Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Haematologica ; 108(9): 2316-2330, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-36475518

RESUMEN

Mono-allelic germline disruptions of the transcription factor GATA2 result in a propensity for developing myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML), affecting more than 85% of carriers. How a partial loss of GATA2 functionality enables leukemic transformation years later is unclear. This question has remained unsolved mainly due to the lack of informative models, as Gata2 heterozygote mice do not develop hematologic malignancies. Here we show that two different germline Gata2 mutations (TgErg/Gata2het and TgErg/Gata2L359V) accelerate AML in mice expressing the human hematopoietic stem cell regulator ERG. Analysis of Erg/Gata2het fetal liver and bone marrow-derived hematopoietic cells revealed a distinct pre-leukemic phenotype. This was characterized by enhanced transition from stem to progenitor state, increased proliferation, and a striking mitochondrial phenotype, consisting of highly expressed oxidative-phosphorylation-related gene sets, elevated oxygen consumption rates, and notably, markedly distorted mitochondrial morphology. Importantly, the same mitochondrial gene-expression signature was observed in human AML harboring GATA2 aberrations. Similar to the observations in mice, non-leukemic bone marrows from children with germline GATA2 mutation demonstrated marked mitochondrial abnormalities. Thus, we observed the tumor suppressive effects of GATA2 in two germline Gata2 genetic mouse models. As oncogenic mutations often accumulate with age, GATA2 deficiency-mediated priming of hematopoietic cells for oncogenic transformation may explain the earlier occurrence of MDS/AML in patients with GATA2 germline mutation. The mitochondrial phenotype is a potential therapeutic opportunity for the prevention of leukemic transformation in these patients.


Asunto(s)
Deficiencia GATA2 , Leucemia Mieloide Aguda , Síndromes Mielodisplásicos , Niño , Humanos , Ratones , Animales , Deficiencia GATA2/genética , Síndromes Mielodisplásicos/patología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Médula Ósea/patología , Células Madre Hematopoyéticas/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo
2.
Blood ; 135(25): 2271-2285, 2020 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-32202636

RESUMEN

SETD2, the histone H3 lysine 36 methyltransferase, previously identified by us, plays an important role in the pathogenesis of hematologic malignancies, but its role in myelodysplastic syndromes (MDSs) has been unclear. In this study, low expression of SETD2 correlated with shortened survival in patients with MDS, and the SETD2 levels in CD34+ bone marrow cells of those patients were increased by decitabine. We knocked out Setd2 in NUP98-HOXD13 (NHD13) transgenic mice, which phenocopies human MDS, and found that loss of Setd2 accelerated the transformation of MDS into acute myeloid leukemia (AML). Loss of Setd2 enhanced the ability of NHD13+ hematopoietic stem and progenitor cells (HSPCs) to self-renew, with increased symmetric self-renewal division and decreased differentiation and cell death. The growth of MDS-associated leukemia cells was inhibited though increasing the H3K36me3 level by using epigenetic modifying drugs. Furthermore, Setd2 deficiency upregulated hematopoietic stem cell signaling and downregulated myeloid differentiation pathways in the NHD13+ HSPCs. Our RNA-seq and chromatin immunoprecipitation-seq analysis indicated that S100a9, the S100 calcium-binding protein, is a target gene of Setd2 and that the addition of recombinant S100a9 weakens the effect of Setd2 deficiency in the NHD13+ HSPCs. In contrast, downregulation of S100a9 leads to decreases of its downstream targets, including Ikba and Jnk, which influence the self-renewal and differentiation of HSPCs. Therefore, our results demonstrated that SETD2 deficiency predicts poor prognosis in MDS and promotes the transformation of MDS into AML, which provides a potential therapeutic target for MDS-associated acute leukemia.


Asunto(s)
Anemia Refractaria con Exceso de Blastos/patología , Calgranulina B/fisiología , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/fisiología , Leucemia Mieloide Aguda/etiología , Anemia Refractaria con Exceso de Blastos/genética , Anemia Refractaria con Exceso de Blastos/metabolismo , Animales , Calgranulina B/biosíntesis , Calgranulina B/genética , Transformación Celular Neoplásica , Células Cultivadas , Decitabina/farmacología , Regulación hacia Abajo , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/patología , Código de Histonas/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/biosíntesis , N-Metiltransferasa de Histona-Lisina/genética , Proteínas de Homeodominio/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/mortalidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Síndromes Mielodisplásicos/patología , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Fusión Oncogénica/genética , Pronóstico , Proteínas Recombinantes/uso terapéutico , Factores de Tiempo , Análisis de Matrices Tisulares , Transcriptoma
3.
Proc Natl Acad Sci U S A ; 116(3): 890-899, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30593567

RESUMEN

The AML1-ETO fusion protein, generated by the t(8;21) chromosomal translocation, is causally involved in nearly 20% of acute myeloid leukemia (AML) cases. In leukemic cells, AML1-ETO resides in and functions through a stable protein complex, AML1-ETO-containing transcription factor complex (AETFC), that contains multiple transcription (co)factors. Among these AETFC components, HEB and E2A, two members of the ubiquitously expressed E proteins, directly interact with AML1-ETO, confer new DNA-binding capacity to AETFC, and are essential for leukemogenesis. However, the third E protein, E2-2, is specifically silenced in AML1-ETO-expressing leukemic cells, suggesting E2-2 as a negative factor of leukemogenesis. Indeed, ectopic expression of E2-2 selectively inhibits the growth of AML1-ETO-expressing leukemic cells, and this inhibition requires the bHLH DNA-binding domain. RNA-seq and ChIP-seq analyses reveal that, despite some overlap, the three E proteins differentially regulate many target genes. In particular, studies show that E2-2 both redistributes AETFC to, and activates, some genes associated with dendritic cell differentiation and represses MYC target genes. In AML patients, the expression of E2-2 is relatively lower in the t(8;21) subtype, and an E2-2 target gene, THPO, is identified as a potential predictor of relapse. In a mouse model of human t(8;21) leukemia, E2-2 suppression accelerates leukemogenesis. Taken together, these results reveal that, in contrast to HEB and E2A, which facilitate AML1-ETO-mediated leukemogenesis, E2-2 compromises the function of AETFC and negatively regulates leukemogenesis. The three E proteins thus define a heterogeneity of AETFC, which improves our understanding of the precise mechanism of leukemogenesis and assists development of diagnostic/therapeutic strategies.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Leucemia Mieloide Aguda/etiología , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Humanos , Leucemia Mieloide Aguda/metabolismo , Recurrencia
4.
J Immunol ; 199(5): 1817-1826, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28739877

RESUMEN

PALLD is an actin cross-linker supporting cellular mechanical tension. However, its involvement in the regulation of phagocytosis, a cellular activity essential for innate immunity and physiological tissue turnover, is unclear. We report that PALLD is highly induced along with all-trans-retinoic acid-induced maturation of myeloid leukemia cells, to promote Ig- or complement-opsonized phagocytosis. PALLD mechanistically facilitates phagocytic receptor clustering by regulating actin polymerization and c-Src dynamic activation during particle binding and early phagosome formation. PALLD is also required at the nascent phagosome to recruit phosphatase oculocerebrorenal syndrome of Lowe, which regulates phosphatidylinositol-4,5-bisphosphate hydrolysis and actin depolymerization to complete phagosome closure. Collectively, our results show a new function for PALLD as a crucial regulator of the early phase of phagocytosis by elaborating dynamic actin polymerization and depolymerization.


Asunto(s)
Actinas/metabolismo , Proteínas del Citoesqueleto/metabolismo , Células Dendríticas/inmunología , Leucemia Mieloide Aguda/inmunología , Células Madre Neoplásicas/fisiología , Síndrome Oculocerebrorrenal/inmunología , Fagocitosis , Fosfoproteínas/metabolismo , Animales , Diferenciación Celular , Línea Celular Tumoral , Autorrenovación de las Células , Proteínas del Citoesqueleto/genética , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Fagosomas/metabolismo , Fosfoproteínas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Polimerizacion , Agregación de Receptores , Tretinoina/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(16): 6459-64, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23553835

RESUMEN

Retinoic acid inducible gene I (RIG-I) senses viral RNAs and triggers innate antiviral responses through induction of type I IFNs and inflammatory cytokines. However, whether RIG-I interacts with host cellular RNA remains undetermined. Here we report that Rig-I interacts with multiple cellular mRNAs, especially Nf-κb1. Rig-I is required for NF-κB activity via regulating Nf-κb1 expression at posttranscriptional levels. It interacts with the multiple binding sites within 3'-UTR of Nf-κb1 mRNA. Further analyses reveal that three distinct tandem motifs enriched in the 3'-UTR fragments can be recognized by Rig-I. The 3'-UTR binding with Rig-I plays a critical role in normal translation of Nf-κb1 by recruiting the ribosomal proteins [ribosomal protein L13 (Rpl13) and Rpl8] and rRNAs (18S and 28S). Down-regulation of Rig-I or Rpl13 significantly reduces Nf-κb1 and 3'-UTR-mediated luciferase expression levels. These findings indicate that Rig-I functions as a positive regulator for NF-κB signaling and is involved in multiple biological processes in addition to host antivirus immunity.


Asunto(s)
ARN Helicasas DEAD-box/metabolismo , Regulación de la Expresión Génica/fisiología , FN-kappa B/metabolismo , ARN Mensajero/metabolismo , Regiones no Traducidas 3'/genética , Animales , Northern Blotting , Western Blotting , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/genética , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Luciferasas , Ratones , Ratones Noqueados , Análisis por Micromatrices , Simulación de Dinámica Molecular , FN-kappa B/genética , Interferencia de ARN , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Ribosómicas/metabolismo
6.
Dev Biol ; 392(2): 233-44, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24892953

RESUMEN

As the primary driving forces of gastrulation, convergence and extension (C&E) movements lead to a medio-lateral narrowing and an anterior-posterior elongation of the embryonic body axis. Histone methylation as a post-translational modification plays a critical role in early embryonic development, but its functions in C&E movements remain largely unknown. Here, we show that the setdb2-dvr1 transcriptional cascade plays a critical role in C&E movements during zebrafish gastrulation. Knockdown of Setdb2, a SET domain-containing protein possessing a potential histone H3K9 methyltransferase activity, induced abnormal C&E movements, resulting in anterior-posterior shortening and medio-lateral expansion of the embryonic axis, as well as abnormal notochord cell polarity. Furthermore, we found that Setdb2 functions through fine-tuning the expression of dvr1, a ligand of the TGF-ß superfamily, to an appropriate level to ensure proper C&E movements in a non-cell-autonomous manner. In addition, both overexpression and knockdown of Dvr1 at the one-cell stage resulted in defects at epiboly and C&E. These data demonstrate that Setdb2 is a novel regulator for C&E movements and acts by modulating the expression level of dvr1, suggesting that Dvr1 acts as a direct and essential mediator for C&E cell movements.


Asunto(s)
Movimiento Celular/fisiología , Gastrulación/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Animales , Western Blotting , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Hibridación in Situ , Análisis por Micromatrices , Morfolinos/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Crecimiento Transformador beta/genética , Proteínas de Pez Cebra/genética
7.
Am J Nephrol ; 39(2): 110-21, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24503548

RESUMEN

OBJECTIVES: Acute kidney injury (AKI) is a common complication in hospitalized patients and the incidence of AKI is rapidly increasing. Despite the advances in treatment of AKI, many patients still progress to end-stage renal disease and depend on dialysis. Therefore, early diagnosis and adequate treatment of AKI could improve prognosis. METHODS: We established rat models of AKI induced by cisplatin nephrotoxicity and renal ischemia-reperfusion (I/R). Urine samples were collected, labeled with isobaric tags for relative and absolute quantification agents, and then subjected to nano-LC-MS/MS-based proteomic analysis. Results of the proteomic study were confirmed by Western blot. We also performed RNAi to silence nestin and investigate its role in renal I/R injury. We then validated its clinical application by studying urine nestin levels in AKI patients with cardiovascular surgeries. RESULTS: Our proteomic analysis showed that fetuin-A, nestin, hamartin and T-kininogen were differentially expressed in the urine samples of rats after cisplatin or I/R treatment. Western blot confirmed the differential expression of these proteins in animal models and ELISA confirmed the differential expression of nestin in human urine samples. To explore the expression of nestin in the development of AKI, our results showed that nestin was primarily detected in the glomeruli and barely detected in tubular cells but increased in tubular cells during I/R- and cisplatin-induced AKI. The urine nestin-to-creatinine ratio increased earlier than serum creatinine in AKI patients with postcardiovascular surgeries. The role of nestin in AKI might be related to the p53 signaling pathway. CONCLUSIONS: Thus, our results demonstrated that urinary nestin could be a urinary biomarker for patients with AKI and its role in AKI might be related to the p53 signaling pathway.


Asunto(s)
Lesión Renal Aguda/diagnóstico , Lesión Renal Aguda/orina , Nestina/orina , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/orina , Lesión Renal Aguda/inducido químicamente , Adulto , Anciano , Animales , Antineoplásicos/farmacología , Biomarcadores/orina , Cisplatino/farmacología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Nestina/genética , Proteómica/métodos , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Sensibilidad y Especificidad , Transducción de Señal/fisiología , Adulto Joven
8.
FEBS J ; 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38652546

RESUMEN

l-threonine dehydrogenase (Tdh) is an enzyme that links threonine metabolism to epigenetic modifications and mitochondria biogenesis. In vitro studies show that it is critical for the regulation of trimethylation of histone H3 lysine 4 (H3K4me3) levels and cell fate determination of mouse embryonic stem cells (mESCs). However, whether Tdh regulates a developmental process in vivo and, if it does, whether it also primarily regulates H3K4me3 levels in this process as it does in mESCs, remains elusive. Here, we revealed that, in zebrafish hematopoiesis, tdh is preferentially expressed in neutrophils. Knockout of tdh causes a decrease in neutrophil number and slightly suppresses their acute injury-induced migration, but, unlike the mESCs, the level of H3K4me3 is not evidently reduced in neutrophils sorted from the kidney marrow of adult tdh-null zebrafish. These phenotypes are dependent on the enzymatic activity of Tdh. Importantly, a soluble supplement of nutrients that are able to fuel the acetyl-CoA pool, such as pyruvate, glucose and branched-chain amino acids, is sufficient to rescue the reduction in neutrophils caused by tdh deletion. In summary, our study presents evidence for the functional requirement of Tdh-mediated threonine metabolism in a developmental process in vivo. It also provides an animal model for investigating the nutritional regulation of myelopoiesis and immune response, as well as a useful tool for high-throughput drug/nutrition screening.

9.
Proc Natl Acad Sci U S A ; 107(50): 21683-8, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21118980

RESUMEN

Arsenic, a curative agent for acute promyelocytic leukemia, induces cell apoptosis and degradation of BCR-ABL in chronic myelogenous leukemia (CML). We demonstrated that ubiquitination and degradation of BCR-ABL was mediated by c-CBL, a RING-type E3 ligase that was also shown to be involved in ubiquitination for many other receptor/protein tyrosine kinases. Our data showed that c-CBL protein was considerably up-regulated by arsenic sulfide (As(4)S(4)). Interestingly, arsenic directly bound the RING finger domain of c-CBL to inhibit its self-ubiquitination/degradation without interfering with the enhancement of ubiquitination and subsequent proteolysis of its substrate BCR-ABL. Degradation of BCR-ABL due to c-CBL induction as a result of arsenic treatment was also observed in vivo in CML mice. These findings provide insight into the molecular mechanisms of arsenic and further support its therapeutic applications in CML in combination with tyrosine kinase inhibitors and potentially also in other malignancies involving aberrant receptor/protein tyrosine kinase signaling.


Asunto(s)
Arsenicales/uso terapéutico , Proteínas de Fusión bcr-abl/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Sulfuros/uso terapéutico , Secuencia de Aminoácidos , Animales , Proteínas de Fusión bcr-abl/genética , Células HeLa , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Complejos Multiproteicos/metabolismo , Conformación Proteica , Proteínas Proto-Oncogénicas c-cbl/química , Proteínas Proto-Oncogénicas c-cbl/genética , Distribución Aleatoria , Ubiquitinación
10.
Proc Natl Acad Sci U S A ; 107(7): 2956-61, 2010 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-20133625

RESUMEN

HYPB is a human histone H3 lysine 36 (H3K36)-specific methyltransferase and acts as the ortholog of yeast Set2. This study explored the physiological function of mammalian HYPB using knockout mice. Homozygous disruption of Hypb impaired H3K36 trimethylation but not mono- or dimethylation, and resulted in embryonic lethality at E10.5-E11.5. Severe vascular defects were observed in the Hypb(-/-) embryo, yolk sac, and placenta. The abnormally dilated capillaries in mutant embryos and yolk sacs could not be remodeled into large blood vessels or intricate networks, and the aberrantly rounded mesodermal cells exhibited weakened interaction with endothelial cells. The embryonic vessels failed to invade the labyrinthine layer of placenta, which impaired the embryonic-maternal vascular connection. These defects could not be rescued by wild-type tetraploid blastocysts, excluding the possibility that they were caused by the extraembryonic tissues. Consistent with these phenotypes, gene expression profiling in wild-type and Hypb(-/-) yolk sacs revealed that the Hypb disruption altered the expression of some genes involved in vascular remodeling. At the cellular level, Hypb(-/-) embryonic stem cell-derived embryonic bodies, as well as in vitro-cultured human endothelial cells with siRNA-mediated suppression of HYPB, showed obvious defects in cell migration and invasion during vessel formation, suggesting an intrinsic role of Hypb in vascular development. Taken together, these results indicate that Hypb is required for embryonic vascular remodeling and provide a tool to study the function of H3K36 methylation in vasculogenesis/angiogenesis.


Asunto(s)
Embrión de Mamíferos/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , N-Metiltransferasa de Histona-Lisina/metabolismo , Neovascularización Fisiológica/fisiología , Animales , Células Cultivadas , Embrión de Mamíferos/metabolismo , Perfilación de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Metilación , Ratones , Ratones Noqueados , Neovascularización Fisiológica/genética , Interferencia de ARN
11.
Proc Natl Acad Sci U S A ; 106(9): 3378-83, 2009 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-19208803

RESUMEN

In this study, we show that combined use of Imatinib (IM) and arsenic sulfide [As(4)S(4) (AS)] exerts more profound therapeutic effects in a BCR/ABL-positive mouse model of chronic myeloid leukemia (CML) than either drug as a single agent. A systematic analysis of dynamic changes of the proteome, phosphoproteome, and transcriptome in K562 cells after AS and/or IM treatment was performed to address the mechanisms underlying this synergy. Our data indicate that AS promotes the activities of the unfolded protein reaction (UPR) and ubiquitination pathway, which could form the biochemical basis for the pharmacological effects of this compound. In this CML model, AS targets BCR/ABL through the ubiquitination of key lysine residues, leading to its proteasomal degradation, whereas IM inhibits the PI3K/AKT/mTOR pathway. Combination of the 2 agents synergistically arrests the cell cycle, decreases activity of BCR/ABL, and leads to activation of intrinsic and extrinsic apoptosis pathways through complex modifications to both transcription and protein levels. Thus, these results suggest potential clinical benefits of IM/AS combination therapy for human CML.


Asunto(s)
Arsenicales/uso terapéutico , Proteínas de Fusión bcr-abl/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Piperazinas/uso terapéutico , Pirimidinas/uso terapéutico , Sulfuros/uso terapéutico , Biología de Sistemas , Animales , Benzamidas , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Proteínas de Fusión bcr-abl/química , Perfilación de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Mesilato de Imatinib , Ratones , Modelos Moleculares , Trasplante de Neoplasias , Fosfatidilinositol 3-Quinasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Tasa de Supervivencia , Serina-Treonina Quinasas TOR , Ubiquitinas/metabolismo
12.
Front Cell Dev Biol ; 10: 992714, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36158200

RESUMEN

The ETO-family transcriptional corepressors, including ETO, ETO2, and MTGR1, are all involved in leukemia-causing chromosomal translocations. In every case, an ETO-family corepressor acquires a DNA-binding domain (DBD) to form a typical transcription factor-the DBD binds to DNA, while the ETO moiety manifests transcriptional activity. A directly comparative study of these "homologous" fusion transcription factors may clarify their similarities and differences in regulating transcription and leukemogenesis. Here, we performed a side-by-side comparison between AML1-ETO and ETO2-GLIS2, the most common fusion proteins in M2-and M7-subtypes of acute myeloid leukemia, respectively, by inducible expression of them in U937 leukemia cells. We found that, although AML1-ETO and ETO2-GLIS2 can use their own DBDs to bind DNA, they share a large proportion of genome-wide binding regions dependent on other cooperative transcription factors, including the ETS-, bZIP- and bHLH-family proteins. AML1-ETO acts as either transcriptional repressor or activator, whereas ETO2-GLIS2 mainly acts as activator. The repressor-versus-activator functions of AML1-ETO might be determined by the abundance of cooperative transcription factors/cofactors on the target genes. Importantly, AML1-ETO and ETO2-GLIS2 differentially regulate key transcription factors in myeloid differentiation including PU.1 and C/EBPß. Consequently, AML1-ETO inhibits, but ETO2-GLIS2 facilitates, myeloid differentiation of U937 cells. This function of ETO2-GLIS2 is reminiscent of a similar effect of MLL-AF9 as previously reported. Taken together, this directly comparative study between AML1-ETO and ETO2-GLIS2 in the same cellular context provides insights into context-dependent transcription regulatory mechanisms that may underlie how these seemingly "homologous" fusion transcription factors exert distinct functions to drive different subtypes of leukemia.

13.
Blood ; 113(6): 1340-9, 2009 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-18941117

RESUMEN

Precise transcriptional control of developmental stage-specific expression and switching of alpha- and beta-globin genes is significantly important to understand the general principles controlling gene expression and the pathogenesis of thalassemia. Although transcription factors regulating beta-globin genes have been identified, little is known about the microRNAs and trans-acting mechanism controlling alpha-globin genes transcription. Here, we show that an erythroid lineage-specific microRNA gene, miR-144, expressed at specific developmental stages during zebrafish embryogenesis, negatively regulates the embryonic alpha-globin, but not embryonic beta-globin, gene expression, through physiologically targeting klfd, an erythroid-specific Krüppel-like transcription factor. Klfd selectively binds to the CACCC boxes in the promoters of both alpha-globin and miR-144 genes to activate their transcriptions, thus forming a negative feedback circuitry to fine-tune the expression of embryonic alpha-globin gene. The selective effect of the miR-144-Klfd pathway on globin gene regulation may thereby constitute a novel therapeutic target for improving the clinical outcome of patients with thalassemia.


Asunto(s)
Embrión no Mamífero/metabolismo , Eritropoyesis/fisiología , Regulación del Desarrollo de la Expresión Génica , MicroARNs/genética , Globinas alfa/genética , Animales , Animales Modificados Genéticamente , Apoptosis , Northern Blotting , Western Blotting , Biología Computacional , Embrión no Mamífero/citología , Células Precursoras Eritroides/citología , Células Precursoras Eritroides/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Técnicas para Inmunoenzimas , Hibridación in Situ , Factores de Transcripción de Tipo Kruppel/antagonistas & inhibidores , Factores de Transcripción de Tipo Kruppel/fisiología , MicroARNs/metabolismo , Oligonucleótidos/farmacología , Regiones Promotoras Genéticas/genética , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética , Pez Cebra , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/fisiología , Globinas alfa/metabolismo , Globinas beta/genética , Globinas beta/metabolismo
14.
Proc Natl Acad Sci U S A ; 105(6): 2076-81, 2008 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-18250304

RESUMEN

Acquisition of additional genetic and/or epigenetic abnormalities other than the BCR/ABL fusion gene is believed to cause disease progression in chronic myeloid leukemia (CML) from chronic phase to blast crisis (BC). To gain insights into the underlying mechanisms of progression to BC, we screened DNA samples from CML patients during blast transformation for mutations in a number of transcription factor genes that are critical for myeloid-lymphoid development. In 85 cases of CML blast transformation, we identified two new mutations in the coding region of GATA-2, a negative regulator of hematopoietic stem/progenitor cell differentiation. A L359V substitution within zinc finger domain (ZF) 2 of GATA-2 was found in eight cases with myelomonoblastic features, whereas an in-frame deletion of 6 aa (delta341-346) spanning the C-terminal border of ZF1 was detected in one patient at myeloid BC with eosinophilia. Further studies indicated that L359V not only increased transactivation activity of GATA-2 but also enhanced its inhibitory effects on the activity of PU.1, a major regulator of myelopoiesis. Consistent with the myelomonoblastic features of CML transformation with the GATA-2 L359V mutant, transduction of the GATA-2 L359V mutant into HL-60 cells or BCR/ABL-harboring murine cells disturbed myelomonocytic differentiation/proliferation in vitro and in vivo, respectively. These data strongly suggest that GATA-2 mutations may play a role in acute myeloid transformation in a subset of CML patients.


Asunto(s)
Factor de Transcripción GATA2/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mieloide Aguda/genética , Mutación , Animales , Secuencia de Bases , Células COS , Línea Celular , Chlorocebus aethiops , Cartilla de ADN , Progresión de la Enfermedad , Humanos , Inmunoprecipitación , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Mieloide Aguda/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
Proc Natl Acad Sci U S A ; 105(30): 10553-8, 2008 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-18650396

RESUMEN

RIG-I has been implicated in innate immunity by sensing intracellular viral RNAs and inducing type I IFN production. However, we have found a significant RIG-I induction in a biological setting without active viral infection-namely, during RA-induced terminal granulocytic differentiation of acute myeloid leukemias. Here, we present evidence that a significant Rig-I induction also occurs during normal myelopoiesis and that the disruption of the Rig-I gene in mice leads to the development of a progressive myeloproliferative disorder. The initiation of progressive myeloproliferative disorder is mainly due to an intrinsic defect of Rig-I(-/-) myeloid cells, which are characterized by a reduced expression of IFN consensus sequence binding protein, a major regulator of myeloid differentiation. Thus, our study reveals a critical regulatory role of Rig-I in modulating the generation and differentiation of granulocytes.


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Regulación de la Expresión Génica , Granulocitos/citología , Receptores de Ácido Retinoico/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Proteína 58 DEAD Box , Exones , Granulocitos/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Inmunidad Innata , Ratones , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/citología , Trastornos Mieloproliferativos/metabolismo
16.
Yi Chuan ; 33(8): 879-85, 2011 Aug.
Artículo en Zh | MEDLINE | ID: mdl-21831804

RESUMEN

To investigate the variance in exogenous gene expression driven by the different promoters in leukemia cells, four GFP reporter lentivirus vectors carrying different promoters, including EF1α, PGK, Ubiquitin and CMV, were selected. Leukemia cell lines NB4, THP1, HL60 and Kasumi were infected with lentivirus produced from these reporter vectors, respectively. Then, fluorescence microscope, flow cytometry and fluorescence quantitative PCR were used to detect the GFP expression strength. The results of this study clearly showed that the expression levels of the reporter genes with four different promoters were significantly different. Among them, EF1α drove the highest level of GFP expression, while CMV promoter induced the lowest level. Our results suggested that promoters should be carefully chosen in order to get the appropriate exogenous expression level in leukemia cells according to the study need.


Asunto(s)
Expresión Génica , Leucemia/genética , Regiones Promotoras Genéticas , Línea Celular Tumoral , Genes Reporteros , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Leucemia/metabolismo
17.
Cell Death Dis ; 12(6): 568, 2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34078881

RESUMEN

GATA2, a key transcription factor in hematopoiesis, is frequently mutated in hematopoietic malignancies. How the GATA2 mutants contribute to hematopoiesis and malignant transformation remains largely unexplored. Here, we report that Gata2-L359V mutation impeded hematopoietic differentiation in murine embryonic and adult hematopoiesis and blocked murine chronic myeloid leukemia (CML) cell differentiation. We established a Gata2-L359V knockin mouse model in which the homozygous Gata2-L359V mutation caused major defects in primitive erythropoiesis with an accumulation of erythroid precursors and severe anemia, leading to embryonic lethality around E11.5. During adult life, the Gata2-L359V heterozygous mice exhibited a notable decrease in bone marrow (BM) recovery under stress induction with cytotoxic drug 5-fluorouracil. Using RNA sequencing, it was revealed that homozygous Gata2-L359V suppressed genes related to embryonic hematopoiesis in yolk sac, while heterozygous Gata2-L359V dysregulated genes related to cell cycle and proliferation in BM Lin-Sca1+c-kit+ cells. Furthermore, through chromatin immunoprecipitation sequencing and transactivation experiments, we found that this mutation enhanced the DNA-binding capacity and transcriptional activities of Gata2, which was likely associated with the altered expression of some essential genes during embryonic and adult hematopoiesis. In mice model harboring BCR/ABL, single-cell RNA-sequencing demonstrated that Gata2-L359V induced additional gene expression profile abnormalities and partially affected cell differentiation at the early stage of myelomonocytic lineage, evidenced by the increase of granulocyte-monocyte progenitors and monocytosis. Taken together, our study unveiled that Gata2-L359V mutation induces defective hematopoietic development and blocks the differentiation of CML cells.


Asunto(s)
Factor de Transcripción GATA2/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Animales , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Factor de Transcripción GATA2/genética , Hematopoyesis , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación
18.
Cell Discov ; 7(1): 98, 2021 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-34697290

RESUMEN

The amino acid response (AAR) and unfolded protein response (UPR) pathways converge on eIF2α phosphorylation, which is catalyzed by Gcn2 and Perk, respectively, under different stresses. This close interconnection makes it difficult to specify different functions of AAR and UPR. Here, we generated a zebrafish model in which loss of threonyl-tRNA synthetase (Tars) induces angiogenesis dependent on Tars aminoacylation activity. Comparative transcriptome analysis of the tars-mutant and wild-type embryos with/without Gcn2- or Perk-inhibition reveals that only Gcn2-mediated AAR is activated in the tars-mutants, whereas Perk functions predominantly in normal development. Mechanistic analysis shows that, while a considerable amount of eIF2α is normally phosphorylated by Perk, the loss of Tars causes an accumulation of uncharged tRNAThr, which in turn activates Gcn2, leading to phosphorylation of an extra amount of eIF2α. The partial switchover of kinases for eIF2α largely overwhelms the functions of Perk in normal development. Interestingly, although inhibition of Gcn2 and Perk in this stress condition both can reduce the eIF2α phosphorylation levels, their functional consequences in the regulation of target genes and in the rescue of the angiogenic phenotypes are dramatically different. Indeed, genetic and pharmacological manipulations of these pathways validate that the Gcn2-mediated AAR, but not the Perk-mediated UPR, is required for tars-deficiency induced angiogenesis. Thus, the interconnected AAR and UPR pathways differentially regulate angiogenesis through selective functions and mutual competitions, reflecting the specificity and efficiency of multiple stress response pathways that evolve integrally to enable an organism to sense/respond precisely to various types of stresses.

19.
Biochem Biophys Res Commun ; 394(1): 18-23, 2010 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-20167204

RESUMEN

Makorin-2, consisting of four highly conserved C(3)H zinc fingers, a Cys-His motif and a C(3)HC(4) RING zinc finger domain, is a putative ribonucleoprotein. We have previously reported that Xenopus makorin-2 (mkrn2) is a neurogenesis inhibitor acting upstream of glycogen synthase kinase-3beta (GSK-3beta) in the phosphatidylinositol 3-kinase/Akt pathway. In an effort to identify the functional domains required for its anti-neurogenic activity, we designed and constructed a series of N- and C-terminal truncation mutants of mkrn2. Concurred with the full-length mkrn2, we showed that overexpression of one of the truncation mutants mkrn2(s)-7, which consists of only the third C(3)H zinc finger, Cys-His motif and C(3)HC(4) RING zinc finger, is essential and sufficient to produce the phenotypical dorso-posterior deficiencies and small-head/short-tail phenotype in tadpoles. In animal cap explant assay, we further demonstrated that mkrn2(s)-7 not only inhibits activin and retinoic acid-induced animal cap neuralization and the expression of a pan-neural marker neural cell adhesion molecule, but also induces GSK-3beta expression. These results collectively suggest that the third C(3)H zinc finger, Cys-His motif and C(3)HC(4) RING zinc finger are indispensable for the anti-neurogenic activity of mkrn2.


Asunto(s)
Neurogénesis , Ribonucleoproteínas/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Secuencias de Aminoácidos/genética , Animales , Secuencia Conservada , Embrión no Mamífero/metabolismo , Glucógeno Sintasa Quinasa 3/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Filogenia , Estructura Terciaria de Proteína/genética , Ribonucleoproteínas/clasificación , Ribonucleoproteínas/genética , Proteínas de Xenopus/clasificación , Proteínas de Xenopus/genética , Xenopus laevis/genética , Xenopus laevis/metabolismo , Dedos de Zinc/genética
20.
Am J Nephrol ; 31(1): 24-35, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19864886

RESUMEN

BACKGROUND: Transforming growth factor-beta (TGF-beta)-induced epithelial-to-mesenchymal transition (EMT) plays an important role in renal fibrosis and progression of chronic kidney disease (CKD). Phosphorylation of proteins is essential to TGF-beta signaling. We applied isobaric tags for relative and absolute quantification (iTRAQ) technology to profile the phosphoproteins in tubular epithelial cells in response to TGF-beta-induced EMT in order to further study molecular events. METHODS: HK-2 cells were treated with TGF-beta1 to induce EMT. The cells were divided into a control group (without TGF-beta1 treatment) and a TGF-beta1-treated group. Phosphoproteins from two groups were extracted and differentially labeled with iTRAQ reagents and processed by 2D-nano-HPLC-MS/MS. Validating of iTRAQ analysis was performed by western blot. Bioinformatic analysis was performed by on-line databases. RESULTS: By iTRAQ-2D-nano-HPLC-MS/MS, 38 differentially expressed phosphoproteins were identified which included 19 up-regulated phosphoproteins and 19 down-regulated phosphoproteins. Western blot confirmed up-regulation of phosphorylated moesin and HSP90alpha. Bioinformatic analysis suggested that the majority of proteins were located in the nucleus and endoplasmic reticulum lumen. The phosphoproteins were categorized into 17 molecular function classifications. Nucleic acid binding protein, cytoskeletal protein and chaperone were the major categories of molecular function. A biological network was built to analyze interaction between up-regulated proteins. CONCLUSION: We demonstrate a TGF-beta1-mediated post-transcriptional regulation of EMT in tubular epithelial cells. Phosphorylation of moesin and HSP90alpha might play a role in TGF-beta-induced EMT.


Asunto(s)
Transdiferenciación Celular , Células Epiteliales/fisiología , Túbulos Renales/citología , Túbulos Renales/fisiología , Mesodermo/citología , Mesodermo/fisiología , Fosfoproteínas/fisiología , Proteómica , Factor de Crecimiento Transformador beta1/fisiología , Células Cultivadas , Humanos , Urotelio/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA