Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Blood ; 137(26): 3591-3594, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33971000

RESUMEN

VEXAS syndrome (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) is a monogenic disease of adulthood caused by somatic mutations in UBA1 in hematopoietic progenitor cells. Patients develop inflammatory and hematologic symptoms. Myeloid-driven autoinflammation and progressive bone marrow failure lead to substantial morbidity and mortality. Effective medical treatments need to be identified. Reports in the current issue of Blood describe novel UBA1 genetic variants, treatment options, and insight into disease pathophysiology. VEXAS syndrome represents a prototype for a new class of diseases.


Asunto(s)
Genes Ligados a X , Enfermedades Genéticas Congénitas , Mutación , Trastornos Mieloproliferativos , Enzimas Activadoras de Ubiquitina/genética , Células Eritroides/enzimología , Enfermedades Genéticas Congénitas/diagnóstico por imagen , Enfermedades Genéticas Congénitas/enzimología , Enfermedades Genéticas Congénitas/genética , Humanos , Masculino , Células Mieloides/enzimología , Trastornos Mieloproliferativos/diagnóstico por imagen , Trastornos Mieloproliferativos/enzimología , Trastornos Mieloproliferativos/genética , Síndrome
3.
Cell Death Dis ; 11(2): 135, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-32075953

RESUMEN

Diamond-Blackfan anemia (DBA) is a rare, inherited bone marrow failure syndrome, characterized by red blood cell aplasia, developmental abnormalities, and enhanced risk of malignancy. However, the underlying pathogenesis of DBA is yet to be understood. Recently, mutations in the gene encoding ribosomal protein (RP) L18 were identified in DBA patients. RPL18 is a crucial component of the ribosomal large subunit but its role in hematopoiesis remains unknown. To genetically model the ribosomal defect identified in DBA, we generated a rpl18 mutant line in zebrafish, using CRISPR/Cas9 system. Molecular characterization of this mutant line demonstrated that Rpl18 deficiency mirrored the erythroid defects of DBA, namely a lack of mature red blood cells. Rpl18 deficiency caused an increase in p53 activation and JAK2-STAT3 activity. Furthermore, we found inhibitors of JAK2 or STAT3 phosphorylation could rescue anemia in rpl18 mutants. Our research provides a new in vivo model of Rpl18 deficiency and suggests involvement of signal pathway of JAK2-STAT3 in the DBA pathogenesis.


Asunto(s)
Anemia de Diamond-Blackfan/enzimología , Células Eritroides/enzimología , Eritropoyesis , Janus Quinasa 2/metabolismo , Proteínas Ribosómicas/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteínas de Pez Cebra/metabolismo , Anemia de Diamond-Blackfan/sangre , Anemia de Diamond-Blackfan/tratamiento farmacológico , Anemia de Diamond-Blackfan/genética , Animales , Animales Modificados Genéticamente , Modelos Animales de Enfermedad , Células Eritroides/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Janus Quinasa 2/antagonistas & inhibidores , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Ribosómicas/genética , Factor de Transcripción STAT3/antagonistas & inhibidores , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/antagonistas & inhibidores
4.
Exp Hematol ; 82: 43-52.e4, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32014431

RESUMEN

Aged hematopoietic stem cells (HSCs) undergo biased lineage priming and differentiation toward production of myeloid cells. A comprehensive understanding of gene regulatory mechanisms causing HSC aging is needed to devise new strategies to sustainably improve immune function in aged individuals. Here, a focused short hairpin RNA screen of epigenetic factors reveals that the histone acetyltransferase Kat6b regulates myeloid cell production from hematopoietic progenitor cells. Within the stem and progenitor cell compartment, Kat6b is highly expressed in long-term (LT)-HSCs and is significantly decreased with aging at the transcript and protein levels. Knockdown of Kat6b in young LT-HSCs causes skewed production of myeloid cells at the expense of erythroid cells both in vitro and in vivo. Transcriptome analysis identifies enrichment of aging and macrophage-associated gene signatures alongside reduced expression of self-renewal and multilineage priming signatures. Together, our work identifies KAT6B as a novel epigenetic regulator of hematopoietic differentiation and a target to improve aged immune function.


Asunto(s)
Envejecimiento/metabolismo , Diferenciación Celular , Células Eritroides/enzimología , Regulación Enzimológica de la Expresión Génica , Histona Acetiltransferasas/biosíntesis , Células Progenitoras Mieloides/enzimología , Envejecimiento/genética , Envejecimiento/patología , Animales , Epigénesis Genética , Células Eritroides/patología , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Histona Acetiltransferasas/genética , Masculino , Ratones , Ratones Transgénicos , Células Progenitoras Mieloides/patología , Transcriptoma
5.
J Clin Invest ; 130(4): 1843-1849, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31895700

RESUMEN

Cancer-related anemia is present in more than 60% of newly diagnosed cancer patients and is associated with substantial morbidity and high medical costs. Drugs that enhance erythropoiesis are urgently required to decrease transfusion rates and improve quality of life. Clinical studies have observed an unexpected improvement in hemoglobin and RBC transfusion-independence in patients with acute myeloid leukemia (AML) treated with the isocitrate dehydrogenase 2 (IDH2) mutant-specific inhibitor enasidenib, leading to improved quality of life without a reduction in AML disease burden. Here, we demonstrate that enasidenib enhanced human erythroid differentiation of hematopoietic progenitors. The phenomenon was not observed with other IDH1/2 inhibitors and occurred in IDH2-deficient CRISPR-engineered progenitors independently of D-2-hydroxyglutarate. The effect of enasidenib on hematopoietic progenitors was mediated by protoporphyrin accumulation, driving heme production and erythroid differentiation in committed CD71+ progenitors rather than hematopoietic stem cells. Our results position enasidenib as a promising therapeutic agent for improvement of anemia and provide the basis for a clinical trial using enasidenib to decrease transfusion dependence in a wide array of clinical contexts.


Asunto(s)
Aminopiridinas/farmacología , Diferenciación Celular/efectos de los fármacos , Células Eritroides/enzimología , Células Madre Hematopoyéticas/enzimología , Isocitrato Deshidrogenasa/antagonistas & inhibidores , Triazinas/farmacología , Células Eritroides/citología , Células Madre Hematopoyéticas/citología , Humanos , Isocitrato Deshidrogenasa/metabolismo , Protoporfirinas/metabolismo
6.
FASEB J ; 32(2): 681-692, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28970249

RESUMEN

PI-PLCß1 is involved in cell proliferation, differentiation, and myelodysplastic syndrome (MDS) pathogenesis. Moreover, the increased activity of PI-PLCß1 reduces the expression of PKC-α, which, in turn, delays the cell proliferation and is linked to erythropoiesis. Lenalidomide is currently used in low-risk patients with MDS and del(5q), where it can suppress the del(5q) clone and restore normal erythropoiesis. In this study, we analyzed the effect of lenalidomide on 16 patients with low-risk del(5q) MDS, as well as del(5q) and non-del(5q) hematopoietic cell lines, mainly focusing on erythropoiesis, cell cycle, and PI-PLCß1/PKC-α signaling. Overall, 11 patients were evaluated clinically, and 10 (90%) had favorable responses; the remaining case had a stable disease. At a molecular level, both responder patients and del(5q) cells showed a specific induction of erythropoiesis, with a reduced γ/ß-globin ratio, an increase in glycophorin A, and a nuclear translocation of PKC-α. Moreover, lenalidomide could induce a selective G0/G1 arrest of the cell cycle in del(5q) cells, slowing down the rate proliferation in those cells. Altogether, our results could not only better explain the role of PI-PLCß1/PKC-α signaling in erythropoiesis but also lead to a better comprehension of the lenalidomide effect on del(5q) MDS and pave the way to innovative, targeted therapies.-Poli, A., Ratti, S., Finelli, C., Mongiorgi, S., Clissa, C., Lonetti, A., Cappellini, A., Catozzi, A., Barraco, M., Suh, P.-G., Manzoli, L., McCubrey, J. A., Cocco, L., Follo, M. Y. Nuclear translocation of PKC-α is associated with cell cycle arrest and erythroid differentiation in myelodysplastic syndromes (MDSs).


Asunto(s)
Diferenciación Celular , Núcleo Celular/enzimología , Células Eritroides/enzimología , Eritropoyesis , Puntos de Control de la Fase G1 del Ciclo Celular , Síndromes Mielodisplásicos/enzimología , Proteína Quinasa C-alfa/metabolismo , Transducción de Señal , Transporte Activo de Núcleo Celular , Anciano , Anciano de 80 o más Años , Línea Celular , Núcleo Celular/genética , Núcleo Celular/patología , Células Eritroides/patología , Femenino , Humanos , Masculino , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Proteína Quinasa C-alfa/genética , Fase de Descanso del Ciclo Celular
7.
Biochem Biophys Res Commun ; 478(3): 1179-84, 2016 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-27544028

RESUMEN

Anemia due to attenuated erythroid terminal differentiation is one of the most common hematological disorders occurring at all stages of life. We previously demonstrated that catalytic subunit α of protein phosphatase 2A (PP2Acα) modulates fetal liver erythropoiesis. However the corresponding PP2A regulatory subunit in this process remains unknown. In this study, we report that chemical inhibition of PP2A activity with okadaic acid impairs hemin-induced erythroid differentiation. Interestingly, B56 family member B56ß is the only regulatory subunit whose expression is induced by both erythropoietin in fetal liver cells and hemin in erythroleukemia K562 cells. Finally, knockdown of B56ß attenuates hemin-induced K562 erythroid differentiation. Collectively, our data identify B56ß as the potential functional regulatory subunit of PP2A in erythroid differentiation, shedding light on new target for precise modulation of PP2A activity for treatment of anemia and related diseases.


Asunto(s)
Diferenciación Celular , Células Eritroides/citología , Células Eritroides/enzimología , Proteínas de la Membrana/metabolismo , Proteína Fosfatasa 2/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Hemina/farmacología , Humanos , Células K562 , Ratones Endogámicos C57BL , Modelos Biológicos , Ácido Ocadaico/farmacología
8.
Blood ; 128(5): 699-709, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27207795

RESUMEN

Human blood cell counts are tightly maintained within narrow physiologic ranges, largely controlled by cytokine-integrated signaling and transcriptional circuits that regulate multilineage hematopoietic specification. Known genetic loci influencing blood cell production account for <10% of platelet and red blood cell variability, and thrombopoietin/cellular myeloproliferative leukemia virus liganding is dispensable for definitive thrombopoiesis, establishing that fundamentally important modifier loci remain unelucidated. In this study, platelet transcriptome sequencing and extended thrombocytosis cohort analyses identified a single loss-of-function mutation (BLVRB(S111L)) causally associated with clonal and nonclonal disorders of enhanced platelet production. BLVRB(S111L) encompassed within the substrate/cofactor [α/ß dinucleotide NAD(P)H] binding fold is a functionally defective redox coupler using flavin and biliverdin (BV) IXß tetrapyrrole(s) and results in exaggerated reactive oxygen species accumulation as a putative metabolic signal leading to differential hematopoietic lineage commitment and enhanced thrombopoiesis. These data define the first physiologically relevant function of BLVRB and implicate its activity and/or heme-regulated BV tetrapyrrole(s) in a unique redox-regulated bioenergetic pathway governing terminal megakaryocytopoiesis; these observations also define a mechanistically restricted drug target retaining potential for enhancing human platelet counts.


Asunto(s)
Hemo/metabolismo , Redes y Vías Metabólicas , Mutación/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/genética , Trombopoyesis/genética , Alelos , Antígenos CD34/metabolismo , Plaquetas/metabolismo , Linaje de la Célula , Estudios de Cohortes , Células Eritroides/citología , Células Eritroides/enzimología , Estudios de Asociación Genética , Hematopoyesis , Humanos , Megacariocitos/citología , Megacariocitos/enzimología , Oxidación-Reducción , Polimorfismo de Nucleótido Simple/genética , Especies Reactivas de Oxígeno/metabolismo , Factores de Riesgo , Análisis de Secuencia de ARN , Trombocitosis/genética
9.
Biochim Biophys Acta ; 1864(5): 441-52, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26854603

RESUMEN

5-Aminolevulinate synthase (ALAS), a pyridoxal 5'-phosphate (PLP)-dependent homodimeric enzyme, catalyzes the initial step of heme biosynthesis in non-plant eukaryotes. The precursor form of the enzyme is translated in the cytosol, and upon mitochondrial import, the N-terminal targeting presequence is proteolytically cleaved to generate mature ALAS. In bone marrow-derived erythroid cells, a mitochondrial- and site-specific endoprotease of yet unknown primary structure, produces a protein shorter than mature erythroid ALAS (ALAS2) found in peripheral blood erythroid cells. This truncated ALAS2 lacks the presequence and the N-terminal sequence (corresponding to ~7 KDa molecular mass) present in ALAS2 from peripheral blood erythroid cells. How the truncation affects the structural topology and catalytic properties of ALAS2 is presently not known. To address this question, we created a recombinant, truncated, murine ALAS2 (ΔmALAS2) devoid of the cleavable N-terminal region and examined its catalytic and biophysical properties. The N-terminal truncation of mALAS2 did not significantly affect the organization of the secondary structure, but a subtle reduction in the rigidity of the tertiary structure was noted. Furthermore, thermal denaturation studies revealed a decrease of 4.3°C in the Tm value of ΔmALAS2, implicating lower thermal stability. While the kcat of ΔmALAS2 is slightly increased over that of the wild-type enzyme, the slowest step in the ΔmALAS2-catalyzed reaction remains dominated by ALA release. Importantly, intrinsic disorder algorithms imply that the N-terminal region of mALAS2 is highly disordered, and thus susceptible to proteolysis. We propose that the N-terminal truncation offers a cell-specific ALAS2 regulatory mechanism without hindering heme synthesis.


Asunto(s)
5-Aminolevulinato Sintetasa/química , 5-Aminolevulinato Sintetasa/genética , Hemo/biosíntesis , Relación Estructura-Actividad , 5-Aminolevulinato Sintetasa/metabolismo , Animales , Células de la Médula Ósea/enzimología , Catálisis , Células Eritroides/enzimología , Hemo/genética , Ratones
10.
Drug Des Devel Ther ; 9: 3153-62, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26124638

RESUMEN

BACKGROUND: The transcription factor GATA-2 is predominantly expressed in hematopoietic stem and progenitor cells and counteracts the erythroid-specific transcription factor GATA-1, to modulate the proliferation and differentiation of hematopoietic cells. During hematopoietic cell differentiation, GATA-2 exhibits dynamic expression patterns, which are regulated by multiple transcription factors. METHODS: Stable LSD1-knockdown cell lines were established by growing murine erythroleukemia (MEL) or mouse embryonic stem cells together with virus particles, in the presence of Polybrene(®) at 4 µg/mL, for 24-48 hours followed by puromycin selection (1 µg/mL) for 2 weeks. Real-time polymerase chain reaction (PCR)-based quantitative chromatin immunoprecipitation (ChIP) analysis was used to test whether the TAL1 transcription factor is bound to 1S promoter in the GATA-2 locus or whether LSD1 colocalizes with TAL1 at the 1S promoter. The sequential ChIP assay was utilized to confirm the role of LSD1 in the regulation of H3K4me2 at the GATA-2 locus during erythroid differentiation. Western blot analysis was employed to detect the protein expression. The alamarBlue(®) assay was used to examine the proliferation of the cells, and the absorbance was monitored at optical density (OD) 570 nm and OD 600 nm. RESULTS: In this study, we showed that LSD1 regulates the expression of GATA-2 during erythroid differentiation. Knockdown of LSD1 results in increased GATA-2 expression and inhibits the differentiation of MEL and embryonic stem cells. Furthermore, we demonstrated that LSD1 binds to the 1S promoter of the GATA-2 locus and suppresses GATA-2 expression, via histone demethylation. CONCLUSION: Our data revealed that LSD1 mediates erythroid differentiation, via epigenetic modification of the GATA-2 locus.


Asunto(s)
Células Madre Embrionarias/enzimología , Células Eritroides/enzimología , Eritropoyesis , Factor de Transcripción GATA2/metabolismo , Histona Demetilasas/metabolismo , Leucemia Eritroblástica Aguda/enzimología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular , Metilación de ADN , Regulación hacia Abajo , Epigénesis Genética , Factor de Transcripción GATA2/genética , Técnicas de Silenciamiento del Gen , Histona Demetilasas/genética , Histonas/metabolismo , Leucemia Eritroblástica Aguda/genética , Ratones , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Tiempo
11.
Genes Dev ; 29(2): 123-8, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25593305

RESUMEN

The methyltransferase activity of the trithorax group (TrxG) protein MLL1 found within its COMPASS (complex associated with SET1)-like complex is allosterically regulated by a four-subunit complex composed of WDR5, RbBP5, Ash2L, and DPY30 (also referred to as WRAD). We report structural evidence showing that in WRAD, a concave surface of the Ash2L SPIa and ryanodine receptor (SPRY) domain binds to a cluster of acidic residues, referred to as the D/E box, in RbBP5. Mutational analysis shows that residues forming the Ash2L/RbBP5 interface are important for heterodimer formation, stimulation of MLL1 catalytic activity, and erythroid cell terminal differentiation. We also demonstrate that a phosphorylation switch on RbBP5 stimulates WRAD complex formation and significantly increases KMT2 (lysine [K] methyltransferase 2) enzyme methylation rates. Overall, our findings provide structural insights into the assembly of the WRAD complex and point to a novel regulatory mechanism controlling the activity of the KMT2/COMPASS family of lysine methyltransferases.


Asunto(s)
Histonas/metabolismo , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Animales , Diferenciación Celular , Línea Celular Tumoral , Cristalización , Análisis Mutacional de ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Activación Enzimática/genética , Células Eritroides/citología , Células Eritroides/enzimología , N-Metiltransferasa de Histona-Lisina/metabolismo , Metilación/efectos de los fármacos , Metiltransferasas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Factores de Transcripción/química , Factores de Transcripción/metabolismo
12.
J Cell Sci ; 127(Pt 12): 2761-70, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24727614

RESUMEN

Protein tyrosine phosphatases (PTPs) are involved in hematopoiesis, but the function of many PTPs is not well characterized in vivo. Here, we have identified Ptpn9a, an ortholog of human PTPN9, as a crucial regulator of erythroid cell development in zebrafish embryos. ptpn9a, but not ptpn9b, was expressed in the posterior lateral plate mesoderm and intermediate cell mass - two primitive hematopoietic sites during zebrafish embryogenesis. Morpholino-mediated knockdown of ptpn9a caused erythrocytes to be depleted by inhibiting erythroid cell maturation without affecting erythroid proliferation and apoptosis. Consistently, both dominant-negative PTPN9 (with mutation C515S) and siRNA against PTPN9 inhibited erythroid differentiation in human K562 cells. Mechanistically, depletion of ptpn9 in zebrafish embryos in vivo or in K562 cells in vitro increased phosphorylated STAT3, and the hyper-phosphorylated STAT3 entrapped and prevented the transcription factors GATA1 and ZBP-89 (also known as ZNF148) from regulating erythroid gene expression. These findings imply that PTPN9 plays an important role in erythropoiesis by disrupting an inhibitory complex of phosphorylated STAT3, GATA1 and ZBP-89, providing new cellular and molecular insights into the role of ptpn9a in developmental hematopoiesis.


Asunto(s)
Células Eritroides/enzimología , Procesamiento Proteico-Postraduccional , Proteínas Tirosina Fosfatasas no Receptoras/fisiología , Factor de Transcripción STAT3/metabolismo , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/fisiología , Pez Cebra/fisiología , Animales , Embrión no Mamífero/citología , Embrión no Mamífero/enzimología , Eritropoyesis , Factor de Transcripción GATA1/metabolismo , Gastrulación , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Células K562 , Fosforilación , Factores de Transcripción/metabolismo , Pez Cebra/embriología
13.
Int J Biochem Cell Biol ; 50: 112-22, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24594363

RESUMEN

To clarify the role of HDACs in erythropoiesis, expression, activity and function of class I (HDAC1, HDAC2, HDAC3) and class IIa (HDAC4, HDAC5) HDACs during in vitro maturation of human erythroblasts were compared. During erythroid maturation, expression of HDAC1, HDAC2 and HDAC3 remained constant and activity and GATA1 association (its partner of the NuRD complex), of HDAC1 increased. By contrast, HDAC4 content drastically decreased and HDAC5 remained constant in content but decreased in activity. In erythroid cells, pull down experiments identified the presence of a novel complex formed by HDAC5, GATA1, EKLF and pERK which was instead undetectable in cells of the megakaryocytic lineage. With erythroid maturation, association among HDAC5, GATA1 and EKLF persisted but levels of pERK sharply decreased. Treatment of erythroleukemic cells with inhibitors of ERK phosphorylation reduced by >90% the total and nuclear content of HDAC5, GATA1 and EKLF, suggesting that ERK phosphorylation is required for the formation of this complex. Based on the function of class IIa HDACs as chaperones of other proteins to the nucleus and the erythroid-specificity of HDAC5 localization, this novel HDAC complex was named nuclear remodeling shuttle erythroid (NuRSERY). Exposure of erythroid cells to the class II-selective HDAC inhibitor (HDACi) APHA9 increased γ/(γ+ß) globin expression ratios (Mai et al., 2007), suggesting that NuRSERY may regulate globin gene expression. In agreement with this hypothesis, exposure of erythroid cells to APHA9 greatly reduced the association among HDAC5, GATA1 and EKLF. Since exposure to APHA9 did not affect survival rates or p21 activation, NuRSERY may represent a novel, possibly less toxic, target for epigenetic therapies of hemoglobinopaties and other disorders.


Asunto(s)
Células Eritroides/metabolismo , Factor de Transcripción GATA1/metabolismo , Histona Desacetilasas/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , eIF-2 Quinasa/metabolismo , Diferenciación Celular/fisiología , Núcleo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Eritroblastos/citología , Eritroblastos/enzimología , Eritroblastos/patología , Células Eritroides/citología , Células Eritroides/enzimología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/enzimología , Células Madre Hematopoyéticas/metabolismo , Humanos , Células K562 , Megacariocitos/citología , Megacariocitos/enzimología , Megacariocitos/metabolismo , Fosforilación
14.
Nature ; 504(7478): 158-62, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24196717

RESUMEN

Newborn infants are highly susceptible to infection. This defect in host defence has generally been ascribed to the immaturity of neonatal immune cells; however, the degree of hyporesponsiveness is highly variable and depends on the stimulation conditions. These discordant responses illustrate the need for a more unified explanation for why immunity is compromised in neonates. Here we show that physiologically enriched CD71(+) erythroid cells in neonatal mice and human cord blood have distinctive immunosuppressive properties. The production of innate immune protective cytokines by adult cells is diminished after transfer to neonatal mice or after co-culture with neonatal splenocytes. Neonatal CD71(+) cells express the enzyme arginase-2, and arginase activity is essential for the immunosuppressive properties of these cells because molecular inhibition of this enzyme or supplementation with L-arginine overrides immunosuppression. In addition, the ablation of CD71(+) cells in neonatal mice, or the decline in number of these cells as postnatal development progresses parallels the loss of suppression, and restored resistance to the perinatal pathogens Listeria monocytogenes and Escherichia coli. However, CD71(+) cell-mediated susceptibility to infection is counterbalanced by CD71(+) cell-mediated protection against aberrant immune cell activation in the intestine, where colonization with commensal microorganisms occurs swiftly after parturition. Conversely, circumventing such colonization by using antimicrobials or gnotobiotic germ-free mice overrides these protective benefits. Thus, CD71(+) cells quench the excessive inflammation induced by abrupt colonization with commensal microorganisms after parturition. This finding challenges the idea that the susceptibility of neonates to infection reflects immune-cell-intrinsic defects and instead highlights processes that are developmentally more essential and inadvertently mitigate innate immune protection. We anticipate that these results will spark renewed investigation into the need for immunosuppression in neonates, as well as improved strategies for augmenting host defence in this vulnerable population.


Asunto(s)
Antígenos CD/metabolismo , Células Eritroides/inmunología , Infecciones por Escherichia coli/inmunología , Tolerancia Inmunológica/inmunología , Listeriosis/inmunología , Receptores de Transferrina/metabolismo , Animales , Animales Recién Nacidos , Arginasa/genética , Arginasa/metabolismo , Susceptibilidad a Enfermedades/inmunología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Células Eritroides/enzimología , Escherichia coli/inmunología , Femenino , Sangre Fetal/citología , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Tolerancia Inmunológica/genética , Listeria monocytogenes/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Necrosis Tumoral alfa/metabolismo
15.
J Clin Invest ; 123(8): 3614-23, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23863711

RESUMEN

The unique sensitivity of early red cell progenitors to iron deprivation, known as the erythroid iron restriction response, serves as a basis for human anemias globally. This response impairs erythropoietin-driven erythropoiesis and underlies erythropoietic repression in iron deficiency anemia. Mechanistically, the erythroid iron restriction response results from inactivation of aconitase enzymes and can be suppressed by providing the aconitase product isocitrate. Recent studies have implicated the erythroid iron restriction response in anemia of chronic disease and inflammation (ACDI), offering new therapeutic avenues for a major clinical problem; however, inflammatory signals may also directly repress erythropoiesis in ACDI. Here, we show that suppression of the erythroid iron restriction response by isocitrate administration corrected anemia and erythropoietic defects in rats with ACDI. In vitro studies demonstrated that erythroid repression by inflammatory signaling is potently modulated by the erythroid iron restriction response in a kinase-dependent pathway involving induction of the erythroid-inhibitory transcription factor PU.1. These results reveal the integration of iron and inflammatory inputs in a therapeutically tractable erythropoietic regulatory circuit.


Asunto(s)
Anemia/tratamiento farmacológico , Células Eritroides/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Deficiencias de Hierro , Isocitratos/farmacología , Aconitato Hidratasa/metabolismo , Anemia/metabolismo , Anemia/patología , Animales , Células Cultivadas , Células Eritroides/enzimología , Femenino , Humanos , Interferón gamma/fisiología , Isocitratos/uso terapéutico , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Ratas Endogámicas Lew , Transducción de Señal , Transactivadores/metabolismo , Activación Transcripcional
16.
PLoS One ; 8(4): e61939, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23613979

RESUMEN

Expression of oncogenic Bcr-Abl inhibits cell differentiation of hematopoietic stem/progenitor cells in chronic myeloid leukemia (CML). Differentiation therapy is considered to be a new strategy for treating this type of leukemia. Aclacinomycin A (ACM) is an antitumor antibiotic. Previous studies have shown that ACM induced erythroid differentiation of CML cells. In this study, we investigate the effect of ACM on the sensitivity of human CML cell line K562 to Bcr-Abl specific inhibitor imatinib (STI571, Gleevec). We first determined the optimal concentration of ACM for erythroid differentiation but not growth inhibition and apoptosis in K562 cells. Then, pretreatment with this optimal concentration of ACM followed by a minimally toxic concentration of imatinib strongly induced growth inhibition and apoptosis compared to that with simultaneous co-treatment, indicating that ACM-induced erythroid differentiation sensitizes K562 cells to imatinib. Sequential treatment with ACM and imatinib induced Bcr-Abl down-regulation, cytochrome c release into the cytosol, and caspase-3 activation, as well as decreased Mcl-1 and Bcl-xL expressions, but did not affect Fas ligand/Fas death receptor and procaspase-8 expressions. ACM/imatinib sequential treatment-induced apoptosis was suppressed by a caspase-9 inhibitor and a caspase-3 inhibitor, indicating that the caspase cascade is involved in this apoptosis. Furthermore, we demonstrated that ACM induced erythroid differentiation through the p38 mitogen-activated protein kinase (MAPK) pathway. The inhibition of erythroid differentiation by p38MAPK inhibitor SB202190, p38MAPK dominant negative mutant or p38MAPK shRNA knockdown, reduced the ACM/imatinib sequential treatment-mediated growth inhibition and apoptosis. These results suggest that differentiated K562 cells induced by ACM-mediated p38MAPK pathway become more sensitive to imatinib and result in down-regulations of Bcr-Abl and anti-apoptotic proteins, growth inhibition and apoptosis. These results provided a potential management by which ACM might have a crucial impact on increasing sensitivity of CML cells to imatinib in the differentiation therapeutic approaches.


Asunto(s)
Aclarubicina/farmacología , Benzamidas/farmacología , Diferenciación Celular/efectos de los fármacos , Células Eritroides/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Piperazinas/farmacología , Pirimidinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Aclarubicina/uso terapéutico , Apoptosis/efectos de los fármacos , Benzamidas/uso terapéutico , Caspasa 3/metabolismo , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Ensayos de Selección de Medicamentos Antitumorales , Activación Enzimática/efectos de los fármacos , Células Eritroides/efectos de los fármacos , Células Eritroides/enzimología , Proteínas de Fusión bcr-abl/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Mesilato de Imatinib , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Pirimidinas/uso terapéutico , Proteína bcl-X/metabolismo , Receptor fas/metabolismo
17.
PLoS One ; 8(3): e56715, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23483889

RESUMEN

Protein arginine methylation is emerging as a pivotal posttranslational modification involved in regulating various cellular processes; however, its role in erythropoiesis is still elusive. Erythropoiesis generates circulating red blood cells which are vital for body activity. Deficiency in erythroid differentiation causes anemia which compromises the quality of life. Despite extensive studies, the molecular events regulating erythropoiesis are not fully understood. This study showed that the increase in protein arginine methyltransferase 1 (PRMT1) levels, via transfection or protein transduction, significantly promoted erythroid differentiation in the bipotent human K562 cell line as well as in human primary hematopoietic progenitor CD34(+) cells. PRMT1 expression enhanced the production of hemoglobin and the erythroid surface marker glycophorin A, and also up-regulated several key transcription factors, GATA1, NF-E2 and EKLF, which are critical for lineage-specific differentiation. The shRNA-mediated knockdown of PRMT1 suppressed erythroid differentiation. The methyltransferase activity-deficient PRMT1G80R mutant failed to stimulate differentiation, indicating the requirement of arginine methylation of target proteins. Our results further showed that a specific isoform of p38 MAPK, p38α, promoted erythroid differentiation, whereas p38ß did not play a role. The stimulation of erythroid differentiation by PRMT1 was diminished in p38α- but not p38ß-knockdown cells. PRMT1 appeared to act upstream of p38α, since expression of p38α still promoted erythroid differentiation in PRMT1-knockdown cells, and expression of PRMT1 enhanced the activation of p38 MAPK. Importantly, we showed for the first time that PRMT1 was associated with p38α in cells by co-immunoprecipitation and that PRMT1 directly methylated p38α in in vitro methylation assays. Taken together, our findings unveil a link between PRMT1 and p38α in regulating the erythroid differentiation program and provide evidence suggesting a novel regulatory mechanism for p38α through arginine methylation.


Asunto(s)
Diferenciación Celular , Células Eritroides/citología , Células Eritroides/enzimología , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Antígenos CD34/metabolismo , Diferenciación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Células Eritroides/efectos de los fármacos , Eritropoyetina/farmacología , Técnicas de Silenciamiento del Gen , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/enzimología , Humanos , Células K562 , Metilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos
18.
J Biochem ; 152(6): 509-19, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22923740

RESUMEN

Human erytholeukemia K562 cells are induced to differentiate along the erythroid lineage by a variety of chemical compounds, including hemin, sodium butyrate and 1-ß-d-arabinofuranosylcytosine. We have investigated the induction of erythroid differentiation of K562 cells by glutamine depletion. When K562 cells were cultured in glutamine-minus medium, the induction of hemoglobin synthesis, accompanied by those of heme-biosynthetic enzymes and erythroid transcriptional factors, was observed. This induction was dependent on the temporally marked decrease of intracellular level of glutathione, followed by the marked activation of p38MAPK and SAPK/JNK, but not ERK. Under glutamine-deficient conditions, the treatment of K562 cells with sodium butyrate resulted in the marked enhancement of the induction of heme biosynthesis. Glutamine depletion also accelerated the expressions of erythroid-related factors including α-globin and heme-biosynthetic enzymes, GATA-1 and NF-E2, in sodium butyrate-induced K562 cells. The transcriptional activity of ß-globin gene promoter-reporter was markedly enhanced by these treatments, indicating that glutamine deficiency in combination with sodium butyrate treatment gives high efficiency of chemical-induced differentiation in the hematopoiesis process.


Asunto(s)
Butiratos/farmacología , Diferenciación Celular/efectos de los fármacos , Células Eritroides/citología , Glutamina/deficiencia , Hematopoyesis , Células Eritroides/enzimología , Células Eritroides/metabolismo , Factor de Transcripción GATA1/metabolismo , Genes Reporteros , Glutatión/metabolismo , Hemo/biosíntesis , Humanos , Células K562 , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , MAP Quinasa Quinasa 4/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Subunidad p45 del Factor de Transcripción NF-E2/metabolismo , Regiones Promotoras Genéticas , Activación Transcripcional , Globinas alfa/metabolismo , Globinas beta/genética , Globinas beta/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
19.
PLoS One ; 6(7): e21358, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21765894

RESUMEN

In mammalian cells, heme can be degraded by heme-oxygenases (HO). Heme-oxygenase 1 (HO-1) is known to be the heme inducible isoform, whereas heme-oxygenase 2 (HO-2) is the constitutive enzyme. Here we investigated the presence of HO during erythroid differentiation in human bone marrow erythroid precursors and K562 cells. HO-1 mRNA and protein expression levels were below limits of detection in K562 cells. Moreover, heme was unable to induce HO-1, at the protein and mRNA profiles. Surprisingly, HO-2 expression was inhibited upon incubation with heme. To evaluate the physiological relevance of these findings, we analyzed HO expression during normal erythropoiesis in human bone marrow. Erythroid precursors were characterized by lack of significant expression of HO-1 and by progressive reduction of HO-2 during differentiation. FLVCR expression, a recently described heme exporter found in erythroid precursors, was also analyzed. Interestingly, the disruption in the HO detoxification system was accompanied by a transient induction of FLVCR. It will be interesting to verify if the inhibition of HO expression, that we found, is preventing a futile cycle of concomitant heme synthesis and catabolism. We believe that a significant feature of erythropoiesis could be the replacement of heme breakdown by heme exportation, as a mechanism to prevent heme toxicity.


Asunto(s)
Células de la Médula Ósea/enzimología , Eritropoyesis , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/metabolismo , Adulto , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Células Eritroides/citología , Células Eritroides/efectos de los fármacos , Células Eritroides/enzimología , Eritropoyesis/efectos de los fármacos , Glicoforinas/metabolismo , Hemo/farmacología , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemoglobinas/biosíntesis , Humanos , Células K562 , Proteínas de Transporte de Membrana/metabolismo , Ratones , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/enzimología , Receptores Virales/metabolismo
20.
Biochemistry ; 50(7): 1194-202, 2011 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-21207956

RESUMEN

Hypoxia-inducible factor 1 (HIF1) is a heterodimeric basic helix-loop-helix transcription factor that regulates many key genes. δ-Aminolevulinate synthase (ALAS) catalyzes the first and rate-limiting reaction in the heme biosynthetic pathway. In this study, we show that hypoxia-induced expression of erythroid-specific ALAS2 is mediated by HIF1 in erythroid cells. Under hypoxic conditions, significantly increased ALAS2 mRNA and protein levels were detected in K562 cells and erythroid induction cultures of CD34+ hematopoietic stem/progenitor cells. Enforced HIF1α expression increased the level of ALAS2 expression, while HIF1α knockdown by RNA interference decreased the level of ALAS2 expression. In silico analysis revealed three potential hypoxia-response elements (HREs) that are located 611, 621, and 741 bp downstream of the ALAS2 gene. The results from reporter gene and mutation analysis suggested that these elements are necessary for a maximal hypoxic response. Chromatin immunoprecipitation and polymerase chain reaction showed that the HREs could be recognized and bound by HIF1α in vivo. These results demonstrate that the upregulation of ALAS2 during hypoxia is directly mediated by HIF1. We hypothesize that HIF1-mediated ALAS2 upregulation promotes erythropoiesis to satisfy the needs of an organism under hypoxic conditions. This may be accomplished via increased heme levels and an interaction between ALAS2 and erythropoietin.


Asunto(s)
5-Aminolevulinato Sintetasa/biosíntesis , Células Eritroides/enzimología , Factor 1 Inducible por Hipoxia/fisiología , 5-Aminolevulinato Sintetasa/genética , Secuencia de Bases , Sitios de Unión/genética , Hipoxia de la Célula/genética , Hipoxia de la Célula/fisiología , Células Cultivadas , Inducción Enzimática/fisiología , Células Eritroides/metabolismo , Eritropoyesis/genética , Eritropoyesis/fisiología , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Células K562 , Modelos Biológicos , Especificidad de Órganos/genética , Unión Proteica , Elementos de Respuesta/genética , Elementos de Respuesta/fisiología , Estudios de Validación como Asunto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA