Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
BMC Genomics ; 24(1): 304, 2023 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-37280519

RESUMEN

BACKGROUND: Hereditary spherocytosis (HS) is a common inherited hemolytic anemia, caused by mutations in five genes that encode erythrocyte membrane skeleton proteins. The red blood cell (RBC) lifespan could directly reflect the degree of hemolysis. In the present cohort of 23 patients with HS, we performed next-generation sequencing (NGS) and Levitt's carbon monoxide (CO) breath test to investigate the potential genotype-degree of hemolysis correlation. RESULTS: In the present cohort, we identified 8 ANK1,9 SPTB,5 SLC4A1 and 1 SPTA1 mutations in 23 patients with HS, and the median RBC lifespan was 14(8-48) days. The median RBC lifespan of patients with ANK1, SPTB and SLC4A1 mutations was 13 (8-23), 13 (8-48) and 14 (12-39) days, respectively, with no statistically significant difference (P = 0.618). The median RBC lifespan of patients with missense, splice and nonsense/insertion/deletion mutations was 16.5 (8-48), 14 (11-40) and 13 (8-20) days, respectively, with no significant difference (P = 0.514). Similarly, we found no significant difference in the RBC lifespan of patients with mutations located in the spectrin-binding domain and the nonspectrin-binding domain [14 (8-18) vs. 12.5 (8-48) days, P = 0.959]. In terms of the composition of mutated genes, 25% of patients with mild hemolysis carried ANK1 or SPTA1 mutations, while 75% of patients with mild hemolysis carried SPTB or SLC4A1 mutations. In contrast, 46.7% of patients with severe hemolysis had ANK1 or SPTA1 mutations and 53.3% of patients with severe hemolysis had SPTB or SLC4A1 mutations. However, there was no statistically significant difference in the distribution of mutated genes between the two groups (P = 0.400). CONCLUSION: The present study is the first to investigate the potential association between genotype and degree of hemolysis in HS. The present findings indicated that there is no significant correlation between genotype and degree of hemolysis in HS.


Asunto(s)
Hemólisis , Esferocitosis Hereditaria , Humanos , Ancirinas/genética , Ancirinas/metabolismo , Espectrina/genética , Espectrina/metabolismo , Esferocitosis Hereditaria/genética , Esferocitosis Hereditaria/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas de la Membrana/genética , Mutación , Genotipo
2.
Mol Genet Genomics ; 298(2): 427-439, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36598564

RESUMEN

Hereditary Spherocytosis (HS) is a common cause of hemolytic anemia varying from mild to severe hemolysis due to defects in red cell membrane protein genes, namely ANK1, SPTB, SPTA1, SLC4A1, and EPB42. These genes are considerably very large spaning 40-50 exons making gene-by-gene analysis costly and laborious by conventional methods. In this study, we explored 26 HS patients harboring 21 ANK1 variants identified by next-generation sequencing (NGS), characteristics and spectrum of the detected ANK1variants were analyzed in this study. Clinically, all the HS patients showed moderate to severe transfusion-dependent hemolytic anemia, some requiring splenectomy. We identified 13 novel and 8 reported variants, mainly 9 frameshifts, 2 missense, 6 nonsense, and 4 splice site ANK1 variants, using NGS technology. Frameshifts were remarkably the most common variant type seen in Indian HS patients with ANK1 gene defects. We have also explored expression levels of red cell membrane ankyrin protein by flow cytometry in 14 HS patients with ANK1 gene defects and a significant reduction in ankyrin protein expression has been found. This report mainly illustrates the molecular and phenotypic heterogeneity of ANK1 variants causing HS in Indian patients. Ankyrin-1 mutations are a significant cause of loss of function in dominant HS in the Indian population. Comprehensive genetic and phenotypic evaluation assists in implementing the knowledge of genetic patterns and spectrum of ANK1 gene variants, providing molecular support for HS diagnosis.


Asunto(s)
Ancirinas , Esferocitosis Hereditaria , Humanos , Ancirinas/genética , Ancirinas/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas de la Membrana/genética , Mutación , Esferocitosis Hereditaria/genética , Esferocitosis Hereditaria/diagnóstico , Esferocitosis Hereditaria/metabolismo
3.
Cell Physiol Biochem ; 55(1): 117-129, 2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33667330

RESUMEN

BACKGROUND/AIMS: Hereditary Spherocytosis (HS) is the most common erythrocyte membrane disorder causing hemolytic anemia. The wide heterogeneity of both clinical and laboratory manifestations of HS contributes to difficulties associated with the diagnosis of this disorder. Although massive data previously reported worldwide, there is yet no data on HS among the Tunisian population. Here we aim to characterize HS in Tunisian patients at biochemical and cellular levels, identify the membrane protein deficiency, and compare the accuracy of the diagnostic tests to identify the most appropriate assay for HS diagnosis. METHODS: We investigated 81 patients with hemolytic anemia and 167 normal controls. The exploration of HS based on clinical and family history, physical examination, and the results of laboratory tests: blood smear, osmotic fragility test (OFT), cryohemolysis test (CT), pink test (PT), eosine-5'-maleimide (EMA) test, and erythrocyte membrane protein electrophoresis. RESULTS: We identified 21 patients with HS, classified as severe (6/21;28.5%), moderate (10/21;47.6%), and mild (5/21;23.8%). The most prevalent protein deficiency was the band 3 protein detected in ten Tunisian HS patients. The EMA test showed a high specificity (97.5%) and sensitivity (94.7%) for HS diagnosis compared to the other screening tests. Interestingly, fourteen among sixteen patients presenting with homozygous sickle cells HbSS showed an increase of EMA fluorescence intensity compared to other anemic patients. CONCLUSION: Our study highlights the efficiency of the EMA dye for the detection of HS whatever the nature of the involved protein deficiency. We report for the first time, the most prevalent protein deficiency among Tunisians with HS. Moreover, we found that the combination of the EMA-binding test with PT or incubated OFT improves the diagnosis sensitivity while maintaining a good specificity.


Asunto(s)
Eosina Amarillenta-(YS)/análogos & derivados , Membrana Eritrocítica , Citometría de Flujo , Proteínas de la Membrana/metabolismo , Adolescente , Adulto , Niño , Preescolar , Eosina Amarillenta-(YS)/química , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/patología , Femenino , Humanos , Lactante , Masculino , Fragilidad Osmótica , Proteómica , Esferocitosis Hereditaria/metabolismo , Esferocitosis Hereditaria/patología , Túnez
4.
J Cell Mol Med ; 23(6): 4454-4463, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31016877

RESUMEN

Hereditary spherocytosis (HS) is the most common inherited haemolytic anaemia disorder. ANK1 mutations account for most HS cases, but pathogenicity analysis and functional research have not been widely performed for these mutations. In this study, in order to confirm diagnosis, gene mutation was screened in two unrelated Chinese families with HS by a next-generation sequencing (NGS) panel and then confirmed by Sanger sequencing. Two novel heterozygous mutations (c.C841T, p.R281X and c.T290G, p.L97R) of the ANK1 gene were identified in the two families respectively. Then, the pathogenicity of the two new mutations and two previously reported ANK1 mutations (c.C648G, p.Y216X and c.G424T, p.E142X) were studied by in vitro experiments. The four mutations increased the osmotic fragility of cells, reduced the stabilities of ANK1 proteins and prevented the protein from localizing to the plasma membrane and interacting with SPTB and SLC4A1. We classified these four mutations into disease-causing mutations for HS. Thus, conducting the same mutation test and providing genetic counselling for the two families were meaningful and significant. Moreover, the identification of two novel mutations enriches the ANK1 mutation database, especially in China.


Asunto(s)
Ancirinas/genética , Ancirinas/metabolismo , Pueblo Asiatico/genética , Mutación con Pérdida de Función , Esferocitosis Hereditaria/genética , Esferocitosis Hereditaria/patología , Secuencia de Aminoácidos , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Ancirinas/química , Niño , Preescolar , Femenino , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , Masculino , Linaje , Conformación Proteica , Estabilidad Proteica , Homología de Secuencia , Espectrina/metabolismo , Esferocitosis Hereditaria/metabolismo
5.
Acta Haematol ; 139(1): 60-66, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29402830

RESUMEN

With the widespread use of genetic diagnostic technologies, many novel mutations have been identified in hereditary spherocytosis (HS)-related genes, including SPTA1, SPTB, ANK1, SLC4A1, and EPB42. However, mutations in HS-related genes are dispersed and nonspecific in the diagnosis of some HS patients, indicating significant heterogeneity in the molecular deficiency of HS. It is necessary to provide the molecular and genetic characteristics of these 5 genes for clinicians to examine HS. Here, we reviewed the recent proposed molecular genetic mechanisms of HS.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Esferocitosis Hereditaria/genética , Biomarcadores , Humanos , Mutación , Esferocitosis Hereditaria/diagnóstico , Esferocitosis Hereditaria/metabolismo
6.
Haematologica ; 101(9): 1018-27, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27247322

RESUMEN

Ankyrin-R provides a key link between band 3 and the spectrin cytoskeleton that helps to maintain the highly specialized erythrocyte biconcave shape. Ankyrin deficiency results in fragile spherocytic erythrocytes with reduced band 3 and protein 4.2 expression. We use in vitro differentiation of erythroblasts transduced with shRNAs targeting ANK1 to generate erythroblasts and reticulocytes with a novel ankyrin-R 'near null' human phenotype with less than 5% of normal ankyrin expression. Using this model, we demonstrate that absence of ankyrin negatively impacts the reticulocyte expression of a variety of proteins, including band 3, glycophorin A, spectrin, adducin and, more strikingly, protein 4.2, CD44, CD47 and Rh/RhAG. Loss of band 3, which fails to form tetrameric complexes in the absence of ankyrin, alongside GPA, occurs due to reduced retention within the reticulocyte membrane during erythroblast enucleation. However, loss of RhAG is temporally and mechanistically distinct, occurring predominantly as a result of instability at the plasma membrane and lysosomal degradation prior to enucleation. Loss of Rh/RhAG was identified as common to erythrocytes with naturally occurring ankyrin deficiency and demonstrated to occur prior to enucleation in cultures of erythroblasts from a hereditary spherocytosis patient with severe ankyrin deficiency but not in those exhibiting milder reductions in expression. The identification of prominently reduced surface expression of Rh/RhAG in combination with direct evaluation of ankyrin expression using flow cytometry provides an efficient and rapid approach for the categorization of hereditary spherocytosis arising from ankyrin deficiency.


Asunto(s)
Ancirinas/deficiencia , Proteínas Sanguíneas/metabolismo , Eritroblastos/metabolismo , Membrana Eritrocítica/metabolismo , Lisosomas/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteína 1 de Intercambio de Anión de Eritrocito/química , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Diferenciación Celular/genética , Células Cultivadas , Citoesqueleto/genética , Citoesqueleto/metabolismo , Eritroblastos/química , Eritroblastos/citología , Eritropoyesis/genética , Regulación de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Mutación , Unión Proteica , Multimerización de Proteína , Proteolisis , Esferocitosis Hereditaria/genética , Esferocitosis Hereditaria/metabolismo
9.
Haematologica ; 100(1): 133-42, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25344524

RESUMEN

Band 3 is the most abundant protein in the erythrocyte membrane and forms the core of a major multiprotein complex. The absence of band 3 in human erythrocytes has only been reported once, in the homozygous band 3 Coimbra patient. We used in vitro culture of erythroblasts derived from this patient, and separately short hairpin RNA-mediated depletion of band 3, to investigate the development of a band 3-deficient erythrocyte membrane and to specifically assess the stability and retention of band 3 dependent proteins in the absence of this core protein during terminal erythroid differentiation. Further, using lentiviral transduction of N-terminally green fluorescent protein-tagged band 3, we demonstrated the ability to restore expression of band 3 to normal levels and to rescue secondary deficiencies of key proteins including glycophorin A, protein 4.2, CD47 and Rh proteins arising from the absence of band 3 in this patient. By transducing band 3-deficient erythroblasts from this patient with band 3 mutants with absent or impaired ability to associate with the cytoskeleton we also demonstrated the importance of cytoskeletal connectivity for retention both of band 3 and of its associated dependent proteins within the reticulocyte membrane during the process of erythroblast enucleation.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Ancirinas/deficiencia , Citoesqueleto/metabolismo , Eritroblastos/metabolismo , Membrana Eritrocítica/metabolismo , Eritropoyesis/fisiología , Complejos Multiproteicos/metabolismo , Esferocitosis Hereditaria/metabolismo , Proteína 1 de Intercambio de Anión de Eritrocito/antagonistas & inhibidores , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Ancirinas/genética , Ancirinas/metabolismo , Estudios de Casos y Controles , Diferenciación Celular , Células Cultivadas , Eritroblastos/citología , Citometría de Flujo , Homocigoto , Humanos , Fenotipo , Unión Proteica , ARN Interferente Pequeño/genética , Reticulocitos/citología , Reticulocitos/metabolismo , Esferocitosis Hereditaria/genética , Esferocitosis Hereditaria/patología
11.
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
12.
Biometals ; 28(5): 913-28, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26292972

RESUMEN

Iron is an essential element for fundamental cell functions and a catalyst for chemical reactions. Three samples extracted from the human spleen were investigated by scanning (SEM) and transmission electron microscopy (TEM), Mössbauer spectrometry (MS), and SQUID magnetometry. The sample with diagnosis of hemosiderosis (H) differs from that referring to hereditary spherocytosis and the reference sample. SEM reveals iron-rich micrometer-sized aggregate of various structures-tiny fibrils in hereditary spherocytosis sample and no fibrils in hemochromatosis. Hematite and magnetite particles from 2 to 6 µm in TEM with diffraction in all samples were shown. The SQUID magnetometry shows different amount of diamagnetic, paramagnetic and ferrimagnetic structures in the tissues. The MS results indicate contribution of ferromagnetically split sextets for all investigated samples. Their occurrence indicates that at least part of the sample is magnetically ordered below the critical temperature. The iron accumulation process is different in hereditary spherocytosis and hemosiderosis. This fact may be the reason of different iron crystallization.


Asunto(s)
Compuestos Férricos/metabolismo , Hierro/química , Bazo/química , Autopsia , Cristalización , Compuestos Férricos/química , Óxido Ferrosoférrico/química , Hemosiderosis/metabolismo , Hemosiderosis/patología , Humanos , Hierro/metabolismo , Microscopía Electrónica de Transmisión , Espectroscopía de Mossbauer , Esferocitosis Hereditaria/metabolismo , Esferocitosis Hereditaria/patología , Bazo/metabolismo , Bazo/patología , Bazo/ultraestructura
13.
Rinsho Ketsueki ; 56(7): 837-45, 2015 Jul.
Artículo en Japonés | MEDLINE | ID: mdl-26251147

RESUMEN

Band 3 protein accounts for the largest percentage of whole erythrocyte membrane proteins. Abnormalities in this protein are closely associated with pathologies including hereditary spherocytosis (HS), Southeast Asian ovalocytosis and distant renal tubular acidosis. Currently, EMA binding capacity measurement in erythrocytes is the most useful screening test for diagnosing HS. We have also demonstrated reduced EMA binding capacity in patients with HS who have deficiencies of membrane proteins such as ankyrin not directly binding to EMA and who have as yet undetectable abnormalities of membrane proteins. However, even patients with hereditary elliptocytosis, who have a partial spectrin deficiency, were found to show reduced EMA binding capacity. Six of 7 had spherocytic elliptocytosis. Therefore, it is necessary to meticulously diagnose HS by ruling out all other possibilities.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/química , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Ancirinas/deficiencia , Membrana Eritrocítica/química , Membrana Eritrocítica/metabolismo , Esferocitosis Hereditaria/metabolismo , Proteína 1 de Intercambio de Anión de Eritrocito/deficiencia , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Ancirinas/química , Ancirinas/genética , Ancirinas/metabolismo , Humanos , Mutación , Unión Proteica , Esferocitosis Hereditaria/genética
14.
Blood ; 120(17): 3586-93, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22968456

RESUMEN

The human ankyrin-1 gene (ANK1) contains 3 tissue-specific alternative promoters. We have shown previously that the erythroid-specific ankyrin 1 (ANK1E) core promoter contains a 5' DNase I hypersensitive site (HS) with barrier insulator function that prevents gene silencing in vitro and in vivo. Mutations in the ANK1E barrier region lead to decreased ANK1 mRNA levels and hereditary spherocytosis. In this report, we demonstrate a second ANK1E regulatory element located in an adjacent pair of DNase I HS located 5.6 kb 3' of the ANK1E promoter at the 3' boundary of an erythroid-specific DNase I-sensitive chromatin domain. The 3' regulatory element exhibits enhancer activity in vitro and in transgenic mice, and it has the histone modifications associated with an enhancer element. One of the ANK1E 3'HS contains an NF-E2 binding site that is required for enhancer function. We show that a chromatin loop brings the 3' enhancer and NF-E2 into proximity with the 5' barrier region including the ANK1E core promoter. These observations demonstrate a model for the tissue-specific activation of alternative promoters that may be applicable to the ∼ 30% of mammalian genes with alternative promoters that exhibit distinct expression patterns.


Asunto(s)
Ancirinas/genética , Cromatina/genética , Elementos de Facilitación Genéticos , Elementos Aisladores , Subunidad p45 del Factor de Transcripción NF-E2/genética , Regiones Promotoras Genéticas , Esferocitosis Hereditaria/genética , Regiones no Traducidas 3' , Regiones no Traducidas 5' , Animales , Ancirinas/metabolismo , Sitios de Unión , Línea Celular Tumoral , Cromatina/química , Cromatina/metabolismo , Desoxirribonucleasa I/genética , Desoxirribonucleasa I/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Células K562 , Ratones , Ratones Transgénicos , Subunidad p45 del Factor de Transcripción NF-E2/metabolismo , Especificidad de Órganos , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Esferocitosis Hereditaria/metabolismo
15.
J Biol Chem ; 287(23): 19115-21, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22514283

RESUMEN

The conserved TPLH tetrapeptide motif of ankyrin repeats (ARs) plays an important role in stabilizing AR proteins, and histidine (TPLH)-to-arginine (TPLR) mutations in this motif have been associated with a hereditary human anemia, spherocytosis. Here, we used a combination of atomic force microscopy-based single-molecule force spectroscopy and molecular dynamics simulations to examine the mechanical effects of His → Arg substitutions in TPLH motifs in a model AR protein, NI6C. Our molecular dynamics results show that the mutant protein is less mechanically stable than the WT protein. Our atomic force microscopy results indicate that the mechanical energy input necessary to fully unfold the mutant protein is only half of that necessary to unfold the WT protein (53 versus 106 kcal/mol). In addition, the ability of the mutant to generate refolding forces is also reduced. Moreover, the mutant protein subjected to cyclic stretch-relax measurements displays mechanical fatigue, which is absent in the WT protein. Taken together, these results indicate that the His → Arg substitutions in TPLH motifs compromise mechanical properties of ARs and suggest that the origin of hereditary spherocytosis may be related to mechanical failure of ARs.


Asunto(s)
Repetición de Anquirina/genética , Modelos Moleculares , Mutación Missense , Sustitución de Aminoácidos , Humanos , Estructura Terciaria de Proteína , Esferocitosis Hereditaria/genética , Esferocitosis Hereditaria/metabolismo
17.
JCI Insight ; 8(20)2023 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-37676741

RESUMEN

Hereditary spherocytosis (HS) is the most common, nonimmune, hereditary, chronic hemolytic anemia after hemoglobinopathies. The genetic defects in membrane function causing HS lead to perturbation of the RBC metabolome, with altered glycolysis. In mice genetically lacking protein 4.2 (4.2-/-; Epb42), a murine model of HS, we showed increased expression of pyruvate kinase (PK) isoforms in whole and fractioned RBCs in conjunction with abnormalities in the glycolytic pathway and in the glutathione (GSH) system. Mitapivat, a PK activator, metabolically reprogrammed 4.2-/- mouse RBCs with amelioration of glycolysis and the GSH cycle. This resulted in improved osmotic fragility, reduced phosphatidylserine positivity, amelioration of RBC cation content, reduction of Na/K/Cl cotransport and Na/H-exchange overactivation, and decrease in erythroid vesicles release in vitro. Mitapivat treatment significantly decreased erythrophagocytosis and beneficially affected iron homeostasis. In mild-to-moderate HS, the beneficial effect of splenectomy is still controversial. Here, we showed that splenectomy improves anemia in 4.2-/- mice and that mitapivat is noninferior to splenectomy. An additional benefit of mitapivat treatment was lower expression of markers of inflammatory vasculopathy in 4.2-/- mice with or without splenectomy, indicating a multisystemic action of mitapivat. These findings support the notion that mitapivat treatment should be considered for symptomatic HS.


Asunto(s)
Anemia Hemolítica , Esferocitosis Hereditaria , Animales , Ratones , Modelos Animales de Enfermedad , Esferocitosis Hereditaria/genética , Esferocitosis Hereditaria/metabolismo , Eritrocitos/metabolismo , Anemia Hemolítica/genética , Anemia Hemolítica/metabolismo
18.
Zhonghua Xue Ye Xue Za Zhi ; 44(4): 316-320, 2023 Apr 14.
Artículo en Zh | MEDLINE | ID: mdl-37357001

RESUMEN

Objective: To report gene mutations in nine patients with hereditary elliptocytosis (HE) and analyze the characteristics of pathogenic gene mutations in HE. Methods: The clinical and gene mutations of nine patients clinically diagnosed with HE at Institute of Hematology & Blood Diseases Hospital from June 2018 to February 2022 were reported and verified by next-generation sequencing to analyze the relationship between gene mutations and clinical phenotypes. Results: Erythrocyte membrane protein gene mutations were detected among nine patients with HE, including six with SPTA1 mutation, one with SPTB mutation, one with EPB41 mutation, and one with chromosome 20 copy deletion. A total of 11 gene mutation sites were involved, including 6 known mutations and 5 novel mutations. The five novel mutations included SPTA1: c.1247A>C (p. K416T) in exon 9, c.1891delG (p. A631fs*17) in exon 15, E6-E12 Del; SPTB: c.154C>T (p. R52W) ; and EPB41: c.1636A>G (p. I546V) . Three of the six patients with the SPTA1 mutation were SPTA1 exon 9 mutation. Conclusion: SPTA1 is the most common mutant gene in patients with HE.


Asunto(s)
Eliptocitosis Hereditaria , Esferocitosis Hereditaria , Humanos , Mutación , Eliptocitosis Hereditaria/genética , Eliptocitosis Hereditaria/diagnóstico , Eliptocitosis Hereditaria/metabolismo , Membrana Eritrocítica/genética , Membrana Eritrocítica/metabolismo , Exones , Secuenciación de Nucleótidos de Alto Rendimiento , Esferocitosis Hereditaria/genética , Esferocitosis Hereditaria/metabolismo
19.
Blood ; 116(2): 267-9, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20339087

RESUMEN

During erythroblast enucleation, membrane proteins distribute between extruded nuclei and reticulocytes. In hereditary spherocytosis (HS) and hereditary elliptocytosis (HE), deficiencies of membrane proteins, in addition to those encoded by the mutant gene, occur. Elliptocytes, resulting from protein 4.1R gene mutations, lack not only 4.1R but also glycophorin C, which links the cytoskeleton and bilayer. In HS resulting from ankyrin-1 mutations, band 3, Rh-associated antigen, and glycophorin A are deficient. The current study was undertaken to explore whether aberrant protein sorting, during enucleation, creates these membrane-spanning protein deficiencies. We found that although glycophorin C sorts to reticulocytes normally, it distributes to nuclei in 4.1R-deficient HE cells. Further, glycophorin A and Rh-associated antigen, which normally partition predominantly to reticulocytes, distribute to both nuclei and reticulocytes in an ankyrin-1-deficient murine model of HS. We conclude that aberrant protein sorting is one mechanistic basis for protein deficiencies in HE and HS.


Asunto(s)
Eliptocitosis Hereditaria/metabolismo , Eritroblastos/metabolismo , Eritropoyesis/fisiología , Proteínas de la Membrana/metabolismo , Esferocitosis Hereditaria/metabolismo , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Proteínas Sanguíneas/deficiencia , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Núcleo Celular/metabolismo , Eliptocitosis Hereditaria/genética , Eliptocitosis Hereditaria/patología , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/patología , Técnica del Anticuerpo Fluorescente , Glicoforinas/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Mutantes , Proteínas de Microfilamentos , Mutación , Transporte de Proteínas , Esferocitosis Hereditaria/genética , Esferocitosis Hereditaria/patología
20.
Biochem J ; 433(2): 313-22, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-21039340

RESUMEN

AE1 (anion exchanger 1) and protein 4.2 associate in a protein complex bridging the erythrocyte membrane and cytoskeleton; disruption of the complex results in unstable erythrocytes and HS (hereditary spherocytosis). Three HS mutations (E40K, G130R and P327R) in cdAE1 (the cytoplasmic domain of AE1) occur with deficiencies of protein 4.2. The interaction of wild-type AE1, AE1HS mutants, mdEA1 (the membrane domain of AE1), kAE1 (the kidney isoform of AE1) and AE1SAO (Southeast Asian ovalocytosis AE1) with protein 4.2 was examined in transfected HEK (human embryonic kidney)-293 cells. The HS mutants had wild-type expression levels and plasma-membrane localization. Protein 4.2 expression was not dependent on AE1. Protein 4.2 was localized throughout the cytoplasm and co-localized at the plasma membrane with the HS mutants mdAE1 and kAE1, but at the ER (endoplasmic reticulum) with AE1SAO. Pull-down assays revealed diminished levels of protein 4.2 associated with the HS mutants relative to AE1. The mdAE1 did not bind protein 4.2, whereas kAE1 and AE1SAO bound wild-type amounts of protein 4.2. A protein 4.2 fatty acylation mutant, G2A/C173A, had decreased plasma-membrane localization compared with wild-type protein 4.2, and co-expression with AE1 enhanced its plasma-membrane localization. Subcellular fractionation showed the majority of wild-type and G2A/C173A protein 4.2 was associated with the cytoskeleton of HEK-293 cells. The present study shows that cytoplasmic HS mutants cause impaired binding of protein 4.2 to AE1, leaving protein 4.2 susceptible to loss during erythrocyte development.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Citoplasma/metabolismo , Proteínas del Citoesqueleto/química , Proteínas del Citoesqueleto/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Mutación , Esferocitosis Hereditaria/metabolismo , Proteína 1 de Intercambio de Anión de Eritrocito/química , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Citoplasma/química , Proteínas del Citoesqueleto/genética , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Unión Proteica , Esferocitosis Hereditaria/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA