Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Exp Hematol ; 73: 25-37.e8, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30876823

RESUMEN

Krüppel-like factor 1 (KLF1), a transcription factor controlling definitive erythropoiesis, is involved in sequential control of terminal cell division and enucleation via fine regulation of key cell cycle regulator gene expression in erythroid lineage cells. Type IV congenital dyserythropoietic anemia (CDA) is caused by a monoallelic mutation at the second zinc finger of KLF1 (c.973G>A, p.E325K). We recently diagnosed a female patient with type IV CDA with the identical missense mutation. To understand the mechanism underlying the dyserythropoiesis caused by the mutation, we generated induced pluripotent stem cells (iPSCs) from the CDA patient (CDA-iPSCs). The erythroid cells that differentiated from CDA-iPSCs (CDA-erythroid cells) displayed multinucleated morphology, absence of CD44, and dysregulation of the KLF1 target gene expression. In addition, uptake of bromodeoxyuridine by CDA-erythroid cells was significantly decreased at the CD235a+/CD71+ stage, and microarray analysis revealed that cell cycle regulator genes were dysregulated, with increased expression of negative regulators such as CDKN2C and CDKN2A. Furthermore, inducible expression of the KLF1 E325K, but not the wild-type KLF1, caused a cell cycle arrest at the G1 phase in CDA-erythroid cells. Microarray analysis of CDA-erythroid cells and real-time polymerase chain reaction analysis of the KLF1 E325K inducible expression system also revealed altered expression of several KLF1 target genes including erythrocyte membrane protein band 4.1 (EPB41), EPB42, glutathione disulfide reductase (GSR), glucose phosphate isomerase (GPI), and ATPase phospholipid transporting 8A1 (ATP8A1). Our data indicate that the E325K mutation in KLF1 is associated with disruption of transcriptional control of cell cycle regulators in association with erythroid membrane or enzyme abnormalities, leading to dyserythropoiesis.


Asunto(s)
Anemia Diseritropoyética Congénita , Diferenciación Celular/genética , Células Eritroides , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Células Madre Pluripotentes Inducidas , Factores de Transcripción de Tipo Kruppel , Mutación Missense , Adulto , Sustitución de Aminoácidos , Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/metabolismo , Anemia Diseritropoyética Congénita/patología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p18 de las Quinasas Dependientes de la Ciclina/metabolismo , Células Eritroides/metabolismo , Células Eritroides/patología , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo
2.
Mol Genet Metab Rep ; 18: 1-6, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30519529

RESUMEN

OBJECTIVE: The prognosis of adrenoleukodystrophy (ALD)with neurological involvement is generally dismal; however, allogeneic stem cell transplantation (SCT) is recognized as effective to stabilize or improve the clinical symptoms of ALD. Herein, we report the clinical outcomes of patients with ALD who consecutively underwent allogeneic stem cell transplantation with reduced intensity conditioning at our institution. PATIENTS: Sixteen patients with ALD, who were symptomatic (n = 14) or presymptomatic (n = 2), received SCT from 2010 to 2016. The stem cell source was cord blood (n = 14), or bone marrow from a human leukocyte antigen identical sibling (n = 2). The conditioning regimen prior to transplantation was reduced intensity and consisted of fludarabine (125 mg/m2), melphalan (140 mg/m2) and low dose total body irradiation (TBI) of 4Gy (n = 15) or 3Gy (n = 1). RESULTS: Primary engraftment was obtained in 11 patients, and 4 of the 5 patients who lost the primary graft received a second cord blood transplantation and were engrafted. Five years overall and event-free survival were 90.9% and 61.1% respectively, with a median of 45 months (range 16-91). Loes score stabilized or improved by 18 months after transplantation except for patients with internal capsule involvement. CONCLUSION: Allogeneic SCT with reduced intensity conditioning for patients with ALD was safely performed without major transplant-related complications even in symptomatic patients and neurological symptoms were stabilized after SCT in patients without internal capsule involvement.

3.
Int J Hematol ; 108(3): 306-311, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29936674

RESUMEN

Congenital dyserythropoietic anemia (CDA) is a heterogeneous group of rare congenital disorders characterized by ineffective erythropoiesis and dysplastic changes in erythroblasts. Diagnosis of CDA is based primarily on the morphology of bone marrow erythroblasts; however, genetic tests have recently become more important. Here, we performed genetic analysis of 10 Japanese patients who had been diagnosed with CDA based on laboratory findings and morphological characteristics. We examined 10 CDA patients via central review of bone marrow morphology and genetic analysis for congenital bone marrow failure syndromes. Sanger sequencing for CDAN1, SEC23B, and KLF1 was performed for all patients. We performed whole-exome sequencing in patients without mutation in these genes. Three patients carried pathogenic CDAN1 mutations, whereas no SEC23B mutations were identified in our cohort. WES unexpectedly identified gene mutations known to cause congenital hemolytic anemia in two patients: canonical G6PD p.Val394Leu mutation and SPTA1 p.Arg28His mutation. Comprehensive genetic analysis is warranted for more effective diagnosis of patients with suspected CDA.


Asunto(s)
Anemia Diseritropoyética Congénita/diagnóstico , Anemia Hemolítica Congénita/diagnóstico , Exoma/genética , Pruebas Genéticas/métodos , Adulto , Anemia Diseritropoyética Congénita/genética , Anemia Hemolítica Congénita/genética , Pueblo Asiatico , Diagnóstico Diferencial , Femenino , Humanos , Masculino
4.
Pediatr Transplant ; 22(2)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29239087

RESUMEN

In SCT, death from transplant-related complications is the major obstacle hindering improvement of transplant outcomes, and proper supportive care is essential to reduce TRM. The transplant outcomes of 210 pediatric patients with malignant and non-malignant disorders who consecutively underwent SCT in our institution from 2000 to 2013 were analyzed. The transplant years were divided into three periods: A (2000-2004), B (2005-2008), and C (2009-2013), and an improvement in 5-year OS and a decrease in 5-year TRM were observed over these time periods; that is, OS was 61.5%, 60.3%, and 79.5% (P = .062), and TRM was 19.9%, 7.9%, and 0.0% (P < .001) in periods A, B, and C, respectively. On multivariate analysis, the prognostic factor for TRM for all patients was administration of danaparoid (HR = 0.109, 95% CI = 0.033-0.363, P < .001), and for patients with hematological malignancies in allogeneic SCT, the prognostic factors were danaparoid (HR = 0.046, 95% CI = 0.006-0.326, P = .002) and advanced disease at SCT (HR = 4.802, 95% CI = 1.734-13.30, P = .003). A reduction in TRM after SCT was observed over the time periods, and supportive care with danaparoid was found to be significantly effective in reducing TRM in SCT for children.


Asunto(s)
Anticoagulantes/uso terapéutico , Sulfatos de Condroitina/uso terapéutico , Dermatán Sulfato/uso terapéutico , Heparitina Sulfato/uso terapéutico , Trasplante de Células Madre/mortalidad , Adolescente , Niño , Preescolar , Femenino , Estudios de Seguimiento , Humanos , Lactante , Recién Nacido , Masculino , Evaluación de Resultado en la Atención de Salud , Pronóstico , Estudios Retrospectivos , Trasplante de Células Madre/efectos adversos , Análisis de Supervivencia
5.
Genet Med ; 19(7): 796-802, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28102861

RESUMEN

PURPOSE: Precise genetic diagnosis of inherited bone marrow failure syndromes (IBMFS), a heterogeneous group of genetic disorders, is challenging but essential for precise clinical decision making. METHODS: We analyzed 121 IBMFS patients using a targeted sequencing covering 184 associated genes and 250 IBMFS patients using whole-exome sequencing (WES). RESULTS: We achieved successful genetic diagnoses for 53 of 121 patients (44%) using targeted sequencing and for 68 of 250 patients (27%) using WES. In the majority of cases (targeted sequencing: 45/53, 85%; WES: 63/68, 93%), the detected variants were concordant with, and therefore supported, the clinical diagnoses. However, in the remaining 13 cases (8 patients by target sequencing and 5 patients by WES), the clinical diagnoses were incompatible with the detected variants. CONCLUSION: Our approach utilizing targeted sequencing and WES achieved satisfactory diagnostic rates and supported the efficacy of massive parallel sequencing as a diagnostic tool for IBMFS.Genet Med advance online publication 19 January 2017.


Asunto(s)
Anemia Aplásica/diagnóstico , Anemia Aplásica/genética , Enfermedades de la Médula Ósea/diagnóstico , Enfermedades de la Médula Ósea/genética , Hemoglobinuria Paroxística/diagnóstico , Hemoglobinuria Paroxística/genética , Secuenciación de Nucleótidos de Alto Rendimiento/estadística & datos numéricos , Trastornos de Fallo de la Médula Ósea , Exoma/genética , Femenino , Pruebas Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , Mutación/genética , Análisis de Secuencia de ADN/métodos , Secuenciación del Exoma/métodos
6.
Haematologica ; 100(12): 1546-52, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26315930

RESUMEN

Acquired aplastic anemia is an immune-mediated disease characterized by severe defects in stem cell number resulting in hypocellular marrow and peripheral blood cytopenias. Minor paroxysmal nocturnal hemoglobinuria populations and a short telomere length were identified as predictive biomarkers of immunosuppressive therapy responsiveness in aplastic anemia. We enrolled 113 aplastic anemia patients (63 boys and 50 girls) in this study to evaluate their response to immunosuppressive therapy. The paroxysmal nocturnal hemoglobinuria populations and telomere length were detected by flow cytometry. Forty-seven patients (42%) carried a minor paroxysmal nocturnal hemoglobinuria population. The median telomere length of aplastic anemia patients was -0.99 standard deviation (SD) (range -4.01-+3.01 SD). Overall, 60 patients (53%) responded to immunosuppressive therapy after six months. Multivariate logistic regression analysis identified the absence of a paroxysmal nocturnal hemoglobinuria population and a shorter telomere length as independent unfavorable predictors of immunosuppressive therapy response at six months. The cohort was stratified into a group of poor prognosis (paroxysmal nocturnal hemoglobinuria negative and shorter telomere length; 37 patients) and good prognosis (paroxysmal nocturnal hemoglobinuria positive and/or longer telomere length; 76 patients), respectively. The response rates of the poor prognosis and good prognosis groups at six months were 19% and 70%, respectively (P<0.001). The combined absence of a minor paroxysmal nocturnal hemoglobinuria population and a short telomere length is an efficient predictor of poor immunosuppressive therapy response, which should be considered while deciding treatment options: immunosuppressive therapy or first-line hematopoietic stem cell transplantation. The trial was registered in www.umin.ac.jp with number UMIN000017972.


Asunto(s)
Anemia Aplásica , Hemoglobinuria Paroxística , Terapia de Inmunosupresión , Homeostasis del Telómero , Telómero/metabolismo , Adolescente , Anemia Aplásica/diagnóstico , Anemia Aplásica/metabolismo , Anemia Aplásica/terapia , Niño , Preescolar , Femenino , Hemoglobinuria Paroxística/diagnóstico , Hemoglobinuria Paroxística/metabolismo , Hemoglobinuria Paroxística/terapia , Humanos , Lactante , Masculino , Pronóstico
9.
PLoS One ; 10(12): e0145394, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26720758

RESUMEN

Juvenile myelomonocytic leukemia (JMML), an overlap of myelodysplastic / myeloproliferative neoplasm, is an intractable pediatric myeloid neoplasm. Epigenetic regulation of transcription, particularly by CpG methylation, plays an important role in tumor progression, mainly by repressing tumor-suppressor genes. To clarify the clinical importance of aberrant DNA methylation, we studied the hypermethylation status of 16 target genes in the genomes of 92 patients with JMML by bisulfite conversion and the pryosequencing technique. Among 16 candidate genes, BMP4, CALCA, CDKN2A, and RARB exhibited significant hypermethylation in 72% (67/92) of patients. Based on the number of hypermethylated genes, patients were stratified into three cohorts based on an aberrant methylation score (AMS) of 0, 1-2, or 3-4. In the AMS 0 cohort, the 5-year overall survival (OS) and transplantation-free survival (TFS) were good (69% and 76%, respectively). In the AMS 1-2 cohort, the 5-year OS was comparable to that in the AMS 0 cohort (68%), whereas TFS was poor (6%). In the AMS 3-4 cohort, 5-year OS and TFS were markedly low (8% and 0%, respectively). Epigenetic analysis provides helpful information for clinicians to select treatment strategies for patients with JMML. For patients with AMS 3-4 in whom hematopoietic stem cell transplantation does not improve the prognosis, alternative therapies, including DNA methyltransferase inhibitors and new molecular-targeting agents, should be established as treatment options.


Asunto(s)
Metilación de ADN/genética , Leucemia Mielomonocítica Juvenil/genética , Adolescente , Niño , Preescolar , Estudios de Cohortes , Femenino , Estudios de Asociación Genética , Humanos , Lactante , Masculino , Análisis Multivariante , Mutación/genética , Análisis de Supervivencia , Resultado del Tratamiento
10.
Haematologica ; 99(8): 1312-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24816243

RESUMEN

Predicting the response to immunosuppressive therapy could provide useful information to help the clinician define treatment strategies for patients with aplastic anemia. In our current study, we evaluated the relationship between telomere length of lymphocytes at diagnosis and the response to immunosuppressive therapy in 64 children with aplastic anemia, using flow fluorescence in situ hybridization. Median age of patients was ten years (range 1.5-16.2 years). Severity of the disease was classified as very severe in 23, severe in 21, and moderate in 20 patients. All patients were enrolled in multicenter studies using antithymocyte globulin and cyclosporine. The response rate to immunosuppressive therapy at six months was 52% (33 of 64). The probability of 5-year failure-free survival and overall survival were 56% (95% confidence interval (CI): 41-69%) and 97% (95%CI: 87-99%), respectively. Median telomere length in responders was -0.4 standard deviation (SD) (-2.7 to +3.0 SD) and -1.5 SD (-4.0 to +1.6 (SD)) in non-responders (P<0.001). Multivariate analysis showed that telomere length shorter than -1.0 SD (hazard ratio (HR): 22.0; 95%CI: 4.19-115; P<0.001), platelet count at diagnosis less than 25×10(9)/L (HR: 13.9; 95%CI: 2.00-96.1; P=0.008), and interval from diagnosis to immunosuppressive therapy longer than 25 days (HR: 4.81; 95%CI: 1.15-20.1; P=0.031) were the significant variables for poor response to immunosuppressive therapy. Conversely to what has been found in adult patients, measurement of the telomere length of lymphocytes at diagnosis is a promising assay in predicting the response to immunosuppressive therapy in children with aplastic anemia.


Asunto(s)
Anemia Aplásica/diagnóstico , Anemia Aplásica/tratamiento farmacológico , Inmunosupresores/uso terapéutico , Linfocitos/efectos de los fármacos , Homeostasis del Telómero/efectos de los fármacos , Telómero/efectos de los fármacos , Adolescente , Anemia Aplásica/inmunología , Niño , Preescolar , Femenino , Humanos , Inmunosupresores/farmacología , Lactante , Linfocitos/inmunología , Linfocitos/patología , Masculino , Valor Predictivo de las Pruebas , Telómero/inmunología , Telómero/patología , Homeostasis del Telómero/fisiología , Resultado del Tratamiento
11.
Nat Genet ; 45(8): 937-41, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23832011

RESUMEN

Juvenile myelomonocytic leukemia (JMML) is an intractable pediatric leukemia with poor prognosis whose molecular pathogenesis is poorly understood, except for somatic or germline mutations of RAS pathway genes, including PTPN11, NF1, NRAS, KRAS and CBL, in the majority of cases. To obtain a complete registry of gene mutations in JMML, whole-exome sequencing was performed for paired tumor-normal DNA from 13 individuals with JMML (cases), which was followed by deep sequencing of 8 target genes in 92 tumor samples. JMML was characterized by a paucity of gene mutations (0.85 non-silent mutations per sample) with somatic or germline RAS pathway involvement in 82 cases (89%). The SETBP1 and JAK3 genes were among common targets for secondary mutations. Mutations in the latter were often subclonal and may be involved in the progression rather than the initiation of leukemia, and these mutations associated with poor clinical outcome. Our findings provide new insights into the pathogenesis and progression of JMML.


Asunto(s)
Proteínas Portadoras/genética , Exoma , Janus Quinasa 3/genética , Leucemia Mielomonocítica Juvenil/genética , Mutación , Proteínas Nucleares/genética , Niño , Preescolar , Progresión de la Enfermedad , Femenino , Mutación de Línea Germinal , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , Leucemia Mielomonocítica Juvenil/metabolismo , Leucemia Mielomonocítica Juvenil/mortalidad , Masculino , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Transducción de Señal
12.
Int J Hematol ; 97(5): 650-3, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23605369

RESUMEN

The congenital dyserythropoietic anemias (CDAs) are a heterogeneous group of genetic disorders of red cell production. They are characterized by ineffective erythropoiesis and dyserythropoiesis. Here, we present the clinical description and mutation analysis of a Japanese female with CDA type 1. She has long been diagnosed with unclassified congenital hemolytic anemia from the neonatal period. However, bone marrow morphology and genetic testing of the CDAN1 gene at the age of 12 years confirmed the afore-mentioned diagnosis. Thus, we should be aware of the possibility of CDA if the etiology of congenital anemia or jaundice cannot be clearly elucidated.


Asunto(s)
Anemia Diseritropoyética Congénita/diagnóstico , Anemia Diseritropoyética Congénita/genética , Anemia Hemolítica Congénita/diagnóstico , Glicoproteínas/genética , Mutación , Médula Ósea/patología , Niño , Análisis Mutacional de ADN , Diagnóstico Diferencial , Femenino , Heterocigoto , Humanos , Proteínas Nucleares
13.
J Pediatr Hematol Oncol ; 35(5): e219-23, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23588332

RESUMEN

BACKGROUND: Voriconazole is a triazole antifungal agent with potent activity against a broad spectrum of pathogens, including Aspergillus and Candida species. In human adults, allelic polymorphisms of CYP2C19 are known to correlate with significant variation in voriconazole plasma concentration. Here, we report an analysis of CYP2C19 phenotype and voriconazole plasma concentrations in children. METHODS: This retrospective study included 37 children who had voriconazole plasma concentrations measured from May 2006 to June 2011. All had single-nucleotide polymorphisms that define the 3 major CYP2C19 alleles. Patients were classified as follows: normal metabolizers, intermediate metabolizers, poor metabolizers, or hypermetabolizers. RESULTS: The frequencies of the 3 CYP2C19 genetic polymorphisms were similar to those previously reported for Japanese adults. Trough plasma concentrations of voriconazole were significantly higher in the poor metabolizer and intermediate metabolizer groups compared with the normal metabolizer and hypermetabolizer groups (P=0.004). Two patients with high plasma concentrations experienced voriconazole-related severe adverse events (syndrome of inappropriate antidiuretic hormone secretion and cardiac toxicities). CONCLUSIONS: The current study suggests that a significant association exists in children between the voriconazole plasma concentration and the CYP2C19 phenotype. Dose adjustment based on CYP2C19 phenotype may be useful during voriconazole therapy, especially for Japanese children, who as a group have a higher incidence of the poor metabolizer and intermediate metabolizer phenotypes.


Asunto(s)
Antifúngicos/sangre , Hidrocarburo de Aril Hidroxilasas/genética , Pirimidinas/sangre , Triazoles/sangre , Adolescente , Antifúngicos/efectos adversos , Pueblo Asiatico/genética , Niño , Preescolar , Cromatografía Líquida de Alta Presión , Citocromo P-450 CYP2C19 , Femenino , Humanos , Lactante , Masculino , Fenotipo , Polimorfismo de Nucleótido Simple , Pirimidinas/efectos adversos , Estudios Retrospectivos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Triazoles/efectos adversos , Voriconazol
14.
Brain Dev ; 35(7): 690-3, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23141186

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature aging disease, caused by a de novo mutation of lamin-A gene, LMNA G608G. Accumulation of abnormal lamin-A (progerin) compromises nuclear membrane integrity and results in the accelerated senescence. Affected patients show a typical feature of birdlike face, alopecia, sclerotic skin, loss of subcutaneous fat, and short stature with advancing years. Neonatal scleroderma is the first presentation, although early diagnosis is challenging. The leading cause of death is cardio-/cerebro-vascular accidents associated with atherosclerosis. However, not all findings may recapitulate the aging process. We herein report a 9-year-old Japanese male with HGPS who developed cerebral infarction. The genetic study of peripheral blood-derived DNA determined a heterozygous c.1824C>T mutation, p.G608G. Telomere length of lymphocytes was normal. Bilateral stenosis of carotid siphons was prominent, while systemic arteriosclerosis was unremarkable assessed by the ankle-brachial index, carotid ultrasound imaging and funduscopic study. HGPS patients have marked loss and functional defects in vascular smooth muscle cells, leading to the vulnerability to circulatory stress. Symmetrical stenosis of siphons might occur as a distinctive cerebral vasculopathy of HGPS, rather than simple vascular senescence. Peripheral blood study on LMNA G608G and telomere length could screen progerias in infancy for early therapeutic intervention.


Asunto(s)
Estenosis Carotídea/etiología , Estenosis Carotídea/patología , Progeria/patología , Secuencia de Bases , Niño , Humanos , Lamina Tipo A/genética , Masculino , Mutación , Progeria/genética
15.
Blood ; 120(7): 1485-8, 2012 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-22753870

RESUMEN

Juvenile myelomonocytic leukemia (JMML) is a rare pediatric myeloid neoplasm characterized by excessive proliferation of myelomonocytic cells. Somatic mutations in genes involved in GM-CSF signal transduction, such as NRAS, KRAS, PTPN11, NF1, and CBL, have been identified in more than 70% of children with JMML. In the present study, we report 2 patients with somatic mosaicism for oncogenic NRAS mutations (G12D and G12S) associated with the development of JMML. The mutated allele frequencies quantified by pyrosequencing were various and ranged from 3%-50% in BM and other somatic cells (ie, buccal smear cells, hair bulbs, or nails). Both patients experienced spontaneous improvement of clinical symptoms and leukocytosis due to JMML without hematopoietic stem cell transplantation. These patients are the first reported to have somatic mosaicism for oncogenic NRAS mutations. The clinical course of these patients suggests that NRAS mosaicism may be associated with a mild disease phenotype in JMML.


Asunto(s)
Leucemia Mielomonocítica Juvenil/genética , Mosaicismo , Mutación/genética , Oncogenes/genética , Proteínas ras/genética , Secuencia de Bases , Niño , Análisis Mutacional de ADN , Humanos , Lactante , Masculino , Datos de Secuencia Molecular
17.
Intern Med ; 51(8): 917-20, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22504250

RESUMEN

We describe the first case of genetically diagnosed congenital dyserythropoietic anemia (CDA) type 1 in a Japanese man. The patient had hemolytic anemia since he was a child, and he developed diabetes, hypogonadism, and liver dysfunction in his thirties, presumably from systemic iron overload. When he was 48 years old a diagnosis was finally made by genetic analysis that revealed a homozygous mutation of CDAN1 gene (Pro1129Leu). His serum hepcidin-25 level was inappropriately low. We conclude that physicians should be aware of the possibility of CDA in a patient with anemia and systemic iron overload at any age.


Asunto(s)
Anemia Diseritropoyética Congénita/sangre , Anemia Diseritropoyética Congénita/genética , Péptidos Catiónicos Antimicrobianos/sangre , Pueblo Asiatico/genética , Glicoproteínas/genética , Mutación/genética , Adulto , Hepcidinas , Humanos , Masculino , Proteínas Nucleares
18.
Paediatr Drugs ; 14(3): 157-63, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22480363

RESUMEN

Juvenile myelomonocytic leukemia (JMML) is a rare clonal myelodysplastic/myeloproliferative disorder that affects young children. It is characterized by hypersensitivity of JMML cells to granulocyte-macrophage colony-stimulating factor (GM-CSF) in vitro. The pathogenesis of JMML seems to arise from constitutional activation of the GM-CSF/RAS (a GTPase) signaling pathway, a result of mutations in RAS, NF1, PTPN11, and CBL that interfere with downstream components of the pathway. Most patients with JMML usually experience an aggressive clinical course, and hematopoietic stem cell transplantation (HSCT) is currently the only curative treatment, although the high rates of relapses and graft failures are of great concern. In contrast, a certain proportion of patients experience a stable clinical course for a considerable period of time, and sometimes the disease even spontaneously resolves without any treatment. Recent studies have provided us with increased knowledge of genotype-phenotype correlations in JMML, and suggested that differences in clinical courses may reflect genetic status. Thus, genotype-based management is of current international interest, especially for JMML with RAS mutations. Cumulative evidence suggests that RAS mutations can be related to favorable clinical outcomes, and HSCT may not have to be a mandatory therapeutic option for a portion of patients with this mutation, although a consensus regarding genotype-based management has not yet been achieved. Further efforts toward identifying which patients who will do well without HSCT are required.


Asunto(s)
Genes ras/genética , Trasplante de Células Madre Hematopoyéticas/métodos , Leucemia Mielomonocítica Juvenil/genética , Mutación/genética , Enfermedades Raras/genética , Síndrome Linfoproliferativo Autoinmune/genética , Niño , Preescolar , Genotipo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Leucemia Mielomonocítica Juvenil/terapia , Neurofibromina 1/genética , Fenotipo , Proteínas Proto-Oncogénicas p21(ras)/genética , Enfermedades Raras/terapia , Transducción de Señal/genética
19.
Int J Hematol ; 95(6): 675-9, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22527854

RESUMEN

Fanconi anemia (FA) is a disorder characterized by developmental anomalies, bone marrow failure and a predisposition to malignancy. It has recently been shown that hematopoietic stem cell transplantation using fludarabine (FLU)-based reduced-intensity conditioning is an efficient and quite safe therapeutic modality. We retrospectively analyzed the outcome of bone marrow transplantation (BMT) in eight patients with FA performed in two institutes between 2001 and 2011. There were seven females and one male with a median age at diagnosis = 4.5 years (range 2-12 years). The constitutional characteristics associated with FA, such as developmental anomalies, short stature and skin pigmentation, were absent in three of the patients. One patient showed myelodysplastic features at the time of BMT. All patients received BMT using FLU, cyclophosphamide (CY) and rabbit anti-thymocyte globulin (ATG) either from a related donor (n = 4) or an unrelated donor (n = 4). Acute graft-versus-host disease (GVHD) of grade I developed in one patient, while chronic GVHD was not observed in any patient. All patients are alive and achieved hematopoietic recovery at a median follow-up of 72 months (range 4-117 months). BMT using FLU/low-dose CY/ATG -based regimens regardless to the donor is a beneficial therapeutic approach for FA patients.


Asunto(s)
Trasplante de Médula Ósea , Anemia de Fanconi/cirugía , Trasplante de Médula Ósea/efectos adversos , Niño , Preescolar , Anemia de Fanconi/tratamiento farmacológico , Anemia de Fanconi/mortalidad , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Masculino , Acondicionamiento Pretrasplante , Trasplante Homólogo , Resultado del Tratamiento
20.
Int J Hematol ; 95(5): 581-4, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22461033

RESUMEN

Juvenile myelomonocytic leukemia is a rare malignancy that occurs in pediatric patients. Previous reports, have described leukemic cells may infiltrate many organs, such as the lungs, skin, liver, spleen, and intestines, but not the central nervous system, although central nervous system infiltration remains a point of concern in every patient with acute leukemia. Here, we present one case of a boy with juvenile myelomonocytic leukemia who developed multiple lesions in the brain while undergoing chemotherapy with 6-mercaptopurine and cytarabine. We diagnosed the central nervous system involvement by magnetic resonance imaging, cerebrospinal fluid cytology, and the patient's clinical course. He was treated with a high dose of cytarabine and intrathecal chemotherapy, then with unrelated cord blood stem cell transplantation. He has been in a first complete remission for more than 18 months after cord blood stem cell transplantation without any neurological sequelae. In conclusion, we encountered a boy with juvenile myelomonocytic leukemia who developed central nervous system lesions under standard chemotherapy. We subsequently switched treatment to central nervous system-oriented chemotherapy, which resulted in a good clinical condition and successful cord blood stem cell transplantation.


Asunto(s)
Antimetabolitos Antineoplásicos/efectos adversos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Citarabina/efectos adversos , Leucemia Mielomonocítica Juvenil/tratamiento farmacológico , Mercaptopurina/efectos adversos , Humanos , Lactante , Leucemia Mielomonocítica Juvenil/patología , Leucemia Mielomonocítica Juvenil/cirugía , Masculino , Trasplante de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...