Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
J Biol Chem ; 297(3): 100988, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34298020

RESUMO

Nemo-like kinase (NLK) is a member of the mitogen-activated protein kinase family of kinases and shares a highly conserved kinase domain with other mitogen-activated protein kinase family members. The activation of NLK contributes to the pathogenesis of Diamond-Blackfan anemia (DBA), reducing c-myb expression and mechanistic target of rapamycin activity, and is therefore a potential therapeutic target. Unlike other anemias, the hematopoietic effects of DBA are largely restricted to the erythroid lineage. Mutations in ribosomal genes induce ribosomal insufficiency and reduced protein translation, dramatically impacting early erythropoiesis in the bone marrow of patients with DBA. We sought to identify compounds that suppress NLK and increases erythropoiesis in ribosomal insufficiency. We report that the active component of ginseng, ginsenoside Rb1, suppresses NLK expression and improves erythropoiesis in in vitro models of DBA. Ginsenoside Rb1-mediated suppression of NLK occurs through the upregulation of miR-208, which binds to the 3'-UTR of NLK mRNA and targets it for degradation. We also compare ginsenoside Rb1-mediated upregulation of miR-208 with metformin-mediated upregulation of miR-26. We conclude that targeting NLK expression through miRNA binding of the unique 3'-UTR is a viable alternative to the challenges of developing small-molecule inhibitors to target the highly conserved kinase domain of this specific kinase.


Assuntos
Anemia de Diamond-Blackfan/patologia , Eritropoese/efeitos dos fármacos , Ginsenosídeos/farmacologia , Panax/química , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Regiões 3' não Traduzidas , Animais , Humanos
2.
Haematologica ; 107(2): 446-456, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33440921

RESUMO

Diamond-Blackfan anemia (DBA) is an inherited bone marrow failure disorder in which pure red blood cell aplasia is associated with physical malformations and a predisposition to cancer. Twentyfive percent of patients with DBA have mutations in a gene encoding ribosomal protein S19 (RPS19). Our previous proof-of-concept studies demonstrated that DBA phenotype could be successfully treated using lentiviral vectors in Rps19-deficient DBA mice. In our present study, we developed a clinically applicable single gene, self-inactivating lentiviral vector, containing the human RPS19 cDNA driven by the human elongation factor 1a short promoter, which can be used for clinical gene therapy development for RPS19-deficient DBA. We examined the efficacy and safety of the vector in a Rps19-deficient DBA mouse model and in human primary RPS19-deficient CD34+ cord blood cells. We observed that transduced Rps19-deficient bone marrow cells could reconstitute mice long-term and rescue the bone marrow failure and severe anemia observed in Rps19-deficient mice, with a low risk of mutagenesis and a highly polyclonal insertion site pattern. More importantly, the vector can also rescue impaired erythroid differentiation in human primary RPS19-deficient CD34+ cord blood hematopoietic stem cells. Collectively, our results demonstrate the efficacy and safety of using a clinically applicable lentiviral vector for the successful treatment of Rps19-deficient DBA in a mouse model and in human primary CD34+ cord blood cells. These findings show that this vector can be used to develop clinical gene therapy for RPS19-deficient DBA patients.


Assuntos
Anemia de Diamond-Blackfan , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/metabolismo , Anemia de Diamond-Blackfan/terapia , Animais , Sangue Fetal/metabolismo , Terapia Genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Mutação , RNA Interferente Pequeno/genética , Proteínas Ribossômicas/genética
3.
Haematologica ; 105(11): 2561-2571, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-33131245

RESUMO

Massive expansion of erythroid progenitor cells is essential for surviving anemic stress. Research towards understanding this critical process, referred to as stress-erythropoiesis, has been hampered due to lack of specific marker-combinations enabling analysis of the distinct stress-progenitor cells capable of providing radioprotection and enhanced red blood cell production. Here we present a method for precise identification and in vivo validation of progenitor cells contributing to both steady-state and stress-erythropoiesis, enabling for the first time in-depth molecular characterization of these cells. Differential expression of surface markers CD150, CD9 and Sca1 defines a hierarchy of splenic stress-progenitors during irradiation-induced stress recovery in mice, and provides high-purity isolation of the functional stress-BFU-Es with a 100-fold improved enrichment compared to state-of-the-art. By transplanting purified stress-progenitors expressing the fluorescent protein Kusabira Orange, we determined their kinetics in vivo and demonstrated that CD150+CD9+Sca1- stress-BFU-Es provide a massive but transient radioprotective erythroid wave, followed by multi-lineage reconstitution from CD150+CD9+Sca1+ multi-potent stem/progenitor cells. Whole genome transcriptional analysis revealed that stress-BFU-Es express gene signatures more associated with erythropoiesis and proliferation compared to steady-state BFU-Es, and are BMP-responsive. Evaluation of chromatin accessibility through ATAC sequencing reveals enhanced and differential accessibility to binding sites of the chromatin-looping transcription factor CTCF in stress-BFU-Es compared to steady-state BFU-Es. Our findings offer molecular insight to the unique capacity of stress-BFU-Es to rapidly form erythroid cells in response to anemia and constitute an important step towards identifying novel erythropoiesis stimulating agents.


Assuntos
Eritropoetina , Transcriptoma , Animais , Epigênese Genética , Células Eritroides , Células Precursoras Eritroides , Eritropoese/genética , Camundongos
4.
Int J Mol Sci ; 21(19)2020 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-33008134

RESUMO

α1-microglobulin (A1M) is a small protein present in vertebrates including humans. It has several physiologically relevant properties, including binding of heme and radicals as well as enzymatic reduction, that are used in the protection of cells and tissue. Research has revealed that A1M can ameliorate heme and ROS-induced injuries in cell cultures, organs, explants and animal models. Recently, it was shown that A1M could reduce hemolysis in vitro, observed with several different types of insults and sources of RBCs. In addition, in a recently published study, it was observed that mice lacking A1M (A1M-KO) developed a macrocytic anemia phenotype. Altogether, this suggests that A1M may have a role in RBC development, stability and turnover. This opens up the possibility of utilizing A1M for therapeutic purposes in pathological conditions involving erythropoietic and hemolytic abnormalities. Here, we provide an overview of A1M and its potential therapeutic effect in the context of the following erythropoietic and hemolytic conditions: Diamond-Blackfan anemia (DBA), 5q-minus myelodysplastic syndrome (5q-MDS), blood transfusions (including storage), intraventricular hemorrhage (IVH), preeclampsia (PE) and atherosclerosis.


Assuntos
alfa-Globulinas/genética , Eritrócitos/metabolismo , Eritropoese/genética , Síndromes Mielodisplásicas/genética , alfa-Globulinas/metabolismo , Animais , Feminino , Heme/genética , Heme/metabolismo , Hemólise/genética , Homeostase , Humanos , Camundongos , Camundongos Knockout , Síndromes Mielodisplásicas/metabolismo , Síndromes Mielodisplásicas/terapia
5.
J Cell Physiol ; 234(9): 16503-16516, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30793301

RESUMO

Cyclin-dependent kinase 8 (CDK8) is a mediator complex-associated transcriptional regulator that acts depending on context and cell type. While primarily under investigation as potential cancer therapeutics, some inhibitors of CDK8-and its paralog CDK19-have been reported to affect the osteoblast lineage and bone formation. This study investigated the effects of two selective CDK8/19 inhibitors on osteoclastogenesis and osteoblasts in vitro, and further evaluated how local treatment with a CDK8/19 inhibitor affects cancellous bone healing in rats. CDK8/19 inhibitors did not alter the proliferation of neither mouse bone marrow-derived macrophages (BMMs) nor primary mouse osteoblasts. Receptor activator of nuclear factor κΒ (NF-κB) ligand (RANKL)-induced osteoclastogenesis from mouse BMMs was suppressed markedly by inhibition of CDK8/19, concomitant with reduced tartrate-resistant acid phosphatase (TRAP) activity and C-terminal telopeptide of type I collagen levels. This was accompanied by downregulation of PU.1, RANK, NF-κB, nuclear factor of activated T-cells 1 (NFATc1), dendritic cell-specific transmembrane protein (DC-STAMP), TRAP, and cathepsin K in RANKL-stimulated BMMs. Downregulating RANK and its downstream signaling in osteoclast precursors enforce CDK8/19 inhibitors as anticatabolic agents to impede excessive osteoclastogenesis. In mouse primary osteoblasts, CDK8/19 inhibition did not affect differentiation but enhanced osteoblast mineralization by promoting alkaline phosphatase activity and downregulating osteopontin, a negative regulator of mineralization. In rat tibiae, a CDK8/19 inhibitor administered locally promoted cancellous bone regeneration. Our data indicate that inhibitors of CDK8/19 have the potential to develop into therapeutics to restrict osteolysis and enhance bone regeneration.

6.
Nature ; 499(7456): 92-6, 2013 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-23748442

RESUMO

Stem cells and progenitors in many lineages undergo self-renewing divisions, but the extracellular and intracellular proteins that regulate this process are largely unknown. Glucocorticoids stimulate red blood cell formation by promoting self-renewal of early burst-forming unit-erythroid (BFU-E) progenitors. Here we show that the RNA-binding protein ZFP36L2 is a transcriptional target of the glucocorticoid receptor (GR) in BFU-Es and is required for BFU-E self-renewal. ZFP36L2 is normally downregulated during erythroid differentiation from the BFU-E stage, but its expression is maintained by all tested GR agonists that stimulate BFU-E self-renewal, and the GR binds to several potential enhancer regions of ZFP36L2. Knockdown of ZFP36L2 in cultured BFU-E cells did not affect the rate of cell division but disrupted glucocorticoid-induced BFU-E self-renewal, and knockdown of ZFP36L2 in transplanted erythroid progenitors prevented expansion of erythroid lineage progenitors normally seen following induction of anaemia by phenylhydrazine treatment. ZFP36L2 preferentially binds to messenger RNAs that are induced or maintained at high expression levels during terminal erythroid differentiation and negatively regulates their expression levels. ZFP36L2 therefore functions as part of a molecular switch promoting BFU-E self-renewal and a subsequent increase in the total numbers of colony-forming unit-erythroid (CFU-E) progenitors and erythroid cells that are generated.


Assuntos
Divisão Celular , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/metabolismo , Tristetraprolina/metabolismo , Animais , Contagem de Células , Divisão Celular/efeitos dos fármacos , Linhagem da Célula , Regulação para Baixo , Eritropoese/genética , Técnicas de Silenciamento de Genes , Glucocorticoides/farmacologia , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/metabolismo , Estresse Fisiológico , Tristetraprolina/deficiência , Tristetraprolina/genética
7.
Genes Dev ; 25(2): 119-24, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21196494

RESUMO

Using RNA-seq technology, we found that the majority of microRNAs (miRNAs) present in CFU-E erythroid progenitors are down-regulated during terminal erythroid differentiation. Of the developmentally down-regulated miRNAs, ectopic overexpression of miR-191 blocks erythroid enucleation but has minor effects on proliferation and differentiation. We identified two erythroid-enriched and developmentally up-regulated genes, Riok3 and Mxi1, as direct targets of miR-191. Knockdown of either Riok3 or Mxi1 blocks enucleation, and either physiological overexpression of miR-191 or knockdown of Riok3 or Mxi1 blocks chromatin condensation. Thus, down-regulation of miR-191 is essential for erythroid chromatin condensation and enucleation by allowing up-regulation of Riok3 and Mxi1.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regulação para Baixo , Eritroblastos/citologia , MicroRNAs/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular , Linhagem Celular , Núcleo Celular/metabolismo , Células Cultivadas , Eritroblastos/metabolismo , Técnicas de Silenciamento de Genes , Camundongos , Proteínas Supressoras de Tumor/genética , Regulação para Cima
8.
Genes Dev ; 25(24): 2573-8, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22155924

RESUMO

Long noncoding RNAs (lncRNAs) are differentially expressed under both normal and pathological conditions, implying that they may play important biological functions. Here we examined the expression of lncRNAs during erythropoiesis and identified an erythroid-specific lncRNA with anti-apoptotic activity. Inhibition of this lncRNA blocks erythroid differentiation and promotes apoptosis. Conversely, ectopic expression of this lncRNA can inhibit apoptosis in mouse erythroid cells. This lncRNA represses expression of Pycard, a proapoptotic gene, explaining in part the inhibition of programmed cell death. These findings reveal a novel layer of regulation of cell differentiation and apoptosis by a lncRNA.


Assuntos
Apoptose , Diferenciação Celular , Células Eritroides/citologia , Eritropoese/genética , Regulação da Expressão Gênica no Desenvolvimento , RNA não Traduzido/metabolismo , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose , Proteínas Adaptadoras de Sinalização CARD , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Células Eritroides/metabolismo , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL
9.
Mol Ther ; 25(8): 1805-1814, 2017 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-28434866

RESUMO

Diamond-Blackfan anemia is a congenital erythroid hypoplasia and is associated with physical malformations and a predisposition to cancer. Twenty-five percent of patients with Diamond-Blackfan anemia have mutations in a gene encoding ribosomal protein S19 (RPS19). Through overexpression of RPS19 using a lentiviral vector with the spleen focus-forming virus promoter, we demonstrated that the Diamond-Blackfan anemia phenotype can be successfully treated in Rps19-deficient mice. In our present study, we assessed the efficacy of a clinically relevant promoter, the human elongation factor 1α short promoter, with or without the locus control region of the ß-globin gene for treatment of RPS19-deficient Diamond-Blackfan anemia. The findings demonstrate that these vectors rescue the proliferation defect and improve erythroid development of transduced RPS19-deficient bone marrow cells. Remarkably, bone marrow failure and severe anemia in Rps19-deficient mice was cured with enforced expression of RPS19 driven by the elongation factor 1α short promoter. We also demonstrate that RPS19-deficient bone marrow cells can be transduced and these cells have the capacity to repopulate bone marrow in long-term reconstituted mice. Our results collectively demonstrate the feasibility to cure RPS19-deficient Diamond-Blackfan anemia using lentiviral vectors with cellular promoters that possess a reduced risk of insertional mutagenesis.


Assuntos
Anemia de Diamond-Blackfan/genética , Medula Óssea/metabolismo , Medula Óssea/patologia , Vetores Genéticos/genética , Lentivirus/genética , Regiões Promotoras Genéticas , Anemia de Diamond-Blackfan/diagnóstico , Anemia de Diamond-Blackfan/terapia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Diferenciação Celular/genética , Proliferação de Células , Modelos Animais de Doenças , Expressão Gênica , Ordem dos Genes , Terapia Genética , Sobrevivência de Enxerto/genética , Hematopoese/genética , Humanos , Camundongos , Fenótipo , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Ribossômicas/genética , Transdução Genética , Transgenes , Integração Viral
10.
Br J Haematol ; 171(4): 517-29, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26305041

RESUMO

Diamond-Blackfan anaemia (DBA) is a rare congenital disease causing severe anaemia and progressive bone marrow failure. The majority of patients carry mutations in ribosomal proteins, which leads to depletion of erythroid progenitors in the bone marrow. As many as 40% of all DBA patients receive glucocorticoids to alleviate their anaemia. However, despite their use in DBA treatment for more than half a century, the therapeutic mechanisms of glucocorticoids remain largely unknown. Therefore we sought to study disease specific effects of glucocorticoid treatment using a ribosomal protein s19 (Rps19) deficient mouse model of DBA. This study determines for the first time that a mouse model of DBA can respond to glucocorticoid treatment, similar to DBA patients. Our results demonstrate that glucocorticoid treatment reduces apoptosis, rescues erythroid progenitor depletion and premature differentiation of erythroid cells. Furthermore, glucocorticoids prevent Trp53 activation in Rps19-deficient cells- in a disease-specific manner. Dissecting the therapeutic mechanisms behind glucocorticoid treatment of DBA provides indispensible insight into DBA pathogenesis. Identifying mechanisms important for DBA treatment also enables development of more disease-specific treatments of DBA.


Assuntos
Anemia de Diamond-Blackfan/tratamento farmacológico , Eritropoese/efeitos dos fármacos , Prednisolona/uso terapêutico , Proteínas Ribossômicas/deficiência , Proteína Supressora de Tumor p53/fisiologia , Adolescente , Anemia de Diamond-Blackfan/sangue , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Inibidor de Quinase Dependente de Ciclina p21/genética , Dexametasona/farmacologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Células Precursoras Eritroides/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Prednisolona/farmacologia , Quimera por Radiação , Proteínas Ribossômicas/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Regulação para Cima/efeitos dos fármacos , Proteína X Associada a bcl-2/biossíntese , Proteína X Associada a bcl-2/genética
11.
Blood ; 120(11): 2225-8, 2012 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-22791294

RESUMO

Diamond-Blackfan anemia (DBA) is a congenital erythroid hypoplasia caused by a functional haploinsufficiency of genes encoding for ribosomal proteins. Recently, a case study reported a patient who became transfusion-independent in response to treatment with the amino acid L-leucine. Therefore, we have validated the therapeutic effect of L-leucine using our recently generated mouse model for RPS19-deficient DBA. Administration of L-leucine significantly improved the anemia in Rps19-deficient mice (19% improvement in hemoglobin concentration; 18% increase in the number of erythrocytes), increased the bone marrow cellularity, and alleviated stress hematopoiesis. Furthermore, the therapeutic response to L-leucine appeared specific for Rps19-deficient hematopoiesis and was associated with down-regulation of p53 activity. Our study supports the rationale for clinical trials of L-leucine as a therapeutic agent for DBA.


Assuntos
Anemia de Diamond-Blackfan/dietoterapia , Suplementos Nutricionais , Modelos Animais de Doenças , Hematínicos/uso terapêutico , Hematopoese , Leucina/uso terapêutico , Regulação para Cima , Anemia de Diamond-Blackfan/sangue , Anemia de Diamond-Blackfan/metabolismo , Anemia de Diamond-Blackfan/patologia , Animais , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Regulação para Baixo , Contagem de Eritrócitos , Técnicas de Silenciamento de Genes , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Hemoglobinas/análise , Camundongos , Camundongos Transgênicos , Terapia de Alvo Molecular , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Proteínas Ribossômicas/antagonistas & inibidores , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
12.
Haematologica ; 99(12): 1792-8, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25216681

RESUMO

Diamond-Blackfan anemia is a congenital erythroid hypoplasia caused by functional haploinsufficiency of genes encoding ribosomal proteins. Mutations involving the ribosomal protein S19 gene are detected in 25% of patients. Enforced expression of ribosomal protein S19 improves the overall proliferative capacity, erythroid colony-forming potential and erythroid differentiation of hematopoietic progenitors from ribosomal protein S19-deficient patients in vitro and in vivo following xenotransplantation. However, studies using animal models are needed to assess the therapeutic efficacy and safety of the viral vectors. In the present study we have validated the therapeutic potential of gene therapy using mouse models of ribosomal protein S19-deficient Diamond-Blackfan anemia. Using lentiviral gene transfer we demonstrated that enforced expression of ribosomal protein S19 cures the anemia and lethal bone marrow failure in recipients transplanted with ribosomal protein S19-deficient cells. Furthermore, gene-corrected ribosomal protein S19-deficient cells showed an increased pan-hematopoietic contribution over time compared to untransduced cells without signs of vector-mediated toxicity. Our study provides a proof of principle for the development of clinical gene therapy to cure ribosomal protein 19-deficient Diamond-Blackfan anemia.


Assuntos
Anemia de Diamond-Blackfan/prevenção & controle , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos/administração & dosagem , Células-Tronco Hematopoéticas/citologia , Hemoglobinúria Paroxística/prevenção & controle , Proteínas Ribossômicas/fisiologia , Anemia Aplástica , Anemia de Diamond-Blackfan/genética , Anemia de Diamond-Blackfan/patologia , Animais , Doenças da Medula Óssea , Transtornos da Insuficiência da Medula Óssea , Células-Tronco Hematopoéticas/metabolismo , Hemoglobinúria Paroxística/genética , Hemoglobinúria Paroxística/patologia , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , RNA Interferente Pequeno/genética , Proteínas Ribossômicas/antagonistas & inibidores
13.
Nat Commun ; 15(1): 8131, 2024 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-39284836

RESUMO

Hematopoietic stem cells (HSCs) react to various stress conditions. However, it is unclear whether and how HSCs respond to severe anemia. Here, we demonstrate that upon induction of acute anemia, HSCs rapidly proliferate and enhance their erythroid differentiation potential. In severe anemia, lipoprotein profiles largely change and the concentration of ApoE increases. In HSCs, transcription levels of lipid metabolism-related genes, such as very low-density lipoprotein receptor (Vldlr), are upregulated. Stimulation of HSCs with ApoE enhances their erythroid potential, whereas HSCs in Apoe knockout mice do not respond to anemia induction. VldlrhighHSCs show higher erythroid potential, which is enhanced after acute anemia induction. VldlrhighHSCs are epigenetically distinct because of their low chromatin accessibility, and more chromatin regions are closed upon acute anemia induction. Chromatin regions closed upon acute anemia induction are mainly binding sites of Erg. Inhibition of Erg enhanced the erythroid differentiation potential of HSCs. Our findings indicate that lipoprotein metabolism plays an important role in HSC regulation under severe anemic conditions.


Assuntos
Anemia , Apolipoproteínas E , Diferenciação Celular , Células-Tronco Hematopoéticas , Lipoproteínas , Animais , Anemia/metabolismo , Anemia/genética , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Apolipoproteínas E/metabolismo , Apolipoproteínas E/genética , Lipoproteínas/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de LDL/metabolismo , Receptores de LDL/genética , Masculino , Cromatina/metabolismo , Eritropoese/genética , Células Eritroides/metabolismo
14.
Blood ; 118(24): 6258-68, 2011 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-21998215

RESUMO

This article reviews the regulation of production of RBCs at several levels. We focus on the regulated expansion of burst-forming unit-erythroid erythroid progenitors by glucocorticoids and other factors that occur during chronic anemia, inflammation, and other conditions of stress. We also highlight the rapid production of RBCs by the coordinated regulation of terminal proliferation and differentiation of committed erythroid colony-forming unit-erythroid progenitors by external signals, such as erythropoietin and adhesion to a fibronectin matrix. We discuss the complex intracellular networks of coordinated gene regulation by transcription factors, chromatin modifiers, and miRNAs that regulate the different stages of erythropoiesis.


Assuntos
Eritrócitos/citologia , Células Precursoras Eritroides/citologia , Eritropoese , Animais , Proliferação de Células , Cromatina/metabolismo , Epigênese Genética , Eritrócitos/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoetina/metabolismo , Humanos , MicroRNAs/metabolismo , Modelos Biológicos , Transcrição Gênica
15.
Blood ; 117(12): 3435-44, 2011 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-21177435

RESUMO

With the aim of finding small molecules that stimulate erythropoiesis earlier than erythropoietin and that enhance erythroid colony-forming unit (CFU-E) production, we studied the mechanism by which glucocorticoids increase CFU-E formation. Using erythroid burst-forming unit (BFU-E) and CFU-E progenitors purified by a new technique, we demonstrate that glucocorticoids stimulate the earliest (BFU-E) progenitors to undergo limited self-renewal, which increases formation of CFU-E cells > 20-fold. Interestingly, glucocorticoids induce expression of genes in BFU-E cells that contain promoter regions highly enriched for hypoxia-induced factor 1α (HIF1α) binding sites. This suggests activation of HIF1α may enhance or replace the effect of glucocorticoids on BFU-E self-renewal. Indeed, HIF1α activation by a prolyl hydroxylase inhibitor (PHI) synergizes with glucocorticoids and enhances production of CFU-Es 170-fold. Because PHIs are able to increase erythroblast production at very low concentrations of glucocorticoids, PHI-induced stimulation of BFU-E progenitors thus represents a conceptually new therapeutic window for treating erythropoietin-resistant anemia.


Assuntos
Proliferação de Células/efeitos dos fármacos , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/fisiologia , Glucocorticoides/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/agonistas , Aminoácidos Dicarboxílicos/farmacologia , Animais , Sítios de Ligação , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Separação Celular , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Embrião de Mamíferos , Inibidores Enzimáticos/farmacologia , Feto/citologia , Citometria de Fluxo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Fígado/citologia , Fígado/embriologia , Camundongos , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética
16.
Blood ; 118(23): 6087-96, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-21989989

RESUMO

Diamond-Blackfan anemia (DBA) is a congenital erythroid hypoplasia caused by a functional haploinsufficiency of genes encoding for ribosomal proteins. Among these genes, ribosomal protein S19 (RPS19) is mutated most frequently. Generation of animal models for diseases like DBA is challenging because the phenotype is highly dependent on the level of RPS19 down-regulation. We report the generation of mouse models for RPS19-deficient DBA using transgenic RNA interference that allows an inducible and graded down-regulation of Rps19. Rps19-deficient mice develop a macrocytic anemia together with leukocytopenia and variable platelet count that with time leads to the exhaustion of hematopoietic stem cells and bone marrow failure. Both RPS19 gene transfer and the loss of p53 rescue the DBA phenotype implying the potential of the models for testing novel therapies. This study demonstrates the feasibility of transgenic RNA interference to generate mouse models for human diseases caused by haploinsufficient expression of a gene.


Assuntos
Anemia de Diamond-Blackfan/genética , Modelos Animais de Doenças , Hemoglobinúria Paroxística/genética , Camundongos Transgênicos , Proteínas Ribossômicas/genética , Anemia Aplástica , Anemia de Diamond-Blackfan/patologia , Anemia de Diamond-Blackfan/fisiopatologia , Anemia Macrocítica/genética , Anemia Macrocítica/patologia , Anemia Macrocítica/fisiopatologia , Animais , Apoptose/fisiologia , Doenças da Medula Óssea , Transtornos da Insuficiência da Medula Óssea , Transplante de Medula Óssea , Divisão Celular/fisiologia , Células Cultivadas , Expressão Gênica/fisiologia , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/fisiologia , Hemoglobinúria Paroxística/patologia , Hemoglobinúria Paroxística/fisiopatologia , Leucopenia/genética , Leucopenia/patologia , Leucopenia/fisiopatologia , Camundongos , Fenótipo , Contagem de Plaquetas , RNA Interferente Pequeno/farmacologia , Proteínas Ribossômicas/deficiência , Proteína Supressora de Tumor p53/genética
17.
Biomolecules ; 13(6)2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-37371508

RESUMO

177Lu-DOTATATE peptide receptor radionuclide therapy (PRRT) is used clinically to treat metastasized or unresectable neuroendocrine tumors (NETs). Although 177Lu-DOTATATE is mostly well tolerated in patients, bone marrow suppression and long-term renal toxicity are still side effects that should be considered. Amino acids are often used to minimize renal radiotoxicity, however, they are associated with nausea and vomiting in patients. α1-microglobulin (A1M) is an antioxidant with heme- and radical-scavenging abilities. A recombinant form (rA1M) has previously been shown to be renoprotective in preclinical models, including in PRRT-induced kidney damage. Here, we further investigated rA1M's renal protective effect in a mouse 177Lu-DOTATATE model in terms of administration route and dosing regimen and as a combined therapy with amino acids (Vamin). Moreover, we investigated the protective effect of rA1M on peripheral blood and bone marrow cells, as well as circulatory biomarkers. Intravenous (i.v.) administration of rA1M reduced albuminuria levels and circulatory levels of the oxidative stress-related protein fibroblast growth factor-21 (FGF-21). Dual injections of rA1M (i.e., at 0 and 24 h post-177Lu-DOTATATE administration) preserved bone marrow cellularity and peripheral blood reticulocytes. Administration of Vamin, alone or in combination with rA1M, did not show any protection of bone marrow cellularity or peripheral reticulocytes. In conclusion, this study suggests that rA1M, administered i.v. for two consecutive days in conjunction with 177Lu-DOTATATE, may reduce hematopoietic and kidney toxicity during PRRT with 177Lu-DOTATATE.


Assuntos
Octreotida , Compostos Organometálicos , Camundongos , Animais , Octreotida/farmacologia , Octreotida/uso terapêutico , Rim/metabolismo , Modelos Animais de Doenças , Aminoácidos/farmacologia , Aminoácidos/uso terapêutico , Compostos Organometálicos/farmacologia , Compostos Organometálicos/uso terapêutico
18.
ScientificWorldJournal ; 2012: 184362, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22619618

RESUMO

Diamond-Blackfan anemia (DBA) is a congenital erythroid hypoplastic anemia, characterized by macrocytic anemia, reticulocytopenia, and severely reduced numbers of erythroid precursors in the bone marrow. For more than fifty years, glucocorticoids have remained the main option for pharmacological treatment of DBA. While continuous glucocorticoid administration increases hemoglobin levels in a majority of DBA patients, it also causes severe side effects. There is therefore a great need for more specific and effective treatments to boost or replace the use of glucocorticoids. Over the years, many alternative therapies have been tried out, but most of them have shown to be ineffective. Here we review previous and current attempts to develop such alternative therapies for DBA. We further discuss how emerging knowledge regarding the pathological mechanism in DBA and the therapeutic mechanism of glucocorticoids treatment may reveal novel drug targets for DBA treatment.


Assuntos
Anemia de Diamond-Blackfan/terapia , Anemia de Diamond-Blackfan/genética , Ensaios Clínicos como Assunto , Terapia Genética , Glucocorticoides/administração & dosagem , Glucocorticoides/efeitos adversos , Humanos , Mutação
19.
Cell Reprogram ; 24(5): 225-227, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36219716

RESUMO

A combination of a MEK/ERK-signaling inhibitor and three chromatin-remodeling molecules enhances generation of platelet-producing megakaryocytes in vitro, possibly through direct reprogramming.


Assuntos
Cromatina , Megacariócitos , Quinases de Proteína Quinase Ativadas por Mitógeno
20.
Exp Hematol ; 105: 50-61, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34757171

RESUMO

Diamond-Blackfan anemia (DBA) is a rare genetic disorder in which patients present a scarcity of erythroid precursors in an otherwise normocellular bone marrow. Most, but not all, patients carry mutations in ribosomal proteins such as RPS19, suggesting that compromised mRNA translation and ribosomal stress are pathogenic mechanisms causing depletion of erythroid precursors. To gain further insight to disease mechanisms in DBA, we performed a custom short hairpin RNA (shRNA) based screen against 750 genes hypothesized to affect DBA pathophysiology. Among the hits were two shRNAs against the erythroid specific heme-regulated eIF2α kinase (HRI), which is a negative regulator of mRNA translation. This study shows that shRNA-mediated HRI silencing or loss of one HRI allele improves expansion of Rps19-deficient erythroid precursors, as well as improves the anemic phenotype in Rps19-deficient animals. We found that Rps19-deficient erythroblasts have elevated levels of unbound intracellular heme, which is normalized by HRI heterozygosity. Additionally, targeting elevated heme levels by treating cells with the heme scavenger alpha-1-microglobulin (A1M), increased proliferation of Rps19-deficient erythroid precursors and decreased heme levels in a disease-specific manner. HRI heterozygosity, but not A1M treatment, also decreased the elevated p53 activity observed in Rps19-deficient cells, indicating that p53 activation is caused by ribosomal stress and aberrant mRNA translation and not heme overload in Rps19-deficiency. Together, these findings suggest that targeting elevated heme levels is a promising new treatment strategy for DBA.


Assuntos
alfa-Globulinas/uso terapêutico , Anemia de Diamond-Blackfan/terapia , Heme/análise , Anemia de Diamond-Blackfan/sangue , Anemia de Diamond-Blackfan/genética , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Deleção de Genes , Inativação Gênica , Terapia Genética , Heme/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Serina-Treonina Quinases/genética , Proteínas Recombinantes/uso terapêutico , Proteínas Ribossômicas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA