Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Transl Med ; 22(1): 526, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38822352

RESUMO

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


Assuntos
Antígenos CD34 , Neutrófilos , Humanos , Neutrófilos/metabolismo , Neutrófilos/citologia , Antígenos CD34/metabolismo , Células Cultivadas , Espécies Reativas de Oxigênio/metabolismo , Proteômica , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/citologia , Edição de Genes , Degranulação Celular , Células-Tronco/metabolismo , Células-Tronco/citologia , Citocinas/metabolismo , Fenótipo
2.
Cancer Res ; 84(2): 211-225, 2024 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-37921711

RESUMO

Myelodysplastic syndromes with ring sideroblasts (MDS-RS) commonly develop from hematopoietic stem cells (HSC) bearing mutations in the splicing factor SF3B1 (SF3B1mt). Direct studies into MDS-RS pathobiology have been limited by a lack of model systems that fully recapitulate erythroid biology and RS development and the inability to isolate viable human RS. Here, we combined successful direct RS isolation from patient samples, high-throughput multiomics analysis of cells encompassing the SF3B1mt stem-erythroid continuum, and functional assays to investigate the impact of SF3B1mt on erythropoiesis and RS accumulation. The isolated RS differentiated, egressed into the blood, escaped traditional nonsense-mediated decay (NMD) mechanisms, and leveraged stress-survival pathways that hinder wild-type hematopoiesis through pathogenic GDF15 overexpression. Importantly, RS constituted a contaminant of magnetically enriched CD34+ cells, skewing bulk transcriptomic data. Mis-splicing in SF3B1mt cells was intensified by erythroid differentiation through accelerated RNA splicing and decreased NMD activity, and SF3B1mt led to truncations in several MDS-implicated genes. Finally, RNA mis-splicing induced an uncoupling of RNA and protein expression, leading to critical abnormalities in proapoptotic p53 pathway genes. Overall, this characterization of erythropoiesis in SF3B1mt RS provides a resource for studying MDS-RS and uncovers insights into the unexpectedly active biology of the "dead-end" RS. SIGNIFICANCE: Ring sideroblast isolation combined with state-of-the-art multiomics identifies survival mechanisms underlying SF3B1-mutant erythropoiesis and establishes an active role for erythroid differentiation and ring sideroblasts themselves in SF3B1-mutant myelodysplastic syndrome pathogenesis.


Assuntos
Síndromes Mielodisplásicas , Fosfoproteínas , Humanos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/patologia , Fatores de Processamento de RNA/genética , Fatores de Processamento de RNA/metabolismo , Splicing de RNA/genética , Mutação , Fatores de Transcrição/metabolismo , RNA/metabolismo
3.
Immunol Cell Biol ; 101(10): 947-963, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37694300

RESUMO

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


Assuntos
Proteoma , Proteômica , Proteoma/metabolismo , Células Cultivadas , Macrófagos/metabolismo , Monócitos/fisiologia
4.
Semin Hematol ; 59(3): 150-155, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-36115692

RESUMO

Large scale high-throughput DNA sequencing studies have identified clonal hematopoiesis (CH) as a clinical phenomenon characterized by a disproportionately large clonal population in the hematopoietic system with a shared mutational background. CH originates through mutations in hematopoietic stem and progenitor cells (HSPCs) which provide a proliferative advantage over unmutated HSPCs and has been characterized as a risk factor for myeloid neoplasm (MN) development. Large population studies found that CH is an age-related event which is commonly found in association with milder phenotypes such as cytopenia, mild monocytosis, intravascular hemolysis, or chronic inflammation. More importantly, the vast majority of individuals with CH are asymptomatic and healthy people of advanced age, where the impact of CH is thus considered to be of indeterminate potential (CHIP). These conditions are sometimes referred to as benign to facilitate distinction from overt MN but, despite this definition, may still result in severe illness, reduced overall survival, and increased risk of hematologic neoplasms development and all-cause mortality. The purpose of this review is to describe clinical conditions associated with CH, the clinical significance of CH-related clinical phenotypes, and the determinants of progression from CH to overt MN following the paradigmatic example of SF3B1-driven CH.


Assuntos
Neoplasias Hematológicas , Transtornos Mieloproliferativos , Hematopoiese Clonal/genética , Neoplasias Hematológicas/genética , Hematopoese/genética , Células-Tronco Hematopoéticas , Humanos , Transtornos Mieloproliferativos/genética , Fosfoproteínas/genética , Fatores de Processamento de RNA/genética
5.
EJHaem ; 2(2): 175-187, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34124710

RESUMO

The SARS-CoV-2 virus causes COVID-19, an infection capable of causing severe disease and death but which can also be asymptomatic or oligosymptomatic. We investigated whether ABO blood group or secretor status was associated with COVID-19 severity. We investigated secretor status because expression of ABO glycans on secreted proteins and non-erythroid cells are controlled by a fucosyltransferase (FUT2), and inactivating FUT2 mutations result in a non-secretor phenotype which protects against some viral infections. Data combined from healthcare records and our own laboratory tests (n = 275) of hospitalized SARS-CoV-2 polymerase chain reaction positive patients confirmed higher than expected numbers of blood group A individuals compared to O (RR = 1.24, CI 95% [1.05, 1.47], p = 0.0111). There was also a significant association between group A and COVID-19-related cardiovascular complications (RR = 2.56, CI 95% [1.43, 4.55], p = 0.0011) which is independent of gender. Molecular analysis revealed that group A non-secretors are significantly less likely to be hospitalized than secretors. Testing of convalescent plasma donors, among whom the majority displayed COVID-19 symptoms and only a small minority required hospitalization, group A non-secretors were slightly over-represented. Our findings showed that group A non-secretors are not resistant to infection by SARS-CoV-2, but are more likely to experience a less severe form of associated disease.

8.
Blood Adv ; 3(17): 2653-2663, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31506283

RESUMO

The capacity to undergo substantial deformation is a defining characteristic of the red blood cell (RBC), facilitating transit through the splenic interendothelial slits and microvasculature. Establishment of this remarkable property occurs during a process of reticulocyte maturation that begins with egress through micron-wide pores in the bone marrow and is completed within the circulation. The requirement to undertake repeated cycles of deformation necessitates that both reticulocytes and erythrocytes regulate membrane-cytoskeletal protein interactions in order to maintain cellular stability. In the absence of transcriptional activity, modulation of these interactions in RBCs is likely to be achieved primarily through specific protein posttranslational modifications, which at present remain undefined. In this study, we use high-throughput methods to define the processes that underlie the response to deformation and shear stress in both reticulocytes and erythrocytes. Through combination of a bead-based microsphiltration assay with phosphoproteomics we describe posttranslational modification of RBC proteins associated with deformation. Using microsphiltration and microfluidic biochip-based assays, we explore the effect of inhibiting kinases identified using this dataset. We demonstrate roles for GSK3 and Lyn in capillary transit and maintenance of membrane stability following deformation and show that combined inhibition of these kinases significantly decreases reticulocyte capacity to undergo repeated deformation. Finally, we derive a comprehensive and integrative phosphoproteomic dataset that provides a valuable resource for further mechanistic dissection of the molecular pathways that underlie the RBC's response to mechanical stimuli and for the study of reticulocyte maturation.


Assuntos
Deformação Eritrocítica/fisiologia , Eritrócitos/fisiologia , Proteínas de Membrana/metabolismo , Fosforilação/fisiologia , Forma Celular , Células Cultivadas , Membrana Eritrocítica/química , Membrana Eritrocítica/metabolismo , Eritrócitos/citologia , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Processamento de Proteína Pós-Traducional/fisiologia , Proteômica , Reticulócitos/citologia , Reticulócitos/fisiologia , Quinases da Família src/metabolismo
9.
Nat Commun ; 10(1): 3806, 2019 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-31444345

RESUMO

Investigating the role that host erythrocyte proteins play in malaria infection is hampered by the genetic intractability of this anucleate cell. Here we report that reticulocytes derived through in vitro differentiation of an enucleation-competent immortalized erythroblast cell line (BEL-A) support both successful invasion and intracellular development of the malaria parasite Plasmodium falciparum. Using CRISPR-mediated gene knockout and subsequent complementation, we validate an essential role for the erythrocyte receptor basigin in P. falciparum invasion and demonstrate rescue of invasive susceptibility by receptor re-expression. Successful invasion of reticulocytes complemented with a truncated mutant excludes a functional role for the basigin cytoplasmic domain during invasion. Contrastingly, knockout of cyclophilin B, reported to participate in invasion and interact with basigin, did not impact invasive susceptibility of reticulocytes. These data establish the use of reticulocytes derived from immortalized erythroblasts as a powerful model system to explore hypotheses regarding host receptor requirements for P. falciparum invasion.


Assuntos
Engenharia Genética/métodos , Interações Hospedeiro-Parasita , Malária Falciparum/parasitologia , Plasmodium falciparum/patogenicidade , Reticulócitos/parasitologia , Animais , Basigina/genética , Basigina/metabolismo , Sistemas CRISPR-Cas , Diferenciação Celular , Linhagem Celular , Ciclofilinas/genética , Ciclofilinas/metabolismo , Eritroblastos/fisiologia , Técnicas de Inativação de Genes , Vetores Genéticos/genética , Células HEK293 , Humanos , Lentivirus/genética , Plasmodium falciparum/metabolismo , Domínios Proteicos/genética , Proteínas de Protozoários/metabolismo , Reticulócitos/fisiologia , Transdução Genética
10.
Haematologica ; 103(12): 1997-2007, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30076174

RESUMO

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


Assuntos
Vesículas Citoplasmáticas/metabolismo , Eritrócitos/metabolismo , Miosina Tipo II/metabolismo , Reticulócitos/metabolismo , Diferenciação Celular , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Eritrócitos/citologia , Eritropoese , Humanos , Proteínas Motores Moleculares/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Fosforilação , Proteômica/métodos , Reticulócitos/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA