 7, 923]. It seems that MECOM is involved in critical pathways for regulation of regenerative hematopoiesis and B-cell differentiation. RUS (radioulnar synostosis) and B-cell lymphopenia has been observed only in patients with mutations affecting a short region in the C-terminal zinc finger domain of EVI1 [2]. But it is not clear whether B-cell deficiency in MECOM-associated disease is due to a common stem-cell defect or to the specific involvement of MECOM in B-cell development. A specific role in B-cell development is suggested by the fact that a gene amplification of MECOM seems to play a role in persistent polyclonal binucleated B-cell lymphocytosis [24].
Following a thorough review of all reported RUSAT-2 cases, we considered the remarkable features distinct from RUSAT-2 were proximal radioulnar synostosis and thrombocytopenia. However, because the thrombocytopenia is nonspecific and skeletal abnormalities are insidious, and the total number of reported cases is limited, WES technology is increasingly used to identify the pathogenesis and establish a definite diagnosis. We performed trio-based WES and identified a novel de novo missense mutation (NM_001105078.3:c.2285G > A, p.Arg762Lys) of the MECOM gene in our patient. The variant was classified as “likely pathogenic” according to the ACMG criteria, supporting a genetic diagnosis of RUSAT-2 for the proband, with main complaints of pancytopenia and forearms limited movement.
Together with the reported cases, 64 different MECOM variants in 64 unrelated individuals and family members with RUSAT-2 were known. Of the detected pathogenic or likely pathogenic mutations, 6 (9.4%) cases were nonsense mutations, 5 (7.8%) cases were splice mutations, 8 (12.5%) cases were deletion, 6 (9.4%) cases were frameshift mutations, and 35 (54.7%) cases were missense mutations, while 4 patients were unknown the mutation types [14, 7, 923]. The majority of the reported mutations were missense mutations. The region coding for the 10 zinc finger domains are the mutational hotspots. The case described here presented a variant in exon 11 leading to an amino acid exchange at the 9th zinc finger.
As previously reported, the MECOM gene contains 16 exons and encodes a 1051 amino-acid protein which is a zinc finger transcription factor called EVI1, a transcription factor involved in embryogenesis, homeostasis of the hematopoietic stem cell compartment and megakaryocyte differentiation [3] (Fig. 3). Missense pathogenetic variants of EVI1 may reduce its interaction with DNA and/or other transcription factors [25]. Another report suggested MECOM being a candidate gene for hereditary hematological malignancies [26]. The novel variant (c.2285G > A, p.Arg762Lys) detected in our patient’s buccal swab and peripheral blood, and the same mutation was not detected in the parents who were asymptomatic and with normal blood counts. The results strongly suggest that the variant was a germ line mutation and disease-causing mutation. The novel de novo variant of MECOM gene is located in exon 11. Another research reported a splicing variant (NM_001105078:c.2285 + 1G > A) which mutated at the same site. So we speculated that both of the variants induced transcriptional dysregulation, similar to that caused by missense variants at the 8th or 9th ZF motif according to the functional analysis [17]. To clarify whether haploinsufficiency or a dominant-negative effect of the novel variant may be a causative mechanism of RUSAT-2 will require further functional studies (Fig. 3). More cases of RUSAT-2 from different ethnic populations are required for this to be validated.
Definitive treatment is a hematopoetic stem cell transplantation. Of the 38 reported patients with MECOM-associated syndrome transplanted, 34 engrafted while four died of transplant-related complications [14, 7, 923].
In conclusion, we reported a novel de novo missense mutation in (c.2285G > A, p.Arg762Lys) in the MECOM gene in a Chinese boy with RUSAT-2. This finding not only contributed to better genetic counseling, but also expanded the pathogenic mutation spectrum of MECOM gene. We also highlight the genetic analysis in all IBMFS and HSCT is the definitive treatment. Further studies are required to elucidate the molecular pathology underlying the development of RUSAT.

Acknowledgements

The authors would like to acknowledge the patient and his family who fight to overcome disease and allow us to collect much needed information about the causes and treatment of RUSAT. They also acknowledge the nurses who had taken care of the child in HSCT period.

Declarations

Written informed consent was obtained from the parents for the publication of any potentially identifiable images or data included in this article. The study was approved by the Ethics Committee of West China Second University Hospital and complied with the guidelines outlined in the declaration of Helsinki were followed.
N/A.

Conflict of interest

The authors declare that there is no conflict of interest.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Nielsen M, Vermont C, Aten E, et al. Deletion of the 3q26 region including the EVI1 and MDS1 genes in a neonate with congenital thrombocytopenia and subsequent aplastic anaemia. J Med Genet. 2012;49(9):598–600.CrossRefPubMed Nielsen M, Vermont C, Aten E, et al. Deletion of the 3q26 region including the EVI1 and MDS1 genes in a neonate with congenital thrombocytopenia and subsequent aplastic anaemia. J Med Genet. 2012;49(9):598–600.CrossRefPubMed
2.
Zurück zum Zitat Germeshausen M, Ancliff P, Estrada J, et al. MECOM-associated syndrome: a heterogeneous inherited bone marrow failure syn- drome with amegakaryocytic thrombocytopenia. Blood Adv. 2018;2:586–96.CrossRefPubMedPubMedCentral Germeshausen M, Ancliff P, Estrada J, et al. MECOM-associated syndrome: a heterogeneous inherited bone marrow failure syn- drome with amegakaryocytic thrombocytopenia. Blood Adv. 2018;2:586–96.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Niihori T, Ouchi-Uchiyama M, Sasahara Y, et al. Mutations in MECOM, encodingoncoprotein EVI1, cause radioulnar synostosis with amegakaryocytic thrombocytopenia. Am J Hum Genet. 2015;97:848–54.CrossRefPubMedPubMedCentral Niihori T, Ouchi-Uchiyama M, Sasahara Y, et al. Mutations in MECOM, encodingoncoprotein EVI1, cause radioulnar synostosis with amegakaryocytic thrombocytopenia. Am J Hum Genet. 2015;97:848–54.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Bluteau O, Sebert M, Leblanc T, et al. A landscape of germline mutations in a cohort of inherited bone marrow failure patients. Blood. 2018;131:717–32.CrossRefPubMed Bluteau O, Sebert M, Leblanc T, et al. A landscape of germline mutations in a cohort of inherited bone marrow failure patients. Blood. 2018;131:717–32.CrossRefPubMed
5.
Zurück zum Zitat Voit RA, Sankaran VG. MECOM deficiency: from bone marrow failure to impaired B-cell development. J Clin Immunol. 2023;43(6):1052–66.CrossRefPubMed Voit RA, Sankaran VG. MECOM deficiency: from bone marrow failure to impaired B-cell development. J Clin Immunol. 2023;43(6):1052–66.CrossRefPubMed
6.
Zurück zum Zitat Thompson AA, Woodruff K, Feig SA, et al. Congenital thrombocytopenia and radio ulnar synostosis: a new familial syndrome. Br J Haematol. 2001;113:866–70.CrossRefPubMed Thompson AA, Woodruff K, Feig SA, et al. Congenital thrombocytopenia and radio ulnar synostosis: a new familial syndrome. Br J Haematol. 2001;113:866–70.CrossRefPubMed
7.
Zurück zum Zitat Osumi T, Tsujimoto S, Nakabayashi K, et al. Somatic MECOM mosaicism in a patient with congenital bone marrow failure without a radial abnormality. Pediatr Blood Cancer. 2018;65(6):e26959.CrossRefPubMed Osumi T, Tsujimoto S, Nakabayashi K, et al. Somatic MECOM mosaicism in a patient with congenital bone marrow failure without a radial abnormality. Pediatr Blood Cancer. 2018;65(6):e26959.CrossRefPubMed
8.
Zurück zum Zitat Richards S, Aziz N, Bale S, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–24.CrossRefPubMedPubMedCentral Richards S, Aziz N, Bale S, et al. Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med. 2015;17(5):405–24.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Chinga M, Bertuch A, Afify Z, et al. Am J Med Genet A. 2023;191(7):1826–35.CrossRef Chinga M, Bertuch A, Afify Z, et al. Am J Med Genet A. 2023;191(7):1826–35.CrossRef
10.
Zurück zum Zitat Lord SV, Jimenez JE, Kroeger ZA, et al. A MECOM variant in an African American child with radioulnar synostosis and thrombocytopenia. Clin Dysmorphol. 2018;27(1):9–11.CrossRefPubMed Lord SV, Jimenez JE, Kroeger ZA, et al. A MECOM variant in an African American child with radioulnar synostosis and thrombocytopenia. Clin Dysmorphol. 2018;27(1):9–11.CrossRefPubMed
11.
Zurück zum Zitat Kjeldsen E, Veigaard C, Aggerholm A, Hasle H. Congenital hypoplastic bone marrow failure associated with a de novo partial deletion of the MECOM gene at 3q26.2. Gene. 2018;656:86–94.CrossRefPubMed Kjeldsen E, Veigaard C, Aggerholm A, Hasle H. Congenital hypoplastic bone marrow failure associated with a de novo partial deletion of the MECOM gene at 3q26.2. Gene. 2018;656:86–94.CrossRefPubMed
12.
Zurück zum Zitat Ripperger T, Hofmann W, Koch JC, Steinemann D, et al. MDS1 and EVI1 complex locus (MECOM): a novel candidate gene for hereditary hematological malignancies. Haematologica. 2018;103(2):e55–8.CrossRefPubMedPubMedCentral Ripperger T, Hofmann W, Koch JC, Steinemann D, et al. MDS1 and EVI1 complex locus (MECOM): a novel candidate gene for hereditary hematological malignancies. Haematologica. 2018;103(2):e55–8.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Loganathan A, Munirathnam D, Ravikumar T. A novel mutation in the MECOM gene causing radioulnar synostosis with amegakaryocytic thrombocytopenia (RUSAT-2) in an infant. Pediatr Blood Cancer. 2019;66(4):e27574.CrossRefPubMed Loganathan A, Munirathnam D, Ravikumar T. A novel mutation in the MECOM gene causing radioulnar synostosis with amegakaryocytic thrombocytopenia (RUSAT-2) in an infant. Pediatr Blood Cancer. 2019;66(4):e27574.CrossRefPubMed
14.
Zurück zum Zitat Bouman A, Knegt L, Gröschel S, et al. Congenital thrombocytopenia in a neonate with an interstitial microdeletion of 3q26.2q26.31. Am J Med Genet A. 2016;170A(2):504–9.CrossRefPubMed Bouman A, Knegt L, Gröschel S, et al. Congenital thrombocytopenia in a neonate with an interstitial microdeletion of 3q26.2q26.31. Am J Med Genet A. 2016;170A(2):504–9.CrossRefPubMed
15.
Zurück zum Zitat Veken L, Maiburg M, Groenendaal F, et al. Lethal neonatal bone marrow failure syndrome with multiple congenital abnormalities, including limb defects, due to a constitutional deletion of 3’ MECOM. Haematologica. 2018;103(4):e173–6.CrossRefPubMedPubMedCentral Veken L, Maiburg M, Groenendaal F, et al. Lethal neonatal bone marrow failure syndrome with multiple congenital abnormalities, including limb defects, due to a constitutional deletion of 3’ MECOM. Haematologica. 2018;103(4):e173–6.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Weizmann D, Pincez T, Roussy M, et al. New MECOM variant in a child with severe neonatal cytopenias spontaneously resolving. Pediatr Blood Cancer. 2020;67(5):e28215.CrossRefPubMed Weizmann D, Pincez T, Roussy M, et al. New MECOM variant in a child with severe neonatal cytopenias spontaneously resolving. Pediatr Blood Cancer. 2020;67(5):e28215.CrossRefPubMed
17.
Zurück zum Zitat Niihori T, Tanoshim R, Sasahara Y, et al. Phenotypic heterogeneity in individuals with MECOM variants in 2 families. Blood Adv. 2022;6(18):5257–61.CrossRefPubMedPubMedCentral Niihori T, Tanoshim R, Sasahara Y, et al. Phenotypic heterogeneity in individuals with MECOM variants in 2 families. Blood Adv. 2022;6(18):5257–61.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Deliloğlu B, Tüfekçi Ö, Tüzün F, et al. A novel mecom gene mutation associated with amegakaryocytic thrombocytopenia in a premature infant. Turk J Pediatr. 2022;64(4):736–40.CrossRefPubMed Deliloğlu B, Tüfekçi Ö, Tüzün F, et al. A novel mecom gene mutation associated with amegakaryocytic thrombocytopenia in a premature infant. Turk J Pediatr. 2022;64(4):736–40.CrossRefPubMed
19.
Zurück zum Zitat Wall E, Forsyth J, Kinning E, et al. Fetal hydrops caused by a novel pathogenic MECOM variant. Prenat Diagn. 2023;43(6):717–20.CrossRefPubMed Wall E, Forsyth J, Kinning E, et al. Fetal hydrops caused by a novel pathogenic MECOM variant. Prenat Diagn. 2023;43(6):717–20.CrossRefPubMed
20.
Zurück zum Zitat Walne A, Tummala H, Ellison A, et al. Expanding the phenotypic and genetic spectrum of radioulnar synostosis associated hematological disease. Haematologica. 2018;103(7):e284–7.CrossRefPubMedPubMedCentral Walne A, Tummala H, Ellison A, et al. Expanding the phenotypic and genetic spectrum of radioulnar synostosis associated hematological disease. Haematologica. 2018;103(7):e284–7.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Al-Abboh H, Zahra A, Adekile A. A novel MECOM variant associated with congenital amegakaryocytic thrombocytopenia and radioulnar synostosis. Pediatr Blood Cancer. 2022;69(12):e29761.CrossRefPubMed Al-Abboh H, Zahra A, Adekile A. A novel MECOM variant associated with congenital amegakaryocytic thrombocytopenia and radioulnar synostosis. Pediatr Blood Cancer. 2022;69(12):e29761.CrossRefPubMed
22.
Zurück zum Zitat Shen F, Yang Y, Zheng Y, et al. MECOM-related disorder: radioulnar synostosis without hematological aberration due to unique variants. Genet Med. 2022;24(5):1139–47.CrossRefPubMed Shen F, Yang Y, Zheng Y, et al. MECOM-related disorder: radioulnar synostosis without hematological aberration due to unique variants. Genet Med. 2022;24(5):1139–47.CrossRefPubMed
23.
Zurück zum Zitat Datta SS, Basu S, Ghara N, et al. Utility of platelet cross-matching in a case of neonatal alloimmunue thrombocytopenia associated with a de novo MECOM variant. Blood Res. 2021;56(1):53–6.CrossRefPubMedPubMedCentral Datta SS, Basu S, Ghara N, et al. Utility of platelet cross-matching in a case of neonatal alloimmunue thrombocytopenia associated with a de novo MECOM variant. Blood Res. 2021;56(1):53–6.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Cornet E, Mossafa H, Courel K, et al. Persistent polyclonal binucleated B-cell lymphocytosis and MECOM gene amplification. BMC Res Notes. 2016;9:138.CrossRefPubMedPubMedCentral Cornet E, Mossafa H, Courel K, et al. Persistent polyclonal binucleated B-cell lymphocytosis and MECOM gene amplification. BMC Res Notes. 2016;9:138.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Noris P, Pecci A. Hereditary thrombocytopenias: a growing list of disorders. Hematol Am Soc Hematol Educ Program. 2017;1:385–99.CrossRef Noris P, Pecci A. Hereditary thrombocytopenias: a growing list of disorders. Hematol Am Soc Hematol Educ Program. 2017;1:385–99.CrossRef
26.
Zurück zum Zitat Ripperger T, Hofmann W, Koch JC, et al. MDS1 and EVI1 complex locus(MECOM): a novel candidate gene for hereditary hematological malignancies. Haematologica. 2018;103:e55–8.CrossRefPubMedPubMedCentral Ripperger T, Hofmann W, Koch JC, et al. MDS1 and EVI1 complex locus(MECOM): a novel candidate gene for hereditary hematological malignancies. Haematologica. 2018;103:e55–8.CrossRefPubMedPubMedCentral
Metadaten
Titel
A novel missense mutation in the MECOM gene in a Chinese boy with radioulnar synostosis with amegakaryocytic thrombocytopenia
verfasst von
Duowen Huang
Mingyan Jiang
Yiping Zhu
Dongjun Li
Xiaoxi Lu
Ju Gao
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
BMC Pediatrics / Ausgabe 1/2024
Elektronische ISSN: 1471-2431
DOI
https://doi.org/10.1186/s12887-024-04552-1

Weitere Artikel der Ausgabe 1/2024

BMC Pediatrics 1/2024 Zur Ausgabe

Ähnliche Überlebensraten nach Reanimation während des Transports bzw. vor Ort

29.05.2024 Reanimation im Kindesalter Nachrichten

Laut einer Studie aus den USA und Kanada scheint es bei der Reanimation von Kindern außerhalb einer Klinik keinen Unterschied für das Überleben zu machen, ob die Wiederbelebungsmaßnahmen während des Transports in die Klinik stattfinden oder vor Ort ausgeführt werden. Jedoch gibt es dabei einige Einschränkungen und eine wichtige Ausnahme.

Alter der Mutter beeinflusst Risiko für kongenitale Anomalie

28.05.2024 Kinder- und Jugendgynäkologie Nachrichten

Welchen Einfluss das Alter ihrer Mutter auf das Risiko hat, dass Kinder mit nicht chromosomal bedingter Malformation zur Welt kommen, hat eine ungarische Studie untersucht. Sie zeigt: Nicht nur fortgeschrittenes Alter ist riskant.

Begünstigt Bettruhe der Mutter doch das fetale Wachstum?

Ob ungeborene Kinder, die kleiner als die meisten Gleichaltrigen sind, schneller wachsen, wenn die Mutter sich mehr ausruht, wird diskutiert. Die Ergebnisse einer US-Studie sprechen dafür.

Bei Amblyopie früher abkleben als bisher empfohlen?

22.05.2024 Fehlsichtigkeit Nachrichten

Bei Amblyopie ist das frühzeitige Abkleben des kontralateralen Auges in den meisten Fällen wohl effektiver als der Therapiestandard mit zunächst mehrmonatigem Brilletragen.

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.