Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 268
Filtrar
1.
Blood Cells Mol Dis ; 104: 102792, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37633023

RESUMO

Sickle cell disease (SCD) is the most common ß-hemoglobinopathy caused by various mutations in the adult ß-globin gene resulting in sickle hemoglobin production, chronic hemolytic anemia, pain, and progressive organ damage. The best therapeutic strategies to manage the clinical symptoms of SCD is the induction of fetal hemoglobin (HbF) using chemical agents. At present, among the Food and Drug Administration-approved drugs to treat SCD, hydroxyurea is the only one proven to induce HbF protein synthesis, however, it is not effective in all people. Therefore, we evaluated the ability of the novel Bach1 inhibitor, HPP-D to induce HbF in KU812 cells and primary sickle erythroid progenitors. HPP-D increased HbF and decreased Bach1 protein levels in both cell types. Furthermore, chromatin immunoprecipitation assay showed reduced Bach1 and increased NRF2 binding to the γ-globin promoter antioxidant response elements. We also observed increased levels of the active histone marks H3K4Me1 and H3K4Me3 supporting an open chromatin configuration. In primary sickle erythroid progenitors, HPP-D increased γ-globin transcription and HbF positive cells and reduced sickled erythroid progenitors under hypoxia conditions. Collectively, our data demonstrate that HPP-D induces γ-globin gene transcription through Bach1 inhibition and enhanced NRF2 binding in the γ-globin promoter antioxidant response elements.


Assuntos
Anemia Falciforme , gama-Globinas , Humanos , Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , gama-Globinas/genética , Hemoglobina Falciforme/genética , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/uso terapêutico , Ativação Transcricional/efeitos dos fármacos , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo
2.
Biomed Pharmacother ; 142: 112058, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34426256

RESUMO

Reversal of fetal hemoglobin (HbF) silencing is an attractive therapeutic intervention for ß-thalassemia and sickle cell anemia. The current study proposes the therapeutic of repurposing of cilostazol, an FDA-approved antithrombotic agent, as a promising HbF inducer. Preliminary, we report that cilostazol induced erythroid differentiation and hemoglobinization of human erythroleukemia K562 cells. The erythroid differentiation was accompanied by increased expression of γ-globin mRNA transcripts and HbF production. Cilostazol induced erythroid differentiation and HbF production, without significantly affecting proliferation and viability of hemoglobin producing cells at maximum erythroid inducing concentration. Moreover, we investigated the effect of cilostazol on human ß- and γ-globin transgenes in in vivo ß-YAC transgenic mice, harboring human ß-locus along with ß-LCR. A good in vitro correlation was found with substantial up-regulation in fetal globin mRNA; whereas, the ß-globin gene expression was not significantly changed. F-cells, analysis in the peripheral blood of cilostazol-treated mice, revealed a significant increase in the F-cells population as compared with sham control groups. Together, these findings support the potential of cilostazol as an HbF inducer, which can be evaluated further to develop a new HbF inducer.


Assuntos
Cilostazol/farmacologia , Hemoglobina Fetal/biossíntese , Hemoglobinopatias/tratamento farmacológico , Globinas beta/metabolismo , gama-Globinas/metabolismo , Anemia Falciforme/tratamento farmacológico , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cilostazol/uso terapêutico , Reposicionamento de Medicamentos , Células Eritroides/efeitos dos fármacos , Hemoglobina Fetal/efeitos dos fármacos , Hemoglobina Fetal/genética , Hemoglobinas/efeitos dos fármacos , Hemoglobinas/metabolismo , Humanos , Células K562 , Camundongos Transgênicos , Globinas beta/genética , Talassemia beta/tratamento farmacológico , gama-Globinas/genética
3.
Commun Biol ; 4(1): 677, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34083702

RESUMO

Immortalized erythroid cell lines are expected to be a promising source of ex vivo manufactured red blood cells (RBCs), however the induction of enucleation in these cell lines is inefficient at present. We utilized an imaging-based high-throughput system to identify chemical compounds that trigger enucleation of human erythroid cell lines. Among >3,300 compounds, we identified multiple histone deacetylase inhibitors (HDACi) inducing enucleated cells from the cell line, although an increase in membrane fragility of enucleated cells was observed. Gene expression profiling revealed that HDACi treatment increased the expression of cytoskeletal genes, while an erythroid-specific cell membrane protein, SPTA1, was significantly down-regulated. Restoration of SPTA1 expression using CRISPR-activation partially rescued the fragility of cells and thereby improved the enucleation efficiency. Our observations provide a potential solution for the generation of mature cells from erythroid cell lines, contributing to the future realization of the use of immortalized cell lines for transfusion therapies.


Assuntos
Núcleo Celular/efeitos dos fármacos , Eritrócitos/metabolismo , Células Eritroides/efeitos dos fármacos , Perfilação da Expressão Gênica/métodos , Inibidores de Histona Desacetilases/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Núcleo Celular/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Eritrócitos/citologia , Células Eritroides/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Inibidores de Histona Desacetilases/isolamento & purificação , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
4.
Mol Med Rep ; 24(2)2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34109429

RESUMO

GATA binding protein 1 (GATA­1) is one of the most important hematopoietic transcription factors in the production of blood cells, such as platelets, eosinophils, mast cells and erythrocytes. GATA­1 regulates the participation of microRNA (miRNAs/miRs) in erythroid differentiation under normoxia. However, GATA­1 expression and the regulation of miR­210­3p in the context of erythroid differentiation under hypoxia remain unknown. The present study examined the expression levels of GATA­1 and miR­210­3p in the model of erythroid differentiation in K562 cells under hypoxia, and determined the effects of GATA­1, miR­210­3p and SMAD2 on erythroid differentiation through lentivirus transfection experiments. The present study detected increased GATA­1 expression under hypoxia. Moreover, miR­210­3p was identified as a positive regulator of erythroid differentiation, which was upregulated both during erythroid differentiation and in GATA­1 overexpression experiments under hypoxia. Importantly, in the K562 cell model of erythroid differentiation under hypoxia, miR­210­3p was upregulated in a GATA­1­dependent manner. Using a double luciferase reporter assay, miR­210­3p was identified as a downstream target of GATA­1­mediated regulation of erythropoiesis. Gain­ or loss­of­function analysis of miR­210­3p identified its importance in erythroid differentiation. Furthermore, it was found that SMAD2 may be a downstream target gene for miR­210­3p. Bioinformatics predictions suggested that SMAD2 mediated miR­210­3p­induced regulation of erythroid differentiation. Collectively, the present study provides novel insights into the miRNA regulation of erythroid differentiation.


Assuntos
Hipóxia Celular/genética , Células Eritroides/metabolismo , MicroRNAs/genética , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Eritroides/citologia , Células Eritroides/efeitos dos fármacos , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Técnicas de Silenciamento de Genes , Hemina/farmacologia , Humanos , Células K562 , MicroRNAs/metabolismo , Proteína Smad2/genética , Proteína Smad2/metabolismo , Regulação para Cima/genética
5.
Int J Mol Sci ; 22(7)2021 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-33805426

RESUMO

Myeloproliferative neoplasms (MPNs) are a group of disorders characterized by clonal expansion of abnormal hematopoietic stem cells leading to hyperproliferation of one or more myeloid lineages. The main complications in MPNs are high risk of thrombosis and progression to myelofibrosis and leukemia. MPN patients with high risk scores are treated by hydroxyurea (HU), interferon-α, or ruxolitinib, a tyrosine kinase inhibitor. Polycythemia vera (PV) is an MPN characterized by overproduction of red blood cells (RBCs). ABCG2 is a member of the ATP-binding cassette superfamily transporters known to play a crucial role in multidrug resistance development. Proteome analysis showed higher ABCG2 levels in PV RBCs compared to RBCs from healthy controls and an additional increase of these levels in PV patients treated with HU, suggesting that ABCG2 might play a role in multidrug resistance in MPNs. In this work, we explored the role of ABCG2 in the transport of ruxolitinib and HU using human cell lines, RBCs, and in vitro differentiated erythroid progenitors. Using stopped-flow analysis, we showed that HU is not a substrate for ABCG2. Using transfected K562 cells expressing three different levels of recombinant ABCG2, MPN RBCs, and cultured erythroblasts, we showed that ABCG2 potentiates ruxolitinib-induced cytotoxicity that was blocked by the ABCG2-specific inhibitor KO143 suggesting ruxolitinib intracellular import by ABCG2. In silico modeling analysis identified possible ruxolitinib-binding site locations within the cavities of ABCG2. Our study opens new perspectives in ruxolitinib efficacy research targeting cell types depending on ABCG2 expression and polymorphisms among patients.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Eritrócitos/metabolismo , Proteínas de Neoplasias/metabolismo , Policitemia Vera/tratamento farmacológico , Pirazóis/farmacologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/química , Apoptose/efeitos dos fármacos , Sítios de Ligação , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Simulação por Computador , Dicetopiperazinas/farmacologia , Eritrócitos/efeitos dos fármacos , Células Eritroides/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Hidroxiureia/metabolismo , Hidroxiureia/farmacologia , Interferon-alfa/farmacologia , Células K562 , Transtornos Mieloproliferativos/sangue , Transtornos Mieloproliferativos/tratamento farmacológico , Transtornos Mieloproliferativos/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Nitrilas , Fosfatidilserinas/metabolismo , Policitemia Vera/sangue , Policitemia Vera/patologia , Pirazóis/química , Pirazóis/metabolismo , Pirazóis/farmacocinética , Pirimidinas
6.
Cells ; 10(4)2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33810313

RESUMO

Diamond Blackfan Anemia (DBA) is a congenital macrocytic anemia associated with ribosomal protein haploinsufficiency. Ribosomal dysfunction delays globin synthesis, resulting in excess toxic free heme in erythroid progenitors, early differentiation arrest, and pure red cell aplasia. In this study, DBA induced pluripotent stem cell (iPSC) lines were generated from blood mononuclear cells of DBA patients with inactivating mutations in RPS19 and subjected to hematopoietic differentiation to model disease phenotypes. In vitro differentiated hematopoietic cells were used to investigate whether eltrombopag, an FDA-approved mimetic of thrombopoietin with robust intracellular iron chelating properties, could rescue erythropoiesis in DBA by restricting the labile iron pool (LIP) derived from excessive free heme. DBA iPSCs exhibited RPS19 haploinsufficiency, reduction in the 40S/60S ribosomal subunit ratio and early erythroid differentiation arrest in the absence of eltrombopag, compared to control isogenic iPSCs established by CRISPR/Cas9-mediated correction of the RPS19 point mutation. Notably, differentiation of DBA iPSCs in the presence of eltrombopag markedly improved erythroid maturation. Consistent with a molecular mechanism based on intracellular iron chelation, we observed that deferasirox, a clinically licensed iron chelator able to permeate into cells, also enhanced erythropoiesis in our DBA iPSC model. In contrast, erythroid maturation did not improve substantially in DBA iPSC differentiation cultures supplemented with deferoxamine, a clinically available iron chelator that poorly accesses LIP within cellular compartments. These findings identify eltrombopag as a promising new therapeutic to improve anemia in DBA.


Assuntos
Anemia de Diamond-Blackfan/tratamento farmacológico , Anemia de Diamond-Blackfan/patologia , Benzoatos/uso terapêutico , Diferenciação Celular , Células Eritroides/patologia , Hidrazinas/uso terapêutico , Células-Tronco Pluripotentes Induzidas/patologia , Modelos Biológicos , Pirazóis/uso terapêutico , Anemia de Diamond-Blackfan/genética , Animais , Sequência de Bases , Benzoatos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Células Eritroides/efeitos dos fármacos , Eritropoese , Humanos , Hidrazinas/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Espaço Intracelular/metabolismo , Ferro/metabolismo , Camundongos Endogâmicos NOD , Camundongos SCID , Mutação/genética , Pirazóis/farmacologia
7.
Nat Commun ; 12(1): 1248, 2021 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-33623001

RESUMO

Mutations in human equilibrative nucleoside transporter 3 (ENT3) encoded by SLC29A3 results in anemia and erythroid hypoplasia, suggesting that ENT3 may regulate erythropoiesis. Here, we demonstrate that lysosomal ENT3 transport of taurine-conjugated bile acids (TBA) facilitates TBA chemical chaperone function and alleviates endoplasmic reticulum (ER) stress in expanding mouse hematopoietic stem and progenitor cells (HSPCs). Slc29a3-/- HSPCs accumulate less TBA despite elevated levels of TBA in Slc29a3-/- mouse plasma and have elevated basal ER stress, reactive oxygen species (ROS), and radiation-induced apoptosis. Reintroduction of ENT3 allows for increased accumulation of TBA into HSPCs, which results in TBA-mediated alleviation of ER stress and erythroid apoptosis. Transplanting TBA-preconditioned HSPCs expressing ENT3 into Slc29a3-/- mice increase bone marrow repopulation capacity and erythroid pool size and prevent early mortalities. Together, these findings suggest a putative role for a facilitative lysosomal transporter in the bile acid regulation of ER stress in mouse HSPCs which may have implications in erythroid biology, the treatment of anemia observed in ENT3-mutated human genetic disorders, and nucleoside analog drug therapy.


Assuntos
Ácidos e Sais Biliares/metabolismo , Estresse do Retículo Endoplasmático , Células-Tronco Hematopoéticas/metabolismo , Lisossomos/metabolismo , Animais , Apoptose/efeitos dos fármacos , Ácidos e Sais Biliares/sangue , Transporte Biológico/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Transplante de Células-Tronco Hematopoéticas , Concentração de Íons de Hidrogênio , Lisossomos/efeitos dos fármacos , Metabolômica , Camundongos , Proteínas de Transporte de Nucleosídeos/metabolismo , Taurina/metabolismo , Ácido Tauroquenodesoxicólico/farmacologia
8.
Anal Sci ; 37(2): 229-232, 2021 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-33390466

RESUMO

The electrorotation (ROT) rates of K562 cells accompanied by erythroid differentiation were estimated to identify the differentiation status by using a novel electrorotation device with a microwell arranged on polynomial electrodes. Successive estimations of individual cells were achieved by sequential manipulations which involve trapping of the cell by positive dielectrophoresis (DEP), rotating by ROT, and removing by negative DEP. The ROT rate increased with the differentiation of K562 cells, because the cytoplasm conductivity would increase with an increase of the concentration of iron ions to produce hemoglobin. The ROT rate could be utilized to estimate the stage of cell differentiation without labeling.


Assuntos
Ácido Butírico/farmacologia , Técnicas Eletroquímicas , Células Eritroides/efeitos dos fármacos , Ácido Butírico/química , Diferenciação Celular/efeitos dos fármacos , Condutividade Elétrica , Humanos , Células K562
9.
Reprod Toxicol ; 99: 27-38, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33249232

RESUMO

Rat developmental toxicity including embryolethality and teratogenicity (mainly ventricular septal defects and wavy ribs) were produced by S-53482, an N-phenylimide herbicide that inhibits protoporphyrinogen oxidase (PPO) common to chlorophyll and heme biosynthesis. The sequence of key biological events in the mode of action has been elucidated as follows: inhibition of PPO interferes with normal heme synthesis, which causes loss of blood cells leading to fetal anemia, embryolethality and the development of malformations. In this study we investigated whether the rat is a relevant model for the assessment of the human hazard of the herbicide. To study effects on heme biosynthesis, human erythroleukemia, human cord blood, and rat erythroleukemia cells were treated with the herbicide during red cell differentiation. Protoporphyrin IX, a marker of PPO inhibition, and heme were determined. We investigated whether synchronous maturation of primitive erythropoiesis, which can contribute to massive losses of embryonic blood, occurs in rats. The population of primitive erythroblasts was observed on gestational days 11 through 14. Heme production was suppressed in rat erythroid cells. In contrast, heme reduction was not seen in both human erythroid cells when PPO was inhibited. Rats underwent synchronous maturation in primitive erythropoiesis. Our results combined with epidemiological findings that patients with deficient PPO are not anemic led us to conclude that human erythroblasts are resistant to the herbicide. It is suggested that the rat would be an inappropriate model for assessing the developmental toxicity of S-53482 in humans as rats are specifically sensitive to PPO inhibition by the herbicide.


Assuntos
Benzoxazinas/toxicidade , Células Eritroides/efeitos dos fármacos , Heme/metabolismo , Herbicidas/toxicidade , Ftalimidas/toxicidade , Protoporfirinogênio Oxidase/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Células Eritroides/metabolismo , Feminino , Sangue Fetal , Humanos , Gravidez , Protoporfirinas/metabolismo , Ratos Sprague-Dawley , Especificidade da Espécie
10.
Artigo em Inglês | MEDLINE | ID: mdl-33035680

RESUMO

Hematopoiesis, the complex developmental process that forms blood components and replenishes the blood system, involves multiple intracellular and extracellular mechanisms. We previously demonstrated that lysophosphatidic acid (LPA), a lipid growth factor, has opposing regulatory effects on erythrocyte differentiation through activation of LPA receptors 2 and 3; yet the mechanisms underlying this process remain unclear. In this study, LPA2 is observed that highly expressed in common myeloid progenitors (CMP) in murine myeloid cells, whereas the expression of LPA3 displaces in megakaryocyte-erythroid progenitors (MEP) of later stage of myeloid differentiation. Therefore, we hypothesized that the switching expression of LPA2 and LPA3 determine the hematic homeostasis of mammalian megakaryocytic-erythroid lineage. In vitro colony-forming unit assays of murine progenitors reveal that LPA2 agonist GRI reduces the erythroblast differentiation potential of CMP. In contrast, LPA3 agonist OMPT increases the production of erythrocytes from megakaryocyte-erythrocyte progenitor cells (MEP). In addition, treatment with GRI reduces the erythroid, CMP, and MEP populations in mice, indicating that LPA2 predominantly inhibits myeloid differentiation at an early stage. In contrast, activation of LPA3 increases the production of terminally differentiated erythroid cells through activation of erythropoietic transcriptional factor. We also demonstrate that the LPA3 signaling is essential for restoration of phenylhydrazine (PHZ)-induced acute hemolytic anemia in mice and correlates to erythropoiesis impairment of Hutchinson-Gilford progeria Symptom (HGPS) premature aging expressed K562 model. Our results reveal the distinct roles of LPA2 and LPA3 at different stages of hematopoiesis in vivo, providing potentiated therapeutic strategies of anemia treatment.


Assuntos
Anemia Hemolítica/genética , Células Eritroides/metabolismo , Eritropoese/genética , Células Mieloides/metabolismo , Receptores de Ácidos Lisofosfatídicos/genética , Células-Tronco/metabolismo , Anemia Hemolítica/induzido quimicamente , Anemia Hemolítica/tratamento farmacológico , Anemia Hemolítica/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Linhagem da Célula/genética , Modelos Animais de Doenças , Células Eritroides/citologia , Células Eritroides/efeitos dos fármacos , Eritropoese/efeitos dos fármacos , Regulação da Expressão Gênica , Humanos , Isoquinolinas/farmacologia , Células K562 , Lisofosfolipídeos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Células Mieloides/citologia , Células Mieloides/efeitos dos fármacos , Organotiofosfatos/farmacologia , Fenil-Hidrazinas/administração & dosagem , Ácidos Fosfatídicos/farmacologia , Receptores de Ácidos Lisofosfatídicos/agonistas , Receptores de Ácidos Lisofosfatídicos/metabolismo , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
11.
Med Chem ; 17(2): 121-133, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32407283

RESUMO

BACKGROUND: The use of medicinal agents to augment the fetal hemoglobin (HbF) accretion is an important approach for the treatment of sickle-cell anemia and ß-thalassemia. HbF inducers have the potential to reduce the clinical symptoms and blood transfusion dependence in the patients of ß- hemoglobinopathies. OBJECTIVE: The current study was aimed to examine the erythroid induction potential of newly synthesized thiourea derivatives. METHODS: Thiourea derivatives 1-27 were synthesized by using environmentally friendly methods. Compounds 3, 10 and 22 were found to be new. The structures of synthesized derivatives were deduced by using various spectroscopic techniques. These derivatives were then evaluated for their erythroid induction using the human erythroleukemic K562 cell line, as a model. The benzidine-H2O2 assay was used to evaluate erythroid induction, while HbF expression was studied through immunocytochemistry using the Anti-HbF antibody. Cytotoxicity of compounds 1-27 was also evaluated on mouse fibroblast 3T3 cell line and cancer Hela cell line using MTT assay. RESULT: All the compounds (1-27) have not been reported for their erythroid induction activity previously. Compounds 1, 2, and 3 were found to be the potent erythroid inducing agents with % induction of 45± 6.9, 44± 5.9, and 41± 6.1, at 1.56, 0.78, and 0.78 µM concentrations, respectively, as compared to untreated control (12 ± 1 % induction). Furthermore, compound 1, 2, and 3 significantly induced fetal hemoglobin the expression up to 4.2-fold, 4.06-fold, and 3.52-fold, respectively, as compared to untreated control. Moreover, the compounds 1-4, 6-9, 11, 12, 15, 17, 19, 22, 23, and 25 were found to be non-cytotoxic against the 3T3 cell line. CONCLUSION: This study signifies that the compounds reported here may serve as the starting point for the designing and development of new fetal hemoglobin inducers for the treatment of ß- hemoglobinopathies.


Assuntos
Tioureia/análogos & derivados , Tioureia/síntese química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Células Eritroides/efeitos dos fármacos , Química Verde , Humanos , Camundongos , Estrutura Molecular , Tioureia/química
12.
Biochim Biophys Acta Mol Cell Res ; 1868(3): 118931, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33340546

RESUMO

The CCDC26 gene is considered to encode a functional noncoding RNA associated with acute myeloid leukemia and other cancers. However, investigations into the physiological roles of CCDC26 are rare. Previously, we reported that CCDC26 regulated proliferation and cell death of leukemia cells through KIT, a receptor tyrosine kinase, by using K562 leukemia cells and their derivative CCDC26-knockdown (KD) cells. Here we propose a new role of CCDC26 in the differentiation of erythroid cells. We showed that expression of embryonic (ε- and ζ-) globins was markedly upregulated in CCDC26-KD cells compared with K562 control cells during hemin-induced differentiation. In contrast, expression of fetal-type γ-globin, a major globin expressed in original K562 cells, was decreased. These changes in the expression of globin genes mainly took place at the transcriptional level, with significant suppression of transcription of adult (ß-, δ-) globins in CCDC26-KD cells. Re-introduction of exogenous CCDC26 into the CCDC26-KD cells recovered low-level expression of the embryonal globins. These results suggest CCDC26 has a role in switching transcription of globin genes in the differentiation of erythroid cells. The expression profile of the CCDC26-KD cells and control cells suggests FOG-2, a transcriptional modulator, as a candidate for a mediator of the CCDC26-associated regulation. We showed that both embryonic globins were transcriptionally activated in FOG-2-KD K562 cells. The KIT inhibitor ISCK03 suppressed the production of hemoglobin in K562 cells but did not affect transcription of globin genes. To summarize, FOG-2, but not KIT, is responsible for globin transcriptional regulation by CCDC26.


Assuntos
Proteínas de Ligação a DNA/genética , Células Eritroides/citologia , Globinas/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , RNA Longo não Codificante/genética , Fatores de Transcrição/genética , Diferenciação Celular/efeitos dos fármacos , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Globinas/metabolismo , Hemoglobinas/genética , Hemoglobinas/metabolismo , Humanos , Imidazóis/farmacologia , Células K562 , Sulfonamidas/farmacologia
13.
Blood Cells Mol Dis ; 87: 102528, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33341510

RESUMO

While red blood cells (RBCs) and granulocytes have been more studied, platelets and reticulocytes are not commonly used in paroxysmal nocturnal hemoglobinuria (PNH) flow-cytometry and less is known about susceptibility to complement-mediated destruction and effects of anti-complement therapy on these populations. We performed flow-cytometry of RBCs and granulocytes in 90 PNH patients and of platelets and reticulocytes in a subgroup (N = 36), to unveil perturbations of these populations during PNH disease course before and after anti-complement treatment. We found that platelets and reticulocytes were less sensitive to complement-mediated lysis than RBCs but not as resistant as granulocytes, as shown by mean sensitive fraction (difference in a given PNH population vs. PNH granulocyte clone size). In treated patients, reticulocytes, platelets, RBCs (with differences between type II and III) and granulocytes significantly increased post-treatment, confirming the role of PNH hematopoiesis within the context of anti-complement therapy. Moreover, we found that PNH platelet clone size reflects PNH granulocyte clone size. Finally, we established correlations between sensitive fraction of PNH cell-types and thrombosis. In sum, we applied a flow-cytometry panel for investigation of PNH peripheral blood populations' perturbations before and after eculizumab treatment to explore complement-sensitivity and kinetics of these cells during the disease course.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Células Sanguíneas/efeitos dos fármacos , Inativadores do Complemento/uso terapêutico , Hemoglobinúria Paroxística/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais Humanizados/farmacologia , Células Sanguíneas/citologia , Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Inativadores do Complemento/farmacologia , Eritrócitos/citologia , Eritrócitos/efeitos dos fármacos , Células Eritroides/citologia , Células Eritroides/efeitos dos fármacos , Feminino , Citometria de Fluxo , Granulócitos/citologia , Granulócitos/efeitos dos fármacos , Hemoglobinúria Paroxística/sangue , Humanos , Masculino , Pessoa de Meia-Idade , Reticulócitos/citologia , Reticulócitos/efeitos dos fármacos , Adulto Jovem
14.
Int J Mol Sci ; 21(23)2020 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-33255601

RESUMO

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.


Assuntos
Apoptose/efeitos dos fármacos , Eritropoese/genética , Eritropoetina/genética , Macrófagos/metabolismo , Animais , Diferenciação Celular/genética , Ácido Clodrônico/farmacologia , Eritroblastos/efeitos dos fármacos , Eritroblastos/metabolismo , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Eritropoese/efeitos dos fármacos , Eritropoetina/farmacologia , Lipossomos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos
15.
Stem Cell Reports ; 15(3): 721-734, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32763160

RESUMO

FAM122A is a highly conserved housekeeping gene, but its physiological and pathophysiological roles remain greatly elusive. Based on the fact that FAM122A is highly expressed in human CD71+ early erythroid cells, herein we report that FAM122A is downregulated during erythroid differentiation, while its overexpression significantly inhibits erythrocytic differentiation in primary human hematopoietic progenitor cells and erythroleukemia cells. Mechanistically, FAM122A directly interacts with the C-terminal zinc finger domain of GATA1, a critical transcriptional factor for erythropoiesis, and reduces GATA1 chromatin occupancy on the promoters of its target genes, thus resulting in the decrease of GATA1 transcriptional activity. The public datasets show that FAM122A is abnormally upregulated in patients with ß-thalassemia. Collectively, our results demonstrate that FAM122A plays an inhibitory role in the regulation of erythroid differentiation, and it would be a potentially therapeutic target for GATA1-related dyserythropoiesis or an important regulator for amplifying erythroid cells ex vivo.


Assuntos
Diferenciação Celular , Células Eritroides/citologia , Células Eritroides/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfoproteínas/metabolismo , Antígenos CD34/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , DNA/metabolismo , Regulação para Baixo/genética , Células Eritroides/efeitos dos fármacos , Eritropoetina/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Células K562 , Fosfoproteínas/química , Ligação Proteica/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Dedos de Zinco
16.
Bull Exp Biol Med ; 169(3): 332-337, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32737724

RESUMO

The hemostimulating effects of c-Jun N-terminal kinase (JNK) inhibitor were examined on the mouse model of myelosuppression provoked by 5-fluorouracil. Blockade of JNK during postcytostatic period accelerated recovery of granulomonocytopoiesis and erythropoiesis. It also increased the content of neutrophilic granulocytes and erythroid cells in the hematopoietic tissue and elevated the counts of neutrophils and reticulocytes in the peripheral blood. The development of these phenomena resulted from elevated content and up-regulated functional activity of bone marrow hematopoietic progenitors associated with the direct action of JNK inhibitor on these progenitors and enhanced secretion of hemopoietins by stromal elements of the hematopoiesis-inducing microenvironment.


Assuntos
Citostáticos/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Animais , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Fluoruracila/farmacologia , Granulócitos/efeitos dos fármacos , Granulócitos/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Reticulócitos/efeitos dos fármacos , Reticulócitos/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Exp Cell Res ; 394(2): 112168, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32653411

RESUMO

Pharmacologic induction of fetal hemoglobin (HbF) is an effective strategy for treating ß-hemoglobinopathies like ß-thalassemia and sickle cell anemia by ameliorating disease severity. Hydroxyurea is the only FDA-approved agent that induces HbF, but significant nonresponders and toxicity limit its clinical usefulness. This study relates preclinical investigation of Tenofovir disoproxil fumarate (TDF) as a potential HbF inducing agent, using human erythroleukemia cell line and a ß-YAC mouse model. Erythroid induction of K562 cells was studied by the benzidine/H2O2 reaction, total hemoglobin production was estimated by plasma hemoglobin assay kit, and γ-globin gene expression by RT-qPCR, whereas, fetal hemoglobin production was estimated by flow cytometry and immunofluorescence microscopy. We observed significantly increased γ- globin gene transcription and HbF expression mediated by TDF in K562 cells. Subsequent treatment of ß-YAC transgenic mice with TDF confirmed HbF induction in vivo through an increase in γ-globin gene expression and in the percentage of HbF positive red blood cells. Moreover, TDF showed no cytotoxic effect at HbF inducing concentrations. These data support the potential development of TDF for the treatment of hematological disorders, including ß-thalassemia and sickle cell anemia.


Assuntos
Hemoglobina Fetal/biossíntese , Tenofovir/farmacologia , gama-Globinas/biossíntese , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Eritroides/citologia , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Células K562 , Camundongos Transgênicos , Tenofovir/química , Transcrição Gênica/efeitos dos fármacos , gama-Globinas/genética
18.
FASEB J ; 34(9): 11672-11684, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32667087

RESUMO

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.


Assuntos
Anemia Ferropriva/metabolismo , Eritropoese/efeitos dos fármacos , Deficiências de Ferro , Testosterona/administração & dosagem , Androgênios/administração & dosagem , Anemia Ferropriva/sangue , Anemia Ferropriva/genética , Animais , Eritroblastos/citologia , Eritroblastos/efeitos dos fármacos , Eritroblastos/metabolismo , Contagem de Eritrócitos , Células Eritroides/citologia , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Eritropoese/genética , Feminino , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Expressão Gênica/efeitos dos fármacos , Ferro/fisiologia , Camundongos Endogâmicos C57BL , Receptores da Transferrina/genética , Receptores da Transferrina/metabolismo
19.
Blood ; 136(21): 2457-2468, 2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-32678895

RESUMO

Congenital erythropoietic porphyria (CEP) is an inborn error of heme synthesis resulting from uroporphyrinogen III synthase (UROS) deficiency and the accumulation of nonphysiological porphyrin isomer I metabolites. Clinical features are heterogeneous among patients with CEP but usually combine skin photosensitivity and chronic hemolytic anemia, the severity of which is related to porphyrin overload. Therapeutic options include symptomatic strategies only and are unsatisfactory. One promising approach to treating CEP is to reduce the erythroid production of porphyrins through substrate reduction therapy by inhibiting 5-aminolevulinate synthase 2 (ALAS2), the first and rate-limiting enzyme in the heme biosynthetic pathway. We efficiently reduced porphyrin accumulation after RNA interference-mediated downregulation of ALAS2 in human erythroid cellular models of CEP disease. Taking advantage of the physiological iron-dependent posttranscriptional regulation of ALAS2, we evaluated whether iron chelation with deferiprone could decrease ALAS2 expression and subsequent porphyrin production in vitro and in vivo in a CEP murine model. Treatment with deferiprone of UROS-deficient erythroid cell lines and peripheral blood CD34+-derived erythroid cultures from a patient with CEP inhibited iron-dependent protein ALAS2 and iron-responsive element-binding protein 2 expression and reduced porphyrin production. Furthermore, porphyrin accumulation progressively decreased in red blood cells and urine, and skin photosensitivity in CEP mice treated with deferiprone (1 or 3 mg/mL in drinking water) for 26 weeks was reversed. Hemolysis and iron overload improved upon iron chelation with full correction of anemia in CEP mice treated at the highest dose of deferiprone. Our findings highlight, in both mouse and human models, the therapeutic potential of iron restriction to modulate the phenotype in CEP.


Assuntos
Anemia Hemolítica/tratamento farmacológico , Deferiprona/uso terapêutico , Quelantes de Ferro/uso terapêutico , Sobrecarga de Ferro/tratamento farmacológico , Transtornos de Fotossensibilidade/tratamento farmacológico , Porfiria Eritropoética/tratamento farmacológico , 5-Aminolevulinato Sintetase/antagonistas & inibidores , 5-Aminolevulinato Sintetase/biossíntese , 5-Aminolevulinato Sintetase/genética , Adulto , Anemia Hemolítica/etiologia , Animais , Sistemas CRISPR-Cas , Linhagem Celular , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Feminino , Técnicas de Introdução de Genes , Humanos , Ferro/metabolismo , Sobrecarga de Ferro/etiologia , Leucemia Eritroblástica Aguda/patologia , Camundongos , Células-Tronco de Sangue Periférico/efeitos dos fármacos , Células-Tronco de Sangue Periférico/metabolismo , Transtornos de Fotossensibilidade/etiologia , Porfiria Aguda Intermitente/metabolismo , Porfiria Eritropoética/complicações , Porfirinas/biossíntese , Interferência de RNA , RNA Interferente Pequeno/farmacologia
20.
J Pharmacol Exp Ther ; 374(3): 469-478, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32631869

RESUMO

The complex pathophysiology of sickle cell anemia (SCA) involves intravascular hemolytic processes and recurrent vaso-occlusion, driven by chronic vascular inflammation, which result in the disease's severe clinical complications, including recurrent painful vaso-occlusive episodes. Hydroxyurea, the only drug frequently used for SCA therapy, is a cytostatic agent, although it appears to exert nitric oxide/soluble guanylyl cyclase (sGC) modulating activity. As new drugs that can complement or replace the use of hydroxyurea are sought to further reduce vaso-occlusive episode frequency in SCA, we investigated the effects of the sGC agonists BAY 60-2770 (sGC activator) and BAY 41-2272 (sGC stimulator) in the presence or absence of hydroxyurea on SCA vaso-occlusive mechanisms and cell recruitment both ex vivo and in vivo. These agents significantly reduced stimulated human SCA neutrophil adhesive properties ex vivo in association with the inhibition of surface ß2-integrin activation. A single administration of BAY 60-2770 or BAY 41-2272 decreased tumor necrosis factor cytokine-induced leukocyte recruitment in a mouse model of SCA vaso-occlusion. Importantly, the in vivo actions of both agonists were significantly potentiated by the coadministration of hydroxyurea. Erythroid cell fetal hemoglobin (HbF) elevation is also a major goal for SCA therapy. BAY 41-2272 but not BAY 60-2770 at the concentrations employed significantly induced γ-globin gene transcription in association with HbF production in cultured erythroleukemic cells. In conclusion, sGC agonist drugs could represent a promising approach as therapy for SCA, for use either as stand-alone treatments or in combination with hydroxyurea. SIGNIFICANCE STATEMENT: This preclinical study demonstrates that stimulators and activators of sGC are potent inhibitors of the adhesion and recruitment of leukocytes from humans and in mice with sickle cell anemia (SCA) and may represent a promising approach for diminishing vaso-occlusive episode frequency in SCA. Hydroxyurea, a drug already frequently used for treating SCA, was found to potentiate the beneficial effects of sGC agonists in in vivo studies, implying that these classes of compounds could be used alone or in combination therapy.


Assuntos
Anemia Falciforme/tratamento farmacológico , Anemia Falciforme/metabolismo , Hidroxiureia/farmacocinética , Guanilil Ciclase Solúvel/metabolismo , Animais , Benzoatos/farmacologia , Compostos de Bifenilo/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Células Eritroides/efeitos dos fármacos , Células Eritroides/metabolismo , Hemoglobina Fetal/metabolismo , Humanos , Hidrocarbonetos Fluorados/farmacologia , Células K562 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pirazóis/farmacologia , Piridinas/farmacologia , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/metabolismo , Vasodilatadores/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...