Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 287(6): 4129-38, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22147703

RESUMEN

Current models of the erythrocyte membrane depict three populations of band 3: (i) a population tethered to spectrin via ankyrin, (ii) a fraction attached to the spectrin-actin junctional complex via adducin, and (iii) a freely diffusing population. Because many studies of band 3 diffusion also distinguish three populations of the polypeptide, it has been speculated that the three populations envisioned in membrane models correspond to the three fractions observed in diffusion analyses. To test this hypothesis, we characterized band 3 diffusion by single-particle tracking in wild-type and ankyrin- and adducin-deficient erythrocytes. We report that ∼40% of total band 3 in wild-type murine erythrocytes is attached to ankyrin, whereas ∼33% is immobilized by adducin, and ∼27% is not attached to any cytoskeletal anchor. More detailed analyses reveal that mobilities of individual ankyrin- and adducin-tethered band 3 molecules are heterogeneous, varying by nearly 2 orders of magnitude and that there is considerable overlap in diffusion coefficients for adducin and ankyrin-tethered populations. Taken together, the data suggest that although the ankyrin- and adducin-immobilized band 3 can be monitored separately, significant heterogeneity still exists within each population, suggesting that structural and compositional properties likely vary considerably within each band 3 complex.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Ancirinas/metabolismo , Proteínas de Unión a Calmodulina/metabolismo , Membrana Eritrocítica/metabolismo , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Ancirinas/genética , Proteínas de Unión a Calmodulina/genética , Membrana Eritrocítica/genética , Humanos , Ratones , Ratones Mutantes , Unión Proteica/fisiología
2.
Exp Hematol ; 39(3): 305-20, 320.e1-2, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21193012

RESUMEN

OBJECTIVE: Hereditary spherocytosis (HS) is a heterogeneous group of spontaneously arising and inherited red blood cell disorders ranging from very mild subclinical cases to severe and life-threatening cases, with symptoms linked directly to the severity of the mutation at the molecular level. We investigated a novel mouse model in which the heterozygotes present with the diagnostic hallmarks of mild HS and surviving homozygotes phenocopy severe hemolytic HS. MATERIALS AND METHODS: We used N-ethyl-N-nitrosourea mutagenesis to generate random point mutations in the mouse genome and a dominant screen to identify mouse models of human hematopoietic disease. Gene mapping of the HS strain revealed a unique in-frame nonsense mutation arising from a single base transversion in exon 27 of Ank1 (strain designation: Ank1(E924X)). Employing conventional hematopoietic, pathological, biochemical, and cell biology assays, we characterized heterozygous and homozygous Ank1(E924X) mice at the biochemical, cellular, and pathophysiological levels. RESULTS: Although Ank1(E924X/E924X) red blood cell ghosts lack abundant full-length ankyrin-1 isoforms, N-terminal epitope ankyrin-1 antibodies reveal a band consistent with the theoretical size of a truncated mutant ankyrin-1. Using domain-specific antibodies, we further show that this protein lacks both a spectrin-binding domain and a C-terminal regulatory domain. Finally, using antisera that detect C-terminal residues of the products of alternative Ank1 transcripts, we find unique immunoreactive bands not observed in red blood cell ghosts from wild-type or Ank1(E924X) heterozygous mice, including a band similar in size to full-length ankyrin-1. CONCLUSIONS: The Ank1(E924X) strain provides a novel tool to study Ank1 and model HS.


Asunto(s)
Alquilantes/efectos adversos , Ancirinas , Codón sin Sentido , Eritrocitos/metabolismo , Etilnitrosourea/efectos adversos , Esferocitosis Hereditaria , Alquilantes/farmacología , Secuencia de Aminoácidos , Animales , Ancirinas/genética , Ancirinas/metabolismo , Modelos Animales de Enfermedad , Etilnitrosourea/farmacología , Femenino , Humanos , Masculino , Ratones , Ratones Mutantes , Unión Proteica , Eliminación de Secuencia , Esferocitosis Hereditaria/inducido químicamente , Esferocitosis Hereditaria/genética , Esferocitosis Hereditaria/metabolismo
3.
Blood ; 115(9): 1804-14, 2010 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-20056793

RESUMEN

Five spontaneous, allelic mutations in the alpha-spectrin gene, Spna1, have been identified in mice (spherocytosis [sph], sph(1J), sph(2J), sph(2BC), sph(Dem)). All cause severe hemolytic anemia. Here, analysis of 3 new alleles reveals previously unknown consequences of red blood cell (RBC) spectrin deficiency. In sph(3J), a missense mutation (H2012Y) in repeat 19 introduces a cryptic splice site resulting in premature termination of translation. In sph(Ihj), a premature stop codon occurs (Q1853Stop) in repeat 18. Both mutations result in markedly reduced RBC membrane spectrin content, decreased band 3, and absent beta-adducin. Reevaluation of available, previously described sph alleles reveals band 3 and adducin deficiency as well. In sph(4J), a missense mutation occurs in the C-terminal EF hand domain (C2384Y). Notably, an equally severe hemolytic anemia occurs despite minimally decreased membrane spectrin with normal band 3 levels and present, although reduced, beta-adducin. The severity of anemia in sph(4J) indicates that the highly conserved cysteine residue at the C-terminus of alpha-spectrin participates in interactions critical to membrane stability. The data reinforce the notion that a membrane bridge in addition to the classic protein 4.1-p55-glycophorin C linkage exists at the RBC junctional complex that involves interactions between spectrin, adducin, and band 3.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/deficiencia , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Proteínas de Unión a Calmodulina/deficiencia , Proteínas de Unión a Calmodulina/genética , Mutación , Espectrina/deficiencia , Espectrina/genética , Esferocitosis Hereditaria/sangre , Esferocitosis Hereditaria/genética , Alelos , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Proteínas de Unión a Calmodulina/sangre , Codón sin Sentido , Cartilla de ADN/genética , Membrana Eritrocítica/metabolismo , Eritrocitos Anormales/metabolismo , Eritrocitos Anormales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Mutantes , Datos de Secuencia Molecular , Mutación Missense , Estabilidad del ARN/genética , Homología de Secuencia de Aminoácido , Esferocitosis Hereditaria/patología
4.
Proc Natl Acad Sci U S A ; 101(49): 17222-7, 2004 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-15572441

RESUMEN

Rhesus (Rh) antigens are carried by a membrane complex that includes Rh proteins (D and CcEe), Rh-associated glycoproteins (RhAG), and accessory chains (LW and CD47) associated by noncovalent bonds. In heterologous expression systems, RhAG and its kidney orthologs function as ammonium transporters. In red blood cells (RBCs), it is generally accepted that NH(3) permeates by membrane lipid diffusion. We have revisited these issues by studying RBC and ghosts from human and mouse genetic variants with defects of proteins that comprise the Rh complex. In both normal and mutant cells, stopped-flow analyses of intracellular pH changes in the presence of inwardly directed methylammonium (CH(3)NH(+)(3)+CH(3)NH(2)) or ammonium (NH(+)(4)+NH(3)) gradients showed a rapid alkalinization phase. Cells from human and mouse variants exhibited a decrease in their kinetic rate constants that was strictly correlated to the degree of reduction of their RhAG/Rhag expression level. Rate constants were not affected by a reduction of Rh, CD47, or LW. CH(3)NH(2)/NH(3) transport was characterized by (i) a sensitivity to mercurials that is reversible by 2-mercaptoethanol and (ii) a reduction of alkalinization rate constants after bromelain digestion, which cleaves RhAG. The results show that RhAG facilitates CH(3)NH(2)/NH(3) movement across the RBC membrane and represents a potential example of a gas channel in mammalian cells. In RBCs, RhAG may transport NH(3) to detoxifying organs, like kidney and liver, and together with nonerythroid tissue orthologs may contribute to the regulation of the systemic acid-base balance.


Asunto(s)
Amoníaco/metabolismo , Proteínas Sanguíneas/fisiología , Eritrocitos/metabolismo , Glicoproteínas de Membrana/fisiología , Equilibrio Ácido-Base , Animales , Proteínas Sanguíneas/genética , Permeabilidad de la Membrana Celular , Expresión Génica , Variación Genética , Humanos , Concentración de Iones de Hidrógeno , Cinética , Glicoproteínas de Membrana/genética , Metilaminas/metabolismo , Ratones
5.
J Pathol ; 204(4): 450-9, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15495268

RESUMEN

Although the haemolytic anaemia may be the primary concern for hereditary spherocytosis and elliptocytosis patients, it is clear that their situation can be compromised by primary and secondary defects in erythroid and non-erythroid systems of the body. All seven of the red cell membrane skeletal proteins discussed in this review are also expressed in non-erythroid tissues, and mutations in their genes have the potential to cause non-erythroid defects. In some instances, such as the protein 4.1R and ANK1 neurological deficits, the diagnosis is clear. In other instances, because of the complex expression patterns involved, the non-erythroid effects may be difficult to assess. An example is the large multidomain, multifunctional band 3 protein. In this case, the location of the mutation can cause defects in one functional domain or isoform and not the other. In other cases, such as the beta-adducin null mutation, other isoforms may partially compensate for the primary deficiency. In such cases, it may be that the effects of the deficit are subtle but could increase under stress or with age. To be completely successful, treatment strategies must address both primary and secondary effects of the anaemia. If gene replacement therapy is to be used, the more that is known about the underlying genetic mechanisms producing the multiple isoforms the better we will be able to design the best replacement gene. The various animal models that are now available should be invaluable in this regard. They continue to contribute to our understanding of both the primary and the secondary effects and their treatment.


Asunto(s)
Anemia Hemolítica Congénita/genética , Proteínas Sanguíneas/genética , Eritrocitos/fisiología , Proteínas de la Membrana/genética , Ancirinas/genética , Proteínas Sanguíneas/metabolismo , Proteínas de Unión a Calmodulina/genética , Colelitiasis/genética , Colelitiasis/metabolismo , Proteínas del Citoesqueleto/genética , Citoesqueleto/genética , Citoesqueleto/metabolismo , Humanos , Leucocitosis/genética , Leucocitosis/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Espectrina/genética , Esplenomegalia/genética , Esplenomegalia/metabolismo , Trombosis/genética , Trombosis/metabolismo
6.
Hematol J ; 4(6): 445-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14671619

RESUMEN

Ankyrin deficiency is one of the most common causes of hereditary spherocytosis in humans. A spontaneous mutation, normoblastosis (Ank1nb), discovered in 1969 in a mouse stock maintained at the Jackson Laboratory, provides an important animal model for these human ankyrin-deficient anemias. Study of this model has led to the finding of multiple isoforms of Ank1 as well as Ank1nb-related pathology in nonerythroid tissues. To enhance the usefulness of this model, we have identified the Ank1nb mutation as the deletion of a guanosine residue in exon 36 of the erythroid ankyrin gene (Ank1). This results in a frame shift that introduces a stop 13 codons downstream and predicts a 157 kDa nb-ankyrin lacking the regulatory domain but including intact membrane- and spectrin-binding domains. By epitope scanning on immunoblots, we show that a previously reported protein (p150) found in nb reticulocytes is the predicted nb-ankyrin. Existing evidence indicates that this protein is functional, making the normoblastosis mutation a hypomorph rather than a null as originally thought. The nb-ankyrin provides an explanation for the milder phenotype displayed by nb/nb animals relative to the murine spectrin-deficient anemias, spherocytosis (Spna1(sph), Spna1(sph-1J), Spna1(sph-2BC), Spna1(sph-DEM)) and jaundiced (Spnb1(ja)), and suggests that truncated ankyrins could be useful in gene replacement therapy.


Asunto(s)
Anemia Hemolítica/genética , Ancirinas/genética , Empalme Alternativo , Animales , Ancirinas/deficiencia , Secuencia de Bases , Cartilla de ADN , Modelos Animales de Enfermedad , Exones/genética , Intrones/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/genética , Eliminación de Secuencia
7.
J Biol Chem ; 278(28): 25526-33, 2003 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-12719424

RESUMEN

Several studies suggest that the Rh complex represents a major interaction site between the membrane lipid bilayer and the red cell skeleton, but little is known about the molecular basis of this interaction. We report here that ankyrin-R is capable of interacting directly with the C-terminal cytoplasmic domain of Rh and RhAG polypeptides. We first show that the primary defect of ankyrin-R in normoblastosis (nb/nb) spherocytosis mutant mice is associated with a sharp reduction of RhAG and Rh polypeptides. Secondly, our flow cytometric analysis of the Triton X-100 extractability of recombinant fusion proteins expressed in erythroleukemic cell lines suggests that the C-terminal cytoplasmic domains of Rh and RhAG are sufficient to mediate interaction with the erythroid membrane skeleton. Using the yeast two-hybrid system, we demonstrate a direct interaction between the cytoplasmic tails of Rh and RhAG and the second repeat domain (D2) of ankyrin-R. This finding is supported by the demonstration that the substitution of Asp-399 in the cytoplasmic tail of RhAG, a mutation associated with the deficiency of the Rh complex in one Rhnull patient, totally impaired interaction with domain D2 of ankyrin-R. These results identify the Rh/RhAG-ankyrin complex as a new interaction site between the red cell membrane and the spectrin-based skeleton, the disruption of which might result in the stomato-spherocytosis typical of Rhnull red cells.


Asunto(s)
Ancirinas/química , Ancirinas/metabolismo , Proteínas Sanguíneas , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Animales , Western Blotting , Proteínas de Unión a Calmodulina/metabolismo , Citoplasma/metabolismo , Detergentes/farmacología , Electroforesis en Gel de Poliacrilamida , Eritrocitos/metabolismo , Citometría de Flujo , Glutatión Transferasa/metabolismo , Humanos , Células K562 , Membrana Dobles de Lípidos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Mutantes , Modelos Biológicos , Octoxinol/farmacología , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Sistema del Grupo Sanguíneo Rh-Hr/química , Transfección , Técnicas del Sistema de Dos Híbridos
8.
Blood ; 101(1): 325-30, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12393645

RESUMEN

Tetramers of alpha- and beta-spectrin heterodimers, linked by intermediary proteins to transmembrane proteins, stabilize the red blood cell cytoskeleton. Deficiencies of either alpha- or beta-spectrin can result in severe hereditary spherocytosis (HS) or hereditary elliptocytosis (HE) in mice and humans. Four mouse mutations, sph, sph(Dem), sph(2BC), and sph(J), affect the erythroid alpha-spectrin gene, Spna1, on chromosome 1 and cause severe HS and HE. Here we describe the molecular alterations in alpha-spectrin and their consequences in sph(2BC)/sph(2BC) and sph(J)/sph(J) erythrocytes. A splicing mutation, sph(2BC) initiates the skipping of exon 41 and premature protein termination before the site required for dimerization of alpha-spectrin with beta-spectrin. A nonsense mutation in exon 52, sph(J) eliminates the COOH-terminal 13 amino acids. Both defects result in instability of the red cell membrane and loss of membrane surface area. In sph(2BC)/sph(2BC), barely perceptible levels of messenger RNA and consequent decreased synthesis of alpha-spectrin protein are primarily responsible for the resultant hemolysis. By contrast, sph(J)/sph(J) mice synthesize the truncated alpha-spectrin in which the 13-terminal amino acids are deleted at higher levels than normal, but they cannot retain this mutant protein in the cytoskeleton. The sph(J) deletion is near the 4.1/actin-binding region at the junctional complex providing new evidence that this 13-amino acid segment at the COOH-terminus of alpha-spectrin is crucial to the stability of the junctional complex.


Asunto(s)
Eritrocitos/metabolismo , Mutación , Espectrina/genética , Animales , Secuencia de Bases , Sitios de Unión , Deformación Eritrocítica/genética , Membrana Eritrocítica/genética , Membrana Eritrocítica/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Datos de Secuencia Molecular , Unión Proteica/genética , ARN Mensajero/metabolismo , Reticulocitos/ultraestructura , Espectrina/metabolismo , Espectrina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...