Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 139(21): 3181-3193, 2022 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-35040907

RESUMO

Anemia of inflammation, also known as anemia of chronic disease, is refractory to erythropoietin (EPO) treatment, but the mechanisms underlying the EPO refractory state are unclear. Here, we demonstrate that high mobility group box-1 protein (HMGB1), a damage-associated molecular pattern molecule recently implicated in anemia development during sepsis, leads to reduced expansion and increased death of EPO-sensitive erythroid precursors in human models of erythropoiesis. HMGB1 significantly attenuates EPO-mediated phosphorylation of the Janus kinase 2/STAT5 and mTOR signaling pathways. Genetic ablation of receptor for advanced glycation end products, the only known HMGB1 receptor expressed by erythroid precursors, does not rescue the deleterious effects of HMGB1 on EPO signaling, either in human or murine precursors. Furthermore, surface plasmon resonance studies highlight the ability of HMGB1 to interfere with the binding between EPO and the EPOR. Administration of a monoclonal anti-HMGB1 antibody after sepsis onset in mice partially restores EPO signaling in vivo. Thus, HMGB1-mediated restriction of EPO signaling contributes to the chronic phase of anemia of inflammation.


Assuntos
Anemia , Eritropoetina , Proteína HMGB1 , Sepse , Anemia/genética , Animais , Eritropoese/genética , Eritropoetina/metabolismo , Inflamação , Camundongos , Receptores da Eritropoetina/metabolismo , Sepse/complicações
2.
Biophys J ; 120(17): 3588-3599, 2021 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-34352252

RESUMO

Spectrin tetramers of the membranes of enucleated mammalian erythrocytes play a critical role in red blood cell survival in circulation. One of the spectrins, αI, emerged in mammals with enucleated red cells after duplication of the ancestral α-spectrin gene common to all animals. The neofunctionalized αI-spectrin has moderate affinity for ßI-spectrin, whereas αII-spectrin, expressed in nonerythroid cells, retains ancestral characteristics and has a 10-fold higher affinity for ßI-spectrin. It has been hypothesized that this adaptation allows for rapid make and break of tetramers to accommodate membrane deformation. We have tested this hypothesis by generating mice with high-affinity spectrin tetramers formed by exchanging the site of tetramer formation in αI-spectrin (segments R0 and R1) for that of αII-spectrin. Erythrocytes with αIIßI presented normal hematologic parameters yet showed increased thermostability, and their membranes were significantly less deformable; under low shear forces, they displayed tumbling behavior rather than tank treading. The membrane skeleton is more stable with αIIßI and shows significantly less remodeling under deformation than red cell membranes of wild-type mice. These data demonstrate that spectrin tetramers undergo remodeling in intact erythrocytes and that this is required for the normal deformability of the erythrocyte membrane. We conclude that αI-spectrin represents evolutionary optimization of tetramer formation: neither higher-affinity tetramers (as shown here) nor lower affinity (as seen in hemolytic disease) can support the membrane properties required for effective tissue oxygenation in circulation.


Assuntos
Deformação Eritrocítica , Espectrina , Animais , Evolução Biológica , Membrana Eritrocítica , Eritrócitos , Camundongos
3.
Am J Hematol ; 96(9): 1064-1076, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34021930

RESUMO

Identification of stage-specific erythroid cells is critical for studies of normal and disordered human erythropoiesis. While immunophenotypic strategies have previously been developed to identify cells at each stage of terminal erythroid differentiation, erythroid progenitors are currently defined very broadly. Refined strategies to identify and characterize BFU-E and CFU-E subsets are critically needed. To address this unmet need, a flow cytometry-based technique was developed that combines the established surface markers CD34 and CD36 with CD117, CD71, and CD105. This combination allowed for the separation of erythroid progenitor cells into four discrete populations along a continuum of progressive maturation, with increasing cell size and decreasing nuclear/cytoplasmic ratio, proliferative capacity and stem cell factor responsiveness. This strategy was validated in uncultured, primary erythroid cells isolated from bone marrow of healthy individuals. Functional colony assays of these progenitor populations revealed enrichment of BFU-E only in the earliest population, transitioning to cells yielding BFU-E and CFU-E, then CFU-E only. Utilizing CD34/CD105 and GPA/CD105 profiles, all four progenitor stages and all five stages of terminal erythroid differentiation could be identified. Applying this immunophenotyping strategy to primary bone marrow cells from patients with myelodysplastic syndrome, identified defects in erythroid progenitors and in terminal erythroid differentiation. This novel immunophenotyping technique will be a valuable tool for studies of normal and perturbed human erythropoiesis. It will allow for the discovery of stage-specific molecular and functional insights into normal erythropoiesis as well as for identification and characterization of stage-specific defects in inherited and acquired disorders of erythropoiesis.


Assuntos
Células Eritroides/citologia , Células Precursoras Eritroides/citologia , Eritropoese , Antígenos CD/análise , Antígenos CD34/análise , Células da Medula Óssea/citologia , Células Cultivadas , Endoglina/análise , Citometria de Fluxo/métodos , Humanos , Imunofenotipagem/métodos
4.
Blood ; 132(22): 2406-2417, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30254129

RESUMO

Myelodysplastic syndromes (MDSs) are clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis. Anemia is the defining cytopenia of MDS patients, yet the molecular mechanisms for dyserythropoiesis in MDSs remain to be fully defined. Recent studies have revealed that heterozygous loss-of-function mutation of DNA dioxygenase TET2 is 1 of the most common mutations in MDSs and that TET2 deficiency disturbs erythroid differentiation. However, mechanistic insights into the role of TET2 on disordered erythropoiesis are not fully defined. Here, we show that TET2 deficiency leads initially to stem cell factor (SCF)-dependent hyperproliferation and impaired differentiation of human colony-forming unit-erythroid (CFU-E) cells, which were reversed by a c-Kit inhibitor. We further show that this was due to increased phosphorylation of c-Kit accompanied by decreased expression of phosphatase SHP-1, a negative regulator of c-Kit. At later stages, TET2 deficiency led to an accumulation of a progenitor population, which expressed surface markers characteristic of normal CFU-E cells but were functionally different. In contrast to normal CFU-E cells that require only erythropoietin (EPO) for proliferation, these abnormal progenitors required SCF and EPO and exhibited impaired differentiation. We termed this population of progenitors "marker CFU-E" cells. We further show that AXL expression was increased in marker CFU-E cells and that the increased AXL expression led to increased activation of AKT and ERK. Moreover, the altered proliferation and differentiation of marker CFU-E cells were partially rescued by an AXL inhibitor. Our findings document an important role for TET2 in erythropoiesis and have uncovered previously unknown mechanisms by which deficiency of TET2 contributes to ineffective erythropoiesis.


Assuntos
Proteínas de Ligação a DNA/genética , Células Precursoras Eritroides/patologia , Mutação com Perda de Função , Síndromes Mielodisplásicas/genética , Proteínas Proto-Oncogênicas/genética , Linhagem Celular Tumoral , Proliferação de Células , Metilação de DNA , Dioxigenases , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Eritropoese , Deleção de Genes , Técnicas de Silenciamento de Genes , Humanos , Síndromes Mielodisplásicas/patologia , Proteínas Proto-Oncogênicas c-kit/genética , Regulação para Cima
5.
Crit Rev Food Sci Nutr ; 60(17): 2926-2937, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31556313

RESUMO

Probiotics are defined as live microorganisms, which, when administered in adequate amounts, confer health benefits to the host. Traditionally, probiotic food research has heavily focused on the genera Bifidobacteria and Lactobacilli, along with their benefits for gut health. Recently with the identification of new probiotic strains specifically intended for oral health applications, the development of probiotic foods for oral health benefits has garnered interest, with a renewed focus on identifying new food formats for delivering probiotics. The development of novel oral probiotic foods is highly complex, as the composition of a food matrix dictates: (1) bacterial viability during production and shelf life and (2) how bacteria partition with components within a food matrix and subsequently adhere to oral cavity surfaces. At present, virtually no information is available on oral probiotic strains such as Streptococcus salivarius; specifically, how orally-derived strains survive under different food parameters. Furthermore, limited information exists on the partition behavior of probiotics with food components, governed by physico-chemical interactions and adhesion phenomena. This review aspires to examine this framework by providing a foundation with existing literature related to the common probiotic genera, in order to inform and drive future attempts of designing new oral probiotic food formats.


Assuntos
Probióticos , Bactérias , Aderência Bacteriana , Bifidobacterium , Lactobacillus , Viabilidade Microbiana
6.
Proc Natl Acad Sci U S A ; 114(18): E3669-E3678, 2017 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-28416691

RESUMO

Although sentences unfold sequentially, one word at a time, most linguistic theories propose that their underlying syntactic structure involves a tree of nested phrases rather than a linear sequence of words. Whether and how the brain builds such structures, however, remains largely unknown. Here, we used human intracranial recordings and visual word-by-word presentation of sentences and word lists to investigate how left-hemispheric brain activity varies during the formation of phrase structures. In a broad set of language-related areas, comprising multiple superior temporal and inferior frontal sites, high-gamma power increased with each successive word in a sentence but decreased suddenly whenever words could be merged into a phrase. Regression analyses showed that each additional word or multiword phrase contributed a similar amount of additional brain activity, providing evidence for a merge operation that applies equally to linguistic objects of arbitrary complexity. More superficial models of language, based solely on sequential transition probability over lexical and syntactic categories, only captured activity in the posterior middle temporal gyrus. Formal model comparison indicated that the model of multiword phrase construction provided a better fit than probability-based models at most sites in superior temporal and inferior frontal cortices. Activity in those regions was consistent with a neural implementation of a bottom-up or left-corner parser of the incoming language stream. Our results provide initial intracranial evidence for the neurophysiological reality of the merge operation postulated by linguists and suggest that the brain compresses syntactically well-formed sequences of words into a hierarchy of nested phrases.


Assuntos
Encéfalo/fisiologia , Lobo Frontal/fisiologia , Modelos Neurológicos , Fala/fisiologia , Lobo Temporal/fisiologia , Feminino , Humanos , Masculino
7.
Blood ; 129(14): 2002-2012, 2017 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-28167661

RESUMO

The ten-eleven translocation (TET) family of proteins plays important roles in a wide range of biological processes by oxidizing 5-methylcytosine (5mC) to 5-hydroxy-methylcytosine. However, their function in erythropoiesis has remained unclear. We show here that TET2 and TET3 but not TET1 are expressed in human erythroid cells, and we explore the role of these proteins in erythropoiesis. Knockdown experiments revealed that TET2 and TET3 have different functions. Suppression of TET3 expression in human CD34+ cells markedly impaired terminal erythroid differentiation, as reflected by increased apoptosis, the generation of bi/multinucleated polychromatic/orthochromatic erythroblasts, and impaired enucleation, although without effect on erythroid progenitors. In marked contrast, TET2 knockdown led to hyper-proliferation and impaired differentiation of erythroid progenitors. Surprisingly, knockdown of neither TET2 nor TET3 affected global levels of 5mC. Thus, our findings have identified distinct roles for TET2 and TET3 in human erythropoiesis, and provide new insights into their role in regulating human erythroid differentiation at distinct stages of development. Moreover, because knockdown of TET2 recapitulates certain features of erythroid development defects characteristic of myelodysplastic syndromes (MDSs), and the TET2 gene mutation is one of the most common mutations in MDS, our findings may be relevant for improved understanding of dyserythropoiesis of MDS.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Dioxigenases/metabolismo , Eritropoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Antígenos CD34/genética , Antígenos CD34/metabolismo , Proteínas de Ligação a DNA/genética , Dioxigenases/genética , Técnicas de Silenciamento de Genes , Células-Tronco Hematopoéticas/citologia , Humanos , Oxigenases de Função Mista/genética , Oxigenases de Função Mista/metabolismo , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Proteínas Proto-Oncogênicas/genética
8.
Blood ; 127(11): 1481-92, 2016 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-26679864

RESUMO

Current therapeutic strategies for sickle cell anemia are aimed at reactivating fetal hemoglobin. Pomalidomide, a third-generation immunomodulatory drug, was proposed to induce fetal hemoglobin production by an unknown mechanism. Here, we report that pomalidomide induced a fetal-like erythroid differentiation program, leading to a reversion of γ-globin silencing in adult human erythroblasts. Pomalidomide acted early by transiently delaying erythropoiesis at the burst-forming unit-erythroid/colony-forming unit-erythroid transition, but without affecting terminal differentiation. Further, the transcription networks involved in γ-globin repression were selectively and differentially affected by pomalidomide including BCL11A, SOX6, IKZF1, KLF1, and LSD1. IKAROS (IKZF1), a known target of pomalidomide, was degraded by the proteasome, but was not the key effector of this program, because genetic ablation of IKZF1 did not phenocopy pomalidomide treatment. Notably, the pomalidomide-induced reprogramming was conserved in hematopoietic progenitors from individuals with sickle cell anemia. Moreover, multiple myeloma patients treated with pomalidomide demonstrated increased in vivo γ-globin levels in their erythrocytes. Together, these data reveal the molecular mechanisms by which pomalidomide reactivates fetal hemoglobin, reinforcing its potential as a treatment for patients with ß-hemoglobinopathies.


Assuntos
Células-Tronco Hematopoéticas/efeitos dos fármacos , Talidomida/análogos & derivados , Transcrição Gênica/efeitos dos fármacos , gama-Globinas/genética , Adulto , Anemia Falciforme/sangue , Anemia Falciforme/genética , Proteínas de Transporte/sangue , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/metabolismo , Eritropoese/efeitos dos fármacos , Hemoglobina Fetal/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/metabolismo , Histona Desmetilases/sangue , Humanos , Fator de Transcrição Ikaros/sangue , Fator de Transcrição Ikaros/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/sangue , Lentivirus/genética , Mieloma Múltiplo/sangue , Mieloma Múltiplo/genética , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas Nucleares/sangue , Complexo de Endopeptidases do Proteassoma/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Repressoras , Fatores de Transcrição SOXD/sangue , Talidomida/farmacologia , Globinas beta/biossíntese , Globinas beta/genética , gama-Globinas/biossíntese
9.
Am J Hematol ; 93(4): 494-503, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29274096

RESUMO

Studies of human erythropoiesis have relied, for the most part, on the in vitro differentiation of hematopoietic stem and progenitor cells (HSPC) from different sources. Here, we report that despite the common core erythroid program that exists between cord blood (CB)- and peripheral blood (PB)-HSPC induced toward erythroid differentiation in vitro, significant functional differences exist. We undertook a comparative analysis of human erythropoiesis using these two different sources of HSPC. Upon in vitro erythroid differentiation, CB-derived cells proliferated 4-fold more than PB-derived cells. However, CB-derived cells exhibited a delayed kinetics of differentiation, resulting in an increased number of progenitors, notably colony-forming unit (CFU-E). The phenotypes of early erythroid differentiation stages also differed between the two sources with a significantly higher percentage of IL3R- GPA- CD34+ CD36+ cells generated from PB- than CB-HSPCs. This subset was found to generate both burst-forming unit (BFU-E) and CFU-E colonies in colony-forming assays. To further understand the differences between CB- and PB-HSPC, cells at eight stages of erythroid differentiation were sorted from each of the two sources and their transcriptional profiles were compared. We document differences at the CD34, BFU-E, poly- and orthochromatic stages. Genes exhibiting the most significant differences in expression between HSPC sources clustered into cell cycle- and autophagy-related pathways. Altogether, our studies provide a qualitative and quantitative comparative analysis of human erythropoiesis, highlighting the impact of the developmental origin of HSPCs on erythroid differentiation.


Assuntos
Envelhecimento/sangue , Células Precursoras Eritroides/citologia , Eritropoese/fisiologia , Adulto , Antígenos CD34/análise , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Células Precursoras Eritroides/efeitos dos fármacos , Eritropoese/genética , Eritropoetina/farmacologia , Sangue Fetal/citologia , Humanos , Recém-Nascido , Transcriptoma
10.
Biochemistry ; 55(25): 3504-3513, 2016 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-27267274

RESUMO

An asymmetric distribution of phospholipids in the membrane bilayer is inseparable from physiological functions, including shape preservation and survival of erythrocytes, and by implication other cells. Aminophospholipids, notably phosphatidylserine (PS), are confined to the inner leaflet of the erythrocyte membrane lipid bilayer by the ATP-dependent flippase enzyme, ATP11C, counteracting the activity of an ATP-independent scramblase. Phospholipid scramblase 1 (PLSCR1), a single-transmembrane protein, was previously reported to possess scrambling activity in erythrocytes. However, its function was cast in doubt by the retention of scramblase activity in erythrocytes of knockout mice lacking this protein. We show that in the human erythrocyte PLSCR1 is the predominant scramblase and by reconstitution into liposomes that its activity resides in the transmembrane domain. At or below physiological intracellular calcium concentrations, total suppression of flippase activity nevertheless leaves the membrane asymmetry undisturbed. When liposomes or erythrocytes are depleted of cholesterol (a reversible process in the case of erythrocytes), PS quickly appears at the outer surface, implying that cholesterol acts in the cell as a powerful scramblase inhibitor. Thus, our results bring to light a previously unsuspected function of cholesterol in regulating phospholipid scrambling.


Assuntos
Adenosina Trifosfatases/metabolismo , Colesterol/metabolismo , Membrana Eritrocítica/metabolismo , Eritrócitos/metabolismo , Bicamadas Lipídicas/metabolismo , Lipídeos de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Transferência de Fosfolipídeos/metabolismo , Animais , Transporte Biológico , Cálcio/metabolismo , Células Cultivadas , Eritrócitos/citologia , Humanos , Camundongos , Fosfatidilserinas/metabolismo
11.
Biochim Biophys Acta ; 1848(7): 1619-1628, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25883090

RESUMO

During development inside red blood cells (RBCs), Plasmodium falciparum malaria parasites export proteins that associate with the RBC membrane skeleton. These interactions cause profound changes to the biophysical properties of RBCs that underpin the often severe and fatal clinical manifestations of falciparum malaria. P. falciparum erythrocyte membrane protein 1 (PfEMP1) is one such exported parasite protein that plays a major role in malaria pathogenesis since its exposure on the parasitised RBC surface mediates their adhesion to vascular endothelium and placental syncytioblasts. En route to the RBC membrane skeleton, PfEMP1 transiently associates with Maurer's clefts (MCs), parasite-derived membranous structures in the RBC cytoplasm. We have previously shown that a resident MC protein, skeleton-binding protein 1 (SBP1), is essential for the placement of PfEMP1 onto the RBC surface and hypothesised that the function of SBP1 may be to target MCs to the RBC membrane. Since this would require additional protein interactions, we set out to identify binding partners for SBP1. Using a combination of approaches, we have defined the region of SBP1 that binds specifically to defined sub-domains of two major components of the RBC membrane skeleton, protein 4.1R and spectrin. We show that these interactions serve as one mechanism to anchor MCs to the RBC membrane skeleton, however, while they appear to be necessary, they are not sufficient for the translocation of PfEMP1 onto the RBC surface. The N-terminal domain of SBP1 that resides within the lumen of MCs clearly plays an essential, but presently unknown role in this process.


Assuntos
Proteínas de Transporte/metabolismo , Membrana Eritrocítica/metabolismo , Eritrócitos/metabolismo , Proteínas de Membrana/metabolismo , Plasmodium falciparum/metabolismo , Proteínas de Protozoários/metabolismo , Antígenos de Protozoários/genética , Antígenos de Protozoários/metabolismo , Sítios de Ligação , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas do Citoesqueleto/metabolismo , Eritrócitos/parasitologia , Eritrócitos/ultraestrutura , Interações Hospedeiro-Parasita , Humanos , Immunoblotting , Proteínas de Membrana/química , Proteínas de Membrana/genética , Microscopia Confocal , Microscopia Eletrônica de Varredura , Plasmodium falciparum/genética , Plasmodium falciparum/fisiologia , Ligação Proteica , Proteínas de Protozoários/química , Proteínas de Protozoários/genética , Espectrina/metabolismo , Ressonância de Plasmônio de Superfície
12.
BMC Microbiol ; 16(1): 225, 2016 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-27681377

RESUMO

BACKGROUND: Pneumococcal adherence to the nasopharyngeal epithelium is a critical step in colonisation and disease. The probiotic bacterium, Streptococcus salivarius, can inhibit pneumococcal adherence to epithelial cells in vitro. We investigated the mechanism(s) of inhibition using a human pharyngeal epithelial cell line (Detroit 562) following pre-administration of two different strains of S. salivarius. RESULTS: Whilst the bacteriocin-encoding megaplasmids of S. salivarius strains K12 and M18 were essential to prevent pneumococcal growth on solid media, they were not required to inhibit pneumococcal adherence. Experiments testing S. salivarius K12 and two pneumococcal isolates (serotypes 19F and 6A) showed that inhibition of 19F may involve S. salivarius-mediated blocking of pneumococcal binding sites: a negative correlation was observed between adherence of K12 and 19F, and no inhibition occurred when K12 was prevented from contacting epithelial cells. K12-mediated inhibition of adherence by 6A may involve additional mechanisms, since no correlation was observed between adherence of K12 and 6A, and K12 could inhibit 6A adherence in the absence of cell contact. CONCLUSIONS: These results suggest that S. salivarius employs several mechanisms, including blocking pneumococcal binding sites, to reduce pneumococcal adherence to pharyngeal epithelial cells. These findings extend our understanding of how probiotics may inhibit pneumococcal adherence and could assist with the development of novel strategies to prevent pneumococcal colonisation in the future.

13.
Blood ; 124(24): 3636-45, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25339359

RESUMO

Burst-forming unit-erythroid (BFU-E) and colony-forming unit-erythroid (CFU-E) cells are erythroid progenitors traditionally defined by colony assays. We developed a flow cytometry-based strategy for isolating human BFU-E and CFU-E cells based on the changes in expression of cell surface markers during in vitro erythroid cell culture. BFU-E and CFU-E are characterized by CD45(+)GPA(-)IL-3R(-)CD34(+)CD36(-)CD71(low) and CD45(+)GPA(-)IL-3R(-)CD34(-)CD36(+)CD71(high) phenotypes, respectively. Colony assays validated phenotypic assignment giving rise to BFU-E and CFU-E colonies, both at a purity of ∼90%. The BFU-E colony forming ability of CD45(+)GPA(-)IL-3R(-)CD34(+)CD36(-)CD71(low) cells required stem cell factor and erythropoietin, while the CFU-E colony forming ability of CD45(+)GPA(-)IL-3R(-)CD34(-)CD36(+)CD71(high) cells required only erythropoietin. Bioinformatic analysis of the RNA-sequencing data revealed unique transcriptomes at each differentiation stage. The sorting strategy was validated in uncultured primary cells isolated from bone marrow, cord blood, and peripheral blood, indicating that marker expression is not an artifact of in vitro cell culture, but represents an in vivo characteristic of erythroid progenitor populations. The ability to isolate highly pure human BFU-E and CFU-E progenitors will enable detailed cellular and molecular characterization of these distinct progenitor populations and define their contribution to disordered erythropoiesis in inherited and acquired hematologic disease. Our data provides an important resource for future studies of human erythropoiesis.


Assuntos
Antígenos de Diferenciação/biossíntese , Células Precursoras Eritroides/metabolismo , Eritropoese/fisiologia , Eritropoetina/metabolismo , Regulação da Expressão Gênica/fisiologia , Transcriptoma/fisiologia , Separação Celular/métodos , Células Precursoras Eritroides/citologia , Feminino , Humanos , Masculino
14.
Haematologica ; 101(5): 559-65, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26944472

RESUMO

Phosphatidylserine is localized exclusively to the inner leaflet of the membrane lipid bilayer of most cells, including erythrocytes. This asymmetric distribution is critical for the survival of erythrocytes in circulation since externalized phosphatidylserine is a phagocytic signal for splenic macrophages. Flippases are P-IV ATPase family proteins that actively transport phosphatidylserine from the outer to inner leaflet. It has not yet been determined which of the 14 members of this family of proteins is the flippase in human erythrocytes. Herein, we report that ATP11C encodes a major flippase in human erythrocytes, and a genetic mutation identified in a male patient caused congenital hemolytic anemia inherited as an X-linked recessive trait. Phosphatidylserine internalization in erythrocytes with the mutant ATP11C was decreased 10-fold compared to that of the control, functionally establishing that ATP11C is a major flippase in human erythrocytes. Contrary to our expectations phosphatidylserine was retained in the inner leaflet of the majority of mature erythrocytes from both controls and the patient, suggesting that phosphatidylserine cannot be externalized as long as scramblase is inactive. Phosphatidylserine-exposing cells were found only in the densest senescent cells (0.1% of total) in which scramblase was activated by increased Ca(2+) concentration: the percentage of these phosphatidylserine-exposing cells was increased in the patient's senescent cells accounting for his mild anemia. Furthermore, the finding of similar extents of phosphatidylserine exposure by exogenous Ca(2+)-activated scrambling in both control erythrocytes and the patient's erythrocytes implies that suppressed scramblase activity rather than flippase activity contributes to the maintenance of phosphatidylserine in the inner leaflet of human erythrocytes.


Assuntos
Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Anemia Hemolítica Congênita/genética , Anemia Hemolítica Congênita/metabolismo , Eritrócitos/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Adenosina Trifosfatases/deficiência , Adolescente , Anemia Hemolítica Congênita/diagnóstico , Transporte Biológico Ativo , Biomarcadores , Cálcio/metabolismo , Índices de Eritrócitos , Eritrócitos/efeitos dos fármacos , Humanos , Masculino , Proteínas de Membrana Transportadoras/deficiência , Mutação , Fosfatidilserinas/metabolismo , Fosfatidilserinas/farmacologia
15.
J Biol Chem ; 289(9): 5925-37, 2014 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-24381168

RESUMO

Protein 4.1B is a member of protein 4.1 family, adaptor proteins at the interface of membranes and the cytoskeleton. It is expressed in most mammalian tissues and is known to be required in formation of nervous and cardiac systems; it is also a tumor suppressor with a role in metastasis. Here, we explore functions of 4.1B using primary mouse embryonic fibroblasts (MEF) derived from wild type and 4.1B knock-out mice. MEF cells express two 4.1B isoforms: 130 and 60-kDa. 130-kDa 4.1B was absent from 4.1B knock-out MEF cells, but 60-kDa 4.1B remained, suggesting incomplete knock-out. Although the 130-kDa isoform was predominantly located at the plasma membrane, the 60-kDa isoform was enriched in nuclei. 130-kDa-deficient 4.1B MEF cells exhibited impaired cell adhesion, spreading, and migration; they also failed to form actin stress fibers. Impaired cell spreading and stress fiber formation were rescued by re-expression of the 130-kDa 4.1B but not the 60-kDa 4.1B. Our findings document novel, isoform-selective roles for 130-kDa 4.1B in adhesion, spreading, and migration of MEF cells by affecting actin organization, giving new insight into 4.1B functions in normal tissues as well as its role in cancer.


Assuntos
Citoesqueleto de Actina/metabolismo , Movimento Celular/fisiologia , Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Proteínas dos Microfilamentos/metabolismo , Citoesqueleto de Actina/genética , Animais , Adesão Celular/fisiologia , Membrana Celular/genética , Membrana Celular/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Células Cultivadas , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Camundongos , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Neoplasias/genética , Neoplasias/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
16.
Infect Immun ; 83(9): 3438-44, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26077762

RESUMO

Streptococcus agalactiae (group B streptococcus [GBS]) colonizes the rectovaginal tract in 20% to 30% of women and during pregnancy can be transmitted to the newborn, causing severe invasive disease. Current routine screening and antibiotic prophylaxis have fallen short of complete prevention of GBS transmission, and GBS remains a leading cause of neonatal infection. We have investigated the ability of Streptococcus salivarius, a predominant member of the native human oral microbiota, to control GBS colonization. Comparison of the antibacterial activities of multiple S. salivarius strains by use of a deferred-antagonism test showed that S. salivarius strain K12 exhibited the broadest spectrum of activity against GBS. K12 effectively inhibited all GBS strains tested, including disease-implicated isolates from newborns and colonizing isolates from the vaginal tract of pregnant women. Inhibition was dependent on the presence of megaplasmid pSsal-K12, which encodes the bacteriocins salivaricin A and salivaricin B; however, in coculture experiments, GBS growth was impeded by K12 independently of the megaplasmid. We also demonstrated that K12 adheres to and invades human vaginal epithelial cells at levels comparable to GBS. Inhibitory activity of K12 was examined in vivo using a mouse model of GBS vaginal colonization. Mice colonized with GBS were treated vaginally with K12. K12 administration significantly reduced GBS vaginal colonization in comparison to nontreated controls, and this effect was partially dependent on the K12 megaplasmid. Our results suggest that K12 may have potential as a preventative therapy to control GBS vaginal colonization and thereby prevent its transmission to the neonate during pregnancy.


Assuntos
Infecções Estreptocócicas/microbiologia , Infecções Estreptocócicas/transmissão , Streptococcus agalactiae/crescimento & desenvolvimento , Streptococcus/fisiologia , Vagina/microbiologia , Animais , Linhagem Celular , Técnicas de Cocultura , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Camundongos
17.
Anal Chem ; 87(20): 10627-34, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26399599

RESUMO

Protein therapeutics are rapidly transforming the pharmaceutical industry. Unlike for small molecule therapeutics, current technologies are challenged to provide the rapid, high-resolution analyses of protein higher order structures needed to ensure drug efficacy and safety. Consequently, significant attention has turned to developing new methods that can quickly, accurately, and reproducibly characterize the three-dimensional structure of protein therapeutics. In this work, we describe a method that uses diethylpyrocarbonate (DEPC) labeling and mass spectrometry to detect three-dimensional structural changes in therapeutic proteins that have been exposed to degrading conditions. Using ß2-microglobulin, immunoglobulin G1, and human growth hormone as model systems, we demonstrate that DEPC labeling can identify both specific protein regions that mediate aggregation and those regions that undergo more subtle structural changes upon mishandling of these proteins. Importantly, DEPC labeling is able to provide information for up to 30% of the surface residues in a given protein, thereby providing excellent structural resolution. Given the simplicity of the DEPC labeling chemistry and the relatively straightforward mass spectral analysis of DEPC-labeled proteins, we expect this method should be amenable to a wide range of protein therapeutics and their different formulations.


Assuntos
Dietil Pirocarbonato/química , Hormônio do Crescimento/química , Imunoglobulina G/química , Microglobulina beta-2/química , Humanos , Espectrometria de Massas , Modelos Moleculares , Estrutura Molecular
18.
Gastroenterology ; 144(1): 50-52.e5, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22995675

RESUMO

Reductions in levels of the hunger-stimulating hormone ghrelin have been proposed to mediate part of the effects of vertical sleeve gastrectomy (VSG) and Roux-en-Y gastric bypass surgeries for obesity. We studied circulating levels of acyl and desacyl ghrelin in rats after these surgeries. We found that blood levels of ghrelin were reduced after VSG, but not after Roux-en-Y gastric bypass, based on enzyme-linked immunosorbent assay and mass-spectrometry analyses. We compared the effects of VSG in ghrelin-deficient mice and wild-type mice on food intake, body weight, dietary fat preference, and glucose tolerance. We found that VSG produced comparable outcomes in each strain. Reduced ghrelin signaling therefore does not appear to be required for these effects of VSG.


Assuntos
Ingestão de Alimentos , Comportamento Alimentar , Gastrectomia , Grelina/sangue , Animais , Peso Corporal , Gorduras na Dieta , Genótipo , Grelina/deficiência , Grelina/genética , Teste de Tolerância a Glucose , Masculino , Camundongos , Camundongos Knockout , Ratos , Ratos Long-Evans , Transdução de Sinais
19.
A A Pract ; 17(2): e01658, 2023 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-36800513

RESUMO

Platelet storage pool disorders (PSPDs) constitute a diverse group of hematologic abnormalities, which share the common feature of a deficiency in the ability of platelets to aggregate. Parturients with PSPD can present management challenges to their care team when they present for childbirth, especially with regard to neuraxial anesthesia. We report a series of 2 deliveries from unrelated patients affected by PSPD. In particular, we highlight the utility of rotational thromboelastometry (ROTEM) and the need for a multidisciplinary approach to the care of patients with this hematologic abnormality.


Assuntos
Técnicas de Apoio para a Decisão , Tromboelastografia , Gravidez , Feminino , Humanos
20.
Cogn Sci ; 47(7): e13312, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37417470

RESUMO

To model behavioral and neural correlates of language comprehension in naturalistic environments, researchers have turned to broad-coverage tools from natural-language processing and machine learning. Where syntactic structure is explicitly modeled, prior work has relied predominantly on context-free grammars (CFGs), yet such formalisms are not sufficiently expressive for human languages. Combinatory categorial grammars (CCGs) are sufficiently expressive directly compositional models of grammar with flexible constituency that affords incremental interpretation. In this work, we evaluate whether a more expressive CCG provides a better model than a CFG for human neural signals collected with functional magnetic resonance imaging (fMRI) while participants listen to an audiobook story. We further test between variants of CCG that differ in how they handle optional adjuncts. These evaluations are carried out against a baseline that includes estimates of next-word predictability from a transformer neural network language model. Such a comparison reveals unique contributions of CCG structure-building predominantly in the left posterior temporal lobe: CCG-derived measures offer a superior fit to neural signals compared to those derived from a CFG. These effects are spatially distinct from bilateral superior temporal effects that are unique to predictability. Neural effects for structure-building are thus separable from predictability during naturalistic listening, and those effects are best characterized by a grammar whose expressive power is motivated on independent linguistic grounds.


Assuntos
Encéfalo , Idioma , Humanos , Encéfalo/diagnóstico por imagem , Linguística , Mapeamento Encefálico , Percepção Auditiva , Compreensão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA