Alterations in the band3/Rh Protein Complex in Erythrocyte Membranes of Protein 4.2 (Chartres) deficient Cells Occur Late during Erythropoiesis

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3460-3460
Author(s):  
Emi le van den Akker ◽  
Timothy J Satchwell ◽  
Jo F Flatt ◽  
Stephanie Pellegrin ◽  
M. Maigre ◽  
...  

Abstract We report on a 40 year old patient with mild hereditary spherocytosis (RBC: 4.43×1012/dL; Reticulocyte count: 253×109/dL; Hb: 14g/dL), whose red blood cells completely lack protein 4.2. Genetic analysis showed that the patient was a double heterozygote for EPB42 deletions; one allele lacked exon 9 but the sequence remained in frame (protein 4.2 Chartres I) and the other allele contained a di-nucleotide deletion resulting in a premature stop signal (protein 4.2 Chartres II). Homology modelling showed that the hairpin region that forms the proposed band 3 binding site is still present in both mutants. However, the deletion of exon 9 removes a large portion of Domain 2 (core domain) of protein 4.2, potentially removing a band 3 binding groove, and the truncation mutant lacks a portion of the core domain and the whole domains 3 and 4. Therefore, these mutations are likely to destabilize protein 4.2 either directly, or indirectly by disturbing the interaction of protein 4.2 with band 3. Flow cytometry, SDS-PAGE and Western blotting of erythrocyte membranes showed a significant reduction of 70–80 % in CD47 levels, altered Rh associated glycoprotein (RhAG) mobility, reduced GPA/GPB heterodimers, and a 3 fold increase in CD44 levels as reported previously for protein 4.2 null red cells. We stored mature red cells at 4 degrees Celsius over 35 days and found that CD47 continues to be lost in microvesicles as the red cell ages, consistent with a weaker link of CD47 with the cytoskeleton. We investigated band 3 complex stability by performing co-immunoprecipitations and found that lower amounts of band 3 were co-immunoprecipitated using an anti-ankyrin antibody in Chartres red cells compared to wild type, suggesting that the association of band 3 with the cytoskeleton is severely affected. Furthermore, less band 3 was co-immunoprecipitated with an anti-RhAG antibody, consistent with a disturbance of the association of the Rh complex with band 3. We next investigated the stage during erythropoiesis at which the observed changes in band 3 macrocomplex proteins occur. To this end we expanded and differentiated erythroid progenitors from peripheral blood of wild type and the Chartres patient using a three culture system modified from Leberbauer et al. (2005). Synchronous differentiation of a pure erythroid progenitor pool (60% enucleation) demonstrated that protein 4.2 co-immunoprecipitated with band 3 early on in erythroid progenitor differentiation. However, in protein 4.2 Chartres progenitors the mutant forms of protein 4.2 were not expressed at any stage during erythropoiesis, demonstrating that both protein 4.2 mutants are unstable and rapidly degraded. Surprisingly, flow cytometry and western blot analysis revealed that CD47, RhAG, band 3, CD44, and GPA/GPB levels are all similar compared to wild type during erythroid differentiation. Thus, despite the absence of protein 4.2 throughout erythropoiesis, the final changes in the Rh/band3 complex observed in patient’s erythrocytes are not observed. Overall our results suggest that protein 4.2 Chartres is unstable probably due to specific 4.2 mutations that either cause disruption of the band 3 binding sites or an intrinsic instability of these individual mutant proteins. The association of band 3 and ankyrin also appears to be altered in protein 4.2 Chartres suggestive of a weakening of the band 3 cytoskeleton linkage, which could also contribute to the HS phenotype. Importantly, the absence of protein 4.2 not only disturbs ankyrin recruitment to band 3 but also affects association of band 3 with RhAG and disturbs GPA/GPB complexes, which demonstrates the importance of protein 4.2 in the process of band 3 complex formation. Most strikingly, our work demonstrates that the loss of CD47 and the other alterations observed in the band 3/Rh complex in protein 4.2 Chartres must occur late during red blood cell progenitor maturation, presumably after enucleation.

2016 ◽  
Vol 39 (1) ◽  
pp. 331-340 ◽  
Author(s):  
Richard S. Hoehn ◽  
Peter L. Jernigan ◽  
Alex L. Chang ◽  
Michael J. Edwards ◽  
Charles C. Caldwell ◽  
...  

Background/Aims: During storage, units of human red blood cells (pRBCs) experience membrane destabilization and hemolysis which may cause harm to transfusion recipients. This study investigates whether inhibition of acid sphingomyelinase could stabilize erythrocyte membranes and prevent hemolysis during storage. Methods: Human and murine pRBCs were stored under standard blood banking conditions with and without the addition of amitriptyline, a known acid sphingomyelinase inhibitor. Hemoglobin was measured with an electronic hematology analyzer and flow cytometry was used to measure erythrocyte size, complexity, phosphatidylserine externalization, and band 3 protein expression. Results: Cell-free hemoglobin, a marker of hemolysis, increased during pRBC storage. Amitriptyline treatment decreased hemolysis in a dose-dependent manner. Standard pRBC storage led to loss of erythrocyte size and membrane complexity, increased phosphatidylserine externalization, and decreased band 3 protein integrity as determined by flow cytometry. Each of these changes was reduced by treatment with amitriptyline. Transfusion of amitriptyline-treated pRBCs resulted in decreased circulating free hemoglobin. Conclusion: Erythrocyte storage is associated with changes in cell size, complexity, membrane molecular composition, and increased hemolysis. Acid sphingomyelinase inhibition reduced these changes in a dose-dependent manner. Our data suggest a novel mechanism to attenuate the harmful effects after transfusion of aged blood products.


Blood ◽  
1978 ◽  
Vol 51 (3) ◽  
pp. 385-395 ◽  
Author(s):  
J Palek ◽  
SC Liu ◽  
LM Snyder

Abstract The discocyte-echinocyte transformation and the decrease in deformability associated with red cell ATP depletion have been attributed to changes in the physical properties of spectrin and actin, membrane proteins located at the membrane-cytosol interface. We investigated the spontaneous formation of spectrin-rich complexes in human erythrocyte membranes, employing two-dimensional SDS- polyacrylamide gel electrophoresis. Membranes of red cells depleted in ATP under aerobic conditions exhibited (1) an increase in components 4.5 and 8 and globin subunits, (2) a spontaneous formation of heterodimers of spectrin 1 + 2 and spectrin 2 + component 4.9, and (3) a large molecular weight (greater than 10(6) daltons) protein complex with a high spectrin to band 3 ratio. These complexes were dissociated with dithiothreitol and were prevented by anaerobic incubation or the maintenance of red cell ATP and GSH levels with glucose, adenine, and inosine. The complexes 1 + 2 and 2 + 4.9 were also seen in acetylphenylhydrazine-treated, glucose-6-phosphate dehydrogenase- deficient fresh erythrocytes that showed marked GSH depletion but preserved greater than 70% of the original ATP level. However, membranes of these cells did not contain the greater 10(6) dalton aggregate with a high spectrin to band 3 ratio. We concluded that the formation of the latter complex results from rearrangement of spectrin and other polypeptides in membranes of ATP-depleted red cells. Under aerobic conditions, the rearranged proteins undergo spontaneous intermolecular crosslinkings through disulfide couplings.


Blood ◽  
1996 ◽  
Vol 88 (7) ◽  
pp. 2745-2753 ◽  
Author(s):  
AC Rybicki ◽  
RS Schwartz ◽  
EJ Hustedt ◽  
CE Cobb

Band 3 (anion-exchange protein 1-[AE1]) is the major integral membrane protein of human erythrocytes and links the membrane to the underlying cytoskeleton via high-affinity binding to ankyrin. It is unclear whether other cytoskeletal proteins participate in strengthening the ankyrin-band 3 linkage, but a putative role for protein 4.2 (P4.2) has been proposed based on the increased osmotic fragility and spherocytic morphology of P4.2-deficient red blood cells (RBCs). The present study was designed to investigate the hypothesis that P4.2 has a direct role in strengthening the band 3-cytoskeleton linkage in human RBCs, by measuring independent features of this interaction in normal and P4.2-deficient RBCs. The features examined were the rotational mobility of band 3 assayed by time-resolved phosphorescence emission anisotropy (TPA), and the extractability of band 3 by octyl-beta-glucoside, the latter being a nonionic detergent that selectively extracts only band 3 that is not anchored to the cytoskeleton. We find that the amplitude of the most rapidly rotating population of band 3 (correlation time, approximately 30 to 60 microseconds) is increased 81% and 67% in P4.2-deficient ghosts (P4.2NIPPON and band 3MONTEFIORE, respectively) compared with control ghosts. The amplitude of the intermediate speed rotating population of band 3 (correlation time, approximately 200 to 500 microseconds) is increased 23% and 8% in P4.2-deficient ghosts (P4.2NIPPON and band 3MONTEFIORE, respectively) compared with control ghosts, at the expense of the slowly rotating component (correlation time, approximately 2,000 to 3,000 microseconds, amplitude decreased 43% and 39% in P4.2NIPPON and band 3MONTEFIORE, respectively) and immobile component (immobile on this experimental time scale; amplitude decreased 26% and 10% in P4.2NIPPON and band 3MONTEFIORE, respectively) of band 3. These results demonstrate that P4.2 deficiency partially removes band 3 rotational constraints, ie, it increases band 3 rotational mobility. The nonionic detergent octyl-beta-glucoside, which does not disturb band 3-cytoskeleton associations, ie, it extracts only band 3 that is not attached to the cytoskeleton, extracted 30% and 61% more band 3 from P4.2NIPPON and band 3MONTEFIORE ghost membranes, respectively, compared with control ghosts. The octyl-beta-glucoside ghost extracts from both P4.2-deficient phenotypes were enriched in band 3 oligomeric species (tetramers, higher-order oligomers, and aggregates) compared with controls. Since band 3 oligomers selectively associate with the cytoskeleton, these results are consistent with a weakened band 3-cytoskeleton linkage in P4.2-deficient RBC membranes. P4.2 deficiency does not affect band 3 anion transport activity, since uptake of radiolabeled sulfate was similar for control and P4.2-deficient RBCs. Thus, we propose that P4.2 directly participates in strengthening the band 3-cytoskeleton linkage.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 3572-3572
Author(s):  
Colleen E. Annesley ◽  
Rachel E. Rau ◽  
Daniel Magoon ◽  
Gregory McCarty ◽  
David Loeb ◽  
...  

Abstract Background: The WT1 gene encodes for a zinc finger-containing transcription factor involved in differentiation, cell cycle regulation and apoptosis. WT1 expression is developmentally regulated and tissue-specific, with expression maintained in the kidney and in CD34+ hematopoietic progenitor cells. WT1 mutations are reported in approximately 10-15% of both adult and pediatric patients with acute myeloid leukemia (AML), and have been associated with treatment failure and a poor prognosis. Reported mutations consist of insertions, deletions or point mutations; and occur primarily in exon 7 or exon 9 of the WT1 gene. These mutations are thought to alter WT1 DNA-binding ability or result in a loss of function. Despite these observations, the functional contribution of WT1 mutations in leukemogenesis is still largely undetermined. Results and Methods: We have shown that transduction and expression of wild type WT1 in murine 32D cells enhances granulocytic differentiation upon treatment with G-CSF, and that expression of mutant WT1 inhibits this effect. To investigate this in a human AML cell model, we transduced U937 cells with the same WT1 vectors. Strikingly, shortly after transduction, U937 cells expressing wild type WT1 spontaneously differentiate towards a mature monocytic phenotype, but U937 cells expressing mutant WT1 do not differentiate and maintain an immature phenotype (Fig A). This relative block in U937 differentiation with mutant WT1 expression was overcome with differentiation-inducing treatment with all-trans retinoic acid (ATRA). These results suggest that mutant WT1 alters the ability of myeloid cells to terminally differentiate. We obtained a novel knock-in WT1 mutant (WT1mut) mouse model that is heterozygous for the missense mutation R394W in exon 9, and homologous to exon 9 mutations observed in human AML. We evaluated cohorts of two-month old mice and noted an expansion of lineage negative cells and various progenitor cell compartments; particularly, the megakaryocyte-erythroid progenitor (MEP) compartment; in WT1mut bone marrow (BM) relative to wild type. We also found that lineage negative WT1mut BM cells from two-month old mice show higher in vitro colony-forming capacity and an increased ability to serially replate in methylcellulose culture compared to wild type BM cells. Flow cytometry of WT1mut cells at tertiary replating revealed an immature, largely c-kit+ population, suggesting an aberrantly enhanced self-renewal capability of myeloid progenitors in WT1mut mice. Furthermore, survival analysis of the WT1mut mice demonstrates inferior survival compared to wild type, and several WT1mut mice were found to have anemia and myelodysplasia. To address the possibility of germ line WT1mut syndromes causing renal failure and anemia, and thereby influencing survival, we transplanted BM from each genotype into lethally irradiated congenic mice. Upon engraftment with donor marrow, the expression of WT1mut is confined to the hematopoietic system in this model. The Kaplan-Meier survival curve, based on absolute age of the BM, shows statistically significant decreased survival of WT1mut BM transplant recipients compared to wild type BM recipients (Fig B). Anemia and dysplasia were also seen in these WT1mut BM recipients; findings that are suggestive of dysfunctional hematopoiesis, and may be secondary to the changes in progenitor cell self-renewal and differentiation we have observed. Conclusions: Leukemogenic WT1 mutations confer enhanced self-renewal of hematopoietic progenitor cells and a block in terminal myeloid differentiation in vitro, which could potentially prime cells for leukemic transformation upon acquisition of cooperative events. Mice with WT1 mutant bone marrow develop anemia and evidence of myelodysplasia, which may contribute to their decreased survival. These data provide new and important insights into the aberrant functional effects of WT1 mutations on hematopoiesis, and are the first to characterize the hematopoietic phenotype of a WT1 mutation in vivo. Figure: (A) U937 cells expressing wild type WT1 spontaneously differentiate, demonstrated here by gain of monocytic markers CD11a and CD11b as measured by flow cytometry, whereas cells expressing mutant WT1 vectors 101 and 126 remain undifferentiated. (B) Mice transplanted with WT1mut bone marrow have inferior survival compared to mice transplanted with wild type bone marrow. Figure:. (A) U937 cells expressing wild type WT1 spontaneously differentiate, demonstrated here by gain of monocytic markers CD11a and CD11b as measured by flow cytometry, whereas cells expressing mutant WT1 vectors 101 and 126 remain undifferentiated. (B) Mice transplanted with WT1mut bone marrow have inferior survival compared to mice transplanted with wild type bone marrow. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4003-4003
Author(s):  
Timothy J Satchwell ◽  
Bethan R Hawley ◽  
Amanda J Bell ◽  
Maria Leticia Ribeiro ◽  
Ashley M Toye

Abstract The bicarbonate/chloride exchanger protein band 3 is the most abundant protein in the erythrocyte membrane and forms the core of a major multiprotein complex required for vertical association between the plasma membrane and the underlying spectrin cytoskeleton. A wealth of knowledge, derived from a host of varied studies including in vitro binding assays, work on mature erythrocytes and in other cellular systems have identified a number of binding partners including ankyrin, adducin and protein 4.2 amongst others. However, studies of the role that band 3 and the establishment of its connectivity with the cytoskeleton play both in assembly of multiprotein complexes during erythropoiesis and in particular in protein retention during enucleation have been understandably limited by the technical challenges associated with study of this protein within its unique native cellular context. The complete absence of band 3 in human erythrocytes has only been reported once, in a Portuguese patient with severe hereditary spherocytosis and distal renal tubular acidosis resulting from homozygosity for a V488M band 3 mutation (band 3 Coimbra). In this study, we used in vitro culture of erythroblasts derived from this patient as well as shRNA mediated depletion of band 3 to investigate the development of a band 3 deficient erythrocyte membrane and to specifically assess the formation, stability and retention of band 3 dependent protein complexes in the absence of this core protein during erythropoiesis and erythroblast enucleation. We demonstrate that the mutant band 3 Coimbra protein is expressed at very low but detectable levels during erythropoiesis but does not reach the cell surface and is not rescued by interaction with wild type protein. Failure to traffic to the plasma membrane and rapid degradation during erythropoiesis accounts for the absence of band 3 in Coimbra erythrocytes. The absence of plasma membrane expression of band 3 results in secondary deficiencies of a host of band 3 associated membrane proteins that we quantitatively show result predominantly from reduced plasma membrane expression during erythropoiesis compounded by impaired retention in the nascent reticulocyte membrane during erythroblast enucleation. In order to explore the importance of the capacity of band 3 to associate with the cytoskeleton for surface expression of this protein and its associated multiprotein complex binding proteins, immature band 3 Coimbra patient erythroblasts were lentivirally transduced with N terminally GFP-tagged wild type band 3 or band 3 mutants with absent or impaired ability to associate with the cytoskeleton. We demonstrate for the first time the ability to restore expression of band 3 to normal levels in this uniquely compromised patient and to rescue key secondary protein deficiencies arising from the absence of band 3 in reticulocytes. Exogenous expression levels of band 3, monitored by GFP intensity, correlate directly with degree of rescue of a variety of band 3 associated proteins. When expressed in band 3 deficient Coimbra erythroblasts, the band 3 membrane domain, which is unable to associate with the cytoskeleton, exhibits an increased partitioning to the plasma membrane surrounding the extruded nuclei compared to wild type band 3 and fails to rescue reticulocyte membrane retention of band 3 associated proteins. Expression of the kidney isoform of band 3, which is unable to bind ankyrin but retains the binding site for the cytoskeletal accessory protein, protein 4.2 results in partial rescue of the protein 4.2 dependent CD47 only. This demonstrates the importance of band 3 association with the cytoskeleton for efficient retention of band 3 associated proteins during erythroblast enucleation. Interestingly, whilst both exhibit reduced reticulocyte membrane retention relative to wild type, a significant proportion of both band 3 membrane domain and kidney band 3 is retained in the reticulocyte membrane following erythroblast enucleation indicating that cytoskeletal attachment of band 3 is not the sole determinant of partitioning during this complex process. This study advances our understanding of the mechanisms by which the properties of band 3 influence the sculpting and composition of the erythrocyte membrane and highlights the role of this protein as a core for assembly and stabilisation of key membrane proteins in both the early and late stages of terminal erythroid differentiation. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1192-1192
Author(s):  
Yoann Vial ◽  
Jeannette Nardelli ◽  
Justine Rousselot ◽  
Emilie Lepeu ◽  
Michele Souyri ◽  
...  

Introduction: Microcephaly is a recurrent feature in patients with inherited bone marrow failure (iBMF) and DNA damage response (DDR) disorders suggesting that common cellular pathways regulate the proliferation and differentiation of neural and hematopoietic progenitors. However, while several studies addressed the role of iBFM or DDR genes in brain development, a possible role for microcephaly genes in hematopoietic development has not been investigated. To address this issue, we studied a mouse model of primary microcephaly with biallelic loss-of-function in Mcph1.MCPH1 mutations are found in 10% of patients with isolated forms of genetic microcephaly (MCPH). Interestingly, MCPH1 helps to maintain genomic integrity during cell division by interacting with proteins involved in cell cycle progression, apoptosis or DNA repair, all cellular processes being also involved in iBMF and DDR syndromes. Methods: Mcph1 null mice were generated by germline deletion of Mcph1 exon 2 (Mcph1tm1.2Kali) (Liang et al., PLoS Genet., 2010). The subpopulations of erythroid progenitors S0 to S5 were phenotypically defined and sorted by flow cytometry according to CD71 and Ter119 expression in the Lin- compartment from mouse liver obtained at birth and during fetal development (E12.5). RNA sequencing (RNA-Seq) was performed on sorted erythroid progenitor fractions obtained from E12.5 fetal livers (SMARTer® Stranded Total RNA-Seq Kit V2-Pico Input library preparation kit). Cell division was studied by multiplexing erythroid specific antibodies with EdU flow cytometry cell proliferation assay. Results: Null mice recapitulated the microcephaly phenotype seen in MCPH patients, but also showed a striking anemic pallor. Numeration and cytomorphologic examination of peripheral blood at birth confirmed macrocytic anemia with low red blood cell count and anisopoikilocytosis. These observations were consistent with congenital dyserythropoiesis in Mcph1-/- mice and prompted us to further characterize the erythroid lineage. Quantification of erythroid progenitor populations in liver at birth showed a significant decreased from the S3 subset (Lin-, CD71High, Ter119High) suggesting impaired terminal differentiation. Similar results were obtained in fetal livers at E12.5 indicating that the defect arose early in hematopoietic ontogeny. Transcriptome analysis of wild-type progenitor populations (S0 to S3) confirmed that Mcph1 is expressed during normal erythropoiesis following a Gata1-like expression profile. This is consistent with the presence in the Mcph1 gene promoter of a binding site for Gata1 and Ldb1 (ENCODE project), supporting an activation by the main erythroid differentiation complex. Strikingly, RNA-Seq analysis revealed deregulation of p53 pathway associated genes in all subsets of Mcph1-/- erythroid progenitors as compared to their wild-type counterparts. Two transcriptional p53 targets involved in cell cycle control, Cdkn1a coding the cyclin-dependent kinase inhibitor (p21) and Ccng1 coding Cyclin G1, were among the most upregulated genes. Cell cycle analysis performed on sorted erythroid progenitors revealed an endoreduplication phenomenon restricted to the S3 subset with subsequent accumulation of tetraploid cells. Interestingly, physiological endoreduplication is initiated by p21 and E2Fs transcription factors, and Mcph1 functionally interacts with E2f1. Our findings suggest that, in the absence of Mcph1, Cdkn1a overexpression possibly combined to a decreased E2f1 activity may lead to endoreduplication in S3 progenitors, impairing further differentiation into mature red blood cells. Few data are available for patients with MCPH1 mutations, hematological defects being possibly outlooked due to the severity of the neurological phenotype. However, CBC performed in one of our patients revealed a macrocytosis consistent with dyserythropoiesis evidenced in mice. Conclusion: We demonstrate for the first time that Mcph1 expression is critical during terminal erythroid differentiation in mice. Altogether our findings provide additional evidence of a unique link between hematopoiesis and neuronal development. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
1978 ◽  
Vol 51 (3) ◽  
pp. 385-395
Author(s):  
J Palek ◽  
SC Liu ◽  
LM Snyder

The discocyte-echinocyte transformation and the decrease in deformability associated with red cell ATP depletion have been attributed to changes in the physical properties of spectrin and actin, membrane proteins located at the membrane-cytosol interface. We investigated the spontaneous formation of spectrin-rich complexes in human erythrocyte membranes, employing two-dimensional SDS- polyacrylamide gel electrophoresis. Membranes of red cells depleted in ATP under aerobic conditions exhibited (1) an increase in components 4.5 and 8 and globin subunits, (2) a spontaneous formation of heterodimers of spectrin 1 + 2 and spectrin 2 + component 4.9, and (3) a large molecular weight (greater than 10(6) daltons) protein complex with a high spectrin to band 3 ratio. These complexes were dissociated with dithiothreitol and were prevented by anaerobic incubation or the maintenance of red cell ATP and GSH levels with glucose, adenine, and inosine. The complexes 1 + 2 and 2 + 4.9 were also seen in acetylphenylhydrazine-treated, glucose-6-phosphate dehydrogenase- deficient fresh erythrocytes that showed marked GSH depletion but preserved greater than 70% of the original ATP level. However, membranes of these cells did not contain the greater 10(6) dalton aggregate with a high spectrin to band 3 ratio. We concluded that the formation of the latter complex results from rearrangement of spectrin and other polypeptides in membranes of ATP-depleted red cells. Under aerobic conditions, the rearranged proteins undergo spontaneous intermolecular crosslinkings through disulfide couplings.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 167-167
Author(s):  
Thomas M. Renaud ◽  
Stefano Rivella

Abstract Abstract 167 Anemia of inflammation is the second most common form of anemia in the general population, and its impact on patient well-being is largely underestimated. Anemia cause by inflammation is multi-factorial and includes hepcidin-induced iron restricted erythropoiesis as well as direct cytokine effects on the bone marrow, erythropoietin production and efficacy, and on the lifespan of red cells. Many murine models of anemia of inflammation are unreliable or cumbersome, but a new model introduced by Sasu et al (Blood, 2010) using a single intraperitoneal injection of heat-killed brucella abortus antigen (HKBA) has proven reproducible and robust. We have used this model to explore the role of interleukin-6 and bone marrow derived cell production of hepcidin in anemia of inflammation (AI). First, we sought to explore the effect this model of AI in wild type mice, iterleukin-6 knockout mice (IL6-KO) and hepcidin knockout mice (Hamp-KO) (n≥15 for each group). We followed these mice for 7 weeks with weekly CBC's to observe the severity and time to recovery from anemia. Wild type mice were most affected 2 weeks after injection and slowly recovered over 7 weeks (HgB at 2 week = 6.4g/dl ± 1.2). IL6-KO mice were equally affected initially, with similar hemoglobin values at 2 weeks (6.9g/dl ± 1.3) and recovered by 6 weeks. Hamp-KO mice were less affected throughout the course of anemia, with hemoglobin values of 10.8g/dl ± 0.7 at 2 weeks with resolution by week 4. IL6-KO mice began to recover more quickly than wild type mice by week three, with hemoglobin values of 10.9g/dl ± 1.5 at that time, compared to wild type mice at 3 weeks with hemoglobin values of 7.4g/dl ± 0.7 (p= 0.0001). We believe that this demonstrates that interleukin-6 and hepcidin do coordinate to contribute to anemia of inflammation, but that there may be independent effects or additional factors. To address these questions, we are currently evaluating iron-related gene expression in these groups of mice as well as evaluating iron stores at multiple time points. We also evaluated serum cytokine levels in each of these groups of mice. We found similar elevations TNF-alpha and interferon gamma in all three groups at 6 and 24 hours. We found similar elevations of IL-6 in wild type and Hamp-KO mice at 6 and 24 hours. Bone marrows and spleens form each group of mice were evaluated at 2 weeks by flow cytometry using ter119 and CD44 to evaluate specific effects on erythroid maturation. This evaluation demonstrated a a profound inhibitory effect on erythropoiesis and, in particular, on the production of erythroid progenitor cells, showing a similar profile by flow cytometry between the three groups. In vitro studies have suggested that macrophage production of hepcidin is important in the development of AI (Theurl et al 2008). We evaluated the importance of bone marrow derived cell production of hepcidin on the development of AI using bone marrow chimeras. Using 600cGy × 2 as a preparative regimen, we transplanted wild type mice with bone marrow from Hamp-KO mice. We also irradiated Hamp-KO mice and transplanted them with wild type marrow. We injected these two groups of mice as well as wild type and Hamp-KO controls, we followed them for a period of 4 weeks with weekly CBC's to evaluate the degree of anemia. Hemoglobin values of wild type mice transplanted with Hamp-KO marrow were statistically indistinguishable from those of non-transplanted wild type mice during the follow-up period (HgB values at 1 week = 6.8g ± 0.7 vs 7.29g ± 1.1; at 2 weeks = 7.3 ± 0.6 vs 6.4 ± 1.2; at 3 weeks = 8.5 ± 1.8 vs 7.4 ± 0.5; at 4 weeks = 9.1 v 1.9 vs 8.6 ± 0.5; p<0.02 for all time points). Hamp-KO mice with wild-type bone marrow were statistically indistinguishable from non-transplanted Hamp-KO mice (Hgb values at 1 week = 9.9 ± 2.4 vs 10.8 ± 0.7; at 2 weeks = 10.6 ± 1.5 vs 10.3 ± 1.0; at 3 weeks = 12.8 ± 1.2 10.8 ± 0.7; at 4 weeks 12.6 ± 1.2 vs. 13.6 ± 1.0; p<0.02 for all time points). This suggests that the production of hepcidin by bone marrow derived cells dose not play a physiologically important role in the development of anemia of inflammation. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 2665-2665
Author(s):  
Kathryn E Mordue ◽  
Timothy J Satchwell ◽  
Ashley M Toye

Abstract CD47 is a ubiquitously expressed ‘Marker of Self’ that protects cells from phagocytosis, through recognition by SIRPα on macrophages (Oldenborg et al Science 2000). CD47 was originally isolated on ovarian tumour cells (Poels et al J Natl Cancer Inst 1986) and has subsequently been detected on leukemic stem cells, where increased CD47 levels ensure immune evasion (Jaiswal et al Cell 2009). CD47 is also a ‘Marker of Self’ on red cells, but is reduced at the cell surface in certain patients with Hereditary Spherocytosis. In red cells, ~60% of CD47 is connected to the cytoskeleton (Dahl et al Blood 2004). Cytoskeletal connectivity of CD47 in the red cell membrane is dependent on the band 3 complex associated protein 4.2, demonstrated by an ~80% reduction in CD47 levels in protein 4.2 null red cells (Mouro-Chanteloup et al Blood 2003). Previous work (van den Akker et al Haematologica 2009) established that CD47 becomes dependent on protein 4.2 at the basophilic erythroblast stage (48 hours post-differentiation), but it is unknown what interactions support CD47 membrane stability prior to protein 4.2 expression during expansion and early erythroid differentiation. CD47 mRNA is alternatively spliced giving rise to four potential isoforms. The most abundant isoforms are form 2, expressed in all bone-marrow derived cells, and form 4 (and form 3), found predominantly in neural tissues (Reinhold et al J Cell Sci 1995). CD47 isoform 2 is the only form expressed on mature red cells, but we hypothesized that expression of other CD47 isoforms with different trafficking or binding characteristics could explain the independence of CD47 prior to band 3 complex assembly. Using specific polyclonal antibodies to multiple CD47 isoforms, we demonstrate that isoform 2 is expressed prior to and throughout in vitroerythroid differentiation. CD47 isoforms 3 and 4 were detected by western blotting until the late polychromatic erythroblast stage (96 hours post-differentiation), but only CD47 isoform 2 was detected at the cell surface. Therefore, we next hypothesised that CD47 must interact with another protein or exhibit different trafficking characteristics to maintain its membrane stability early during terminal differentiation. To identify a candidate protein or associated protein complex, CD47 was immunoprecipitated from expanding erythroblasts (Exp), proerythroblasts (T0), and basophilic erythroblasts (T48), and analysed via Nano-LC mass spectroscopy. In Exp and T0 erythroblasts, CD47 pulled down actin and multiple actin-associated proteins. These interactions were not observed in T48 erythroblasts, corresponding to the time during terminal differentiation when CD47 is dependent on protein 4.2. To confirm a dependence on actin for CD47 membrane stability, well-characterised drugs that disrupt actin dynamics were employed. CD47 expression at the cell membrane, as judged by flow cytometry, was markedly reduced within 30 minutes using actin stabilising drugs (Cytochalasin D (5µM): Exp 13.7±5.4% versus T48 0.5±5.7%; Latrunculin A (1µM): Exp 18.9±3.5% versus T48 9.9±5.9%, of the DMSO control), and destabilising drug (Jasplakinolide (1µM): Exp 24.2±1.9% versus T48 -6±1.8%, of the DMSO control), until the basophilic erythroblast stage. In K562 cells, which predominantly express CD47 isoforms 3 and 4, a larger actin dependency is observed (37±14.9% reduction in CD47 with Cytochalasin D versus a DMSO control) suggesting that dependence on actin by CD47 is not isoform specific. In summary, we propose a role for actin in the maintenance of CD47 at the cell surface before and during early erythroid differentiation. We have shown that CD47 isoform 2 is the major isoform present at the cell surface and that this version is initially dependent on the actin cytoskeleton for its membrane stability by an as yet undetermined mechanism. Once band 3 complex assembly initiates at the surface of the basophilic erythroblast (48 hours post-differentiation), CD47 is selectively incorporated via an interaction with protein 4.2, and is preferentially retained whilst the actin cytoskeleton remodels. In addition to explaining how CD47 expression is maintained during the formation of the red cell membrane, this work raises the possibility that the dependence on actin by CD47 for its membrane stability in hematopoietic stem cells may be exploited for the development of therapeutics that render the leukemic cells susceptible to phagocytosis. Disclosures No relevant conflicts of interest to declare.


1997 ◽  
Vol 35 (11-12) ◽  
pp. 451-453
Author(s):  
F. X. Abad ◽  
A. Bosch ◽  
J. Comas ◽  
D. Villalba ◽  
R. M. Pintó

A method has been developed for the detection of infectious human rotavirus (HRV), based on infection of MA104 and CaCo-2 cell monolayers and ulterior flow cytometry. The sensitivity of the flow cytometry procedure for the cell-adapted HRV enabled the detection of 200 and 2 MPNCU in MA104 and CaCo-2 cells, respectively. Flow cytometry performed five days after infection of CaCo-2 enabled the detection of naturally occurring wild-type HRV in faecal samples and concentrated water samples.


Sign in / Sign up

Export Citation Format

Share Document