Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Oncol ; 13: 1148936, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37035206

RESUMEN

DDX3X is an RNA helicase with many functions in RNA metabolism such as mRNA translation, alternative pre-mRNA splicing and mRNA stability, but also plays a role as a regulator of transcription as well as in the Wnt/beta-catenin- and Nf-κB signaling pathways. The gene encoding DDX3X is located on the X-chromosome, but escapes X-inactivation. Hence females have two active copies and males only one. However, the Y chromosome contains the gene for the male DDX3 homologue, called DDX3Y, which has a very high sequence similarity and functional redundancy with DDX3X, but shows a more restricted protein expression pattern than DDX3X. High throughput sequencing of germinal center (GC)-derived B-cell malignancies such as Burkitt Lymphoma (BL) and Diffuse large B-cell lymphoma (DLBCL) samples showed a high frequency of loss-of-function (LOF) mutations in the DDX3X gene revealing several features that distinguish this gene from others. First, DDX3X mutations occur with high frequency particularly in those GC-derived B-cell lymphomas that also show translocations of the c-MYC proto-oncogene, which occurs in almost all BL and a subset of DLBCL. Second, DDX3X LOF mutations occur almost exclusively in males and is very rarely found in females. Third, mutations in the male homologue DDX3Y have never been found in any type of malignancy. Studies with human primary GC B cells from male donors showed that a loss of DDX3X function helps the initial process of B-cell lymphomagenesis by buffering the proteotoxic stress induced by c-MYC activation. However, full lymphomagenesis requires DDX3 activity since an upregulation of DDX3Y expression is invariably found in GC derived B-cell lymphoma with DDX3X LOF mutation. Other studies with male transgenic mice that lack Ddx3x, but constitutively express activated c-Myc transgenes in B cells and are therefore prone to develop B-cell malignancies, also showed upregulation of the DDX3Y protein expression during the process of lymphomagenesis. Since DDX3Y is not expressed in normal human cells, these data suggest that DDX3Y may represent a new cancer cell specific target to develop adjuvant therapies for male patients with BL and DLBCL and LOF mutations in the DDX3X gene.

2.
Cancer Res ; 82(17): 3172-3186, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-35815807

RESUMEN

The X-linked gene DDX3X encodes an RNA helicase that is mutated at high frequencies in several types of human B-cell lymphoma. Females have two active DDX3X alleles and males carry a DDX3Y homolog on the Y chromosome. We show here that pan-hematopoietic, homozygous deletion of Ddx3x in female mice perturbs erythropoiesis, causing early developmental arrest. However, both hemizygous male and heterozygous female embryos develop normally, suggesting that one Ddx3x allele is sufficient for fetal hematopoietic development in females and that the Ddx3y allele can compensate for the loss of Ddx3x in males. In adult mice, DDX3X deficiency altered hematopoietic progenitors, early lymphoid development, marginal zone and germinal center B cells, and lymphomagenesis in a sex-dependent manner. Loss of both Ddx3x alleles abrogated MYC-driven lymphomagenesis in females, whereas Ddx3x deletion in males did not affect the formation of B-cell lymphoma in both mouse models. Moreover, tumors that appeared in male mice lacking DDX3X showed upregulated expression of DDX3Y, indicating a critical requirement for DDX3 activity for lymphomagenesis. These data reveal sex-specific roles of DDX3X in erythro- and lymphopoiesis as well as in MYC-driven lymphomagenesis. SIGNIFICANCE: The sex-dependent effects of DDX3X deficiency in malignant transformation of B cells and the compensatory role of DDX3Y support inhibition of DDX3 as a treatment strategy for MYC-driven B-cell lymphoma.


Asunto(s)
ARN Helicasas DEAD-box , Genes Ligados a X , Linfoma de Células B , Animales , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Femenino , Homocigoto , Humanos , Linfoma de Células B/genética , Masculino , Ratones , Antígenos de Histocompatibilidad Menor , Eliminación de Secuencia
4.
Front Genet ; 11: 591099, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33193732

RESUMEN

Growth factor independence 1 (GFI1) and the closely related protein GFI1B are small nuclear proteins that act as DNA binding transcriptional repressors. Both recognize the same consensus DNA binding motif via their C-terminal zinc finger domains and regulate the expression of their target genes by recruiting chromatin modifiers such as histone deacetylases (HDACs) and demethylases (LSD1) by using an N-terminal SNAG domain that comprises only 20 amino acids. The only region that is different between both proteins is the region that separates the zinc finger domains and the SNAG domain. Both proteins are co-expressed in hematopoietic stem cells (HSCs) and, to some extent, in multipotent progenitors (MPPs), but expression is specified as soon as early progenitors and show signs of lineage bias. While expression of GFI1 is maintained in lymphoid primed multipotent progenitors (LMPPs) that have the potential to differentiate into both myeloid and lymphoid cells, GFI1B expression is no longer detectable in these cells. By contrast, GFI1 expression is lost in megakaryocyte precursors (MKPs) and in megakaryocyte-erythrocyte progenitors (MEPs), which maintain a high level of GFI1B expression. Consequently, GFI1 drives myeloid and lymphoid differentiation and GFI1B drives the development of megakaryocytes, platelets, and erythrocytes. How such complementary cell type- and lineage-specific functions of GFI1 and GFI1B are maintained is still an unresolved question in particular since they share an almost identical structure and very similar biochemical modes of actions. The cell type-specific accessibility of GFI1/1B binding sites may explain the fact that very similar transcription factors can be responsible for very different transcriptional programming. An additional explanation comes from recent data showing that both proteins may have additional non-transcriptional functions. GFI1 interacts with a number of proteins involved in DNA repair and lack of GFI1 renders HSCs highly susceptible to DNA damage-induced death and restricts their proliferation. In contrast, GFI1B binds to proteins of the beta-catenin/Wnt signaling pathway and lack of GFI1B leads to an expansion of HSCs and MKPs, illustrating the different impact that GFI1 or GFI1B has on HSCs. In addition, GFI1 and GFI1B are required for endothelial cells to become the first blood cells during early murine development and are among those transcription factors needed to convert adult endothelial cells or fibroblasts into HSCs. This role of GFI1 and GFI1B bears high significance for the ongoing effort to generate hematopoietic stem and progenitor cells de novo for the autologous treatment of blood disorders such as leukemia and lymphoma.

5.
Haematologica ; 105(10): 2457-2470, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33054086

RESUMEN

GFI1B-related thrombocytopenia (GFI1B-RT) is a rare bleeding disorder mainly caused by the presence of truncated GFI1B proteins with dominant-negative properties. The disease is characterized by low platelet counts, the presence of abnormal platelets, a megakaryocytic expansion and mild erythroid defects. However, no animal models faithfully reproducing the GFI1B-RT phenotype observed in patients exist. We had previously generated mice with floxed Gfi1b alleles that can be eliminated by Cre recombinase, but those animals developed a much more severe phenotype than GFI1B-RT patients and were of limited interest in assessing the disease. Using CRISPR/Cas9 technology, we have now established three independent mouse lines that carry mutated Gfi1b alleles producing proteins lacking DNA binding zinc fingers and thereby acting in a dominant negative (DN) manner. Mice heterozygous for these Gfi1b-DN alleles show reduced platelet counts and an expansion of megakaryocytes similar to features of human GFI1B-RT but lacking the distinctively large agranular platelets. In addition, Gfi1b-DN mice exhibit an expansion of erythroid precursors indicative of a mildly abnormal erythropoiesis but without noticeable red blood cell defects. When associated with megakaryocyte-specific ablation of the remaining allele, the Gfi1b-DN alleles triggered erythroid-specific deleterious defects. Gfi1b-DN mice also showed a delayed recovery from platelet depletion, indicating a defect in stress thrombopoiesis. However, injecting Gfi1b-DN mice with romiplostim, a thrombopoietin receptor super agonist, increased platelet numbers even beyond normal levels. Thus, our data support a causal link between DN mutations in GFI1B and thrombocytopenia and suggest that patients with GFI1B-RT could be treated successfully with thrombopoietin agonists.


Asunto(s)
Trombocitopenia , Trombopoyesis , Animales , Plaquetas , Eritropoyesis , Humanos , Megacariocitos , Ratones , Mutación , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Trombocitopenia/genética , Trombopoyesis/genética
6.
Nat Commun ; 10(1): 1270, 2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30894540

RESUMEN

Gfi1b is a transcriptional repressor expressed in hematopoietic stem cells (HSCs) and megakaryocytes (MKs). Gfi1b deficiency leads to expansion of both cell types and abrogates the ability of MKs to respond to integrin. Here we show that Gfi1b forms complexes with ß-catenin, its co-factors Pontin52, CHD8, TLE3 and CtBP1 and regulates Wnt/ß-catenin-dependent gene expression. In reporter assays, Gfi1b can activate TCF-dependent transcription and Wnt3a treatment enhances this activation. This requires interaction between Gfi1b and LSD1 and suggests that a tripartite ß-catenin/Gfi1b/LSD1 complex exists, which regulates Wnt/ß-catenin target genes. Consistently, numerous canonical Wnt/ß-catenin target genes, co-occupied by Gfi1b, ß-catenin and LSD1, have their expression deregulated in Gfi1b-deficient cells. When Gfi1b-deficient cells are treated with Wnt3a, their normal cellularity is restored and Gfi1b-deficient MKs regained their ability to spread on integrin substrates. This indicates that Gfi1b controls both the cellularity and functional integrity of HSCs and MKs by regulating Wnt/ß-catenin signaling pathway.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Megacariocitos/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Vía de Señalización Wnt , Proteína Wnt3A/genética , beta Catenina/genética , Oxidorreductasas de Alcohol/genética , Oxidorreductasas de Alcohol/metabolismo , Animales , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Ontología de Genes , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células Madre Hematopoyéticas/citología , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Humanos , Células K562 , Megacariocitos/citología , Ratones , Ratones Noqueados , Anotación de Secuencia Molecular , Cultivo Primario de Células , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Represoras/deficiencia , Tamoxifeno , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo
7.
Leukemia ; 33(1): 110-121, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29925903

RESUMEN

Growth factor independent 1 (Gfi1) controls myeloid differentiation by regulating gene expression and limits the activation of p53 by facilitating its de-methylation at Lysine 372. In human myeloid leukemia, low GFI1 levels correlate with an inferior prognosis. Here, we show that knockdown (KD) of Gfi1 in mice causes a fatal myeloproliferative disease (MPN) that could progress to leukemia after additional mutations. Both KO and KD mice accumulate myeloid cells that show signs of metabolic stress and high levels of reactive oxygen species. However, only KO cells have elevated levels of Lysine 372 methylated p53. This suggests that in contrast to absence of GFI1, KD of GFI1 leads to the accumulation of myeloid cells because sufficient amount of GFI1 is present to impede p53-mediated cell death, leading to a fatal MPN. The combination of myeloid accumulation and the ability to counteract p53 activity under metabolic stress could explain the role of reduced GF1 expression in human myeloid leukemia.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN/fisiología , Leucemia Mieloide/patología , Células Mieloides/patología , Trastornos Mieloproliferativos/patología , Factores de Transcripción/fisiología , Animales , Leucemia Mieloide/etiología , Leucemia Mieloide/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/metabolismo , Trastornos Mieloproliferativos/etiología , Trastornos Mieloproliferativos/metabolismo , Estrés Oxidativo , Canales Catiónicos TRPC/fisiología
8.
Stem Cell Res ; 18: 26-28, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28395797

RESUMEN

Mobilized peripheral blood (MPB) CD34+ cells were cultured to CD41+/CD34+ megakaryoblasts. Cells were sorted to obtain a pure megakaryoblast population that was reprogramed by a hOKSM self-silencing polycistronic vector using lentiviral delivery. The generated induced pluripotent stem cell (iPSC) lines were tested for silencing of the reprogramming construct by flow cytometry. Pluripotency of MML-6838-Cl2 iPSC line was confirmed by expression of associated markers and by in vivo spontaneous differentiation towards the 3 germ layers. The genomic integrity of iPSC line was shown by karyotyping. The MML-6838-Cl2 iPSC is, to our knowledge, the first to be generated from megakaryoblasts.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/citología , Células Progenitoras de Megacariocitos/citología , Animales , Antígenos CD34/metabolismo , Línea Celular , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/trasplante , Cariotipo , Lentivirus/genética , Masculino , Células Progenitoras de Megacariocitos/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Teratoma/metabolismo , Teratoma/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
9.
J Autoimmun ; 79: 74-83, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28237724

RESUMEN

The C1858T single nucleotide polymorphism (SNP) in PTPN22 (protein tyrosine phosphatase nonreceptor 22) leads to the 620 Arg to Trp polymorphism in its encoded human protein LYP. This allelic variant is associated with multiple autoimmune diseases, including type 1 diabetes (T1D), Crohn's disease, rheumatoid arthritis and systemic lupus erythematosus. However, the underlying mechanisms are poorly understood. To study how this polymorphism influences the immune system, we generated a mouse strain with a knock-in of the Trp allele, imitating the human disease-associated variant. We did not find significant difference between the polymorphic and the wild type mice on the proportion of total CD4 T cell, CD8 T cell, NK cell, memory T lymphocyte, macrophage, dendritic cells in both peripheral lymph nodes and spleen. However, macrophages from Trp/Trp mice showed altered morphology and enhanced function, including higher expression of MHCII and B7 molecules and increased phagocytic ability, which further leads to a higher T-cell activation by specific antigen. Our model shows no alteration in immune cell profile by the Trp allele, but brings up macrophages as an important player to consider in explaining the PTPN22 Trp allele effect on autoimmune disease risk.


Asunto(s)
Sustitución de Aminoácidos , Autoinmunidad/genética , Codón , Macrófagos/inmunología , Macrófagos/metabolismo , Mutación , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Alelos , Animales , Antígenos B7/genética , Antígenos B7/inmunología , Citocinas/genética , Citocinas/metabolismo , Expresión Génica , Predisposición Genética a la Enfermedad , Genotipo , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Macrófagos/citología , Masculino , Ratones , Ratones Transgénicos , Fagocitosis/genética , Fagocitosis/inmunología , Polimorfismo de Nucleótido Simple , Proteína Tirosina Fosfatasa no Receptora Tipo 22/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
10.
Haematologica ; 102(3): 484-497, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28082345

RESUMEN

Mutations in GFI1B are associated with inherited bleeding disorders called GFI1B-related thrombocytopenias. We show here that mice with a megakaryocyte-specific Gfi1b deletion exhibit a macrothrombocytopenic phenotype along a megakaryocytic dysplasia reminiscent of GFI1B-related thrombocytopenia. GFI1B deficiency increases megakaryocyte proliferation and affects their ploidy, but also abrogates their responsiveness towards integrin signaling and their ability to spread and reorganize their cytoskeleton. Gfi1b-null megakaryocytes are also unable to form proplatelets, a process independent of integrin signaling. GFI1B-deficient megakaryocytes exhibit aberrant expression of several components of both the actin and microtubule cytoskeleton, with a dramatic reduction of α-tubulin. Inhibition of FAK or ROCK, both important for actin cytoskeleton organization and integrin signaling, only partially restored their response to integrin ligands, but the inhibition of PAK, a regulator of the actin cytoskeleton, completely rescued the responsiveness of Gfi1b-null megakaryocytes to ligands, but not their ability to form proplatelets. We conclude that Gfi1b controls major functions of megakaryocytes such as integrin-dependent cytoskeleton organization, spreading and migration through the regulation of PAK activity whereas the proplatelet formation defect in GFI1B-deficient megakaryocytes is due, at least partially, to an insufficient α-tubulin content.


Asunto(s)
Citoesqueleto/metabolismo , Integrinas/metabolismo , Megacariocitos/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Actinas/química , Actinas/metabolismo , Animales , Diferenciación Celular/genética , Células Cultivadas , Perfilación de la Expresión Génica , Estudios de Asociación Genética , Megacariocitos/efectos de los fármacos , Megacariocitos/patología , Megacariocitos/ultraestructura , Ratones , Ratones Noqueados , Microtúbulos/metabolismo , Fenotipo , Recuento de Plaquetas , Inhibidores de Proteínas Quinasas/farmacología , Multimerización de Proteína , Trombocitopenia/sangre , Trombocitopenia/diagnóstico , Trombocitopenia/genética , Trombocitopenia/metabolismo , Transcriptoma
11.
PLoS One ; 9(5): e96636, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24800817

RESUMEN

Growth factor independence 1b (GFI1B) is a DNA binding repressor of transcription with vital functions in hematopoiesis. Gfi1b-null embryos die at midgestation very likely due to defects in erythro- and megakaryopoiesis. To analyze the full functionality of Gfi1b, we used conditionally deficient mice that harbor floxed Gfi1b alleles and inducible (Mx-Cre, Cre-ERT) or erythroid specific (EpoR-Cre) Cre expressing transgenes. In contrast to the germline knockout, EpoR-Cre mediated erythroid specific ablation of Gfi1b allows full gestation, but causes perinatal lethality with very few mice surviving to adulthood. Both the embryonic deletion of Gfi1b by EpoR-Cre and the deletion in adult mice by Mx-Cre or Cre-ERT leads to reduced numbers of erythroid precursors, perturbed and delayed erythroid maturation, anemia and extramedullary erythropoiesis. Global expression analyses showed that the Hba-x, Hbb-bh1 and Hbb-y embryonic globin genes were upregulated in Gfi1b deficient TER119+ fetal liver cells over the gestation period from day 12.5-17.5 p.c. and an increased level of Hbb-bh1 and Hbb-y embryonic globin gene expression was even maintained in adult Gfi1b deficient mice. While the expression of Bcl11a, a regulator of embryonic globin expression was not affected by Gfi1b deficiency, the expression of Gata1 was reduced and the expression of Sox6, also involved in globin switch, was almost entirely lost when Gfi1b was absent. These findings establish Gfi1b as a regulator of embryonic globin expression and embryonic and adult erythroid maturation.


Asunto(s)
Células Eritroides/metabolismo , Globinas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Animales , Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN , Embrión de Mamíferos/metabolismo , Eritropoyesis , Factor de Transcripción GATA1/metabolismo , Perfilación de la Expresión Génica , Técnicas de Sustitución del Gen , Globinas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Factores de Transcripción SOXD/metabolismo
12.
Autoimmunity ; 46(4): 259-68, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23256897

RESUMEN

Type 1 diabetes (T1D) results from a T cell dependent, autoimmune destruction of insulin producing beta cells in pancreatic islets of Langerhans, which results in insulin deficiency despite attempts at beta cell replacement by the emergence of newly-differentiated beta cells throughout T1D development. The origin of these cells has been difficult to assess as these are rapidly destroyed by the underlying autoimmunity. The identification of islets of Langerhans is typically assessed by either immunochemistry or immunofluorescence using antibodies directed against the different signature hormones and surface markers of various endocrine cells. However, the limited number of markers that can be used simultaneously and the uneven spatial distribution of endocrine cells within islets, limit the use of these histological analyses. To circumvent these caveats, we developed a novel approach using multi-parametric flow cytometry to assess the phenotype and function of pancreatic islet cell populations throughout T1D development. Using such strategy, we show that while beta cells undergo autoimmune destruction, insulin-producing cells arise from trans-differentiated alpha or delta cells, an outcome that was not solely the result of beta cell self-renewal. Moreover, we show that CD4(+)T cell-mediated inflammation correlates with the emergence of this insulin-producing beta cell-like cell.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Fenotipo , Animales , Linfocitos T CD4-Positivos/inmunología , Citocinas/biosíntesis , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Células Secretoras de Glucagón/metabolismo , Inmunofenotipificación , Inflamación/inmunología , Inflamación/metabolismo , Células Secretoras de Insulina/inmunología , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/inmunología , Ratones , Ratones Endogámicos NOD , Células Secretoras de Somatostatina/metabolismo
13.
Epigenetics ; 5(1): 50-60, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-20026906

RESUMEN

DNA methylation patterns are often poorly conserved through cell culturing. To determine the effect of cell immortalization and culture on DNA methylation profiles, we analyzed methylation in the differentially methylated regions (DMR) of five imprinted domains: the intergenic (IG) DMR on chromosome 14q32; potassium voltage-gated channel, KQT-like subfamily, member 1, (KCNQ1); small nuclear ribonucleoprotein polypeptide N (SNRPN), mesoderm specific transcript homolog (MEST); and H19 in lymphoblastoid cell lines (LCLs). In the IG DMR we found an aberrant methylation pattern that was consistent through all the cell lines tested and significantly different from that of noncultured peripheral blood cells. Using a generalized linear mixed model to compare methylation profiles, we show that recently derived LCLs significantly differ from the CEPH LCLs. This implies a gradual cell-culture related deterioration of DNA methylation in the IG DMR with at least two steps that may be identified: loss of methylation at CG sites 1 and 8; and loss of allelic differences in DNA methylation. The IG DMR methylation profile also confirms the high level of clonality of the CEPH LCLs. We conclude that non-transformed primary cells may be less susceptible to epigenetic anomalies and therefore may provide a more accurate reflection of gene expression in vivo.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Metilación de ADN , Epigénesis Genética , Impresión Genómica , Linfocitos/citología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Línea Celular , Cromosomas Humanos Par 14 , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Silenciador del Gen , Variación Genética , Humanos , Persona de Mediana Edad
14.
Mol Cell Biol ; 29(6): 1635-48, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19114559

RESUMEN

During development, human beta-globin locus regulation undergoes two critical switches, the embryonic-to-fetal and fetal-to-adult hemoglobin switches. To define the role of the fetal (A)gamma-globin promoter in switching, human beta-globin-YAC transgenic mice were produced with the (A)gamma-globin promoter replaced by the erythroid porphobilinogen deaminase (PBGD) promoter (PBGD(A)gamma-YAC). Activation of the stage-independent PBGD(A)gamma-globin strikingly stimulated native (G)gamma-globin expression at the fetal and adult stages, identifying a fetal gene pair or bigenic cooperative mechanism. This impaired fetal silencing severely suppressed both delta- and beta-globin expression in PBGD(A)gamma-YAC mice from fetal to neonatal stages and altered kinetics and delayed switching of adult beta-globin. This regulation evokes the two human globin switching patterns in the mouse. Both patterns of DNA demethylation and chromatin immunoprecipitation analysis correlated with gene activation and open chromatin. Locus control region (LCR) interactions detected by chromosome conformation capture revealed distinct spatial fetal and adult LCR bigenic subdomains. Since both intact fetal promoters are critical regulators of fetal silencing at the adult stage, we concluded that fetal genes are controlled as a bigenic subdomain rather than a gene-autonomous mechanism. Our study also provides evidence for LCR complex interaction with spatial fetal or adult bigenic functional subdomains as a niche for transcriptional activation and hemoglobin switching.


Asunto(s)
Cromatina/metabolismo , Hemoglobina Fetal/metabolismo , Globinas beta/metabolismo , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos , Cromatina/genética , Metilación de ADN , Embrión de Mamíferos/metabolismo , Sangre Fetal/metabolismo , Desarrollo Fetal , Hemoglobina Fetal/genética , Regulación del Desarrollo de la Expresión Génica , Humanos , Hidroximetilbilano Sintasa/genética , Hidroximetilbilano Sintasa/metabolismo , Región de Control de Posición , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Globinas beta/genética , gamma-Globinas/genética , gamma-Globinas/metabolismo
15.
Nucleic Acids Res ; 34(18): 5232-7, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17003054

RESUMEN

Beta protein 1 (BP1), a human homeotic transcription factor, is expressed during hematopoeisis in the erythroid lineage. To determine the in vivo role of BP1 in erythropoiesis, we have undertaken two complementary approaches using enforced BP1 expression in both transgenic mice and embryonic stem (ES) cells. Despite repeated attempts, only one adult transgenic BP1 founder mouse among 121 mice was obtained. This mouse presumably survived due to transgene mosaicism because the transgene could not be transmitted. This mouse expressed BP1 and displayed splenomegaly, extramedullary erythropoiesis and severe amyloidosis A in the kidney, a phenotype compatible with thalassemia. Consistently, the presence of BP1 transgene in fetuses was associated with paleness and lethality. In ES cells, BP1 expression in primary differentiation appeared to antagonize adult beta-globin expression. In secondary differentiation, BP1 expression reduced significantly beta-globin gene expression in both primitive and definitive erythroid cells, whereas it impaired only the definitive erythroid cell differentiation. These studies showed that BP1 can negatively modulate adult beta-globin gene expression and definitive erythroid cell differentiation, and suggest that BP1 could play a role in thalassemia.


Asunto(s)
Eritropoyesis , Proteínas de Homeodominio/fisiología , Factores de Transcripción/fisiología , Amiloidosis/patología , Animales , Línea Celular , Células Madre Embrionarias/metabolismo , Células Precursoras Eritroides/metabolismo , Genes Letales , Globinas/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Enfermedades Renales/patología , Ratones , Ratones Transgénicos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
16.
J Biol Chem ; 279(19): 19471-80, 2004 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-15004025

RESUMEN

Characterization of hematopoiesis/erythropoiesis in thalassemias from multipotent primitive cells to mature erythrocytes is of fundamental importance and clinical relevance. We investigated this process in alpha- and beta-globin hemizygous mice, lacking the two adult tandemly organized genes from either the alpha- or beta-globin locus. Although both mice backcrossed on a homogeneous background exhibited similar reduced red blood cell (RBC) survival, beta-globin hemizygous mice had less severe reticulocyte loss and globin chain imbalance, suggesting an apparently milder thalassemia than for alpha-globin hemizygous mice. In contrast, however, beta-globin hemizygous mice displayed a more marked perturbation of hematologic parameters. Quantification of erythroid precursor subpopulations in marrow and spleen of beta-globin hemizygous mice showed more severely impaired maturation from the basophilic to orthochromatophilic erythroblasts and substantial loss of these late precursors probably as a consequence of a greater susceptibility to an excess of free alpha-chain than beta-chain. Hence, only erythroid precursors exhibiting stochastically moderate chain imbalance would escape death and mature to reticulocyte/RBC stage, leading to survival and minimal loss of reticulocytes in the beta-globin hemizygous mice. Furthermore, in response to the ineffective erythropoiesis in beta-globin hemizygous mice, a dynamic compensatory hematopoiesis was observed at earlier differentiation stage as evidenced by a significant increase of erythroid progenitors (erythroid colony-forming units approximately 100-fold) as well as of multipotent primitive cells (day 12 spleen colony-forming units approximately 7-fold). This early compensatory mechanism was less pronounced in alpha-globin hemizygous mice. The expansion of multipotent primitive and potentially stem cell populations, taken together with ineffective erythropoiesis and increased reticulocyte/RBC destruction could confer major cumulative advantage for gene targeting/bone marrow transplantation. Therefore, this study not only corroborated the strong potential effectiveness of transplantation for thalassemic hematopoietic therapy but also demonstrated the existence of a differential regulatory response for alpha- and beta-thalassemia.


Asunto(s)
Eritropoyesis/fisiología , Globinas/genética , Hematopoyesis/fisiología , Animales , Biotinilación , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Diferenciación Celular , Separación Celular , Supervivencia Celular , Ensayo de Unidades Formadoras de Colonias , Cruzamientos Genéticos , Membrana Eritrocítica/metabolismo , Eritrocitos/citología , Citometría de Flujo , Globinas/fisiología , Heterocigoto , Ratones , Ratones Endogámicos C57BL , Reticulocitos/citología , Bazo/citología , Factores de Tiempo , Talasemia alfa/patología , Talasemia beta/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...