Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Neonatology ; 120(5): 661-665, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37473739

RESUMEN

Thrombocytopenia is a common laboratory abnormality encountered in critically ill neonates. The broad differential for thrombocytopenia, and its association with potentially severe neonatal pathology, often presents a diagnostic dilemma prompting extensive evaluation. Hemolysis due to red cell enzymopathies is a rare cause of neonatal thrombocytopenia that is typically brief and self-limiting. Here, we present a case of thrombocytopenia, refractory to transfusion, associated with anemia and hyperbilirubinemia in a neonate with pyruvate kinase deficiency (PKD) arising from compound heterozygous PKLR mutations. The nature of the thrombocytopenia in this patient created considerable diagnostic uncertainty, which was ultimately resolved by whole-exome sequencing. This case emphasizes that inherited red cell defects, such as PKD, are important to consider in cases of neonatal thrombocytopenia.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica , Anemia , Enfermedades del Recién Nacido , Errores Innatos del Metabolismo del Piruvato , Trombocitopenia Neonatal Aloinmune , Recién Nacido , Humanos , Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/diagnóstico , Anemia Hemolítica Congénita no Esferocítica/genética , Errores Innatos del Metabolismo del Piruvato/diagnóstico , Errores Innatos del Metabolismo del Piruvato/genética , Errores Innatos del Metabolismo del Piruvato/complicaciones , Piruvato Quinasa/genética
2.
Transfusion ; 63(1): 257-262, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36349479

RESUMEN

BACKGROUND: Pyruvate Kinase (PK) deficiency is the most common enzyme defect of glycolysis, leading to congenital hemolytic anemia, which can occur during the neonatal period. STUDY DESIGN AND METHODS: We report the prenatal management of fetal anemia related to PK deficiency in a family with a severe proband. RESULTS: The couple had a first child born with hydrops, whose PK deficiency was diagnosed at 18 months of life. He was treated with allogeneic bone marrow transplantation. The second child was free from disease. For the third pregnancy, the amniocentesis revealed a PK deficiency. Weekly ultrasound monitoring of the middle cerebral artery velocity allowed the detection of severe fetal anemia. Two intrauterine red blood cell transfusions (IUTs) were performed, raising the fetal hemoglobin from 6.6 to 14.5 g/dl at 28 weeks' gestation and from 8.9 to 15.3 g/dl at 31 weeks. A hematopoietic stem cell allograft was discussed prenatally but not chosen, as it would not have significantly changed the perinatal prognosis. The patient delivered a 2730 g girl at 37 weeks, with hemoglobin of 13.6 g/dl. The child presented with neonatal jaundice treated with phototherapy and received postnatal transfusions. DISCUSSION: When a proband is identified in a family, fetal investigation is warranted, to set up third-trimester ultrasound surveillance and perinatal management. In case of fetal severe anemia of unknown etiology, the workup on fetal blood sampling before IUT should comprise the search for erythrocytes enzymopathies, such as PK deficiency. IUTs allow safer full-term delivery in cases with PK deficiency.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica , Anemia , Enfermedades Fetales , Embarazo , Recién Nacido , Masculino , Niño , Femenino , Humanos , Piruvato Quinasa , Transfusión de Sangre Intrauterina/efectos adversos , Anemia/etiología , Anemia/terapia , Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/terapia , Anemia Hemolítica Congénita no Esferocítica/diagnóstico , Enfermedades Fetales/diagnóstico por imagen , Enfermedades Fetales/terapia
3.
BMC Pediatr ; 22(1): 461, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35915427

RESUMEN

BACKGROUND: Glucose phosphate isomerase (GPI) deficiency is a rare autosomal recessive disorder that causes hereditary nonspherocytic hemolytic anemia (HNSHA). Homozygous or compound heterozygous mutation of the GPI gene on chromosome 19q13 is the cause of GPI deficiency. Fifty-seven GPI mutations have been reported at the molecular level. CASE PRESENTATION: A 5-month-old boy was presented with repeated episodes of jaundice after birth. He suffered from moderate hemolytic anemia (hemoglobin levels ranging from 62 to 91 g/L) associated with macrocytosis, reticulocytosis, neutropenia, and hyperbilirubinemia. Whole-exome sequencing showed that he has a missense mutation c.301G > A (p.Val101Met) in exon 4 and a frameshift mutation c.812delG (p.Gly271Glufs*131) in exon 10. Mutation p.Gly271Glufs*131 is a novel frameshift null mutation in GPI deficiency. CONCLUSION: In a patient with recurrent jaundice since birth, mutations in the GPI gene associated with HNSHA should be evaluated. The c.812delG (p.Gly271Glufs*131) variant may be a novel mutation of the GPI gene. Compound heterozygous mutations c.301G > A (p.Val101Met) and c.812delG (p.Gly271Glufs*131) are not relevant to neurological impairment.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica , Anemia Hemolítica , Errores Innatos del Metabolismo , Anemia Hemolítica/genética , Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/diagnóstico , Anemia Hemolítica Congénita no Esferocítica/genética , China , Glucosa-6-Fosfato Isomerasa/genética , Homocigoto , Humanos , Lactante , Masculino
4.
Pediatr Blood Cancer ; 69(9): e29837, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35695473

RESUMEN

Glucose-6-phosphate dehydrogenase (G6PD) deficiency is an X-linked genetic disease caused by a pathogenic G6PD mutation. An 8-year-old Chinese male child was investigated because of chronic nonspherocytic hemolytic anemia (CNSHA) associated with hepatosplenomegaly. Genetic analysis unraveled co-inheritance of a hemizygous mutation c.1225C>T (p.Pro409Ser) in G6PD (G6PD Utrecht, previously reported only in The Netherlands) and heterozygote HBB mutation c.316-197C>T (IVS-Ⅱ-654 C>T). Because IVS-Ⅱ-654 C>T on its own does not cause CNSHA, we believe that the clinical manifestations in this patient are essentially due to the G6PD c.1225C>T mutation. The boy gained transfusion independence after splenectomy.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica , Deficiencia de Glucosafosfato Deshidrogenasa , Talasemia beta , Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/genética , Niño , Glucosafosfato Deshidrogenasa/genética , Deficiencia de Glucosafosfato Deshidrogenasa/complicaciones , Deficiencia de Glucosafosfato Deshidrogenasa/genética , Humanos , Masculino , Esplenectomía , Talasemia beta/complicaciones , Talasemia beta/genética , Talasemia beta/cirugía
5.
Cells ; 11(7)2022 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-35406697

RESUMEN

Background: Hereditary spherocytosis (HS) and pyruvate kinase deficiency (PKD) are the most common causes of hereditary chronic hemolytic anemia. Here, we describe clinical and genetic characteristics of a Spanish family with concomitant ß-spectrin (SPTB) c.647G>A variant and pyruvate kinase (PKLR) c.1706G>A variant. Methods: A family of 11 members was studied. Hematological investigation, hemolysis tests, and specific red cell studies were performed in all family members, according to conventional procedures. An ektacytometric study was performed using the osmoscan module of the Lorca ektacytometer (MaxSis. RR Mechatronics). The presence of the SPTB and PKLR variants was confirmed by t-NGS. Results: The t-NGS genetic characterization of the 11 family members showed the presence of a heterozygous mutation for the ß-spectrin (SPTB; c.647G>A) in seven members with HS, three of them co-inherited the PKLR variant c.1706G>A. In the remaining four members, no gene mutation was found. Ektacytometry allowed a clear diagnostic orientation of HS, independently from the PKLR variant. Conclusions: This family study allows concluding that the SPTB mutation, (c.647G>A) previously described as likely pathogenic (LP), should be classified as pathogenic (P), according to the recommendations for pathogenicity of the American College of Medical Genetics and the Association for Molecular Pathology. In addition, after 6 years of clinical follow-up of the patients with HS, it can be inferred that the chronic hemolytic anemia may be attributable to the SPTB mutation only, without influence of the concomitant PKLR. Moreover, only the family members with the SPTB mutation exhibited an ektacytometric profile characteristic of HS.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica , Esferocitosis Hereditaria , Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/diagnóstico , Anemia Hemolítica Congénita no Esferocítica/genética , Humanos , Rayos Láser , Piruvato Quinasa/deficiencia , Piruvato Quinasa/genética , Errores Innatos del Metabolismo del Piruvato , Espectrina/genética , Esferocitosis Hereditaria/complicaciones , Esferocitosis Hereditaria/diagnóstico , Esferocitosis Hereditaria/genética
6.
Blood Adv ; 6(6): 1844-1853, 2022 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-34470054

RESUMEN

Pyruvate kinase deficiency (PKD) is the most common cause of congenital nonspherocytic hemolytic anemia. Although recognition of the disease spectrum has recently expanded, data describing its impact on health-related quality of life (HRQoL) are limited. In this prospective international cohort of 254 patients (131 adults and 123 children) with PKD, we used validated measures to assess the impact of disease on HRQoL (EuroQol 5-Dimension Questionnaire, Pediatric Quality of Life Inventory Generic Core Scale version 4.0, and Functional Assessment of Cancer Therapy-Anemia) and fatigue (Patient Reported Outcomes Measurement Information System Fatigue and Pediatric Functional Assessment of Chronic Illness Therapy-Fatigue). Significant variability in HRQoL and fatigue was reported for adults and children, although individual scores were stable over a 2-year interval. Although adults who were regularly transfused reported worse HRQoL and fatigue compared with those who were not (EuroQol-visual analog scale, 58 vs 80; P = .01), this difference was not seen in children. Regularly transfused adults reported lower physical, emotional, and functional well-being and more anemia symptoms. HRQoL and fatigue significantly differed in children by genotype, with the worst scores in those with 2 severe PKLR mutations; this difference was not seen in adults. However, iron chelation was associated with significantly worse HRQoL scores in children and adults. Pulmonary hypertension was also associated with significantly worse HRQoL. Additionally, 59% of adults and 35% of children reported that their jaundice upset them, identifying this as an important symptom for consideration. Although current treatments for PKD are limited to supportive care, new therapies are in clinical trials. Understanding the impact of PKD on HRQoL is important to assess the utility of these treatments. This trial was registered at www.clinicaltrials.gov as #NCT02053480.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica , Adulto , Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/diagnóstico , Anemia Hemolítica Congénita no Esferocítica/terapia , Niño , Fatiga/etiología , Humanos , Estudios Prospectivos , Piruvato Quinasa/deficiencia , Errores Innatos del Metabolismo del Piruvato , Calidad de Vida
7.
J Pediatr Hematol Oncol ; 43(4): e605-e607, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32590423

RESUMEN

We present the case of a successful liver transplant in a young adult patient with cholestasis and cirrhosis secondary to severe pyruvate kinase (PK) deficiency. Liver transplant resulted in resolution of liver dysfunction, decreased need for blood transfusions and eligibility for bone marrow transplantation. This case represents the third reported patient in the literature with severe PK deficiency who successfully underwent liver transplant as a result of profound cholestasis and liver failure. Explant pathology demonstrated a lack of significant iron deposition indicating that PK deficiency predisposes the liver to injury independent of transfusion-related iron overload.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica/complicaciones , Cirrosis Hepática/etiología , Cirrosis Hepática/terapia , Trasplante de Hígado , Piruvato Quinasa/deficiencia , Errores Innatos del Metabolismo del Piruvato/complicaciones , Adolescente , Anemia Hemolítica Congénita no Esferocítica/patología , Colestasis/etiología , Colestasis/patología , Colestasis/terapia , Femenino , Humanos , Cirrosis Hepática/patología , Errores Innatos del Metabolismo del Piruvato/patología , Resultado del Tratamiento
9.
J Appl Lab Med ; 5(1): 54-61, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32445340

RESUMEN

BACKGROUND: Pyruvate kinase (PK) deficiency is the most common cause of nonspherocytic hemolytic anemia owing to defective glycolysis. This study developed and validated an automated method to measure PK activity in red blood cells (RBCs). METHODS: PK catalyzes the reaction of phosphoenolpyruvate with ADP to form pyruvate and ATP. The pyruvate is reduced in the presence of lactate dehydrogenase and NADH to produce lactate and NAD+. The rate of absorbance decrease at 340 nm is proportional to PK activity. PK and hemoglobin (Hb) measurements were performed on a Roche cobas c501 analyzer. After establishing a k-factor, accuracy, linearity, imprecision, sensitivity, and stability were validated and the reference interval was verified. RESULTS: The k-factor was -9477. Accuracy was evaluated by method comparison (n = 56). Linear regression yielded y = 1.0x - 0.57, and R2 of 0.93. Linearity was determined by combining a high sample with hemolyzing solution in 6 different ratios. Linear regression analysis yielded y = 1.02x - 2.68, and R2 of 1.0. The assay was linear to 87 U/dL. Precision was evaluated by testing hemolysates in 3 replicates/day for 10 days. Within-run imprecision was 1.9% and 2.5% and total imprecision was 4.0% and 5.6% at 14.0 and 8.1 U/g Hb, respectively. The limit of blank was 0.0, and the limit of detection was 1.0 U/dL. Stability was determined in 4 sample types at 3 different temperatures; the changes were all <10% when compared with t0. The current PK reference interval of 4.6 to 11.2 U/g Hb was verified. CONCLUSIONS: This automated assay for quantifying PK in RBCs has acceptable performance characteristics and is fit for intended use.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica/sangre , Anemia Hemolítica/diagnóstico , Eritrocitos/enzimología , Hemoglobinas/análisis , Piruvato Quinasa/análisis , Piruvato Quinasa/deficiencia , Errores Innatos del Metabolismo del Piruvato/sangre , Anemia Hemolítica/etiología , Anemia Hemolítica Congénita no Esferocítica/complicaciones , Automatización de Laboratorios/métodos , Técnicas de Química Analítica , Humanos , Límite de Detección , Piruvato Quinasa/sangre , Errores Innatos del Metabolismo del Piruvato/complicaciones , Reproducibilidad de los Resultados
10.
Br J Haematol ; 184(5): 721-734, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30681718

RESUMEN

Novel therapies in development have brought a new focus on pyruvate kinase deficiency (PKD), the most common congenital haemolytic anaemia due to a glycolytic enzyme deficiency. With an improved recognition of its clinical presentation and understanding of the diagnostic pathway, more patients are likely to be identified with this anaemia. Complications, including gallstones and non-transfusion-related iron overload, require monitoring for early diagnosis and management. Current management remains supportive with red cell transfusions, chelation and splenectomy. Decisions to transfuse and/or splenectomise must be individualised. Haematopoietic stem cell transplant has been pursued in a small number of patients with mixed outcomes. Novel treatment approaches, which range from a small molecule pyruvate kinase activator to gene therapy, may transform the way in which PKD is managed in the future. In this review, we discuss the pathophysiology of PKD and present our approaches to diagnosis, monitoring and management of patients with this anaemia.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica/terapia , Transfusión de Eritrocitos , Piruvato Quinasa/deficiencia , Errores Innatos del Metabolismo del Piruvato/terapia , Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/diagnóstico , Cálculos Biliares/etiología , Cálculos Biliares/terapia , Humanos , Sobrecarga de Hierro/etiología , Sobrecarga de Hierro/terapia , Errores Innatos del Metabolismo del Piruvato/complicaciones , Errores Innatos del Metabolismo del Piruvato/diagnóstico
11.
Ann Biol Clin (Paris) ; 76(5): 493-503, 2018 10 01.
Artículo en Francés | MEDLINE | ID: mdl-30226192

RESUMEN

The most frequent causes of hemolytic anemias are immune or infectious diseases, drug induced hemolysis, thrombotic microangiopathies, hereditary spherocytosis, glucose-6-phosphate dehydrogenase or pyruvate kinase deficiencies, thalassemia's and sickle cell disease. Sometimes no cause is found because a rarer etiology is involved. The goal of this review is to remember some unfrequent constitutional or acquired causes and to point out difficulties to avoid wrong interpretations of analysis results.


Asunto(s)
Anemia Hemolítica/diagnóstico , Anemia Hemolítica/etiología , Anemia Hemolítica/sangre , Anemia Hemolítica Congénita no Esferocítica/sangre , Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/diagnóstico , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/complicaciones , Anemia de Células Falciformes/diagnóstico , Diagnóstico Diferencial , Deficiencia de Glucosafosfato Deshidrogenasa/sangre , Deficiencia de Glucosafosfato Deshidrogenasa/complicaciones , Deficiencia de Glucosafosfato Deshidrogenasa/diagnóstico , Pruebas Hematológicas/métodos , Pruebas Hematológicas/normas , Humanos , Piruvato Quinasa/sangre , Piruvato Quinasa/deficiencia , Errores Innatos del Metabolismo del Piruvato/sangre , Errores Innatos del Metabolismo del Piruvato/complicaciones , Errores Innatos del Metabolismo del Piruvato/diagnóstico , Esferocitosis Hereditaria/sangre , Esferocitosis Hereditaria/complicaciones , Esferocitosis Hereditaria/diagnóstico , Microangiopatías Trombóticas/sangre , Microangiopatías Trombóticas/complicaciones , Microangiopatías Trombóticas/diagnóstico
12.
Dis Model Mech ; 11(5)2018 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-29720471

RESUMEN

In this study, we performed a genome-wide N-ethyl-N-nitrosourea (ENU) mutagenesis screen in mice to identify novel genes or alleles that regulate erythropoiesis. Here, we describe a recessive mouse strain, called RBC19, harbouring a point mutation within the housekeeping gene, Tpi1, which encodes the glycolysis enzyme, triosephosphate isomerase (TPI). A serine in place of a phenylalanine at amino acid 57 severely diminishes enzyme activity in red blood cells and other tissues, resulting in a macrocytic haemolytic phenotype in homozygous mice, which closely resembles human TPI deficiency. A rescue study was performed using bone marrow transplantation of wild-type donor cells, which restored all haematological parameters and increased red blood cell enzyme function to wild-type levels after 7 weeks. This is the first study performed in a mammalian model of TPI deficiency, demonstrating that the haematological phenotype can be rescued.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/genética , Anemia Hemolítica/complicaciones , Anemia Hemolítica/terapia , Trasplante de Médula Ósea , Errores Innatos del Metabolismo de los Carbohidratos/complicaciones , Errores Innatos del Metabolismo de los Carbohidratos/genética , Mutagénesis , Triosa-Fosfato Isomerasa/deficiencia , Anemia Hemolítica/sangre , Anemia Hemolítica Congénita no Esferocítica/sangre , Animales , Errores Innatos del Metabolismo de los Carbohidratos/sangre , Modelos Animales de Enfermedad , Eritrocitos/metabolismo , Etilnitrosourea , Glucólisis , Homocigoto , Ratones , Ratones Mutantes , Mutación Missense/genética , Fenotipo , Triosa-Fosfato Isomerasa/sangre , Triosa-Fosfato Isomerasa/genética
13.
Pediatrics ; 141(Suppl 5): S385-S389, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29610156

RESUMEN

Pyruvate kinase deficiency (PKD) is the most common cause of congenital nonspherocytic chronic hemolytic anemia, and patients normally present with mild to severe anemia, unconjugated hyperbilirubinemia, and splenomegaly. Only a few reports of PKD have documented its association with severe, progressive liver failure. In all those cases, the patients died before liver transplant (LT) or immediately after transplant. We report 2 case patients with liver failure associated with PKD who successfully underwent LT and splenectomy: an infant who presented with neonatal cholestasis and a young adult with a severe form of PKD and having been transfusion dependent during childhood. After transplant, both patients have normal liver function test results and have considerably decreased their need for blood transfusion despite ongoing, mild hemolysis. We suggest that PKD can lead to severe liver dysfunction and that LT and splenectomy can be life-saving procedures in such cases.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/cirugía , Fallo Hepático/etiología , Fallo Hepático/cirugía , Trasplante de Hígado , Piruvato Quinasa/deficiencia , Errores Innatos del Metabolismo del Piruvato/complicaciones , Errores Innatos del Metabolismo del Piruvato/cirugía , Esplenectomía , Anemia Neonatal/etiología , Transfusión Sanguínea , Colestasis/etiología , Humanos , Lactante , Masculino , Adulto Joven
14.
Haematologica ; 103(2): e82-e86, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29242305
16.
Pediatrics ; 136(5): e1366-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26459649

RESUMEN

Unexpected severe cholestasis is part of the presentation in some neonates with hemolytic anemia but is usually self-resolving. Here we report the case of a neonate with pyruvate kinase deficiency (PKD) who presented severe hemolytic anemia at birth, characterized by a rapidly progressive and severe cholestasis with normal γ-glutamyl transpeptidase level associated with hepatic failure. After an extensive investigation to rule out contributing conditions explaining the severity of this patient's clinical presentation, PKD has remained the sole identified etiology. The patient abruptly died of sepsis at 3 months of age before a planned splenectomy and ongoing evaluation for liver transplantation. To the best of our knowledge, only a few similar cases of severe neonatal presentation of PKD complicated with severe hepatic failure and cholestasis have been reported.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica/complicaciones , Colestasis/etiología , Fallo Hepático/etiología , Piruvato Quinasa/deficiencia , Errores Innatos del Metabolismo del Piruvato/complicaciones , Humanos , Recién Nacido , Masculino , Índice de Severidad de la Enfermedad
18.
Am J Hematol ; 90(3): E35-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25388786

RESUMEN

In a family with mild dominant spherocytosis, affected members showed partial band 3 deficiency. The index patient showed more severe clinical symptoms than his relatives, and his red blood cells displayed concomitant low pyruvate kinase activity. We investigated the contribution of partial PK deficiency to the phenotypic expression of mutant band 3 in this family. Pyruvate kinase deficiency and band 3 deficiency were characterized by DNA analysis. Results of red cell osmotic fragility testing, the results of cell deformability obtained by the Automated Rheoscope and Cell Analyzer and the results obtained by Osmotic Gradient Ektacytometry, which is a combination of these tests, were related to the red cell ATP content. Spherocytosis in this family was due to a novel heterozygous mutation in SLC4A1, the gene for band 3. Reduced PK activity of the index patient was attributed to a novel mutation in PKLR inherited from his mother, who was without clinical symptoms. Partial PK deficiency was associated with decreased red cell ATP content and markedly increased osmotic fragility. This suggests an aggravating effect of low ATP levels on the phenotypic expression of band 3 deficiency.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica/genética , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Ancirinas/deficiencia , Mutación , Fenotipo , Piruvato Quinasa/deficiencia , Piruvato Quinasa/genética , Errores Innatos del Metabolismo del Piruvato/genética , Esferocitosis Hereditaria/genética , Adenosina Trifosfato/metabolismo , Adulto , Anciano , Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/metabolismo , Anemia Hemolítica Congénita no Esferocítica/patología , Proteína 1 de Intercambio de Anión de Eritrocito/deficiencia , Ancirinas/genética , Ancirinas/metabolismo , Deformación Eritrocítica , Eritrocitos/metabolismo , Eritrocitos/patología , Femenino , Expresión Génica , Genotipo , Heterocigoto , Humanos , Patrón de Herencia , Masculino , Persona de Mediana Edad , Fragilidad Osmótica , Linaje , Piruvato Quinasa/metabolismo , Errores Innatos del Metabolismo del Piruvato/complicaciones , Errores Innatos del Metabolismo del Piruvato/metabolismo , Errores Innatos del Metabolismo del Piruvato/patología , Esferocitosis Hereditaria/complicaciones , Esferocitosis Hereditaria/metabolismo , Esferocitosis Hereditaria/patología
19.
Fetal Pediatr Pathol ; 33(4): 234-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24840153

RESUMEN

We have reported the first Tunisian case of triosephosphate isomerase (TPI) deficiency in a 2-year-old girl. She was the first child of a nonconsanguineous couple. The disease included a neonatal onset of chronic hemolytic anemia, recurrent low-respiratory infections then progressive neurological involvement. The diagnosis was made after her death from the TPI values of her parents who exhibited intermediate enzyme deficiency. Molecular study of TPI genes showed that the father and the mother are heterozygous for Glu105Asp mutation. Pediatricians must be alert to the differential diagnosis in patients having hemolytic anemia and other concomitant manifestations.


Asunto(s)
Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/diagnóstico , Errores Innatos del Metabolismo de los Carbohidratos/complicaciones , Errores Innatos del Metabolismo de los Carbohidratos/diagnóstico , Enfermedades Neuromusculares/etiología , Triosa-Fosfato Isomerasa/deficiencia , Sustitución de Aminoácidos , Anemia Hemolítica Congénita no Esferocítica/genética , Errores Innatos del Metabolismo de los Carbohidratos/genética , Preescolar , Diagnóstico Diferencial , Resultado Fatal , Femenino , Genes Recesivos , Humanos , Lactante , Recién Nacido , Masculino , Mutación Missense , Padres , Triosa-Fosfato Isomerasa/genética , Túnez
20.
Pediatr Blood Cancer ; 61(8): 1463-5, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24481986

RESUMEN

Pyruvate kinase (PK) deficiency is the commonest enzyme deficiency in the glycolytic pathway leading to hemolytic anemia secondary to decreased Adenosine Triphosphate (ATP) synthesis in the red cells. synthesis. PK deficiency due to mutations in the PKLR (1q21) gene leads to highly variable clinical presentation ranging from severe fetal anemia to well compensated anemia in adults. We describe dyserythropoiesis in the bone marrow of a child with transfusion dependent anemia and unilateral multicystic dysplastic kidney (MCDK) mimicking Congenital Dyserythropoietic Anemia type I (CDA type I). Persistently low erythrocyte PK levels and double heterozygous mutations present in the PKLR gene confirmed the diagnosis of PK deficiency.


Asunto(s)
Anemia Diseritropoyética Congénita , Anemia Hemolítica Congénita no Esferocítica , Riñón Displástico Multiquístico , Mutación , Piruvato Quinasa/deficiencia , Errores Innatos del Metabolismo del Piruvato , Adulto , Anemia Diseritropoyética Congénita/complicaciones , Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/patología , Anemia Hemolítica Congénita no Esferocítica/complicaciones , Anemia Hemolítica Congénita no Esferocítica/genética , Anemia Hemolítica Congénita no Esferocítica/patología , Femenino , Humanos , Recién Nacido , Masculino , Riñón Displástico Multiquístico/complicaciones , Riñón Displástico Multiquístico/genética , Riñón Displástico Multiquístico/patología , Piruvato Quinasa/genética , Errores Innatos del Metabolismo del Piruvato/complicaciones , Errores Innatos del Metabolismo del Piruvato/genética , Errores Innatos del Metabolismo del Piruvato/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA