scholarly journals Ex vivo Engineering of Red Blood Cells

Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. SCI-5-SCI-5
Author(s):  
Jan Frayne ◽  
Deborah E Daniels ◽  
Jan Frayne ◽  
Timothy J Satchwell ◽  
Joseph Hawksworth ◽  
...  

Culture systems for human in vitro erythropoiesis are now well established. Using our 3-stage feeder-free erythroid culture system we can efficiently differentiate erythroid cells from adult and cord blood (CB) CD34+ cells with >105 fold expansion, enucleation rates of up to 95% and producing packed reticulocyte yields of >12ml post leukofiltration1. The final preparations for a first in man clinical trial of adult cultured reticulocytes produced under good manufacturing practice (RESTORE) are underway. Although we have shown that it is possible to modify the CD34+ derived cells using lentivirus for reengineering or additions to the medium, the finite proliferative capacity of CD34+ cells in culture currently limits yield. We therefore took the alternative approach of immortalising early erythroid cells differentiated from adult bone marrow (BM) CD34+ cells, creating the BEL-A (Bristol Erythroid Line Adult) line2, a sustainable erythroid cell source that recapitulates normal adult erythropoiesis, terminally differentiating to generate enucleated reticulocytes that express normal levels of adult globin. We have created a further 13 lines from BM, adult peripheral blood, CB and iPSC CD34+ cells, demonstrating reproducibility of the approach and its application to create lines from more accessible stem cell sources. Analysis of surface marker and globin profiles demonstrate the lines follow a similar differentiation profile to their respective primary cell source, with comparative proteomics confirming cell source representation of the lines. Lines always established at the pro-erythroblast/early basophilic stage, even when later stage erythroid cells were present in populations. As well as proof of principal as an alternative transfusion product and improved tools for studying erythropoiesis, such lines have far reaching additional applications, a number of which we are now exploring: Diagnostic and 'Universal' transfusion products: Presently serological testing by blood group reference laboratories relies on donated blood, which represent a finite resource and for some blood group phenotypes can be difficult to source. We used CRISPR-Cas9 gene editing to remove blood group antigens in order to generate a sustainable bank of cell lines with useful blood group phenotypes for diagnostic purposes3. Building on this, with the aim of developing a more compatible "universal" transfusion product to meet the needs of chronically transfused patients and those with rare blood group phenotypes, we used combinatorial gene targeting to create sublines deficient in multiple antigens responsible for the most common transfusion incompatibilities (ABO [Bombay], Rh [Rhnull], Kell [K0], Duffy [Fynull], GPB [S-s-U-]). Individual and multiple blood group knockout lines retained the ability to undergo terminal differentiation and enucleation, also illustrating the capacity for coexistence of multiple rare blood group phenotypes within viable reticulocytes3. Cytokine independent lines Cytokines represent a substantial cost contribution to erythroid culture systems. We therefore exploited activating mutations found in patient c-Kit and EPOR that cause hypersensitivity to ligand, to create cytokine independent lines thus increasing economic viability of cultured red cells. Bi-allelic EPOR or c-kit edits were introduced into BEL-A with confirmation and exploration of mechanism on differentiation in the absence, or with substantially reduced levels of cytokines. Model disease systems In addition to potential therapeutic applications we are also creating lines as model cellular disease systems for studying molecular mechanisms and as drug screening platforms, via CRISPR-Cas9 gene editing of BEL-A and by directly immortalising patient stem cells. To date we have made b-thalassemia major, HbE thalassemia and lines with KLF1 mutations. Furthermore, we have shown BEL-A reticulocytes support invasion and growth of Plasmodium falciparum and are utilising the line to study mechanisms of malaria parasite invasion4. Kupzig S, Parsons SF, Curnow E, Anstee DJ, Blair A. Superior survival of ex vivo cultured human reticulocytes following transfusion into mice. 2016;102:476-483Trakarnsanga K, Griffiths RE, Wilson MC, et al. An immortalized adult human erythroid line facilitates sustainable and scalable generation of functional red cells. Nat. Commun. 2017;8:14750Hawksworth J, Satchwell TJ, Meinders M, et al. Enhancement of red blood cell transfusion compatibility using CRISPR-mediated erythroblast gene editing. EMBO Mol. Med. 2018; 10:e8454Satchwell TJ, Wright K, Haydn-Smith K, et al. Genetic manipulation of cell line derived reticulocytes enables dissection of host malaria invasion requirements. Nat. Comm. 2019;10:3806 Disclosures No relevant conflicts of interest to declare.

Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. SCI-46-SCI-46
Author(s):  
Anna Rita F Migliaccio ◽  
Carolyn Whitsett ◽  
Giovanni Migliaccio

Abstract Abstract SCI-46 Blood transfusion, the earliest form of cell replacement therapy, has become indispensable for modern medicine making the safety and adequacy of the blood supply a national priority. The US blood supply is adequate overall because in 2006 the number of blood units collected exceed by 7.8% the number of those transfused. However, issues surrounding blood transfusion, such as sporadic shortages and potential adverse events to recipients (related to changes in red cell physiology during storage and alloimmunization in chronically transfused patients) prompted past and current efforts to develop alternative transfusion products. Recently, the culture conditions to generate erythroid cells have greatly improved making the production of a transfusion product ex-vivo a theoretically possible, although expensive, proposition. This recognition is inspiring several investigators to develop production processes for ex-vivo generation of red cell transfusion products. A proof-of-concept demonstrating that ex-vivo generated red cells protect mice from experimentally induced lethal anemia has been obtained. Alternative sources of stem cells which include human embryonic stem cells (hESC) and induced pluripotency stem cells (iPS), are being explored. Since red cells do not have a nucleus, safety considerations suggest that they may represent the first cell therapy product to be generated from hESC and iPS. In addition, discarded hematopoietic stem cells present in adult and cord blood donations may theoretically generate numbers of red cells ex-vivo sufficient for transfusion. Affordable clinical grade humanized culture media have also been developed. Possible differences in immunological and biological properties of erythroid cells from different sources are under investigation. These differences include size, levels of activity of glycolytic enzymes and carbonic anhydrase, expression of different isozymes, hemoglobin and antigenic profiles (HLA class II antigens). This last aspect is particularly important because ex-vivo expanded red cells pose the same risk for infection and incompatibility as any transfusion product but pose unique antigenic risks. Since expression of blood group antigens is susceptible to post-transcriptional modifications, the ex-vivo expansion process itself may induce antigenic variability. Therefore, even cells generated from completely matched stem cell sources may induce auto-immunity and/or appear incompatible. Regarding the identity of ex-vivo generated red cell transfusion products, a conservative approach would be to define them as “enucleated red cells”. In principle, however, ex-vivo generated erythroblasts may also serve as transfusion product. Since they undergo 4–64 further divisions and reduce iron overload, they may represent a more potent transfusion product for patients that require chronic transfusion. The clinical use of these cells, however, may involve development of specific procedures to facilitate their homing/maturation in the erythroid niches of the recipients. In summary, on the basis of these cost, logistic and safety considerations we hypothesize that the clinical application of ex-vivo expanded erythroblasts will involve in sequence, drug discovery for personalized therapy, systemic drug delivery, genotypically matched transfusion for alloimmunized patients and then transfusion in the general population. Disclosures: No relevant conflicts of interest to declare.


Transfusion ◽  
1983 ◽  
Vol 23 (1) ◽  
pp. 65-66 ◽  
Author(s):  
V Biro ◽  
G Garratty ◽  
CL Johnson ◽  
WL Marsh

Open Biology ◽  
2014 ◽  
Vol 4 (1) ◽  
pp. 130202 ◽  
Author(s):  
Jafar Mahdavi ◽  
Necmettin Pirinccioglu ◽  
Neil J. Oldfield ◽  
Elisabet Carlsohn ◽  
Jeroen Stoof ◽  
...  

Campylobacter jejuni is an important cause of human foodborne gastroenteritis; strategies to prevent infection are hampered by a poor understanding of the complex interactions between host and pathogen. Previous work showed that C. jejuni could bind human histo-blood group antigens (BgAgs) in vitro and that BgAgs could inhibit the binding of C. jejuni to human intestinal mucosa ex vivo. Here, the major flagella subunit protein (FlaA) and the major outer membrane protein (MOMP) were identified as BgAg-binding adhesins in C. jejuni NCTC11168 . Significantly, the MOMP was shown to be O- glycosylated at Thr 268 ; previously only flagellin proteins were known to be O- glycosylated in C. jejuni . Substitution of MOMP Thr 268 led to significantly reduced binding to BgAgs. The O- glycan moiety was characterized as Gal(β1–3)-GalNAc(β1–4)-GalNAc(β1–4)-GalNAcα1-Thr 268 ; modelling suggested that O- glycosylation has a notable effect on the conformation of MOMP and this modulates BgAg-binding capacity. Glycosylation of MOMP at Thr 268 promoted cell-to-cell binding, biofilm formation and adhesion to Caco-2 cells, and was required for the optimal colonization of chickens by C. jejuni , confirming the significance of this O- glycosylation in pathogenesis.


1978 ◽  
Vol 18 (1) ◽  
pp. 16-23 ◽  
Author(s):  
M. J. McDowall ◽  
P. J. Lincoln ◽  
B. E. Dodd

The incorporation of a low ionic strength solution (LISS) in the micro-elution technique used for the detection of blood group antigens in stains markedly improves the test's sensitivity. This is because LISS increases the amount of antibody taken up by the antigen in the stain which results in a greater yield of antibody recovered from the slain by elution. LISS also enhances the activity of the eluted antibody if it is introduced as a suspension medium for the red cells used to detect the antibody. The introduction of suitably diluted AB serum as diluent when testing the eluates is an additional advantage. The improvement in the sensitivity of the micro-elution technique is great enough in some instances to allow the detection of an antigen in a stain which is undetectable in the absence of LISS. Moreover some doubtful positive reactions are enhanced sufficiently for the presence of an antigen to be definitely established.


Vox Sanguinis ◽  
2014 ◽  
Vol 108 (3) ◽  
pp. 302-309 ◽  
Author(s):  
R. Sano ◽  
M. Nogawa ◽  
T. Nakajima ◽  
Y. Takahashi ◽  
K. Takahashi ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 102-102
Author(s):  
Agostino Tafuri ◽  
Francesca Pedini ◽  
Federica Francescangeli ◽  
Michele Signore ◽  
Robert Foa ◽  
...  

Abstract Malignant hemopoietic cells are often characterized by ineffective death pathways, resulting in enhanced resistance to apoptosis and ultimately in the survival/expansion of the abnormal clone. Mechanistic studies have been undertaken to identify the aberrant signal transduction pathway and to develop small-molecule inhibitors targeting deregulated modules. Regulators of apoptotic pathways play a key role in the control of erythroid cell expansion; in particularl Bcl-XL is essential for erythroid cell development and, together with Bcl-2, it protects erythroblast survival from cytotoxic stimuli. Previous studies on erythroid cells derived from polycythemia vera (PV) patients have revealed an increased expression of Bcl-XL associated with cell survival in the absence of erythropoietin. The discovery of the JAK2V617F mutation in the vast majority of PV patients and its association with an increased resistance to apoptosis induced by death receptors prompted us to investigate the correlation between JAK2V617F and the expression of anti-apoptotic Bcl-2 family members and to explore the activity of a Bcl-2 inhibitor on PV erythroid cells. ABT-737 is a synthetic small-molecule inhibitor that binds with high affinity to Bcl-2 and Bcl-XL, poorly to Mcl-1, promoting apoptosis as single agent in malignant hemopoietic cells. In this study we analyzed pure populations of primary erythroid precursors obtained from CD34+ cells of healthy donors and PV patients to investigate the expression of Bcl-2, Bcl-XL and Mcl-1. We found that the expression of Bcl-XL and Bcl-2 was increased in PV patients compared to controls, while Mcl-1 levels did not significantly differ. Then we analyzed Bcl-2 and Bcl-XL expression in erythroblasts derived from PV patients clustered on the basis of a different JAK2V617F allele burden. We found that on average the expression of both Bcl-2 and Bcl-XL was comparable to controls in PV erythroblasts with low/null mutation rates while both proteins were significantly (P< 0.001 and P<0.05, respectively) overexpressed in those with a high percentage of mutated JAK2. Subsequently, we investigated whether ABT-737 was effective in inducing apoptosis in normal and PV erythroblasts. A dose-response induction of apoptosis was elicited by ABT- 737 in PV erythroblasts with elevated JAK267F allele burden with higher effects than in normal cells starting from 500nM (P<0.05). The preferential effects of ABT-737 on PV erythroblasts were further documented by differences in ABT-737-induced mitochondrial depolarization and caspase cleavage. Treatment with ABT-737 did not induce significant alterations in cell cycle distribution on both PV and normal erythroid cells. The activity of ABT-737 was then studied in semisolid cultures. BFU-E colony formation was inhibited in PV-derived CD34+ cells but not in normal controls. To investigate the mechanisms underlying ABT-737 inhibition of erythroblast expansion we analyzed GATA-1 levels that resulted significantly (P<0.05) decreased in PV erytroblasts but not in normal cells, indicating a differential extent of caspase-mediated degradation of GATA-1. Taken together these results indicate that ABT-737 promotes apoptosis of mutated JAK2 PV erythroid cells with a preferential activity compared to normal erythroid cells, supporting a potential therapeutic role of this molecule in patients with PV.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1027-1027 ◽  
Author(s):  
Barbara Ghinassi ◽  
Maria Themeli ◽  
Kai-Hsin Chang ◽  
Gregory Halverson ◽  
Ghazala Hashmi ◽  
...  

Abstract Abstract 1027 Red blood cells (RBC) survive shear forces in the microvasculature because trans-membrane complexes embedded in the lipid bilayer attach their membrane to the cytoskeleton assuring its flexibility. The expression of clinically relevant red blood cell antigens present on these complexes is determined by genetic polymorphisms and their developmental regulation. Therefore, flow cytometry studies of blood group antigens may provide insights both on potential immunogenicity and on membrane structure of ex-vivo generated EBs. Blood group antigen profiles of EBs expanded ex vivo from one AB (three experiments), three CB, the H1 hESC line and one iPS line derived from mononuclear cells from a healthy donor were compared by flow cytometry using commercially available antibodies recognizing antigens present on proteins in the 4.1R [Duffy (Fya and Fy3), Kell (Kell prot, K/k, Kpa/Kpb, Jsb) and glycophorin C (GPC, Ge2)] and ankyrin R [glycophorin A (GPA, CD235a, M and EnaFS) RhAG and band 3 (Wrb)] complexes and on other important membrane proteins [glycophorin B (GPB, s and U), urea transporter (Kidd, Jk3), the complement receptor (CD35) and inhibitors of complement-mediated lysis (CD55 and CD59)]. Controls included DNA genotyping (CB, AB and H1-hESC) (HEA-Bead Chip, Immunocor, Norcross, GA) and immunophenotyping of blood red cells from the same AB and CB. Antigen expression similar to that observed on in vivo generated RBC was considered normal. EBs were generated from AB and CB at day 10 in HEMAser cultures whereas EBs from hESC and iPS were derived using previously optimized protocols. The maturation state was determined by morphological analyses and CD36/CD235a profiles. Irrespective of the stem cell source, the immunophenotype of ex-vivo expanded EBs was consistent with that predicted by genotyping. However, source specific differences in the magnitude of antigen expression and in the changes with maturation were observed (see Figure). Immature EBs from AB expressed normal levels of the antigens present on both the 4.1R (Duffy, Kell, GPC) and ankyrin R (GPA, M/N, EnaFS, RhAG and band 3) complexes. With maturation, expression of 4.1R-associated antigens remained normal while that of ankyrin R associated antigens varied (M decreased and RhAG increased). EBs from CB expressed normal levels of antigens present on the ankyrin R complex and of some of those present on the 4.1R complex (Duffy, Kell protein and GPA). However, expression of epitopes on Kell protein varied with some antigens expressed at normal levels (k and Jsb) and others (Kpa/Kpb) at levels 2x greater than normal. With maturation, CB-derived EBs maintained normal levels of ankyrin R associated antigens while those associated with complex 4.1R became barely detectable. EB from hESC expressed unbalanced levels of proteins associated with both ankyrin R (2x levels of GPA and barely detectable levels of RhAG) and 4.1R [3x levels of Duffy and 2x levels of Jsb (Kell) with normal levels of k and Kpb (Kell) antigens] complexes. The variegation in expression of different epitopes on the same protein observed with CB- and hESC-derived EBs likely reflect altered structural conformation of the complexes rather than differences in protein concentration on the membrane. EBs from iPS, as those from AB, expressed normal levels of antigens present on Ankyrin R and 4.1R complexes which increased with maturation. Irrespective of stem cell sources, EBs expressed normal levels of GPB and Kidd. EBs from AB expressed normal levels of the complement regulatory proteins tested which in the case of CD59 CD59 decreased with maturation. EBs from CB expressed normal levels of CD35 and CD59 but 2x levels of CD55 with expression of CD35 and CD55 decreasing with maturation. EBs from iPS expressed 2x levels of CD35 and CD55 and expression of these antigens was not affected by maturation. The observation that blood group antigenic profiles of ex-vivo generated EBs are consistent with those predicted by DNA-genotyping suggests that these cells are unlikely to be immunogenic for known epitopes. However, the antigen profiles of ankyrin R and 4.1R complexes were normal only for AB and iPS-derived EBs raising the possibility that antigenic deviations seen in EBs derived from CB and hESC may have immunologic or functional consequences in vivo. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 638-638 ◽  
Author(s):  
Daniel E. Bauer ◽  
Matthew C. Canver ◽  
Elenoe C. Smith ◽  
Falak Sher ◽  
Luca Pinello ◽  
...  

Abstract Common genetic variation associated with fetal hemoglobin (HbF) level and β-hemoglobin disorder clinical severity marks an erythroid enhancer within the BCL11A gene. The 12 kb intronic enhancer contains three ~1 kb erythroid DNase I hypersensitive sites (DHSs), termed +55, +58, and +62. Here we utilized a human adult-stage erythroid cell line to show by CRISPR-Cas9 mediated targeted deletion that the composite enhancer is required both for BCL11A expression and HbF repression. Because deletion of the entire enhancer is currently too inefficient to consider for a gene editing approach to hemoglobin disorders, we sought to define the critical features of the enhancer in its natural genomic context. We designed and synthesized a tiling pooled guide RNA (gRNA) library to conduct saturating mutagenesis of the enhancer sequences in situ using the CRISPR-Cas9 gene editing platform. The gRNAs direct Cas9 cleavage and non-homologous end-joining repair at discrete sites throughout the enhancer. By comparing the representation of lentiviral gRNA integrants in high and low HbF pools of the adult erythroid cells, we generated a functional map approaching nucleotide resolution of sequences within the enhancer influencing BCL11A regulation. We observed several discrete enhancer regions required for maximal expression. The largest effect was observed by producing mutations within a narrow functional core of the +58 DHS. These sequences include a GATA1 motif conserved among vertebrates located within a primate-specific context. This region constitutes an Achilles Heel for functional inactivation of the enhancer. We also identified rare genetic variants within the +58 DHS core in individuals with sickle cell disease that are associated with HbF level, independent of all known associations of common genetic variants. In parallel, we performed a similar saturating CRISPR mutagenesis screen of the corresponding murine Bcl11a enhancer. To our surprise, despite low-resolution evidence of conservation by primary sequence homology, syntenic genomic position, and shared chromatin signature, the mouse enhancer sequence determinants of BCL11A expression showed substantial functional divergence. The +58 orthologous sequences were dispensable whereas the +62 orthologous sequences were critically required in murine adult erythroid cells. These results were validated by producing targeted deletions in mouse and human adult erythroid cell lines. Furthermore we subjected cells to individual gRNAs to correlate individual nucleotide disruptions with loss of BCL11A expression. To substantiate the tissue-restricted effect of the enhancer mutations, we generated transgenic mice with deletion of the Bcl11a enhancer and found these sequences were dispensable for expression in developing neurons and B-lymphocytes (unlike conventional Bcl11a knockout) but essential for appropriate hemoglobin switching in vivo. We showed that in primary CD34+ hematopoietic stem and progenitor derived human erythroid precursors that delivery of an individual gRNA and Cas9 is sufficient to produce robust reinduction of HbF. These results validate the BCL11A erythroid enhancer as a promising therapeutic target. Our findings define the most favorable regions for generation of indel mutations in the BCL11A erythroid enhancer as a therapeutic genome editing strategy for HbF reinduction for the β-hemoglobin disorders. Disclosures Bauer: Biogen: Research Funding; Editas Medicine: Consultancy. Zhang:Editas Medicine: Membership on an entity's Board of Directors or advisory committees; Horizon Discovery: Membership on an entity's Board of Directors or advisory committees. Orkin:Editas Medicine: Membership on an entity's Board of Directors or advisory committees; Biogen: Research Funding; Pfizer: Research Funding; Sangamo Biosciences: Consultancy.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2319-2319
Author(s):  
Maria Claessen ◽  
Eszter Varga ◽  
Steven Heshusius ◽  
Esther Heideveld ◽  
Martin Hansen ◽  
...  

Abstract Transfusion of donor-derived red blood cells to aleviate anemia is the most common form of cellular therapy. In addition, red blood cells hold great promise as delivery agents of e.g. specific drugs or enzymes. However, the source depends on donor availability and carries a potential risk of alloimmunization and blood borne diseases. More than 30 bloodgroup systems encode >300 bloodgroup antigens and bloodgroup matching becomes increasingly challenging in a multiethnic society. Particularly the chronically transfused patients are at risk for alloimmunisation. In vitro cultured, customizable red blood cells (cRBC) would negate these concerns and introduce precision medicine both in transfusion medicine as well as in drug delivery applications. We aim to produce human cRBC at large-scale and cost effective, for which we need to optimize culture conditions and reduce cost-drivers. We adapted our protocols to GMP culture requirements, which reproducibly provided pure human erythroid cultures within 25 days with a 3.4x107 times expansion from peripheral blood mononuclear cells without prior CD34+ isolation. This expansion depended on the serum free medium we produce, which is supplemented with erythropoietin (Epo, 1 U/ml), stem cell factor (SCF) and glucocorticoids. Expanded erythroblasts CD71 highCD235low/- were differentiated for 10 days in medium supplemented with 5% human plasma, heparin and a higher concentration of Epo (10U/ml) yielding CD71dimCD235a+CD44+CD117-DRAQ5- cRBC. More than 90% of the cells enucleated and expressed adult hemoglobin as well as the correct blood group antigens. Passaging cRBC through a leukodepletion filter yielded 100% enucleated, stable cRBC. Deformability was measured by an Automated Rheoscope and Cell Analyser (ARCA), and oxygen equilibrium curves were measured with a Hemox analyzer. Both parameters were similar in cRBC and freshly isolated reticulocytes. RNA sequencing was performed daily during differentiation and revealed expression dynamics of important erythroid processes, e.g. increased expression of genes involved in blood group expression, globin regulation, and erythroid specific metabolic enzymes, concommittant with loss of expression of genes involved in the formation of organelles, and cell proliferation. The culture process is compatible with upscaling using 5L G-Rex bioreactors., Currently we are preparing a clinical study using biotinylated cRBC. Ultimately, however, large scale production requires an immortal source, for which we aim to use human induced pluripotent stem cells (iPSC) established from rare donors that lack most blood group antigens. Using single cell passaging of iPSC and differentiation in colonies, we generate at average 2x105 cRBC per single iPSC. However, the cRBC cultured from iPSC were less stable following enucleation, and expressed embryonic type globins. Comparison of transcriptome data from iPSC-derived erythroid cells at distinct differentiation stages with erythroid cells at similar stages that were cultured from adult- or cord blood mononuclear cells, or from fetal liver confirmed that most iPSC-derived erythroid cells largely express an embryonic RNA profile. In conclusion, our current protocols enable us to test cRBC cultured from adult peripheral blood for their stability after transfusion. Concurrently, we develop novel bioreactors to upscale the production, and we optimise the protocol to generate cRBC from immortal iPSC lines with near 'universal donor' genotypes. Disclosures No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document