Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO J ; 41(24): e111115, 2022 12 15.
Article in English | MEDLINE | ID: mdl-36215693

ABSTRACT

Mitochondria and peroxisomes are closely related metabolic organelles, both in terms of origin and in terms of function. Mitochondria and peroxisomes can also be turned over by autophagy, in processes termed mitophagy and pexophagy, respectively. However, despite their close relationship, it is not known if both organelles are turned over under similar conditions, and if so, how this might be coordinated molecularly. Here, we find that multiple selective autophagy pathways are activated upon iron chelation and show that mitophagy and pexophagy occur in a BNIP3L/NIX-dependent manner. We reveal that the outer mitochondrial membrane-anchored NIX protein, previously described as a mitophagy receptor, also independently localises to peroxisomes and drives pexophagy. We show this process happens in vivo, with mouse tissue that lacks NIX having a higher peroxisomal content. We further show that pexophagy is stimulated under the same physiological conditions that activate mitophagy, including cardiomyocyte and erythrocyte differentiation. Taken together, our work uncovers a dual role for NIX, not only in mitophagy but also in pexophagy, thus illustrating the interconnection between selective autophagy pathways.


Subject(s)
Macroautophagy , Mitophagy , Mice , Animals , Peroxisomes/metabolism , Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism
2.
PLoS Pathog ; 20(2): e1011989, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38315723

ABSTRACT

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.


Subject(s)
Plasmodium falciparum , Symporters , Plasmodium falciparum/metabolism , Basigin/genetics , Basigin/metabolism , Erythrocytes/metabolism , Protein Domains , Symporters/metabolism
3.
Nucleic Acids Res ; 52(6): 3450-3468, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38412306

ABSTRACT

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.


Subject(s)
Baculoviridae , CRISPR-Cas Systems , DNA , Gene Editing , Viral Proteins , Animals , Humans , Baculoviridae/genetics , CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems/genetics , DNA/genetics , Gene Editing/methods , Viral Proteins/genetics , Cell Line
4.
Blood ; 141(2): 135-146, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36122374

ABSTRACT

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.


Subject(s)
Anemia, Hemolytic, Congenital , Blood Group Antigens , Infant, Newborn , Humans , Mutation, Missense , Anemia, Hemolytic, Congenital/genetics , Erythrocytes/metabolism , Ion Channels/chemistry , Blood Group Antigens/metabolism , Mechanotransduction, Cellular
5.
Br J Haematol ; 204(3): 826-838, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38009561

ABSTRACT

Despite significant global morbidity associated with respiratory infection, there is a paucity of data examining the association between severity of non-SARS-CoV-2 respiratory infection and blood group. We analysed a prospective cohort of adults hospitalised in Bristol, UK, from 1 August 2020 to 31 July 2022, including patients with acute respiratory infection (pneumonia [n = 1934] and non-pneumonic lower respiratory tract infection [NP-LRTI] [n = 1184]), a negative SARS-CoV-2 test and known blood group status. The likelihood of cardiovascular complication, survival and hospital admission length was assessed using regression models with group O and RhD-negative status as reference groups. Group A and RhD-positive were over-represented in both pneumonia and NP-LRTI compared to a first-time donor population (p < 0.05 in all); contrastingly, group O was under-represented. ABO group did not influence cardiovascular complication risk; however, RhD-positive patients with pneumonia had a reduced odds ratio (OR) for cardiovascular complications (OR = 0.77 [95% CI = 0.59-0.98]). Compared to group O, group A individuals with NP-LRTI were more likely to be discharged within 60 days (hazard ratio [HR] = 1.17 [95% CI = 1.03-1.33]), while group B with pneumonia was less likely (HR = 0.8 [95% CI = 0.66-0.96]). This analysis provides some evidence that blood group status may influence clinical outcome following respiratory infection, with group A having increased risk of hospitalisation and RhD-positive patients having reduced cardiovascular complications.


Subject(s)
COVID-19 , Pneumonia , Respiratory Tract Infections , Adult , Humans , SARS-CoV-2 , Prospective Studies , ABO Blood-Group System , United Kingdom
6.
J Transl Med ; 22(1): 526, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822352

ABSTRACT

BACKGROUND: Neutrophils are granulocytes with essential antimicrobial effector functions and short lifespans. During infection or sterile inflammation, emergency granulopoiesis leads to release of immature neutrophils from the bone marrow, serving to boost circulating neutrophil counts. Steady state and emergency granulopoiesis are incompletely understood, partly due to a lack of genetically amenable models of neutrophil development. METHODS: We optimised a method for ex vivo production of human neutrophils from CD34+ haematopoietic progenitors. Using flow cytometry, we phenotypically compared cultured neutrophils with native neutrophils from donors experiencing emergency granulopoiesis, and steady state neutrophils from non-challenged donors. We carry out functional and proteomic characterisation of cultured neutrophils and establish genome editing of progenitors. RESULTS: We obtain high yields of ex vivo cultured neutrophils, which phenotypically resemble immature neutrophils released into the circulation during emergency granulopoiesis. Cultured neutrophils have similar rates of ROS production and bacterial killing but altered degranulation, cytokine release and antifungal activity compared to mature neutrophils isolated from peripheral blood. These differences are likely due to incomplete synthesis of granule proteins, as demonstrated by proteomic analysis. CONCLUSION: Ex vivo cultured neutrophils are genetically tractable via genome editing of precursors and provide a powerful model system for investigating the properties and behaviour of immature neutrophils.


Subject(s)
Antigens, CD34 , Neutrophils , Humans , Neutrophils/metabolism , Neutrophils/cytology , Antigens, CD34/metabolism , Cells, Cultured , Reactive Oxygen Species/metabolism , Proteomics , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Gene Editing , Cell Degranulation , Stem Cells/metabolism , Stem Cells/cytology , Cytokines/metabolism , Phenotype
7.
Immunol Cell Biol ; 101(10): 947-963, 2023.
Article in English | MEDLINE | ID: mdl-37694300

ABSTRACT

Macrophages have previously been characterized based on phenotypical and functional differences into suggested simplified subtypes of MØ, M1, M2a and M2c. These macrophage subtypes can be generated in a well-established primary monocyte culture model that produces cells expressing accepted subtype surface markers. To determine how these subtypes retain functional similarities and better understand their formation, we generated all four subtypes from the same donors. Comparative whole-cell proteomics confirmed that four distinct macrophage subtypes could be induced from the same donor material, with > 50% of 5435 identified proteins being significantly altered in abundance between subtypes. Functional assessment highlighted that these distinct protein expression profiles are primed to enable specific cell functions, indicating that this shifting proteome is predictive of meaningful changes in cell characteristics. Importantly, the 2552 proteins remained consistent in abundance across all macrophage subtypes examined, demonstrating maintenance of a stable core proteome that likely enables swift polarity changes. We next explored the cross-polarization capabilities of preactivated M1 macrophages treated with dexamethasone. Importantly, these treated cells undergo a partial repolarization toward the M2c surface markers but still retain the M1 functional phenotype. Our investigation of polarized macrophage subtypes therefore provides evidence of a sliding scale of macrophage functionality, with these data sets providing a valuable benchmark resource for further studies of macrophage polarity, with relevance for cell therapy development and drug discovery.


Subject(s)
Proteome , Proteomics , Proteome/metabolism , Cells, Cultured , Macrophages/metabolism , Monocytes/physiology
8.
EMBO J ; 37(13)2018 07 02.
Article in English | MEDLINE | ID: mdl-29866703

ABSTRACT

Wound angiogenesis is an integral part of tissue repair and is impaired in many pathologies of healing. Here, we investigate the cellular interactions between innate immune cells and endothelial cells at wounds that drive neoangiogenic sprouting in real time and in vivo Our studies in mouse and zebrafish wounds indicate that macrophages are drawn to wound blood vessels soon after injury and are intimately associated throughout the repair process and that macrophage ablation results in impaired neoangiogenesis. Macrophages also positively influence wound angiogenesis by driving resolution of anti-angiogenic wound neutrophils. Experimental manipulation of the wound environment to specifically alter macrophage activation state dramatically influences subsequent blood vessel sprouting, with premature dampening of tumour necrosis factor-α expression leading to impaired neoangiogenesis. Complementary human tissue culture studies indicate that inflammatory macrophages associate with endothelial cells and are sufficient to drive vessel sprouting via vascular endothelial growth factor signalling. Subsequently, macrophages also play a role in blood vessel regression during the resolution phase of wound repair, and their absence, or shifted activation state, impairs appropriate vessel clearance.


Subject(s)
Macrophages/physiology , Neovascularization, Physiologic , Wound Healing/physiology , Animals , Animals, Genetically Modified , Cells, Cultured , Diagnostic Imaging , Fibroblasts , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice, Inbred C57BL , Zebrafish/genetics
9.
Haematologica ; 106(9): 2304-2311, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34042406

ABSTRACT

Patients with inherited anemia and hemoglobinopathies (such as sickle cell disease and ß-thalassemia) are treated with red blood cell (RBC) transfusions to alleviate their symptoms. Some of these patients may have rare blood group types or go on to develop alloimmune reactions, which can make it difficult to source compatible blood in the donor population. Laboratory-grown RBC represent a particularly attractive alternative which could satisfy an unmet clinical need. The challenge, however, is to produce - from a limited number of stem cells - the 2x1012 RBC required for a standard adult therapeutic dose. Encouraging progress has been made in RBC production from adult stem cells under good manufacturing practice. In 2011, the Douay group conducted a successful proof-of-principle mini-transfusion of autologous manufactured RBC in a single volunteer. In the UK, a trial is planned to assess whether manufactured RBC are equivalent to RBC produced naturally in donors, by testing an allogeneic mini-dose of laboratory-grown manufactured RBC in multiple volunteers. This review discusses recent progress in the erythroid culture field as well as opportunities for further scaling up of manufactured RBC production for transfusion practice.


Subject(s)
Anemia, Sickle Cell , Hemoglobinopathies , Anemia, Sickle Cell/therapy , Blood Transfusion , Erythrocyte Transfusion , Erythrocytes , Humans
10.
Haematologica ; 105(4): 914-924, 2020 04.
Article in English | MEDLINE | ID: mdl-31197068

ABSTRACT

Erythropoiesis is one of the most efficient cellular processes in the human body producing approximately 2.5 million red blood cells every second. This process occurs in a bone marrow niche comprised of a central resident macrophage surrounded by differentiating erythroblasts, termed an erythroblastic island. It is not known what initially attracts the macrophage to erythroblasts to form these islands. The ephrin/Eph receptor family are known to regulate heterophilic cell-cell adhesion. We find that human VCAM1+ and VCAM1- bone marrow macrophages and in vitro cultured macrophages are ephrin-B2 positive, whereas differentiating human erythroblasts express EPHB4, EPHB6 and EPHA4. Furthermore, we detect a rise in integrin activation on erythroblasts at the stage at which the cells bind which is independent of EPH receptor presence. Using a live cell imaging assay, we show that specific inhibitory peptides or shRNA depletion of EPHB4 cause a significant reduction in the ability of macrophages to interact with erythroblasts but do not affect integrin activation. This study demonstrates for the first time that EPHB4 expression is required on erythroblasts to facilitate the initial recognition and subsequent interaction with macrophages, alongside the presence of active integrins.


Subject(s)
Ephrins , Erythroblasts/cytology , Macrophages/cytology , Receptor, EphB4/genetics , Erythropoiesis , Humans , Receptors, Eph Family
11.
Curr Opin Hematol ; 25(3): 163-170, 2018 05.
Article in English | MEDLINE | ID: mdl-29438259

ABSTRACT

PURPOSE OF REVIEW: Current research on the human band 3 glycoprotein, the red cell chloride/bicarbonate anion exchanger (AE1), is highlighted and placed within a structural context. RECENT FINDINGS: The determination of the crystal structure of the membrane domain of human band 3, the founding member of the solute carrier 4 (SLC4) family of bicarbonate transporters, is a major breakthrough toward understanding the mechanism of action of this membrane transport protein, its interaction with partner proteins, and how mutations linked to disease affect its ability to fold and function. SUMMARY: Band 3 contains 14 transmembrane segments arranged in a 7+7 transmembrane inverted repeat topology common to all members of the SLC4 family and the unrelated SLC26 anion transporter family. A functional feature of this fold is the presence of a core and a gate domain: the core domain contains two short transmembrane helices (TM3 and 10) that face each other in the middle of the membrane with the positive N-terminal helix dipoles creating the anion-binding site, whereas the gate domain forms the dimer interface. During transport, the movement of these two domains relative to each other provides the intracellular and extracellular compartments with alternating access to the central anion-binding site.


Subject(s)
Anion Exchange Protein 1, Erythrocyte , Bicarbonates/metabolism , Mutation , Animals , Anion Exchange Protein 1, Erythrocyte/chemistry , Anion Exchange Protein 1, Erythrocyte/genetics , Anion Exchange Protein 1, Erythrocyte/metabolism , Crystallography, X-Ray , Humans , Ion Transport/genetics , Protein Domains , Protein Structure, Secondary
12.
Haematologica ; 103(3): 395-405, 2018 03.
Article in English | MEDLINE | ID: mdl-29284682

ABSTRACT

The classical central macrophage found in erythroblastic islands plays an important role in erythroblast differentiation, proliferation and enucleation in the bone marrow. Convenient human in vitro models to facilitate the study of erythroid-macrophage interactions are desired. Recently, we demonstrated that cultured monocytes/macrophages enhance in vitro erythropoiesis by supporting hematopoietic stem and progenitor cell survival. Herein, we describe that these specific macrophages also support erythropoiesis. Human monocytes cultured in serum-free media supplemented with stem cell factor, erythropoietin, lipids and dexamethasone differentiate towards macrophages expressing CD16, CD163, CD169, CD206, CXCR4 and the phagocytic TAM-receptor family. Phenotypically, they resemble both human bone marrow and fetal liver resident macrophages. This differentiation is dependent on glucocorticoid receptor activation. Proteomic studies confirm that glucocorticoid receptor activation differentiates monocytes to anti-inflammatory tissue macrophages with a M2 phenotype, termed GC-macrophages. Proteins involved in migration, tissue residence and signal transduction/receptor activity are upregulated whilst lysosome and hydrolase activity GO-categories are downregulated. Functionally, we demonstrate that GC-macrophages are highly mobile and can interact to form clusters with erythroid cells of all differentiation stages and phagocytose the expelled nuclei, recapitulating aspects of erythroblastic islands. In conclusion, glucocorticoid-directed monocyte differentiation to macrophages represents a convenient model system to study erythroid-macrophage interactions.


Subject(s)
Cell Differentiation/drug effects , Glucocorticoids/pharmacology , Macrophages/cytology , Monocytes/cytology , Cell Communication , Cells, Cultured , Erythroid Cells/cytology , Erythropoiesis , Humans , Monocytes/drug effects , Proteomics
13.
Haematologica ; 103(12): 1997-2007, 2018 12.
Article in English | MEDLINE | ID: mdl-30076174

ABSTRACT

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.


Subject(s)
Cytoplasmic Vesicles/metabolism , Erythrocytes/metabolism , Myosin Type II/metabolism , Reticulocytes/metabolism , Cell Differentiation , Cells, Cultured , Cytoskeletal Proteins/metabolism , Erythrocytes/cytology , Erythropoiesis , Humans , Molecular Motor Proteins/metabolism , Myosin Heavy Chains/metabolism , Phosphorylation , Proteomics/methods , Reticulocytes/cytology
14.
Mol Cell Proteomics ; 15(6): 1938-46, 2016 06.
Article in English | MEDLINE | ID: mdl-27006477

ABSTRACT

Cord blood stem cells are an attractive starting source for the production of red blood cells in vitro for therapy because of additional expansion potential compared with adult peripheral blood progenitors and cord blood banks usually being more representative of national populations than blood donors. Consequently, it is important to establish how similar cord RBCs are to adult cells. In this study, we used multiplex tandem mass tag labeling combined with nano-LC-MS/MS to compare the proteome of adult and cord RBCs and reticulocytes. 2838 unique proteins were identified, providing the most comprehensive compendium of RBC proteins to date. Using stringent criteria, 1674 proteins were quantified, and only a small number differed in amount between adult and cord RBC. We focused on proteins critical for RBC function. Of these, only the expected differences in globin subunits, along with higher levels of carbonic anhydrase 1 and 2 and aquaporin-1 in adult RBCs would be expected to have a phenotypic effect since they are associated with the differences in gaseous exchange between adults and neonates. Since the RBC and reticulocyte samples used were autologous, we catalogue the change in proteome following reticulocyte maturation. The majority of proteins (>60% of the 1671 quantified) reduced in abundance between 2- and 100-fold following maturation. However, ∼5% were at a higher level in RBCs, localized almost exclusively to cell membranes, in keeping with the known clearance of intracellular recycling pools during reticulocyte maturation. Overall, these data suggest that, with respect to the proteome, there is no barrier to the use of cord progenitors for the in vitro generation of RBCs for transfusion to adults other than the expression of fetal, not adult, hemoglobin.


Subject(s)
Erythroid Cells/cytology , Fetal Blood/cytology , Proteome/analysis , Proteomics/methods , Reticulocytes/cytology , Adult , Carbonic Anhydrase I/metabolism , Carbonic Anhydrase II/metabolism , Cell Differentiation , Chromatography, Liquid , Erythroid Cells/metabolism , Fetal Blood/metabolism , Humans , Infant, Newborn , Reticulocytes/metabolism , Tandem Mass Spectrometry
16.
17.
Haematologica ; 101(9): 1018-27, 2016 09.
Article in English | MEDLINE | ID: mdl-27247322

ABSTRACT

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.


Subject(s)
Ankyrins/deficiency , Blood Proteins/metabolism , Erythroblasts/metabolism , Erythrocyte Membrane/metabolism , Lysosomes/metabolism , Membrane Glycoproteins/metabolism , Anion Exchange Protein 1, Erythrocyte/chemistry , Anion Exchange Protein 1, Erythrocyte/metabolism , Cell Differentiation/genetics , Cells, Cultured , Cytoskeleton/genetics , Cytoskeleton/metabolism , Erythroblasts/chemistry , Erythroblasts/cytology , Erythropoiesis/genetics , Gene Expression Regulation , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Biological , Mutation , Protein Binding , Protein Multimerization , Proteolysis , Spherocytosis, Hereditary/genetics , Spherocytosis, Hereditary/metabolism
19.
Haematologica ; 100(1): 133-42, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25344524

ABSTRACT

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.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/metabolism , Ankyrins/deficiency , Cytoskeleton/metabolism , Erythroblasts/metabolism , Erythrocyte Membrane/metabolism , Erythropoiesis/physiology , Multiprotein Complexes/metabolism , Spherocytosis, Hereditary/metabolism , Anion Exchange Protein 1, Erythrocyte/antagonists & inhibitors , Anion Exchange Protein 1, Erythrocyte/genetics , Ankyrins/genetics , Ankyrins/metabolism , Case-Control Studies , Cell Differentiation , Cells, Cultured , Erythroblasts/cytology , Flow Cytometry , Homozygote , Humans , Phenotype , Protein Binding , RNA, Small Interfering/genetics , Reticulocytes/cytology , Reticulocytes/metabolism , Spherocytosis, Hereditary/genetics , Spherocytosis, Hereditary/pathology
SELECTION OF CITATIONS
SEARCH DETAIL