Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
PLoS Pathog ; 20(2): e1011989, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38315723

RESUMEN

Plasmodium falciparum invasion of the red blood cell is reliant upon the essential interaction of PfRh5 with the host receptor protein basigin. Basigin exists as part of one or more multiprotein complexes, most notably through interaction with the monocarboxylate transporter MCT1. However, the potential requirement for basigin association with MCT1 and the wider role of basigin host membrane context and lateral protein associations during merozoite invasion has not been established. Using genetically manipulated in vitro derived reticulocytes, we demonstrate the ability to uncouple basigin ectodomain presentation from its transmembrane domain-mediated interactions, including with MCT1. Merozoite invasion of reticulocytes is unaffected by disruption of basigin-MCT1 interaction and by removal or replacement of the basigin transmembrane helix. Therefore, presentation of the basigin ectodomain at the red blood cell surface, independent of its native association with MCT1 or other interactions mediated by the transmembrane domain, is sufficient to facilitate merozoite invasion.


Asunto(s)
Plasmodium falciparum , Simportadores , Plasmodium falciparum/metabolismo , Basigina/genética , Basigina/metabolismo , Eritrocitos/metabolismo , Dominios Proteicos , Simportadores/metabolismo
2.
Nucleic Acids Res ; 52(6): 3450-3468, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38412306

RESUMEN

CRISPR-based DNA editing technologies enable rapid and accessible genome engineering of eukaryotic cells. However, the delivery of genetically encoded CRISPR components remains challenging and sustained Cas9 expression correlates with higher off-target activities, which can be reduced via Cas9-protein delivery. Here we demonstrate that baculovirus, alongside its DNA cargo, can be used to package and deliver proteins to human cells. Using protein-loaded baculovirus (pBV), we demonstrate delivery of Cas9 or base editors proteins, leading to efficient genome and base editing in human cells. By implementing a reversible, chemically inducible heterodimerization system, we show that protein cargoes can selectively and more efficiently be loaded into pBVs (spBVs). Using spBVs we achieved high levels of multiplexed genome editing in a panel of human cell lines. Importantly, spBVs maintain high editing efficiencies in absence of detectable off-targets events. Finally, by exploiting Cas9 protein and template DNA co-delivery, we demonstrate up to 5% site-specific targeted integration of a 1.8 kb heterologous DNA payload using a single spBV in a panel of human cell lines. In summary, we demonstrate that spBVs represent a versatile, efficient and potentially safer alternative for CRISPR applications requiring co-delivery of DNA and protein cargoes.


Asunto(s)
Baculoviridae , Sistemas CRISPR-Cas , ADN , Edición Génica , Proteínas Virales , Animales , Humanos , Baculoviridae/genética , Proteína 9 Asociada a CRISPR/genética , Sistemas CRISPR-Cas/genética , ADN/genética , Edición Génica/métodos , Proteínas Virales/genética , Línea Celular
3.
Blood ; 141(2): 135-146, 2023 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-36122374

RESUMEN

Despite the identification of the high-incidence red cell antigen Era nearly 40 years ago, the molecular background of this antigen, together with the other 2 members of the Er blood group collection, has yet to be elucidated. Whole exome and Sanger sequencing of individuals with serologically defined Er alloantibodies identified several missense mutations within the PIEZO1 gene, encoding amino acid substitutions within the extracellular domain of the Piezo1 mechanosensor ion channel. Confirmation of Piezo1 as the carrier molecule for the Er blood group antigens was demonstrated using immunoprecipitation, CRISPR/Cas9-mediated gene knockout, and expression studies in an erythroblast cell line. We report the molecular bases of 5 Er blood group antigens: the recognized Era, Erb, and Er3 antigens and 2 novel high-incidence Er antigens, described here as Er4 and Er5, establishing a new blood group system. Anti-Er4 and anti-Er5 are implicated in severe hemolytic disease of the fetus and newborn. Demonstration of Piezo1, present at just a few hundred copies on the surface of the red blood cell, as the site of a new blood group system highlights the potential antigenicity of even low-abundance membrane proteins and contributes to our understanding of the in vivo characteristics of this important and widely studied protein in transfusion biology and beyond.


Asunto(s)
Anemia Hemolítica Congénita , Antígenos de Grupos Sanguíneos , Recién Nacido , Humanos , Mutación Missense , Anemia Hemolítica Congénita/genética , Eritrocitos/metabolismo , Canales Iónicos/química , Antígenos de Grupos Sanguíneos/metabolismo , Mecanotransducción Celular
4.
Proc Natl Acad Sci U S A ; 114(16): 4225-4230, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28373555

RESUMEN

Invasion of the red blood cell (RBC) by the Plasmodium parasite defines the start of malaria disease pathogenesis. To date, experimental investigations into invasion have focused predominantly on the role of parasite adhesins or signaling pathways and the identity of binding receptors on the red cell surface. A potential role for signaling pathways within the erythrocyte, which might alter red cell biophysical properties to facilitate invasion, has largely been ignored. The parasite erythrocyte-binding antigen 175 (EBA175), a protein required for entry in most parasite strains, plays a key role by binding to glycophorin A (GPA) on the red cell surface, although the function of this binding interaction is unknown. Here, using real-time deformability cytometry and flicker spectroscopy to define biophysical properties of the erythrocyte, we show that EBA175 binding to GPA leads to an increase in the cytoskeletal tension of the red cell and a reduction in the bending modulus of the cell's membrane. We isolate the changes in the cytoskeleton and membrane and show that reduction in the bending modulus is directly correlated with parasite invasion efficiency. These data strongly imply that the malaria parasite primes the erythrocyte surface through its binding antigens, altering the biophysical nature of the target cell and thus reducing a critical energy barrier to invasion. This finding would constitute a major change in our concept of malaria parasite invasion, suggesting it is, in fact, a balance between parasite and host cell physical forces working together to facilitate entry.


Asunto(s)
Antígenos de Protozoos/metabolismo , Membrana Celular/patología , Eritrocitos/patología , Glicoforinas/metabolismo , Malaria Falciparum/patología , Plasmodium falciparum/patogenicidad , Proteínas Protozoarias/metabolismo , Antígenos de Protozoos/genética , Biofisica , Membrana Celular/metabolismo , Membrana Celular/parasitología , Citoesqueleto , Eritrocitos/metabolismo , Eritrocitos/parasitología , Glicoforinas/genética , Interacciones Huésped-Parásitos , Humanos , Malaria Falciparum/metabolismo , Malaria Falciparum/parasitología , Plasmodium falciparum/aislamiento & purificación , Unión Proteica , Proteínas Protozoarias/genética , Transducción de Señal
5.
Haematologica ; 103(12): 1997-2007, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30076174

RESUMEN

The process of maturation of reticulocytes into fully mature erythrocytes that occurs in the circulation is known to be characterized by a complex interplay between loss of cell surface area and volume, removal of remnant cell organelles and redundant proteins, and highly selective membrane and cytoskeletal remodeling. However, the mechanisms that underlie and drive these maturational processes in vivo are currently poorly understood and, at present, reticulocytes derived through in vitro culture fail to undergo the final transition to erythrocytes. Here, we used high-throughput proteomic methods to highlight differences between erythrocytes, cultured reticulocytes and endogenous reticulocytes. We identify a cytoskeletal protein, non-muscle myosin IIA (NMIIA) whose abundance and phosphorylation status differs between reticulocytes and erythrocytes and localized it in the proximity of autophagosomal vesicles. An ex vivo circulation system was developed to simulate the mechanical shear component of circulation and demonstrated that mechanical stimulus is necessary, but insufficient for reticulocyte maturation. Using this system in concurrence with non-muscle myosin II inhibition, we demonstrate the involvement of non-muscle myosin IIA in reticulocyte remodeling and propose a previously undescribed mechanism of shear stress-responsive vesicle clearance that is crucial for reticulocyte maturation.


Asunto(s)
Vesículas Citoplasmáticas/metabolismo , Eritrocitos/metabolismo , Miosina Tipo II/metabolismo , Reticulocitos/metabolismo , Diferenciación Celular , Células Cultivadas , Proteínas del Citoesqueleto/metabolismo , Eritrocitos/citología , Eritropoyesis , Humanos , Proteínas Motoras Moleculares/metabolismo , Cadenas Pesadas de Miosina/metabolismo , Fosforilación , Proteómica/métodos , Reticulocitos/citología
7.
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.
Biophys J ; 107(1): 43-54, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24988340

RESUMEN

The blood stage malaria parasite, the merozoite, has a small window of opportunity during which it must successfully target and invade a human erythrocyte. The process of invasion is nonetheless remarkably rapid. To date, mechanistic models of invasion have focused predominantly on the parasite actomyosin motor contribution to the energetics of entry. Here, we have conducted a numerical analysis using dimensions for an archetypal merozoite to predict the respective contributions of the host-parasite interactions to invasion, in particular the role of membrane wrapping. Our theoretical modeling demonstrates that erythrocyte membrane wrapping alone, as a function of merozoite adhesive and shape properties, is sufficient to entirely account for the first key step of the invasion process, that of merozoite reorientation to its apex and tight adhesive linkage between the two cells. Next, parasite-induced reorganization of the erythrocyte cytoskeleton and release of parasite-derived membrane can also account for a considerable energetic portion of actual invasion itself, through membrane wrapping. Thus, contrary to the prevailing dogma, wrapping by the erythrocyte combined with parasite-derived membrane release can markedly reduce the expected contributions of the merozoite actomyosin motor to invasion. We therefore propose that invasion is a balance between parasite and host cell contributions, evolved toward maximal efficient use of biophysical forces between the two cells.


Asunto(s)
Membrana Celular/parasitología , Eritrocitos/parasitología , Interacciones Huésped-Parásitos , Plasmodium falciparum/patogenicidad , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Humanos , Merozoítos/fisiología
12.
Blood Adv ; 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38916993

RESUMEN

The Glucose transporter 1 (GLUT1) is one of the most abundant proteins within the erythrocyte membrane and is required for glucose and dehydroascorbic acid (Vitamin C precursor) transport. It is widely recognized as a key protein for red cell structure, function, and metabolism. Previous reports highlighted the importance of GLUT1 activity within these uniquely glycolysis-dependent cells, in particular for increasing antioxidant capacity needed to avoid irreversible damage from oxidative stress in humans. However, studies of glucose transporter roles in erythroid cells are complicated by species-specific differences between humans and mice. Here, using CRISPR-mediated gene editing of immortalized erythroblasts and adult CD34+ hematopoietic progenitor cells, we generate committed human erythroid cells completely deficient in expression of GLUT1. We show that absence of GLUT1 does not impede human erythroblast proliferation, differentiation, or enucleation. This work demonstrates for the first-time generation of enucleated human reticulocytes lacking GLUT1. The GLUT1-deficient reticulocytes possess no tangible alterations to membrane composition or deformability in reticulocytes. Metabolomic analyses of GLUT1-deficient reticulocytes reveal hallmarks of reduced glucose import, downregulated metabolic processes and upregulated AMPK-signalling, alongside alterations in antioxidant metabolism, resulting in increased osmotic fragility and metabolic shifts indicative of higher oxidant stress. Despite detectable metabolic changes in GLUT1 deficient reticulocytes, the absence of developmental phenotype, detectable proteomic compensation or impaired deformability comprehensively alters our understanding of the role of GLUT1 in red blood cell structure, function and metabolism. It also provides cell biological evidence supporting clinical consensus that reduced GLUT1 expression does not cause anaemia in GLUT1 deficiency syndrome.

13.
Blood ; 128(1): 11-2, 2016 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-27389540
14.
Blood ; 118(1): 182-191, 2011 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-21527529

RESUMEN

Band 3, the major anion transport protein of human erythrocytes, forms the core of a multiprotein complex in the erythrocyte membrane. Here we studied the spatiotemporal mechanisms of band 3 multiprotein complex assembly during erythropoiesis. Significant pools of intracellular band 3 and Rh-associated glycoprotein (RhAG) were found in the basophilic erythroblast. These intracellular pools decreased in the polychromatic erythroblast, whereas surface expression increased and were lowest in the orthochromatic erythroblast and reticulocytes. Protease treatment of intact cells to remove extracellular epitopes recognized by antibodies to band 3 and RhAG was used to study surface delivery kinetics and intracellular complex composition from the proerythroblast stage to the enucleated reticulocyte. Newly synthesized band 3 and protein 4.2 interact initially in the early stages of the secretory pathway and are found associated at the plasma membrane from the basophilic stage of erythropoiesis. Although we could successfully coimmunoprecipitate Rh with RhAG from plasma membrane pools at a similar stage, no intracellular interaction between these proteins was detectable. Knockdown of RhAG during early erythropoiesis was accompanied by a concomitant drop in membrane expression of Rh polypeptides. These data are consistent with assembly of major components of the band 3 macrocomplex at an early stage during erythropoiesis.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Eritroblastos/metabolismo , Membrana Eritrocítica/metabolismo , Eritropoyesis/fisiología , Complejos Multiproteicos/metabolismo , Reticulocitos/metabolismo , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Diferenciación Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , Retículo Endoplásmico/metabolismo , Eritroblastos/citología , Aparato de Golgi/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Péptido Hidrolasas/metabolismo , Unión Proteica/fisiología , ARN Interferente Pequeño , Reticulocitos/citología
15.
Haematologica ; 98(11): 1788-96, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23935019

RESUMEN

Congenital dyserythropoietic anemia type II is an autosomally recessive form of hereditary anemia caused by SEC23B gene mutations. Patients exhibit characteristic phenotypes including multinucleate erythroblasts, erythrocytes with hypoglycosylated membrane proteins and an apparent double plasma membrane. Despite ubiquitous expression of SEC23B, the effects of mutations in this gene are confined to the erythroid lineage and the basis of this erythroid specificity remains to be defined. In addition, little is known regarding the stage at which the disparate phenotypes of this disease manifest during erythropoiesis. We employ an in vitro culture system to monitor the appearance of the defining phenotypes associated with congenital dyserythropoietic anemia type II during terminal differentiation of erythroblasts derived from small volumes of patient peripheral blood. Membrane protein hypoglycosylation was detected by the basophilic stage, preceding the onset of multinuclearity in orthochromatic erythroblasts that occurs coincident with the loss of secretory pathway proteins including SEC23A during erythropoiesis. Endoplasmic reticulum remnants were observed in nascent reticulocytes of both diseased and healthy donor cultures but were lost upon further maturation of normal reticulocytes, implicating a defect of ER clearance during reticulocyte maturation in congenital dyserythropoietic anemia type II. We also demonstrate distinct isoform and species-specific expression profiles of SEC23 during terminal erythroid differentiation and identify a prolonged expression of SEC23A in murine erythropoiesis compared to humans. We propose that SEC23A is able to compensate for the absence of SEC23B in mouse erythroblasts, providing a basis for the absence of phenotype within the erythroid lineage of a recently described SEC23B knockout mouse.


Asunto(s)
Anemia Diseritropoyética Congénita/genética , Anemia Diseritropoyética Congénita/patología , Eritropoyesis/fisiología , Fenotipo , Animales , Células Cultivadas , Humanos , Ratones , Ratones Endogámicos C57BL
16.
Front Cell Infect Microbiol ; 12: 1039520, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36452302

RESUMEN

Parasites of the genus Plasmodium that cause malaria survive within humans by invasion of, and proliferation within, the most abundant cell type in the body, the red blood cell. As obligate, intracellular parasites, interactions between parasite and host red blood cell components are crucial to multiple aspects of the blood stage malaria parasite lifecycle. The requirement for, and involvement of, an array of red blood cell proteins in parasite invasion and intracellular development is well established. Nevertheless, detailed mechanistic understanding of host cell protein contributions to these processes are hampered by the genetic intractability of the anucleate red blood cell. The advent of stem cell technology and more specifically development of methods that recapitulate in vitro the process of red blood cell development known as erythropoiesis has enabled the generation of erythroid cell stages previously inaccessible in large numbers for malaria studies. What is more, the capacity for genetic manipulation of nucleated erythroid precursors that can be differentiated to generate modified red blood cells has opened new horizons for malaria research. This review summarises current methodologies that harness in vitro erythroid differentiation of stem cells for generation of cells that are susceptible to malaria parasite invasion; discusses existing and emerging approaches to generate novel red blood cell phenotypes and explores the exciting potential of in vitro derived red blood cells for improved understanding the broad role of host red blood cell proteins in malaria pathogenesis.


Asunto(s)
Eritrocitos , Malaria , Humanos , Células Madre , Eritropoyesis , Diferenciación Celular
17.
Biochem Cell Biol ; 89(2): 106-14, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21455263

RESUMEN

The bicarbonate/chloride exchanger 1 (AE1, Band 3) is abundantly expressed in the red blood cell membrane, where it is involved in gas exchange and functions as a major site of cytoskeletal attachment to the erythrocyte membrane. A truncated kidney isoform (kAE1) is highly expressed in type A intercalated cells of the distal tubules, where it is vital for urinary acidification. Recently, kAE1 has emerged as a novel physiologically significant protein in the kidney glomerulus. This minireview will discuss the known interactions of kAE1 in the podocytes and the possible mechanisms whereby this important multispanning membrane protein may contribute to the function of the glomerular filtration barrier and prevent proteinuria.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Eritrocitos/metabolismo , Glomérulos Renales/metabolismo , Túbulos Renales Distales/metabolismo , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/química , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Citoesqueleto/metabolismo , Humanos , Glomérulos Renales/citología , Túbulos Renales Distales/citología , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Podocitos/citología , Podocitos/metabolismo , Conformación Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
18.
J Am Soc Nephrol ; 21(9): 1456-67, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20576809

RESUMEN

The central role of the multifunctional protein nephrin within the macromolecular complex forming the glomerular slit diaphragm is well established, but the mechanisms linking the slit diaphragm to the cytoskeleton and to the signaling pathways involved in maintaining the integrity of the glomerular filter remain incompletely understood. Here, we report that nephrin interacts with the bicarbonate/chloride transporter kidney anion exchanger 1 (kAE1), detected by yeast two-hybrid assay and confirmed by immunoprecipitation and co-localization studies. We confirmed low-level glomerular expression of kAE1 in human and mouse kidneys by immunoblotting and immunofluorescence microscopy. We observed less kAE1 in human glomeruli homozygous for the NPHS1(FinMaj) nephrin mutation, whereas kAE1 expression remained unchanged in the collecting duct. We could not detect endogenous kAE1 expression in NPHS1(FinMaj) podocytes in primary culture, but heterologous re-introduction of wild-type nephrin into these podocytes rescued kAE1 expression. In kidneys of Ae1(-/-) mice, nephrin abundance was normal but its distribution was altered along with the reported kAE1-binding protein integrin-linked kinase (ILK). Ae1(-/-) mice had increased albuminuria with glomerular enlargement, mesangial expansion, mesangiosclerosis, and expansion of the glomerular basement membrane. Glomeruli with ILK-deficient podocytes also demonstrated altered AE1 and nephrin expression, further supporting the functional interdependence of these proteins. These data suggest that the podocyte protein kAE1 interacts with nephrin and ILK to maintain the structure and function of the glomerular basement membrane.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/fisiología , Proteínas de la Membrana/fisiología , Podocitos/metabolismo , Adulto , Albuminuria/metabolismo , Secuencia de Aminoácidos , Animales , Proteína 1 de Intercambio de Anión de Eritrocito/análisis , Células Cultivadas , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Glomérulos Renales/patología , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Proteínas Serina-Treonina Quinasas/fisiología , Técnicas del Sistema de Dos Híbridos , Xenopus
19.
Blood Cells Mol Dis ; 45(1): 1-8, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20346715

RESUMEN

The bicarbonate/chloride exchanger band 3 (Anion Exchanger 1, AE1) is the most abundant protein in the erythrocyte membrane, it has an important role in gas exchange and functions as a point of attachment for the cytoskeletons maintaining the mechanistic and osmotic properties of the erythrocyte. Band 3 is found in three distinct protein complexes within the erythrocyte membrane: an ankyrin-dependent tetrameric band 3 complex, a dimeric band 3 complex bound to the protein 4.1-GPC junctional complex and as freely diffusing dimeric band 3 complexes. Much if not all of our present knowledge of these protein complexes is derived from mouse knockout model systems and human variant blood samples. This review will explore what is known about the band 3 complexes of mice and humans, focussing on the observed species differences and their potential functional consequences.


Asunto(s)
Proteína 1 de Intercambio de Anión de Eritrocito/metabolismo , Membrana Eritrocítica/metabolismo , Animales , Eritrocitos/metabolismo , Humanos , Ratones , Complejos Multiproteicos/metabolismo
20.
Haematologica ; 95(9): 1594-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20378567

RESUMEN

The study of human erythropoiesis in health and disease requires a robust culture system that consistently and reliably generates large numbers of immature erythroblasts that can be induced to differentiate synchronously. We describe a culture method modified from Leberbauer et al. (2005) and obtain a homogenous population of erythroblasts from peripheral blood mononuclear cells (PBMC) without prior purification of CD34(+) cells. This pure population of immature erythroblasts can be expanded to obtain 4x10(8) erythroblasts from 1x10(8) PBMC after 13-14 days in culture. Upon synchronized differentiation, high levels of enucleation (80-90%) and low levels of cell death (<10%) are achieved. We compared the yield of erythroblasts obtained from PBMC, CD34(+) cells or PBMC depleted of CD34(+) cells and show that CD34(-) cells represent the most significant early erythroid progenitor population. This culture system may be particularly useful for investigating the pathophysiology of anemic patients where only small blood volumes are available.


Asunto(s)
Antígenos CD34/análisis , Proliferación Celular , Eritroblastos/citología , Eritropoyesis , Técnicas de Cultivo de Célula/métodos , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA