Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Molecules ; 27(23)2022 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-36500236

RESUMEN

Percutaneous coronary intervention (PCI) is a common procedure for the management of coronary artery obstruction. However, it usually causes vascular wall injury leading to restenosis that limits the long-term success of the PCI endeavor. The ultimate objective of this study was to develop the targeting nanoparticles (NPs) that were destined for the injured subendothelium and attract endothelial progenitor cells (EPCs) to the damaged location for endothelium regeneration. Biodegradable poly(lactic-co-glycolic acid) (PLGA) NPs were conjugated with double targeting moieties, which are glycoprotein Ib alpha chain (GPIbα) and human single-chain antibody variable fragment (HuscFv) specific to the cluster of differentiation 34 (CD34). GPIb is a platelet receptor that interacts with the von Willebrand factor (vWF), highly deposited on the damaged subendothelial surface, while CD34 is a surface marker of EPCs. A candidate anti-CD34 HuscFv was successfully constructed using a phage display biopanning technique. The HuscFv could be purified and showed binding affinity to the CD34-positive cells. The GPIb-conjugated NPs (GPIb-NPs) could target vWF and prevent platelet adherence to vWF in vitro. Furthermore, the HuscFv-conjugated NPs (HuscFv-NPs) could capture CD34-positive cells. The bispecific NPs have high potential to locate at the damaged subendothelial surface and capture EPCs for accelerating the vessel repair.


Asunto(s)
Nanopartículas , Intervención Coronaria Percutánea , Humanos , Endotelio Vascular/metabolismo , Factor de von Willebrand/metabolismo , Plaquetas/metabolismo , Anticuerpos/metabolismo
2.
Haematologica ; 106(11): 2859-2873, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-33054117

RESUMEN

Human ZNF648 is a novel poly C-terminal C2H2 zinc finger protein identified amongst the most dysregulated proteins in erythroid cells differentiated from iPSC. Its nuclear localisation and structure indicate it is likely a DNA-binding protein. Using a combination of ZNF648 overexpression in an iPSC line and primary adult erythroid cells, ZNF648 knockdown in primary adult erythroid cells and megakaryocytes, comparative proteomics and transcriptomics we show that ZNF648 is required for both erythroid and megakaryocyte differentiation. Orthologues of ZNF648 were detected across Mammals, Reptilia, Actinopterygii, in some Aves, Amphibia and Coelacanthiformes suggesting the gene originated in the common ancestor of Osteichthyes (Euteleostomi or bony fish). Conservation of the C-terminal zinc finger domain is higher, with some variation in zinc finger number but a core of at least six zinc fingers conserved across all groups, with the N-terminus recognisably similar within but not between major lineages. This suggests the N-terminus of ZNF648 evolves faster than the C-terminus, however this is not due to exon-shuffling as the entire coding region of ZNF648 is within a single exon. As for other such transcription factors, the N-terminus likely carries out regulatory functions, but showed no sequence similarity to any known domains. The greater functional constraint on the zinc finger domain suggests ZNF648 binds at least some similar regions of DNA in the different organisms. However, divergence of the N-terminal region may enable differential expression, allowing adaptation of function in the different organisms.


Asunto(s)
Eritrocitos/citología , Megacariocitos/citología , Factores de Transcripción , Dedos de Zinc , Animales , Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Humanos
4.
Mol Cell Proteomics ; 15(6): 1938-46, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27006477

RESUMEN

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.


Asunto(s)
Células Eritroides/citología , Sangre Fetal/citología , Proteoma/análisis , Proteómica/métodos , Reticulocitos/citología , Adulto , Anhidrasa Carbónica I/metabolismo , Anhidrasa Carbónica II/metabolismo , Diferenciación Celular , Cromatografía Liquida , Células Eritroides/metabolismo , Sangre Fetal/metabolismo , Humanos , Recién Nacido , Reticulocitos/metabolismo , Espectrometría de Masas en Tándem
6.
Blood ; 119(26): 6296-306, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22490681

RESUMEN

The erythrocyte is one of the best characterized human cells. However, studies of the process whereby human reticulocytes mature to erythrocytes have been hampered by the difficulty of obtaining sufficient numbers of cells for analysis. In the present study, we describe an in vitro culture system producing milliliter quantities of functional mature human adult reticulocytes from peripheral blood CD34(+) cells. We show that the final stage of reticulocyte maturation occurs by a previously undescribed mechanism in which large glycophorin A-containing vesicles forming at the cytosolic face of the plasma membrane are internalized and fuse with autophagosomes before expulsion of the autophagosomal contents by exocytosis. Early reticulocyte maturation is characterized by the selective elimination of unwanted plasma membrane proteins (CD71, CD98, and ß1 integrin) through the endosome-exosome pathway. In contrast, late maturation is characterized by the generation of large glycophorin A-decorated vesicles of autophagic origin.


Asunto(s)
Exocitosis/fisiología , Glicoforinas/metabolismo , Fusión de Membrana/fisiología , Fagosomas/fisiología , Reticulocitos/fisiología , Vesículas Transportadoras/fisiología , Adulto , Diferenciación Celular , Membrana Celular/metabolismo , Eritrocitos/fisiología , Eritrocitos/ultraestructura , Humanos , Microscopía Confocal , Oxígeno/metabolismo , Fagosomas/metabolismo , Reticulocitos/metabolismo , Reticulocitos/ultraestructura , Vesículas Transportadoras/metabolismo
7.
Haematologica ; 99(11): 1677-85, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25107887

RESUMEN

A major barrier to the clinical use of erythrocytes generated in vitro from pluripotent stem cells or cord blood progenitors is failure of these erythrocytes to express adult hemoglobin. The key regulators of globin switching KLF1 and BCL11A are absent or at a lower level than in adult cells in K562 and erythroid cells differentiated in vitro from induced pluripotent stem cells and cord blood progenitors. Transfection or transduction of K562 and cord blood erythroid cells with either KLF1 or BCL11A-XL had little effect on ß-globin expression. In contrast, transduction with both transcription factors stimulated ß-globin expression. Similarly, increasing the level of BCL11A-XL in the induced pluripotent stem cell-derived erythroid cell line HiDEP-1, which has levels of endogenous KLF1 similar to adult cells but lacks BCL11A, resulted in levels of ß-globin equivalent to that of adult erythroid cells. Interestingly, this increase in ß-globin was coincident with a decrease in ε- and ζ-, but not γ-globin, implicating BCL11A in repression of embryonic globin expression. The data show that KLF1 and BCL11A-XL together are required, but sufficient to induce adult levels of ß-globin in induced pluripotent stem cell and cord blood-derived erythroid cells that intrinsically express embryonic or fetal globin.


Asunto(s)
Proteínas Portadoras/genética , Células Eritroides/metabolismo , Hemoglobina Fetal/genética , Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Proteínas Nucleares/genética , Transducción Genética , Globinas beta/genética , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Diferenciación Celular/genética , Línea Celular , Células Eritroides/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Células K562 , Fenotipo , Proteínas Represoras , Transfección , Globinas épsilon/genética , gamma-Globinas/genética
8.
MethodsX ; 12: 102714, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38660027

RESUMEN

At present, the numbers of cultured erythroid cells obtained from culture systems are not on a scale that can be used for therapeutics since the cultured erythroid cells have limited proliferation capacity. Stromal cells are believed to play important roles during erythropoiesis. Our previous study shows that factors secreted by stromal cells enhance the proliferation capacity of adult erythroid cells in the culture system. Among the identified factors, angiotensinogen is one of the most abundant proteins secreted by the stromal cells. This study aims to investigate the effect of angiotensin II, an angiotensinogen derivative, on the proliferation of erythroid cells. •The receptor for angiotensin II was first checked by PCR analysis. It was expressed in erythroblasts at all stages during differentiation.•To study the effect of angiotensin II, CD34+ hematopoietic stem cells were cultured in a 3-stage erythroid culture system with and without angiotensin II. The addition of angiotensin II to the culture media, from day 0 to 8, significantly increases the numbers of cultured erythroid cells, whereas no difference in enucleation is observed.

9.
Blood ; 118(11): 3137-45, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21778342

RESUMEN

Mutations in the human erythroid Krüppel-like factor (EKLF) can lead to either anemia or the benign InLu phenotype. To elucidate the relationship between these mutations and the differing phenotypes, we prepared recombinant forms of wild-type and 5 mutant EKLF proteins and quantitated their binding affinity to a range of EKLF-regulated genes. Missense mutants (R328H, R328L, and R331G) from persons with InLu phenotype did not bind DNA. Hence, as with the heterozygous loss of function nonsense (L127X, S270X, and K292X) and frameshift (P190Lfs and R319Efs) EKLF mutations, monoallelic loss of EKLF does not result in haploinsufficiency at all loci. In contrast, K332Q has a slightly reduced DNA binding affinity (∼ 2-fold) for all promoters examined but exhibits a phenotype only in a compound heterozygote with a nonfunctional allele. E325K also has a reduced, but significant, binding affinity, particularly for the ß-globin gene but results in a disease phenotype even with the wild-type allele expressed, although not as a classic dominant-negative mutant. E325K protein may therefore actively interfere with EKLF-dependent processes by destabilizing transcription complexes, providing a rational explanation for the severity of the disease phenotype. Our study highlights the critical role of residues within the second EKLF zinc finger domain.


Asunto(s)
Enfermedad/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/fisiología , Regiones Promotoras Genéticas , Secuencia de Aminoácidos , Sitios de Unión/genética , Células Cultivadas , Humanos , Factores de Transcripción de Tipo Kruppel/química , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación/fisiología , Fenotipo , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/fisiología , Homología de Secuencia de Aminoácido , Índice de Severidad de la Enfermedad , Especificidad por Sustrato/genética , Activación Transcripcional , Dedos de Zinc/genética
10.
Sci Rep ; 12(1): 15551, 2022 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-36114353

RESUMEN

ß-thalassaemia is one of the most common genetic blood diseases worldwide with over 300 mutations in the HBB gene affecting red blood cell functions. Recently, advances in genome editing technology have provided a powerful tool for precise genetic correction. Generation of patient-derived induced pluripotent stem cells (iPSCs) followed by genetic correction of HBB mutations and differentiation into haematopoietic stem/progenitor cells (HSPCs) offers a potential therapy to cure the disease. However, the biggest challenge is to generate functional HSPCs that are capable of self-renewal and transplantable. In addition, functional analyses of iPSC-derived erythroid cells are hampered by poor erythroid expansion and incomplete erythroid differentiation. Previously, we generated an immortalised erythroid cell line (SiBBE) with unique properties, including unlimited expansion and the ability to differentiate into mature erythrocytes. In this study, we report a highly efficient genetic correction of HbE mutation in the SiBBE cells using the CRISPR/Cas9 system. The HbE-corrected clones restored ß-globin production with reduced levels of HbE upon erythroid differentiation. Our approach provides a sustainable supply of corrected erythroid cells and represents a valuable model for validating the therapeutic efficacy of gene editing systems.


Asunto(s)
Hemoglobina E , Talasemia beta , Sistemas CRISPR-Cas/genética , Línea Celular , Hemoglobina E/genética , Hemoglobina E/metabolismo , Humanos , Células Madre Pluripotentes Inducidas , Globinas beta/genética , Globinas beta/metabolismo , Talasemia beta/genética , Talasemia beta/metabolismo , Talasemia beta/terapia
11.
Stem Cell Res Ther ; 13(1): 467, 2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-36076260

RESUMEN

BACKGROUND: Yes-associated protein (YAP) and WW domain-containing transcription regulator protein 1 (WWTR1, also known as TAZ) are two key transcription co-activators of the Hippo pathway. Both were originally characterized as organ size and cell proliferation regulators. Later studies demonstrated that the Hippo pathway may play a role in Drosophila and mammal hematopoiesis. However, the role of the Hippo pathway in human erythropoiesis has not yet been fully elucidated. METHODS: The role of YAP and TAZ was studied in human erythropoiesis and hematopoietic stem cell (HSC) lineage determination by using mobilized peripheral blood (PB) and cord blood (CB)-derived HSC as a model. HSCs were isolated and cultured in an erythroid differentiation medium for erythroid differentiation and culture in methylcellulose assay for HSC lineage determination study. RESULTS: YAP and TAZ were barely detectable in human HSCs, but became highly expressed in pro-erythroblasts and erythroblasts. Depletion or knockdown of YAP and/or TAZ did not affect the ability of HSC lineage specification to erythroid lineage in either methylcellulose assay or liquid culture. However, depletion of YAP and TAZ did impair erythroblast terminal differentiation to erythrocytes and their enucleation. Moreover, ectopic expression of YAP and TAZ in pro-erythroblasts did not exert an apparent effect on erythroid differentiation, expansion, or morphology. CONCLUSIONS: This study demonstrated that YAP/TAZ plays important role in erythroid maturation and enucleation but is dispensable for lineage determination of human HSCs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Transactivadores , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ/metabolismo , Proteínas Señalizadoras YAP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Eritrocitos , Eritropoyesis/genética , Humanos , Mamíferos/metabolismo , Metilcelulosa , Fosfoproteínas/metabolismo , Transducción de Señal , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
12.
Sci Rep ; 11(1): 22483, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34795367

RESUMEN

Polycythaemia vera (PV) is a haematological disorder caused by an overproduction of erythroid cells. To date, the molecular mechanisms involved in the disease pathogenesis are still ambiguous. This study aims to identify aberrantly expressed proteins in erythroblasts of PV patients by utilizing mass spectrometry-based proteomic analysis. Haematopoietic stem cells (HSCs) were isolated from newly-diagnosed PV patients, PV patients who have received cytoreductive therapy, and healthy subjects. In vitro erythroblast expansion confirmed that the isolated HSCs recapitulated the disease phenotype as the number of erythroblasts from newly-diagnosed PV patients was significantly higher than those from the other groups. Proteomic comparison revealed 17 proteins that were differentially expressed in the erythroblasts from the newly-diagnosed PV patients compared to those from healthy subjects, but which were restored to normal levels in the patients who had received cytoreductive therapy. One of these proteins was S-methyl-5'-thioadenosine phosphorylase (MTAP), which had reduced expression in PV patients' erythroblasts. Furthermore, MTAP knockdown in normal erythroblasts was shown to enhance their proliferative capacity. Together, this study identifies differentially expressed proteins in erythroblasts of healthy subjects and those of PV patients, indicating that an alteration of protein expression in erythroblasts may be crucial to the pathology of PV.


Asunto(s)
Policitemia Vera/tratamiento farmacológico , Policitemia Vera/metabolismo , Purina-Nucleósido Fosforilasa , Adulto , Anciano , Proliferación Celular , Eritroblastos/metabolismo , Eritrocitos/citología , Células Precursoras Eritroides/metabolismo , Femenino , Células Madre Hematopoyéticas/citología , Humanos , Técnicas In Vitro , Masculino , Espectrometría de Masas , Persona de Mediana Edad , Proteoma , Proteómica/métodos , Factor de Células Madre/metabolismo
13.
Mol Ther Methods Clin Dev ; 22: 26-39, 2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34485592

RESUMEN

Developing robust methodology for the sustainable production of red blood cells in vitro is essential for providing an alternative source of clinical-quality blood, particularly for individuals with rare blood group phenotypes. Immortalized erythroid progenitor cell lines are the most promising emergent technology for achieving this goal. We previously created the erythroid cell line BEL-A from bone marrow CD34+ cells that had improved differentiation and enucleation potential compared to other lines reported. In this study we show that our immortalization approach is reproducible for erythroid cells differentiated from bone marrow and also from far more accessible peripheral and cord blood CD34+ cells, consistently generating lines with similar improved erythroid performance. Extensive characterization of the lines shows them to accurately recapitulate their primary cell equivalents and provides a molecular signature for immortalization. In addition, we show that only cells at a specific stage of erythropoiesis, predominantly proerythroblasts, are amenable to immortalization. Our methodology provides a step forward in the drive for a sustainable supply of red cells for clinical use and for the generation of model cellular systems for the study of erythropoiesis in health and disease, with the added benefit of an indefinite expansion window for manipulation of molecular targets.

14.
Stem Cell Res Ther ; 11(1): 481, 2020 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-33176890

RESUMEN

BACKGROUND: Human induced pluripotent stem cells (hiPSCs) offer a renewable source of cells for the generation of hematopoietic cells for cell-based therapy, disease modeling, and drug screening. However, current serum/feeder-free differentiation protocols rely on the use of various cytokines, which makes the process very costly or the generation of embryoid bodies (EBs), which are labor-intensive and can cause heterogeneity during differentiation. Here, we report a simple feeder and serum-free monolayer protocol for efficient generation of iPSC-derived multipotent hematoendothelial progenitors (HEPs), which can further differentiate into endothelial and hematopoietic cells including erythroid and T lineages. METHODS: Formation of HEPs from iPSCs was initiated by inhibition of GSK3 signaling for 2 days followed by the addition of VEGF and FGF2 for 3 days. The HEPs were further induced toward mature endothelial cells (ECs) in an angiogenic condition and toward T cells by co-culturing with OP9-DL1 feeder cells. Endothelial-to-hematopoietic transition (EHT) of the HEPs was further promoted by supplementation with the TGF-ß signaling inhibitor. Erythroid differentiation was performed by culturing the hematopoietic stem/progenitor cells (HSPCs) in a three-stage erythroid liquid culture system. RESULTS: Our protocol significantly enhanced the number of KDR+ CD34+ CD31+ HEPs on day 5 of differentiation. Further culture of HEPs in angiogenic conditions promoted the formation of mature ECs, which expressed CD34, CD31, CD144, vWF, and ICAM-1, and could exhibit the formation of vascular-like network and acetylated low-density lipoprotein (Ac-LDL) uptake. In addition, the HEPs were differentiated into CD8+ T lymphocytes, which could be expanded up to 34-fold upon TCR stimulation. Inhibition of TGF-ß signaling at the HEP stage promoted EHT and yielded a large number of HSPCs expressing CD34 and CD43. Upon erythroid differentiation, these HSPCs were expanded up to 40-fold and displayed morphological changes following stages of erythroid development. CONCLUSION: This protocol offers an efficient and simple approach for the generation of multipotent HEPs and could be adapted to generate desired blood cells in large numbers for applications in basic research including developmental study, disease modeling, and drug screening as well as in regenerative medicine.


Asunto(s)
Células Madre Pluripotentes Inducidas , Diferenciación Celular , Células Endoteliales , Glucógeno Sintasa Quinasa 3 , Células Madre Hematopoyéticas , Humanos
15.
Sci Rep ; 10(1): 16798, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-33033327

RESUMEN

The ß-thalassemia syndromes are the most prevalent genetic disorder globally, characterised by reduced or absent ß-globin chain synthesis. HbE/ß-thalassemia is a subtype of ß-thalassemia with extremely high frequency in Asia. Studying molecular defects behind ß-thalassemia is severely impeded by paucity of material from patients and lack of suitable cell lines. Approaches to derive erythroid cells from induced pluripotent stem cells (iPSCs) created from patients are confounded by poor levels of erythroid cell expansion, aberrant or incomplete erythroid differentiation and foetal/embryonic rather than adult globin expression. In this study we generate an immortalised erythroid cell line from peripheral blood stem cells of a HbE/ß-thalassemia patient. Morphological analysis shows the cells are proerythroblasts with some early basophilic erythroblasts, with no change in morphology over time in culture. The line differentiates along the erythroid pathway to orthochromatic erythroblasts and reticulocytes. Importantly, unlike iPSCs, the line maintains the haemoglobin profile of the patient's red blood cells. This is the first human cellular model for ß-thalassemia providing a sustainable source of disease cells for studying underlying disease mechanisms and for use as drug screening platform, particularly for reagents designed to increase foetal haemoglobin expression as we have additionally demonstrated with hydroxyurea.


Asunto(s)
Diferenciación Celular/fisiología , Eritroblastos/citología , Células Eritroides/citología , Células Madre Hematopoyéticas/citología , Talasemia beta/sangre , Línea Celular , Humanos
16.
Stem Cell Res Ther ; 10(1): 130, 2019 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-31036072

RESUMEN

BACKGROUND: Pluripotent stem cells are attractive progenitor cells for the generation of erythroid cells in vitro as have expansive proliferative potential. However, although embryonic (ESC) and induced pluripotent (iPSC) stem cells can be induced to undergo erythroid differentiation, the majority of cells fail to enucleate and the molecular basis of this defect is unknown. One protein that has been associated with the initial phase of erythroid cell enucleation is the intermediate filament vimentin, with loss of vimentin potentially required for the process to proceed. METHODS: In this study, we used our established erythroid culture system along with western blot, PCR and interegation of comparative proteomic data sets to analyse the temporal expression profile of vimentin in erythroid cells differentiated from adult peripheral blood stem cells, iPSC and ESC throughout erythropoiesis. Confocal microscopy was also used to examine the intracellular localisation of vimentin. RESULTS: We show that expression of vimentin is turned off early during normal adult erythroid cell differentiation, with vimentin protein lost by the polychromatic erythroblast stage, just prior to enucleation. In contrast, in erythroid cells differentiated from iPSC and ESC, expression of vimentin persists, with high levels of both mRNA and protein even in orthochromatic erythroblasts. In the vimentin-positive iPSC orthochromatic erythroblasts, F-actin was localized around the cell periphery; however, in those rare cells captured undergoing enucleation, vimentin was absent and F-actin was re-localized to the enucleosome as found in normal adult orthrochromatic erythroblasts. CONCLUSION: As both embryonic and adult erythroid cells loose vimentin and enucleate, retention of vimentin by iPSC and ESC erythroid cells indicates an intrinsic defect. By analogy with avian erythrocytes which naturally retain vimentin and remain nucleated, retention in iPSC- and ESC-derived erythroid cells may impede enucleation. Our data also provide the first evidence that dysregulation of processes in these cells occurs from the early stages of differentiation, facilitating targeting of future studies.


Asunto(s)
Eritropoyesis/fisiología , Células Madre Pluripotentes Inducidas/metabolismo , Proteómica/métodos , Vimentina/metabolismo , Diferenciación Celular , Células Cultivadas , Células Eritroides , Humanos , Células Madre Pluripotentes Inducidas/citología
17.
MethodsX ; 5: 1626-1632, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30560064

RESUMEN

In vitro generation of red blood cells has become a goal for scientists globally. Directly, in vitro-generated red blood cells (RBCs) may close the gap between blood supply obtained through blood donation and high demand for therapeutic uses. In addition, the cells obtained can be used as a model for haematologic disorders to allow the study of their pathophysiology and novel treatment discovery. For those reasons, a number of RBC culture systems have been established and shown to be successful; however, the cost of each millilitre of packed RBC is still extremely high. In order to reduce the cost, we aim to see if we can reduce the number of factors used in the existing culture system. In this study, we examined how well haematopoietic stem cells proliferate and differentiate into mature red blood cells with modified culture system. •Absence of extra heparin or insulin or both from the erythroid differentiation media did not affect haematopoietic stem cell proliferation and differentiation. Therefore, we show that the cost and complexity of erythroid culture can be reduced, which may improve the feasibility of in vitro generation of red blood cells.

18.
Sci Rep ; 8(1): 1983, 2018 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-29386568

RESUMEN

Development of in vitro culture systems for the generation of red blood cells is a goal of scientists globally with the aim of producing clinical grade products for transfusion. Although mature reticulocytes can be efficiently generated by such systems, the numbers produced fall short of that required for therapeutics, due to limited proliferative capacity of the erythroblasts. To overcome this hurdle, approaches are required to increase the expansion potential of such culture systems. The OP9 mouse stromal cell line is known to promote haematopoietic differentiation of pluripotent stem cells, however an effect of OP9 cells on erythropoiesis has not been explored. In this study, we show not only OP9 co-culture, but factors secreted by OP9 cells in isolation increase the proliferative potential of adult erythroid cells by delaying differentiation and hence maintaining self-renewing cells for an extended duration. The number of reticulocytes obtained was increased by approximately 3.5-fold, bringing it closer to that required for a therapeutic product. To identify the factors responsible, we analysed the OP9 cell secretome using comparative proteomics, identifying 18 candidate proteins. These data reveal the potential to increase erythroid cell numbers from in vitro culture systems without the need for genetic manipulation or co-culture.


Asunto(s)
Diferenciación Celular , Células Eritroides/citología , Células Eritroides/metabolismo , Adulto , Animales , Comunicación Celular , Línea Celular , Proliferación Celular , Forma de la Célula , Técnicas de Cocultivo , Medios de Cultivo Condicionados/farmacología , Eritroblastos/citología , Humanos , Espectrometría de Masas , Ratones , Coloración y Etiquetado , Células del Estroma/citología , Células del Estroma/metabolismo
19.
Stem Cell Res Ther ; 9(1): 46, 2018 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-29482624

RESUMEN

BACKGROUND: Thalassemia is the most common genetic disease worldwide; those with severe disease require lifelong blood transfusion and iron chelation therapy. The definitive cure for thalassemia is allogeneic hematopoietic stem cell transplantation, which is limited due to lack of HLA-matched donors and the risk of post-transplant complications. Induced pluripotent stem cell (iPSC) technology offers prospects for autologous cell-based therapy which could avoid the immunological problems. We now report genetic correction of the beta hemoglobin (HBB) gene in iPSCs derived from a patient with a double heterozygote for hemoglobin E and ß-thalassemia (HbE/ß-thalassemia), the most common thalassemia syndrome in Thailand and Southeast Asia. METHODS: We used the CRISPR/Cas9 system to target the hemoglobin E mutation from one allele of the HBB gene by homology-directed repair with a single-stranded DNA oligonucleotide template. DNA sequences of the corrected iPSCs were validated by Sanger sequencing. The corrected clones were differentiated into hematopoietic progenitor and erythroid cells to confirm their multilineage differentiation potential and hemoglobin expression. RESULTS: The hemoglobin E mutation of HbE/ß-thalassemia iPSCs was seamlessly corrected by the CRISPR/Cas9 system. The corrected clones were differentiated into hematopoietic progenitor cells under feeder-free and OP9 coculture systems. These progenitor cells were further expanded in erythroid liquid culture system and developed into erythroid cells that expressed mature HBB gene and HBB protein. CONCLUSIONS: Our study provides a strategy to correct hemoglobin E mutation in one step and these corrected iPSCs can be differentiated into hematopoietic stem cells to be used for autologous transplantation in patients with HbE/ß-thalassemia in the future.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Hemoglobina E , Células Madre Pluripotentes Inducidas/metabolismo , Talasemia beta , Autoinjertos , Femenino , Hemoglobina E/genética , Hemoglobina E/metabolismo , Humanos , Masculino , Mutación , Trasplante de Células Madre , Talasemia beta/genética , Talasemia beta/metabolismo , Talasemia beta/terapia
20.
EMBO Mol Med ; 10(6)2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29700043

RESUMEN

Regular blood transfusion is the cornerstone of care for patients with red blood cell (RBC) disorders such as thalassaemia or sickle-cell disease. With repeated transfusion, alloimmunisation often occurs due to incompatibility at the level of minor blood group antigens. We use CRISPR-mediated genome editing of an immortalised human erythroblast cell line (BEL-A) to generate multiple enucleation competent cell lines deficient in individual blood groups. Edits are combined to generate a single cell line deficient in multiple antigens responsible for the most common transfusion incompatibilities: ABO (Bombay phenotype), Rh (Rhnull), Kell (K0), Duffy (Fynull), GPB (S-s-U-). These cells can be differentiated to generate deformable reticulocytes, illustrating the capacity for coexistence of multiple rare blood group antigen null phenotypes. This study provides the first proof-of-principle demonstration of combinatorial CRISPR-mediated blood group gene editing to generate customisable or multi-compatible RBCs for diagnostic reagents or recipients with complicated matching requirements.


Asunto(s)
Incompatibilidad de Grupos Sanguíneos/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Transfusión de Eritrocitos , Edición Génica/métodos , Antígenos de Grupos Sanguíneos/genética , Línea Celular , Técnicas de Inactivación de Genes , Humanos , Prueba de Estudio Conceptual
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA