Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Haematologica ; 109(2): 422-430, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-37584291

RESUMO

Monosomy 7 is the most common cytogenetic abnormality in pediatric myelodysplastic syndrome (MDS) and associated with a high risk of disease progression. However, in young children, spontaneous loss of monosomy 7 with concomitant hematologic recovery has been described, especially in the presence of germline mutations in SAMD9 and SAMD9L genes. Here, we report on our experience of close surveillance instead of upfront hematopoietic stem cell transplantation (HSCT) in seven patients diagnosed with SAMD9L syndrome and monosomy 7 at a median age of 0.6 years (range, 0.4-2.9). Within 14 months from diagnosis, three children experienced spontaneous hematological remission accompanied by a decrease in monosomy 7 clone size. Subclones with somatic SAMD9L mutations in cis were identified in five patients, three of whom attained hematological remission. Two patients acquired RUNX1 and EZH2 mutations during the observation period, of whom one progressed to myelodysplastic syndrome with excess of blasts (MDS-EB). Four patients underwent allogeneic HSCT at a median time of 26 months (range, 14-40) from diagnosis for MDSEB, necrotizing granulomatous lymphadenitis, persistent monosomy 7, and severe neutropenia. At last follow-up, six patients were alive, while one passed away due to transplant-related causes. These data confirm previous observations that monosomy 7 can be transient in young children with SAMD9L syndrome. However, they also indicate that delaying HSCT poses a substantial risk of severe infection and disease progression. Finally, surveillance of patients with SAMD9L syndrome and monosomy 7 is critical to define the evolving genetic landscape and to determine the appropriate timing of HSCT (clinicaltrials gov. Identifier: NCT00662090).


Assuntos
Deleção Cromossômica , Síndromes Mielodisplásicas , Humanos , Criança , Pré-Escolar , Lactente , Remissão Espontânea , Síndromes Mielodisplásicas/diagnóstico , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/terapia , Progressão da Doença , Fatores de Transcrição/genética , Monossomia , Cromossomos Humanos Par 7/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética
2.
J Pediatr Hematol Oncol ; 45(8): e1010-e1013, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37703450

RESUMO

Paroxysmal cold hemoglobinuria (PCH) is a rare condition in childhood characterized by complement-mediated premature destruction of red blood cells. PCH is associated with intravascular hemolysis causing hemoglobinuria, which may result in acute kidney injury of various severity. We aimed to retrospectively analyze clinical and laboratory features of children with PCH-associated acute kidney injury received at tertiary Pediatric Hematology and Nephrology Center, University Motol Hospital, Prague, Czech Republic during the period 2016 to 2022. We present here 3 children with PCH-associated acute kidney failure requiring renal replacement therapy. We highlight the association of PCH with kidney disease. Renal parameters and urine examination should be regularly tested in all children with PCH.


Assuntos
Injúria Renal Aguda , Hemoglobinúria Paroxística , Humanos , Criança , Hemoglobinúria Paroxística/complicações , Hemoglobinúria Paroxística/diagnóstico , Estudos Retrospectivos , Eritrócitos , Injúria Renal Aguda/complicações , Hemólise , Temperatura Baixa
3.
Ital J Pediatr ; 49(1): 11, 2023 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-36658659

RESUMO

BACKGROUND: Schimke immunoosseous dysplasia (SIOD) is an ultra-rare inherited disease affecting many organ systems. Spondyloepiphyseal dysplasia, T-cell immunodeficiency and steroid resistant nephrotic syndrome are the main symptoms of this disease. CASE PRESENTATION: We aimed to characterize the clinical, pathological and genetic features of SIOD patients received at tertiary Pediatric Nephrology Center, University Hospital Motol, Prague, Czech Republic during the period 2001-2021. The mean age at diagnosis was 21 months (range 18-48 months). All patients presented with growth failure, nephropathy and immunodeficiency. Infections and neurologic complications were present in most of the affected children during the course of the disease. CONCLUSIONS: Although SIOD is a disease characterized by specific features, the individual phenotype may differ. Neurologic signs can severely affect the quality of life; the view on the management of SIOD is not uniform. Currently, new therapeutic methods are required.


Assuntos
Síndromes de Imunodeficiência , Síndrome Nefrótica , Osteocondrodisplasias , Humanos , Síndrome Nefrótica/diagnóstico , Síndrome Nefrótica/genética , Síndrome Nefrótica/complicações , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/genética , Osteocondrodisplasias/terapia , Centros de Atenção Terciária , República Tcheca , Qualidade de Vida , Doenças Raras , Síndromes de Imunodeficiência/diagnóstico , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/complicações
4.
Blood Cells Mol Dis ; 81: 102380, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31855845

RESUMO

Diamond-Blackfan anemia (DBA) is a rare congenital erythroid aplasia, underlied by haploinsufficient mutations in genes coding for ribosomal proteins (RP) in approximately 70% of cases. DBA is frequently associated with somatic malformations, endocrine dysfunction and with an increased predisposition to cancer. Here we present clinical and genetic characteristics of 62 patients from 52 families enrolled in the Czech and Slovak DBA Registry. Whole exome sequencing (WES) and array comparative genomic hybridization (aCGH) were employed to identify causative mutations in newly diagnosed patients and in cases with previously unrecognized molecular pathology. RP mutation detection rate was 81% (50/62 patients). This included 8 novel point mutations and 4 large deletions encompassing some of the RP genes. Malignant or predisposing condition developed in 8/62 patients (13%): myelodysplastic syndrome in 3 patients; breast cancer in 2 patients; colorectal cancer plus ocular tumor, diffuse large B-cell lymphoma and multiple myeloma each in one case. These patients exclusively harbored RPL5, RPL11 or RPS19 mutations. Array CGH is beneficial for detection of novel mutations in DBA due to its capacity to detect larger chromosomal aberrations. Despite the importance of genotype-phenotype correlation in DBA, phenotypic differences among family members harboring an identical mutation were observed.


Assuntos
Anemia de Diamond-Blackfan/genética , Mutação , Proteínas Ribossômicas/genética , Anemia de Diamond-Blackfan/complicações , Anemia de Diamond-Blackfan/epidemiologia , Hibridização Genômica Comparativa , República Tcheca , Família , Estudos de Associação Genética , Humanos , Neoplasias/etiologia , Sistema de Registros , Eslováquia , Sequenciamento do Exoma
5.
Haematologica ; 101(6): 707-16, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27013649

RESUMO

GATA-2 deficiency was recently described as common cause of overlapping syndromes of immunodeficiency, lymphedema, familiar myelodysplastic syndrome or acute myeloid leukemia. The aim of our study was to analyze bone marrow and peripheral blood samples of children with myelodysplastic syndrome or aplastic anemia to define prevalence of the GATA2 mutation and to assess whether mutations in GATA-2 transcription factor exhibit specific immunophenotypic features. The prevalence of a GATA2 mutation in a consecutively diagnosed cohort of children was 14% in advanced forms of myelodysplastic syndrome (refractory anemia with excess blasts, refractory anemia with excess blasts in transformation, and myelodysplasia-related acute myeloid leukemia), 17% in refractory cytopenia of childhood, and 0% in aplastic anemia. In GATA-2-deficient cases, we found the most profound B-cell lymphopenia, including its progenitors in blood and bone marrow, which correlated with significantly diminished intronRSS-Kde recombination excision circles in comparison to other myelodysplastic syndrome/aplastic anemia cases. The other typical features of GATA-2 deficiency (monocytopenia and natural killer cell lymphopenia) were less discriminative. In conclusion, we suggest screening for GATA2 mutations in pediatric myelodysplastic syndrome, preferentially in patients with impaired B-cell homeostasis in bone marrow and peripheral blood (low number of progenitors, intronRSS-Kde recombination excision circles and naïve cells).


Assuntos
Linfócitos B/metabolismo , Fator de Transcrição GATA2/deficiência , Síndromes Mielodisplásicas/diagnóstico , Síndromes Mielodisplásicas/genética , Células Precursoras de Linfócitos B/metabolismo , Adolescente , Anemia Aplástica/diagnóstico , Anemia Aplástica/etiologia , Biomarcadores , Medula Óssea/metabolismo , Medula Óssea/patologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Criança , Pré-Escolar , Diagnóstico Diferencial , Humanos , Imunofenotipagem , Lactente , Contagem de Linfócitos , Linfopenia/diagnóstico , Mutação , Células Mieloides/metabolismo , Fenótipo , Curva ROC , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Adulto Jovem
6.
Eur J Pediatr ; 175(4): 587-92, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26518681

RESUMO

UNLABELLED: Noonan syndrome (NS) is a genetic condition presenting with typical facies, cardiac defects, short stature, variable developmental deficit, cryptorchidism, skeletal, and other abnormalities. Germline mutations in genes involved in the RAS/MAPK signaling have been discovered to underlie NS. Recently, missense mutations in RIT1 have been reported as causative for individuals with clinical signs of NS. We report on a 2.5-year-old boy with NS phenotype with a novel heterozygous change in the RIT1 gene. The patient was born prematurely from pregnancy monitored for polyhydramnios. At 7 months of age, non-immune neutropenia and splenomegaly have been observed. During the severe pneumonia at 10 months, significant progression of hepatosplenomegaly, leukopenia with monocytosis (15-29 %), and thrombocytopenia occurred. Bone marrow evaluation showed myeloid hyperplasia and monocytosis, suggestive of myeloproliferative syndrome. Clinical phenotype (facial dysmorphism, soft hair, short neck, broad chest, widely spaced nipples, mild pectus carinatum, deep palmar creases, unilateral cryptorchidism), and moderate pulmonary valve stenosis with mild psychomotor delay were indicative of NS. DNA analysis identified a de novo heterozygous variant c.69A >T, p.(Lys23Asn) in exon 2 of the RIT1 gene, presumed to be causative. CONCLUSION: We present a patient with a clinical suspicion of NS carrying a novel substitution in RIT1 and hematologic findings not being observed in RIT1 positive patients to date. Thus, the case broadens variability of hematologic symptoms in RIT1 positive NS individuals. WHAT IS KNOWN: • Noonan syndrome is a common genetically heterogeneous disorder of autosomal dominant inheritance characterized by craniofacial dysmorphism, short stature, congenital heart defects, variable cognitive deficit, and other anomalies. What is new: • We report on a 2.5-year-old male patient with clinical signs of NS and hematologic abnormalities, in whom a novel heterozygous substitution in RIT1 with probable pathogenicity was detected.


Assuntos
Leucopenia/genética , Transtornos Mieloproliferativos/genética , Síndrome de Noonan/genética , Proteínas ras/genética , Pré-Escolar , Feminino , Heterozigoto , Humanos , Leucopenia/diagnóstico , Masculino , Mutação de Sentido Incorreto , Transtornos Mieloproliferativos/diagnóstico , Síndrome de Noonan/diagnóstico , Fenótipo , Gravidez
7.
BMC Pulm Med ; 15: 8, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25879889

RESUMO

BACKGROUND: GATA-2 transcription factor deficiency has recently been described in patients with a propensity towards myeloid malignancy associated with other highly variable phenotypic features: chronic leukocytopenias (dendritic cell-, monocyto-, granulocyto-, lymphocytopenia), increased susceptibility to infections, lymphatic vasculature abnormalities, and sensorineural deafness. Patients often suffer from opportunistic respiratory infections; chronic pulmonary changes have been found in advanced disease. CASE PRESENTATION: We present a case of a 17-year-old previously healthy Caucasian male who was admitted to the hospital with fever, malaise, headache, cough and dyspnea. A chest X-ray revealed bilateral interstitial infiltrates and pneumonia was diagnosed. Despite prompt clinical improvement under antibiotic therapy, interstitial changes remained stable. A high resolution computer tomography showed severe diffuse parenchymal lung disease, while the patient's pulmonary function tests were normal and he was asymptomatic. Lung tissue biopsy revealed chronic reparative and resorptive reaction with organizing vasculitis. At the time of the initial presentation to the hospital, serological signs of acute infection with Epstein-Barr virus (EBV) were present; EBV viremia with atypical serological response persisted during two-year follow up. No other infectious agents were found. Marked monocytopenia combined with B-cell lymphopenia led to a suspicion of GATA-2 deficiency. Diagnosis was confirmed by detection of the previously published heterozygous mutation in GATA2 (c.1081 C > T, p.R361C). The patient's brother and father were both carriers of the same genetic defect. The brother had no clinically relevant ailments despite leukocyte changes similar to the index patient. The father suffered from spondylarthritis, and apart from B-cell lymphopenia, no other changes within the leukocyte pool were seen. CONCLUSION: We conclude that a diagnosis of GATA-2 deficiency should be considered in all patients with diffuse parenchymal lung disease presenting together with leukocytopenia, namely monocyto-, dendritic cell- and B-lymphopenia, irrespective of severity of the clinical phenotype. Genetic counseling and screening for GATA2 mutations within the patient's family should be provided as the phenotype is highly variable and carriers without apparent immunodeficiency are still in danger of developing myeloid malignancy. A prompt recognition of this rare condition helps to direct clinical treatment strategies and follow-up procedures.


Assuntos
Infecções por Vírus Epstein-Barr/genética , Fator de Transcrição GATA2/deficiência , Doenças Pulmonares Intersticiais/genética , Pulmão/patologia , Linfopenia/genética , Adolescente , Linfócitos B/imunologia , Infecções por Vírus Epstein-Barr/imunologia , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/imunologia , Humanos , Leucopenia/genética , Leucopenia/imunologia , Pulmão/diagnóstico por imagem , Doenças Pulmonares Intersticiais/diagnóstico , Doenças Pulmonares Intersticiais/imunologia , Linfopenia/imunologia , Masculino , Monócitos/imunologia , Mutação , Radiografia , Síndrome , Vasculite/diagnóstico , Vasculite/genética , Vasculite/imunologia
8.
Oncology ; 86(3): 152-8, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24643197

RESUMO

OBJECTIVE: Individuals with decreased thiopurine methyltransferase (TPMT) activity are at risk of adverse effects of thiopurine administration whereas its increased activity may inactivate drugs faster. We evaluated genotype-phenotype correlations in patients with suspected hematological malignancies and inflammatory bowel disease from our region based on findings of nonlinear TPMT enzyme kinetics previously unreported. PATIENTS AND METHODS: The study group comprised 267 individuals. They were screened for the most common variants of low TPMT activity. TPMT activity was measured in erythrocytes using the HPLC rate-blanked method. RESULTS: Thirty-three patients (12.4%) were heterozygous (26 were TPMT*1/*3A, 5 TPMT*1/*2, 2 TPMT *1/*3C) and 1 was a compound heterozygote (*2/*3A). Normal and low normal TPMT activities substantially overlapped in wild-type and heterozygous individuals, whereas high activities were found in 29 wild-type genotyped patients. Extreme and life-threatening toxicity was observed in the compound heterozygote patient. CONCLUSION: Activity measurement performed at diagnosis provides clinicians with information on immediate pharmacokinetic-related adverse events and/or hypermetabolism, and genotyping may indicate the rate of pharmacodynamic thioguanine nucleotide accumulation due to slower overall thiopurine metabolism.


Assuntos
Metiltransferases/deficiência , Metiltransferases/genética , Adolescente , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Asparaginase/uso terapêutico , Cromatografia Líquida de Alta Pressão , Ciclofosfamida/uso terapêutico , Citarabina/uso terapêutico , República Tcheca , Daunorrubicina/uso terapêutico , Membrana Eritrocítica/enzimologia , Feminino , Estudos de Associação Genética , Humanos , Doenças Inflamatórias Intestinais/sangue , Masculino , Mercaptopurina/uso terapêutico , Metotrexato/uso terapêutico , Metiltransferases/metabolismo , Reação em Cadeia da Polimerase , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Prednisona/uso terapêutico , Eslováquia , Vincristina/uso terapêutico
9.
Br J Haematol ; 165(4): 556-63, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24533562

RESUMO

Pyruvate kinase (PK) deficiency is an iron-loading anaemia characterized by chronic haemolysis, ineffective erythropoiesis and a requirement for blood transfusion in most cases. We studied 11 patients from 10 unrelated families and found nine different disease-causing PKLR mutations. Two of these mutations - the point mutation c.878A>T (p.Asp293Val) and the frameshift deletion c.1553delG (p.(Arg518Leufs*12)) - have not been previously described in the literature. This frameshift deletion was associated with an unusually severe phenotype involving neonatal hyperferritinaemia that is not typical of PK deficiency. No disease-causing mutations in genes associated with haemochromatosis could be found. Inappropriately low levels of hepcidin with respect to iron loading were detected in all PK-deficient patients with increased ferritin, confirming the predominant effect of accelerated erythropoiesis on hepcidin production. Although the levels of a putative hepcidin suppressor, growth differentiation factor-15, were increased in PK-deficient patients, no negative correlation with hepcidin was found. This result indicates the existence of another as-yet unidentified erythroid regulator of hepcidin synthesis in PK deficiency.


Assuntos
Anemia Hemolítica Congênita não Esferocítica/genética , Ferritinas/sangue , Hepcidinas/sangue , Ferro/sangue , Mutação , Piruvato Quinase/deficiência , Piruvato Quinase/genética , Erros Inatos do Metabolismo dos Piruvatos/genética , Adulto , Sequência de Aminoácidos , Anemia Hemolítica Congênita não Esferocítica/sangue , Criança , Pré-Escolar , Análise Mutacional de DNA , Eritropoese , Feminino , Hepcidinas/biossíntese , Humanos , Lactente , Recém-Nascido , Sobrecarga de Ferro/genética , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Piruvato Quinase/sangue , Erros Inatos do Metabolismo dos Piruvatos/sangue , Análise de Sequência de DNA , Reação Transfusional , Adulto Jovem
10.
Haematologica ; 98(12): 1948-55, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23850805

RESUMO

Clinical and genetic heterogeneity renders confirmation or exclusion of autoimmune lymphoproliferative syndrome difficult. To re-evaluate and improve the currently suggested diagnostic approach to patients with suspected FAS mutation, the most frequent cause of autoimmune lymphoproliferative syndrome, we prospectively determined 11 biomarkers in 163 patients with splenomegaly or lymphadenopathy and presumed or proven autoimmune cytopenia(s). Among 98 patients sequenced for FAS mutations in CD3(+)TCRα/ß(+)CD4(-)CD8(-) "double negative" T cells, 32 had germline and six had somatic FAS mutations. The best a priori predictor of FAS mutations was the combination of vitamin B12 and soluble FAS ligand (cut-offs 1255 pg/mL and 559 pg/mL, respectively), which had a positive predictive value of 92% and a negative predictive value of 97%. We used these data to develop a web-based probability calculator for FAS mutations using the three most discriminatory biomarkers (vitamin B12, soluble FAS ligand, interleukin-10) of the 11 tested. Since more than 60% of patients with lymphoproliferation and autoimmune cytopenia(s) in our cohort did not harbor FAS mutations, 15% had somatic FAS mutations, and the predictive value of double-negative T-cell values was rather low (positive and negative predictive values of 61% and 77%, respectively), we argue that the previously suggested diagnostic algorithm based on determination of double-negative T cells and germline FAS sequencing, followed by biomarker analysis, is not efficient. We propose vitamin B12 and soluble FAS ligand assessment as the initial diagnostic step with subsequent decision on FAS sequencing supported by a probability-calculating tool.


Assuntos
Proteína Ligante Fas/sangue , Proteína Ligante Fas/genética , Transtornos Linfoproliferativos/sangue , Transtornos Linfoproliferativos/genética , Vitamina B 12/sangue , Adolescente , Doenças Autoimunes/sangue , Doenças Autoimunes/diagnóstico , Doenças Autoimunes/genética , Biomarcadores/sangue , Estudos de Coortes , Feminino , Humanos , Transtornos Linfoproliferativos/diagnóstico , Masculino
11.
Am J Med Genet A ; 158A(10): 2545-50, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22887642

RESUMO

Jacobsen syndrome (JBS) is a rare chromosomal disorder caused by terminal deletion of the long arm of chromosome 11. We report on four prenatally diagnosed patients with JBS with variable prenatal and postnatal phenotypes and 11q deletions of varying sizes. Precise characterization of the deleted region in three patients was performed by SNP arrays. The severity of both the prenatal and postnatal phenotypes did not correlate with the size of the haploinsufficient region. Despite the large difference in the deletion size (nearly 6 Mb), both of the live-born patients had similar phenotypes corresponding to JBS. However, one of the most prominent features of JBS, thrombocytopenia, was only present in the live-born boy. The girl, who had a significantly longer deletion spanning all four genes suspected of being causative of JBS-related thrombocytopenia (FLI1, ETS1, NFRKB, and JAM3), did not manifest a platelet phenotype. Therefore, our findings do not support the traditional view of deletion size correlation in JBS or the causative role of FLI1, ETS1, NFRKB, and JAM3 deletion per se for the development of disease-related thrombocytopenia.


Assuntos
Deleção Cromossômica , Cromossomos Humanos Par 11/genética , Síndrome da Deleção Distal 11q de Jacobsen/genética , Proteína Proto-Oncogênica c-fli-1/genética , Trombocitopenia/genética , Adulto , Feminino , Estudos de Associação Genética , Humanos , Lactente , Síndrome da Deleção Distal 11q de Jacobsen/fisiopatologia , Masculino , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Adulto Jovem
12.
Pediatr Rheumatol Online J ; 9(1): 27, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21914180

RESUMO

Autoimmunity is often observed among individuals with primary immune deficiencies; however, the frequency and role of autoimmunity in Schimke immuno-osseous dysplasia (SIOD) has not been fully assessed. SIOD, which is caused by mutations of SMARCAL1, is a rare autosomal recessive disease with its prominent features being skeletal dysplasia, T cell deficiency, and renal failure. We present a child with severe SIOD who developed rituximab resistant Evans syndrome (ES). Consistent with observations in several other immunodeficiency disorders, a review of SIOD patients showed that approximately a fifth of SIOD patients have some features of autoimmune disease. To our best knowledge this case represents the first patient with SIOD and rituximab resistant ES and the first study of autoimmune disease in SIOD.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA