Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 361
Filtrar
1.
Nat Commun ; 12(1): 7334, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34921133

RESUMEN

The erythroid terminal differentiation program couples sequential cell divisions with progressive reductions in cell size. The erythropoietin receptor (EpoR) is essential for erythroblast survival, but its other functions are not well characterized. Here we use Epor-/- mouse erythroblasts endowed with survival signaling to identify novel non-redundant EpoR functions. We find that, paradoxically, EpoR signaling increases red cell size while also increasing the number and speed of erythroblast cell cycles. EpoR-regulation of cell size is independent of established red cell size regulation by iron. High erythropoietin (Epo) increases red cell size in wild-type mice and in human volunteers. The increase in mean corpuscular volume (MCV) outlasts the duration of Epo treatment and is not the result of increased reticulocyte number. Our work shows that EpoR signaling alters the relationship between cycling and cell size. Further, diagnostic interpretations of increased MCV should now include high Epo levels and hypoxic stress.


Asunto(s)
Ciclo Celular , Tamaño de la Célula , Eritrocitos/citología , Eritrocitos/metabolismo , Eritropoyesis , Receptores de Eritropoyetina/metabolismo , Adulto , Animales , Antígenos CD/metabolismo , Antígenos CD4/metabolismo , Diferenciación Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Supervivencia Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Embrión de Mamíferos/metabolismo , Eritroblastos/citología , Eritroblastos/efectos de los fármacos , Eritroblastos/metabolismo , Eritropoyetina/administración & dosificación , Eritropoyetina/farmacología , Femenino , Feto/metabolismo , Voluntarios Sanos , Humanos , Hierro/metabolismo , Hígado/embriología , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Transferrina/metabolismo , Reticulocitos/citología , Reticulocitos/efectos de los fármacos , Reticulocitos/metabolismo , Transducción de Señal , Proteína bcl-X/metabolismo
2.
Sci Rep ; 11(1): 22121, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34764389

RESUMEN

Arsenic exposure poses numerous threats to human health. Our previous work in mice has shown that arsenic causes anemia by inhibiting erythropoiesis. However, the impacts of arsenic exposure on human erythropoiesis remain largely unclear. We report here that low-dose arsenic exposure inhibits the erythroid differentiation of human hematopoietic progenitor cells (HPCs). The impacts of arsenic (in the form of arsenite; As3+) on red blood cell (RBC) development was evaluated using a long-term culture of normal human bone marrow CD34+-HPCs stimulated in vitro to undergo erythropoiesis. Over the time course studied, we analyzed the expression of the cell surface antigens CD34, CD71 and CD235a, which are markers commonly used to monitor the progression of HPCs through the stages of erythropoiesis. Simultaneously, we measured hemoglobin content, which is an important criterion used clinically for diagnosing anemia. As compared to control, low-dose As3+ exposure (100 nM and 500 nM) inhibited the expansion of CD34+-HPCs over the time course investigated; decreased the number of committed erythroid progenitors (BFU-E and CFU-E) and erythroblast differentiation in the subsequent stages; and caused a reduction of hemoglobin content. These findings demonstrate that low-dose arsenic exposure impairs human erythropoiesis, likely by combined effects on various stages of RBC formation.


Asunto(s)
Antígenos CD34/metabolismo , Arsenitos/efectos adversos , Diferenciación Celular/efectos de los fármacos , Células Precursoras Eritroides/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Hemoglobinas/metabolismo , Anemia/inducido químicamente , Anemia/metabolismo , Antígenos CD/metabolismo , Células Cultivadas , Eritroblastos/efectos de los fármacos , Eritroblastos/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoyesis/efectos de los fármacos , Glicoforinas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Receptores de Transferrina/metabolismo
3.
Int J Mol Sci ; 22(15)2021 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-34360974

RESUMEN

Erythropoietin (EPO) downregulates hepcidin expression to increase the availability of iron; the downregulation of hepcidin is mediated by erythroferrone (ERFE) secreted by erythroblasts. Erythroblasts also express transferrin receptor 2 (TFR2); however, the possible role of TFR2 in hepcidin downregulation is unclear. The purpose of the study was to correlate liver expression of hepcidin with the expression of ERFE and TFR2 in murine bone marrow and spleen at 4, 16, 24, 48, 72 and 96 h following administration of a single dose of EPO. Splenic Fam132b expression increased 4 h after EPO injection; liver hepcidin mRNA was decreased at 16 h. In the spleen, expression of TFR2 and transferrin receptor (TFR1) proteins increased by an order of magnitude at 48 and 72 h after EPO treatment. The EPO-induced increase in splenic TFR2 and TFR1 was associated with an increase in the number of Tfr2- and Tfr1-expressing erythroblasts. Plasma exosomes prepared from EPO-treated mice displayed increased amount of TFR1 protein; however, no exosomal TFR2 was detected. Overall, the results confirm the importance of ERFE in stress erythropoiesis, support the role of TFR2 in erythroid cell development, and highlight possible differences in the removal of TFR2 and TFR1 from erythroid cell membranes.


Asunto(s)
Eritropoyetina/farmacología , Receptores de Transferrina/genética , Animales , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Eritroblastos/efectos de los fármacos , Eritroblastos/metabolismo , Exosomas/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Receptores de Transferrina/metabolismo , Bazo/metabolismo
4.
Int J Mol Sci ; 22(2)2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33478008

RESUMEN

Bioreactors are increasingly implemented for large scale cultures of various mammalian cells, which requires optimization of culture conditions. Such upscaling is also required to produce red blood cells (RBC) for transfusion and therapy purposes. However, the physiological suitability of RBC cultures to be transferred to stirred bioreactors is not well understood. PIEZO1 is the most abundantly expressed known mechanosensor on erythroid cells. It is a cation channel that translates mechanical forces directly into a physiological response. We investigated signaling cascades downstream of PIEZO1 activated upon transitioning stationary cultures to orbital shaking associated with mechanical stress, and compared the results to direct activation of PIEZO1 by the chemical agonist Yoda1. Erythroblasts subjected to orbital shaking displayed decreased proliferation, comparable to incubation in the presence of a low dose of Yoda1. Epo (Erythropoietin)-dependent STAT5 phosphorylation, and Calcineurin-dependent NFAT dephosphorylation was enhanced. Phosphorylation of ERK was also induced by both orbital shaking and Yoda1 treatment. Activation of these pathways was inhibited by intracellular Ca2+ chelation (BAPTA-AM) in the orbital shaker. Our results suggest that PIEZO1 is functional and could be activated by the mechanical forces in a bioreactor setup, and results in the induction of Ca2+-dependent signaling cascades regulating various aspects of erythropoiesis. With this study, we showed that Yoda1 treatment and mechanical stress induced via orbital shaking results in comparable activation of some Ca2+-dependent pathways, exhibiting that there are direct physiological outcomes of mechanical stress on erythroblasts.


Asunto(s)
Señalización del Calcio/fisiología , Eritroblastos/fisiología , Estrés Mecánico , Calcio/metabolismo , Calcio/farmacología , Técnicas de Cultivo de Célula , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Eritroblastos/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Eritropoyesis/fisiología , Humanos , Canales Iónicos/agonistas , Canales Iónicos/fisiología , Mecanotransducción Celular/efectos de los fármacos , Mecanotransducción Celular/fisiología , Pirazinas/farmacología , Rotación , Tiadiazoles/farmacología
5.
Clin Exp Immunol ; 203(3): 409-423, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33205401

RESUMEN

Biological treatments such as enzyme-replacement therapies (ERT) can generate anti-drug antibodies (ADA), which may reduce drug efficacy and impact patient safety and consequently led to research to mitigate ADA responses. Transient low-dose methotrexate (TLD-MTX) as a prophylactic ITI regimen, when administered concurrently with ERT, induces long-lived reduction of ADA to recombinant human alglucosidase alfa (rhGAA) in mice. In current clinical practice, a prophylactic ITI protocol that includes TLD-MTX, rituximab and intravenous immunoglobulin (optional), successfully induced lasting control of ADA to rhGAA in high-risk, cross-reactive immunological material (CRIM)-negative infantile-onset Pompe disease (IOPD) patients. More recently, evaluation of TLD-MTX demonstrated benefit in CRIM-positive IOPD patients. To more clearly understand the mechanism for the effectiveness of TLD-MTX, non-targeted transcriptional and proteomic screens were conducted and revealed up-regulation of erythropoiesis signatures. Confirmatory studies showed transiently larger spleens by weight, increased spleen cellularity and that following an initial reduction of mature red blood cells (RBCs) in the bone marrow and blood, a significant expansion of Ter-119+ CD71+ immature RBCs was observed in spleen and blood of mice. Histology sections revealed increased nucleated cells, including hematopoietic precursors, in the splenic red pulp of these mice. This study demonstrated that TLD-MTX induced a transient reduction of mature RBCs in the blood and immature RBCs in the bone marrow followed by significant enrichment of immature, nucleated RBCs in the spleen and blood during the time of immune tolerance induction, which suggested modulation of erythropoiesis may be associated with the induction of immune tolerance to rhGAA.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Eritroblastos/efectos de los fármacos , Tolerancia Inmunológica/efectos de los fármacos , Metotrexato/administración & dosificación , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Relación Dosis-Respuesta a Droga , Eritroblastos/citología , Eritroblastos/metabolismo , Eritrocitos/efectos de los fármacos , Eritrocitos/inmunología , Eritrocitos/metabolismo , Eritropoyesis/efectos de los fármacos , Eritropoyesis/genética , Eritropoyesis/inmunología , Femenino , Perfilación de la Expresión Génica/métodos , Humanos , Tolerancia Inmunológica/genética , Tolerancia Inmunológica/inmunología , Inmunosupresores/administración & dosificación , Inmunosupresores/inmunología , Metotrexato/inmunología , Ratones Endogámicos C57BL , Proteómica/métodos , Bazo/efectos de los fármacos , Bazo/inmunología , Bazo/metabolismo , alfa-Glucosidasas/administración & dosificación
6.
Int J Mol Sci ; 21(23)2020 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-33255601

RESUMEN

Erythropoietin (EPO) is an essential hormone for erythropoiesis, protecting differentiating erythroblasts against apoptosis. EPO has been largely studied in stress or pathological conditions but its regulatory role in steady state erythropoiesis has been less documented. Herein, we report production of EPO by bone marrow-derived macrophages (BMDM) in vitro, and its further enhancement in BMDM conditioned with media from apoptotic cells. Confocal microscopy confirmed EPO production in erythroblastic island (EBI)-associated macrophages, and analysis of mice depleted of EBI macrophages by clodronate liposomes revealed drops in EPO levels in bone marrow (BM) cell lysates, and decreased percentages of EPO-responsive erythroblasts in the BM. We hypothesize that EBI macrophages are an in-situ source of EPO and sustain basal erythropoiesis in part through its secretion. To study this hypothesis, mice were injected with clodronate liposomes and were supplied with exogenous EPO (1-10 IU/mouse) to evaluate potential rescue of the deficiency in erythroid cells. Our results show that at doses of 5 and 10 IU, EPO significantly rescues BM steady state erythropoiesis in mice deficient of macrophages. We propose existence of a mechanism by which EBI macrophages secrete EPO in response to apoptotic erythroblasts, which is in turn controlled by the numbers of erythroid precursors generated.


Asunto(s)
Apoptosis/efectos de los fármacos , Eritropoyesis/genética , Eritropoyetina/genética , Macrófagos/metabolismo , Animales , Diferenciación Celular/genética , Ácido Clodrónico/farmacología , Eritroblastos/efectos de los fármacos , Eritroblastos/metabolismo , Células Eritroides/efectos de los fármacos , Células Eritroides/metabolismo , Eritropoyesis/efectos de los fármacos , Eritropoyetina/farmacología , Liposomas/farmacología , Macrófagos/efectos de los fármacos , Ratones
7.
Syst Biol Reprod Med ; 66(6): 378-386, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32972244

RESUMEN

The Origin Replication Complex subunit 4 (ORC4) is one in six subunits of the Origin Replication Complexes (ORCs) which is essential for initiating licensing at DNA replication origins and recruiting adaptor molecules necessary for various cellular processes. Previously, we reported that ORC4 also plays a vital role in polar body extrusion (PBE) during oogenesis in which half the chromosomes are extruded from the oocyte. We hypothesized that ORC4 might play a broader role in chromatin elimination. We tested its role in enucleation during the development of erythrocytes. Murine erythroleukemia (MEL) cells can be propagated in culture indefinitely and can be induced to enucleate their DNA by treatment with Vacuolin-1, thereby mimicking normal erythrocyte enucleation. We found that ORC4 appeared around the nuclei of the MEL cells with Vacuolin-1 treatment, gradually increasing in thickness before enucleation. We then tested whether ORC4 was required for MEL enucleation by down regulating ORC4 with siRNA-ORC4 during Vacuolin-1 treatment and found that this prevented MEL enucleation. These data are consistent with the model that ORC4 is required for erythroblast enucleation just as it is for oocyte PBE. They suggest a new model in which ORC4 expression is a marker for the initiation to the enucleation pathway.


Asunto(s)
Núcleo Celular/metabolismo , Eritroblastos/metabolismo , Leucemia Eritroblástica Aguda/metabolismo , Oogénesis , Complejo de Reconocimiento del Origen/metabolismo , Cuerpos Polares/metabolismo , Animales , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Núcleo Celular/patología , Replicación del ADN , Eritroblastos/efectos de los fármacos , Eritroblastos/patología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patología , Ratones , Complejo de Reconocimiento del Origen/genética
8.
FASEB J ; 34(9): 11672-11684, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32667087

RESUMEN

Testosterone stimulates iron-dependent erythropoiesis and suppresses hepcidin. To clarify the role of iron in mediating testosterone's effects on erythropoiesis, we induced iron deficiency in mice by feeding low iron diet. Iron-replete and iron-deficient mice were treated weekly with testosterone propionate or vehicle for 3 weeks. Testosterone treatment increased red cell count in iron-replete mice, but, surprisingly, testosterone reduced red cell count in iron-deficient mice. Splenic stress erythropoiesis was stimulated in iron-deficient mice relative to iron-replete mice, and further increased by testosterone treatment, as indicated by the increase in red pulp area, the number of nucleated erythroblasts, and expression levels of TfR1, GATA1, and other erythroid genes. Testosterone treatment of iron-deficient mice increased the ratio of early-to-late erythroblasts in the spleen and bone marrow, and serum LDH level, consistent with ineffective erythropoiesis. In iron-deficient mice, erythropoietin levels were higher but erythropoietin-regulated genes were generally downregulated relative to iron-replete mice, suggesting erythropoietin resistance. Conclusion: Testosterone treatment stimulates splenic stress erythropoiesis in iron-replete as well as iron-deficient mice. However, testosterone worsens anemia in iron-deficient mice because of ineffective erythropoiesis possibly due to erythropoietin resistance associated with iron deficiency. Iron plays an important role in mediating testosterone's effects on erythropoiesis.


Asunto(s)
Anemia Ferropénica/metabolismo , Eritropoyesis/efectos de los fármacos , Deficiencias de Hierro , Testosterona/administración & dosificación , Andrógenos/administración & dosificación , Anemia Ferropénica/sangre , Anemia Ferropénica/genética , Animales , Eritroblastos/citología , Eritroblastos/efectos de los fármacos , Eritroblastos/metabolismo , Recuento de Eritrocitos , Células Eritroides/citología , Células Eritroides/efectos de los fármacos , Células Eritroides/metabolismo , Eritropoyesis/genética , Femenino , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Expresión Génica/efectos de los fármacos , Hierro/fisiología , Ratones Endogámicos C57BL , Receptores de Transferrina/genética , Receptores de Transferrina/metabolismo
9.
Nutrients ; 12(5)2020 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-32384595

RESUMEN

Nigella sativa (N. sativa) is a medicinal plant used for its therapeutic pharmacological effects such as anti-inflammatory, antioxidant, anticancer, antidiabetic, and immunomodulation. This study explored the anti-cytotoxic and anti-genotoxic effect of N. sativa through a micronucleus test (MNT) of BALB/c mice peripheral blood. Using 6-to-8-week-old healthy male BALB/c mice, four groups were formed: (1) Control (sterile water), single-dose 2 mg/kg/intraperitoneal (i.p); (2) N. sativa oil, 500 mg/kg/24 h/7 days/i.p; (3) Cisplatin (CP), single-dose 2 mg/kg/subcutaneous (s.c); (4) N. sativa + CP with their respective dosage. When evaluating polychromatic erythrocytes (PCE), a biomarker of cytotoxicity, the group treated with N. sativa + CP experienced an increase in the frequency of PCE, which demonstrated the recovery of bone marrow and modulation of cell proliferation. The analysis of micronucleated polychromatic erythrocytes (MNPCE), an acute genotoxicity biomarker, showed similar frequency of MNPCE within the groups except in CP, but, in the N. sativa + CP group, the frequency of MNPCE decreased and then regulated. Finally, the frequency of micronucleated erythrocytes (MNE), a biomarker of genotoxicity, the supplementation of N. sativa oil did not induce genotoxic damage in this model. Thus, we conclude that N. sativa has both cytoprotective, genoprotective effects and modulates cell proliferation in BALB/c mice.


Asunto(s)
Cisplatino/toxicidad , Citoprotección/efectos de los fármacos , Eritroblastos/efectos de los fármacos , Pruebas de Micronúcleos/métodos , Nigella sativa/química , Aceites de Plantas/farmacología , Animales , Células de la Médula Ósea/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Masculino , Ratones Endogámicos BALB C , Aceites de Plantas/administración & dosificación , Aceites de Plantas/aislamiento & purificación
10.
Blood ; 135(22): 1957-1968, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32268371

RESUMEN

Reversing the developmental switch from fetal hemoglobin (HbF, α2γ2) to adult hemoglobin (HbA, α2ß2) is an important therapeutic approach in sickle cell disease (SCD) and ß-thalassemia. In healthy individuals, SCD patients, and patients treated with pharmacologic HbF inducers, HbF is present only in a subset of red blood cells known as F cells. Despite more than 50 years of observations, the cause for this heterocellular HbF expression pattern, even among genetically identical cells, remains unknown. Adult F cells might represent a reversion of a given cell to a fetal-like epigenetic and transcriptional state. Alternatively, isolated transcriptional or posttranscriptional events at the γ-globin genes might underlie heterocellularity. Here, we set out to understand the heterogeneity of HbF activation by developing techniques to purify and profile differentiation stage-matched late erythroblast F cells and non-F cells (A cells) from the human HUDEP2 erythroid cell line and primary human erythroid cultures. Transcriptional and proteomic profiling of these cells demonstrated very few differences between F and A cells at the RNA level either under baseline conditions or after treatment with HbF inducers hydroxyurea or pomalidomide. Surprisingly, we did not find differences in expression of any known HbF regulators, including BCL11A or LRF, that would account for HbF activation. Our analysis shows that F erythroblasts are not significantly different from non-HbF-expressing cells and that the primary differences likely occur at the transcriptional level at the ß-globin locus.


Asunto(s)
Eritroblastos/metabolismo , Hemoglobina Fetal/biosíntesis , Hemoglobina A/metabolismo , Adulto , Anemia de Células Falciformes/sangre , Anemia de Células Falciformes/genética , Línea Celular , Separación Celular/métodos , Células Cultivadas , Eritroblastos/clasificación , Eritroblastos/efectos de los fármacos , Células Eritroides/clasificación , Células Eritroides/metabolismo , Hemoglobina Fetal/genética , Perfilación de la Expresión Génica , Hemoglobina A/genética , Humanos , Hidroxiurea/farmacología , Proteómica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Talidomida/análogos & derivados , Talidomida/farmacología
11.
Biochem Pharmacol ; 171: 113717, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31751536

RESUMEN

Heritable disorders associated with hemoglobin production are the most common monogenic disorders. These are mainly represented by disorders such as ß-thalassemia and sickle cell disease. Induction of fetal hemoglobin (HbF) has been known to ameliorate the clinical severity of these ß hemoglobinopathies. A high throughput phenotypic screening was used in this study to isolate novel compounds that may enhance the expression of γ-globin, the component of HbF, in human erythroid cell lines and primary erythroid progenitors derived from human CD34+ cells. The effect of lead compounds on epigenetic enzymes and key transcriptional factors was evaluated to identify their mode of action. One hit compound was further evaluated in vivo using monkey models. Among the ~18,000 compounds screened, 18 compounds were selected and tested to determine their ability to induce HbF in human erythroid cell lines and primary erythroid cells. One of these compounds, a 3-phenyl-isoxazole derivative, could potentially induce HbF in monkey bone marrow cells when administered orally. The compound downregulated negative transcriptional regulators of HbF, Bcl11a and LRF without inhibiting the known epigenetic enzymes. These studies demonstrated the advantages associated with phenotype-screening and identified novel fetal globin inducers that may be useful for treating hemoglobinopathies.


Asunto(s)
Hemoglobina Fetal/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hemoglobinopatías/genética , Proteínas Represoras/genética , Xenobióticos/farmacología , Dedos de Zinc , Animales , Antígenos CD34/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Eritroblastos/citología , Eritroblastos/efectos de los fármacos , Eritroblastos/metabolismo , Hemoglobina Fetal/metabolismo , Hemoglobinopatías/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Macaca fascicularis , Fenotipo , Proteínas Represoras/metabolismo
12.
Artículo en Inglés | MEDLINE | ID: mdl-31708078

RESUMEN

Flow cytometry-based phenotypic detection of red blood cells (RBCs) deficient in surface markers anchored by glycosylphosphatidylinositol (GPI) is an efficient tool for monitoring somatic mutation in mammalian species. Biochemical considerations suggest that GPI-anchored marker-deficient RBCs found in peripheral blood are due to mutations in the endogenous X-linked phosphatidylinositolglycan, class A gene (Pig-a gene). Yet the linkage between the detected mutant phenotype and the actual mutation in the Pig-a gene is difficult to establish directly in mammalian RBCs that are naturally free of genomic DNA and may have only traces of heavily degraded mRNA. We have traced the origin of the marker-deficient RBC phenotype in the precursors of peripheral RBCs, bone marrow erythroid cells (BMEs, also known as erythroblasts), in rats treated by gavage with 75 mg/kg of the potent mutagen, 7,12-dimethyl-benz[a]anthracene (DMBA). The frequencies of marker-deficient BMEs were significantly increased in DMBA-treated rats. We identified Pig-a mutations in sorted mutant phenotype BMEs. The spectrum of DMBA-induced Pig-a mutations in erythroid lineage cells was identical to the spectra of mutations previously determined for the Pig-a and for another X-linked reporter gene, hypoxanthine-guanine phosphoribosyltransferase gene, in cells of lymphoid lineage, spleen T-lymphocytes. Our observations lend additional support to the hypothesis that GPI-anchored marker-deficient RBCs are true Pig-a mutants.


Asunto(s)
9,10-Dimetil-1,2-benzantraceno/toxicidad , Médula Ósea/efectos de los fármacos , Eritroblastos/efectos de los fármacos , Proteínas de la Membrana/genética , Mutación , Animales , Médula Ósea/metabolismo , Ratas
13.
Mult Scler Relat Disord ; 36: 101382, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31546224

RESUMEN

BACKGROUND: In natalizumab treated patients several hematopoietic abnormalities including erythroblasts, myeloblasts and neutrophilic precursors in peripheral blood have been described. So far, long term effects of the hematopoietic changes have not been reported. OBJECTIVE: To describe hematopoietic abnormalities in longitudinally monitored MS patients treated with natalizumab. Patients treated with dimethyl fumarate, teriflunomide and fingolimod served as controls. Secondly, the relation between natalizumab serum levels and the occurrence of hematopoietic abnormalities was explored. METHODS: 212 natalizumab treated patients were included, 91 patients with available baseline samples (998 follow-up samples) were compared with patients with dimethyl fumarate (n = 166, 1154 samples), teriflunomide (n = 38, 228 samples) and fingolimod (n = 114, 853 samples). One hundred twenty one patients without baseline samples (1952 follow-up samples) were included in the follow-up group. RESULTS: More than half of all natalizumab treated patients developed hematopoietic abnormalities, almost a quarter had erythroblasts. Natalizumab use was associated with an increased risk of developing abnormalities in comparison to oral treatment, with a corrected hazard ratio of 2.3, 10.0 and 8.1 in comparison to fingolimod, dimethyl fumarate and teriflunomide respectively. No difference in developing abnormalities was observed in relation to natalizumab serum concentrations. None of the patients developed a hematologic malignancy during follow up. CONCLUSION: Hematopoietic abnormalities are common during natalizumab treatment. Given the lack of consequences of this finding during long-term follow-up, it is generally justifiable to refrain from further diagnostic procedures when observing hematopoietic abnormalities in patients using natalizumab.


Asunto(s)
Eritroblastos/efectos de los fármacos , Hematopoyesis/efectos de los fármacos , Factores Inmunológicos/efectos adversos , Esclerosis Múltiple Recurrente-Remitente/sangre , Esclerosis Múltiple Recurrente-Remitente/tratamiento farmacológico , Natalizumab/efectos adversos , Adulto , Femenino , Humanos , Factores Inmunológicos/administración & dosificación , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Natalizumab/administración & dosificación , Estudios Retrospectivos
14.
Mult Scler Relat Disord ; 29: 145-147, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30711880

RESUMEN

Despite having a high rate of occurrence, erythroblast appearance in peripheral blood may not be a recognized adverse effect of natalizumab (NTZ) treatment. Additionally, the time course and cause of erythroblast appearance remain unclear. We report two cases of multiple sclerosis wherein NTZ treatment led to erythroblast appearance in peripheral blood. Erythroblasts appeared after NTZ administration; however, their counts did not increase and the administration of medication was continued. NTZ can inhibit erythroblastic island formation associated with maturing of erythroblast via VLA-4. Clinicians do not need to be afraid; however, careful observation is recommended because some patients may develop anemia.


Asunto(s)
Eritroblastos/efectos de los fármacos , Factores Inmunológicos/efectos adversos , Esclerosis Múltiple/tratamiento farmacológico , Natalizumab/efectos adversos , Neuritis Óptica/tratamiento farmacológico , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
15.
FEBS Open Bio ; 9(2): 291-303, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30761254

RESUMEN

Enucleation is the process whereby the nucleus is extruded from the erythroblast during late stage mammalian erythropoiesis. However, the specific signaling pathways involved in this process remain unclear. To better understand the mechanisms underlying erythroblast enucleation, we investigated erythroblast enucleation using both the spleens of adult mice with phenylhydrazine-induced anemia and mouse fetal livers. Our results indicated that both iron-bound transferrin (holo-Tf) and the small-molecule iron transporter hinokitiol with iron ions (hinokitiol plus iron) promote hemoglobin synthesis and the enucleation of mouse spleen-derived erythroblasts. Although an antitransferrin receptor 1 (TfR1) monoclonal antibody inhibited both enucleation and hemoglobin synthesis promoted by holo-Tf, it inhibited only enucleation, but not hemoglobin synthesis, promoted by hinokitiol plus iron. Furthermore, siRNA against mouse TfR1 were found to suppress the enucleation of mouse fetal liver-derived erythroblasts, and the endocytosis inhibitor MitMAB inhibited enucleation, hemoglobin synthesis, and the internalization of TfR1 promoted by both types of stimuli. Collectively, our results suggest that TfR1, iron ions, and endocytosis play important roles in mouse erythroblast enucleation.


Asunto(s)
Diferenciación Celular , Núcleo Celular/metabolismo , Eritroblastos/citología , Eritroblastos/metabolismo , Receptores de Transferrina/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Eritroblastos/efectos de los fármacos , Citometría de Flujo , Humanos , Células K562 , Ratones , Ratones Endogámicos C57BL , Fenilhidrazinas/farmacología , ARN Interferente Pequeño/farmacología , Receptores de Transferrina/antagonistas & inhibidores , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo , Relación Estructura-Actividad , Compuestos de Trimetilamonio/farmacología
16.
Dev Cell ; 46(5): 581-594.e4, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30122630

RESUMEN

By functioning as an enzyme cofactor, hemoglobin component, and gene regulator, heme is vital for life. One mode of heme-regulated transcription involves amplifying the activity of GATA-1, a key determinant of erythrocyte differentiation. To discover biological consequences of the metal cofactor-transcription factor mechanism, we merged GATA-1/heme-regulated sectors of the proteome and transcriptome. This multi-omic analysis revealed a GATA-1/heme circuit involving hemoglobin subunits, ubiquitination components, and proteins not implicated in erythrocyte biology, including the zinc exporter Slc30a1. Though GATA-1 induced expression of Slc30a1 and the zinc importer Slc39a8, Slc39a8 dominantly increased intracellular zinc, which conferred erythroblast survival. Subsequently, a zinc transporter switch, involving decreased importer and sustained exporter expression, reduced intracellular zinc during terminal differentiation. Downregulating Slc30a1 increased intracellular zinc and, strikingly, accelerated differentiation. This analysis established a conserved paradigm in which a GATA-1/heme circuit controls trace metal transport machinery and trace metal levels as a mechanism governing cellular differentiation.


Asunto(s)
Proteínas Portadoras/metabolismo , Diferenciación Celular/efectos de los fármacos , Eritroblastos/citología , Factor de Transcripción GATA1/metabolismo , Hemo/metabolismo , Zinc/farmacología , Animales , Proteínas Portadoras/genética , Células Cultivadas , Eritroblastos/efectos de los fármacos , Eritroblastos/metabolismo , Eritropoyesis/efectos de los fármacos , Femenino , Factor de Transcripción GATA1/genética , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteoma , Transcriptoma
17.
Chembiochem ; 19(21): 2254-2256, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30136761

RESUMEN

Hepatocellular carcinoma (HCC), a malignancy of the liver, has become the second most lethal cause of cancer death globally. Recently, scientists discovered that a splenic erythroblast-like cell induced by the primary tumor, termed Ter-cell, promoted HCC progression and metastasis. These findings shed light on the inhibition of Ter-cell or artemin that can serve as a new therapeutic target for HCC.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Eritroblastos/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Progresión de la Enfermedad , Descubrimiento de Drogas , Eritroblastos/metabolismo , Eritroblastos/patología , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Terapia Molecular Dirigida , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/prevención & control , Proteínas del Tejido Nervioso/metabolismo
18.
Bioorg Med Chem Lett ; 28(18): 3038-3041, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30097369

RESUMEN

The approval of the erythropoietin (EPO) mimetic peptide drug peginesatide in 2012 was a breakthrough for the treatment of secondary anemia. However, due to severe allergic reactions, peginesatide was recalled a year later. In this study, 12 novel peptides were designed and synthesized by substituting specific amino acids of the monomeric peptide in peginesatide, with the aim of obtaining new EPO mimetic peptides with higher activities and lower side effects than the parent compound. Their cell proliferation activities were evaluated, and the structure-activity relationships were analyzed. Five compounds had equal cell proliferation activity to the control peptide. Among them, one compound showed a higher in vivo activity than the control peptide, with no obvious side effects.


Asunto(s)
Diseño de Fármacos , Eritroblastos/efectos de los fármacos , Péptidos/farmacología , Animales , Peso Corporal/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Inyecciones Subcutáneas , Ratones , Estructura Molecular , Péptidos/administración & dosificación , Péptidos/síntesis química , Relación Estructura-Actividad
19.
Clin Lymphoma Myeloma Leuk ; 18(8): 528-532, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29937400

RESUMEN

BACKGROUND: Recurrent somatic mutations in SF3B1 have been identified in patients with myelodysplastic syndromes (MDS) and are associated with ring sideroblasts (RS) and relatively favorable clinical outcomes. The 2016 World Health Organization classification categorizes patients with ≥ 5% RS and SF3B1 mutation as MDS-RS, in contrast to its prior MDS-RS classification (≥ 15% RS, no genotyping data). Treatment responses in MDS patients with mutated SF3B1 are not well described. PATIENTS AND METHODS: Patients with MDS and known SF3B1 mutational status were identified from MDS Clinical Research Consortium institutions and grouped when possible as 5% to 15% or ≥ 15% RS. Patients with wild-type versus mutated SF3B1 were matched 2:1 to analyze treatment response. RESULTS: Of 471 patients identified, 16% showed SF3B1 mutation. More patients with mutated SF3B1 were lower-risk MDS. We found that 50% were RS-positive compared to 19% of wild-type patients (P < .001). Having the mutation was associated with better overall survival (hazard ratio = 0.48, P = .001) and longer leukemia-free survival (hazard ratio = 0.5, P < .005). Patients with RS and the mutation had the best outcome. Regarding treatment response, 14 (35%) of 40 erythroid-stimulating agent-treated patients with mutation experienced response versus 9 (16%) of 56 wild-type patients (P = .032), with no differences in response to hypomethylating agents or lenalidomide. CONCLUSION: SF3B1 mutations in MDS are commonly associated with RS and show better outcomes, with mutated/positive RS presence being significantly better than isolated RS or presence of mutation or neither. Patients with mutation showed better responses to an erythroid-stimulating agent. A new categorization incorporating SF3B1 mutation status, regardless of RS percentage, shows clinical value.


Asunto(s)
Biomarcadores de Tumor/genética , Eritroblastos/patología , Mutación , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Fosfoproteínas/genética , Factores de Empalme de ARN/genética , Anciano , Antineoplásicos/uso terapéutico , Eritroblastos/efectos de los fármacos , Femenino , Predisposición Genética a la Enfermedad , Fármacos Hematológicos/uso terapéutico , Humanos , Masculino , Síndromes Mielodisplásicos/tratamiento farmacológico , Síndromes Mielodisplásicos/mortalidad , Fenotipo , Supervivencia sin Progresión , Factores de Riesgo , Factores de Tiempo
20.
J Neurochem ; 146(4): 390-402, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29675901

RESUMEN

Acetylcholinesterase (AChE; EC 3.1.1.7) is known to hydrolyze acetylcholine at cholinergic synapses. In mammalian erythrocyte, AChE exists as a dimer (G2 ) and is proposed to play role in erythropoiesis. To reveal the regulation of AChE during differentiation of erythroblast, erythroblast-like cells (TF-1) were induced to differentiate by application of erythropoietin (EPO). The expression of AChE was increased in parallel to the stages of differentiation. Application of EPO in cultured TF-1 cells induced transcriptional activity of ACHE gene, as well as its protein product. This EPO-induced event was in parallel with erythrocytic proteins, for example, α- and ß-globins. The EPO-induced AChE expression was mediated by phosphorylations of Akt and GATA-1; because the application of Akt kinase inhibitor blocked the gene activation. Erythroid transcription factor also known as GATA-1, a downstream transcription factor of EPO signaling, was proposed here to account for regulation of AChE in TF-1 cell. A binding sequence of GATA-1 was identified in ACHE gene promoter, which was further confirmed by chromatin immunoprecipitation (ChIP) assay. Over-expression of GATA-1 in TF-1 cultures induced AChE expression, as well as activity of ACHE promoter tagged with luciferase gene (pAChE-Luc). The deletion of GATA-1 sequence on the ACHE promoter, pAChEΔGATA-1 -Luc, reduced the promoter activity during erythroblastic differentiation. On the contrary, the knock-down of AChE in TF-1 cultures could lead to a reduction in EPO-induced expression of erythrocytic proteins. These findings indicated specific regulation of AChE during maturation of erythroblast, which provided an insight into elucidating possible mechanisms in regulating erythropoiesis.


Asunto(s)
Acetilcolinesterasa/metabolismo , Diferenciación Celular/efectos de los fármacos , Eritroblastos/efectos de los fármacos , Eritroblastos/enzimología , Eritropoyetina/farmacología , Acetilcolinesterasa/genética , Bencenamina, 4,4'-(3-oxo-1,5-pentanodiil)bis(N,N-dimetil-N-2-propenil-), Dibromuro/farmacología , Línea Celular , Inmunoprecipitación de Cromatina , Cromonas/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/metabolismo , Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica , Humanos , Lípidos de la Membrana/metabolismo , Morfolinas/farmacología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA