Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Biol Chem ; 295(34): 12188-12202, 2020 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-32467227

RESUMO

Triple-negative breast cancer (TNBC) is an aggressive cancer subtype for which effective therapies are unavailable. TNBC has a high frequency of tumor protein p53 (Tp53/p53)- and phosphatase and tensin homolog (PTEN) deficiencies, and combined p53- and PTEN-deficiency is associated with poor prognosis and poor response to anticancer therapies. In this study, we discovered that combined p53- and PTEN-deficiency in TNBC activates expression of the transcription factor mesenchyme homeobox 1 (MEOX1). We found that MEOX1 is expressed only in TNBC cells with frequent deficiencies in p53 and PTEN, and that its expression is undetectable in luminal A, luminal B, and HER2+ subtypes, as well as in normal breast cells with wild-type (WT) p53 and PTEN. Notably, siRNA knockdown of both p53 and PTEN activated MEOX1 expression in breast cancer cells, whereas individual knockdowns of either p53 or PTEN had only minimal effects on MEOX1 expression. MEOX1 knockdown abolished cell proliferation of p53- and PTEN-deficient TNBC in vitro and inhibited tumor growth in vivo, but had no effect on the proliferation of luminal and HER2+ cancer cells and normal breast cells. RNA-Seq and immunoblotting analyses showed that MEOX1 knockdown decreased expression of tyrosine kinase 2 (TYK2), signal transducer and activator of transcription 5B (STAT5B), and STAT6 in p53- and PTEN-deficient TNBC cells. These results reveal the effects of combined p53- and PTEN-deficiency on MEOX1 expression and TNBC cell proliferation, suggesting that MEOX1 may serve as a potential therapeutic target for managing p53- and PTEN-deficient TNBC.


Assuntos
Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/biossíntese , PTEN Fosfo-Hidrolase/deficiência , Fatores de Transcrição/biossíntese , Proteína Supressora de Tumor p53/deficiência , Animais , Feminino , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , PTEN Fosfo-Hidrolase/metabolismo , Fator de Transcrição STAT5/biossíntese , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT6/biossíntese , Fator de Transcrição STAT6/genética , TYK2 Quinase/biossíntese , TYK2 Quinase/genética , Fatores de Transcrição/genética , Neoplasias de Mama Triplo Negativas , Proteína Supressora de Tumor p53/metabolismo
2.
J Biol Chem ; 293(6): 2125-2136, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29263092

RESUMO

The transcription factor BCL11A has recently been reported to be a driving force in triple-negative breast cancer (TNBC), contributing to the maintenance of a chemoresistant breast cancer stem cell (BCSC) population. Although BCL11A was shown to suppress γ-globin and p21 and to induce MDM2 expression in the hematopoietic system, its downstream targets in TNBC are still unclear. For its role in transcriptional repression, BCL11A was found to interact with several corepressor complexes; however, the mechanisms underlying these interactions remain unknown. Here, we reveal that BCL11A interacts with histone methyltransferase (PRC2) and histone deacetylase (NuRD and SIN3A) complexes through their common subunit, RBBP4/7. In fluorescence polarization assays, we show that BCL11A competes with histone H3 for binding to the negatively charged top face of RBBP4. To define that interaction, we solved the crystal structure of RBBP4 in complex with an N-terminal peptide of BCL11A (residues 2-16, BCL11A(2-16)). The crystal structure identifies novel interactions between BCL11A and the side of the ß-propeller of RBBP4 that are not seen with histone H3. We next show that BCL11A(2-16) pulls down RBBP4, RBBP7, and other components of PRC2, NuRD, and SIN3A from the cell lysate of the TNBC cell line SUM149. Furthermore, we demonstrate the therapeutic potential of targeting the RBBP4-BCL11A binding by showing that a BCL11A peptide can decrease aldehyde dehydrogenase-positive BCSCs and mammosphere formation capacity in SUM149. Together, our findings have uncovered a previously unidentified mechanism that BCL11A may use to recruit epigenetic complexes to regulate transcription and promote tumorigenesis.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Nucleares/metabolismo , Proteína 4 de Ligação ao Retinoblastoma/metabolismo , Proteína 7 de Ligação ao Retinoblastoma/metabolismo , Carcinogênese , Proteínas de Transporte/química , Linhagem Celular , Cristalografia por Raios X , Epigenômica , Histona Desacetilases/metabolismo , Histona Metiltransferases/metabolismo , Humanos , Proteínas Nucleares/química , Ligação Proteica , Proteínas Repressoras , Proteína 4 de Ligação ao Retinoblastoma/química , Proteína 7 de Ligação ao Retinoblastoma/química , Fatores de Transcrição/metabolismo , Neoplasias de Mama Triplo Negativas/patologia
3.
Mol Cell ; 42(2): 185-98, 2011 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-21504830

RESUMO

It has been suspected that cell-cycle progression might be functionally coupled with RNA processing. However, little is known about the role of the precise splicing control in cell-cycle progression. Here, we report that SON, a large Ser/Arg (SR)-related protein, is a splicing cofactor contributing to efficient splicing of cell-cycle regulators. Downregulation of SON leads to severe impairment of spindle pole separation, microtubule dynamics, and genome integrity. These molecular defects result from inadequate RNA splicing of a specific set of cell-cycle-related genes that possess weak splice sites. Furthermore, we show that SON facilitates the interaction of SR proteins with RNA polymerase II and other key spliceosome components, suggesting its function in efficient cotranscriptional RNA processing. These results reveal a mechanism for controlling cell-cycle progression through SON-dependent constitutive splicing at suboptimal splice sites, with strong implications for its role in cancer and other human diseases.


Assuntos
Ciclo Celular/genética , Proteínas de Ligação a DNA/metabolismo , Genes cdc , Splicing de RNA , Spliceossomos/metabolismo , Segregação de Cromossomos , Citocinese , Proteínas de Ligação a DNA/genética , Instabilidade Genômica , Células HEK293 , Células HeLa , Humanos , Células K562 , Microtúbulos/metabolismo , Antígenos de Histocompatibilidade Menor , Interferência de RNA , RNA Polimerase II/metabolismo , Fuso Acromático/metabolismo , Fatores de Tempo , Transfecção
4.
Blood ; 123(24): 3760-9, 2014 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-24771859

RESUMO

RUNX1 is an important transcription factor for hematopoiesis. There are multiple alternatively spliced isoforms of RUNX1. The best known isoforms are RUNX1a from use of exon 7A and RUNX1b and c from use of exon 7B. RUNX1a has unique functions due to its lack of C-terminal regions common to RUNX1b and c. Here, we report that the ortholog of human RUNX1a was only found in primates. Furthermore, we characterized 3 Runx1 isoforms generated by exon 6 alternative splicing. Runx1bEx6(-) (Runx1b without exon 6) and a unique mouse Runx1bEx6e showed higher colony-forming activity than the full-length Runx1b (Runx1bEx6(+)). They also facilitated the transactivation of Runx1bEx6(+). To gain insight into in vivo functions, we analyzed a knock-in (KI) mouse model that lacks isoforms Runx1b/cEx6(-) and Runx1bEx6e. KI mice had significantly fewer lineage-Sca1(+)c-Kit(+) cells, short-term hematopoietic stem cells (HSCs) and multipotent progenitors than controls. In vivo competitive repopulation assays demonstrated a sevenfold difference of functional HSCs between wild-type and KI mice. Together, our results show that Runx1 isoforms involving exon 6 support high self-renewal capacity in vitro, and their loss results in reduction of the HSC pool in vivo, which underscore the importance of fine-tuning RNA splicing in hematopoiesis.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Hematopoese/genética , Animais , Sequência de Bases , Células Cultivadas , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Éxons , Regulação da Expressão Gênica , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Sítios de Splice de RNA , Homologia de Sequência
5.
PLoS Genet ; 9(10): e1003765, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24130502

RESUMO

Fusion protein RUNX1-ETO (AML1-ETO, RUNX1-RUNX1T1) is expressed as the result of the 8q22;21q22 translocation [t(8;21)], which is one of the most common chromosomal abnormalities found in acute myeloid leukemia. RUNX1-ETO is thought to promote leukemia development through the aberrant regulation of RUNX1 (AML1) target genes. Repression of these genes occurs via the recruitment of the corepressors N-COR and SMRT due to their interaction with ETO. Mechanisms of RUNX1-ETO target gene upregulation remain less well understood. Here we show that RUNX1-ETO9a, the leukemogenic alternatively spliced transcript expressed from t(8;21), upregulates target gene Alox5, which is a gene critically required for the promotion of chronic myeloid leukemia development by BCR-ABL. Loss of Alox5 expression reduces activity of RUNX1-ETO9a, MLL-AF9 and PML-RARα in vitro. However, Alox5 is not essential for the induction of leukemia by RUNX1-ETO9a in vivo. Finally, we demonstrate that the upregulation of Alox5 by RUNX1-ETO9a occurs via the C2H2 zinc finger transcription factor KLF6, a protein required for early hematopoiesis and yolk sac development. Furthermore, KLF6 is specifically upregulated by RUNX1-ETO in human leukemia cells. This identifies KLF6 as a novel mediator of t(8;21) target gene regulation, providing a new mechanism for RUNX1-ETO transcriptional control.


Assuntos
Araquidonato 5-Lipoxigenase/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Fatores de Transcrição Kruppel-Like/genética , Leucemia Mieloide Aguda/genética , Proteínas Proto-Oncogênicas/genética , Processamento Alternativo , Animais , Linhagem Celular Tumoral , Aberrações Cromossômicas , Regulação Leucêmica da Expressão Gênica , Humanos , Fator 6 Semelhante a Kruppel , Leucemia Mieloide Aguda/patologia , Camundongos , Proteínas de Fusão Oncogênica/genética , Proteína 1 Parceira de Translocação de RUNX1 , Fatores de Transcrição/genética
6.
Blood ; 121(15): 2882-90, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23372166

RESUMO

Advancements in human pluripotent stem cell (hPSC) research have potential to revolutionize therapeutic transplantation. It has been demonstrated that transcription factors may play key roles in regulating maintenance, expansion, and differentiation of hPSCs. In addition to its regulatory functions in hematopoiesis and blood-related disorders, the transcription factor RUNX1 is also required for the formation of definitive blood stem cells. In this study, we demonstrated that expression of endogenous RUNX1a, an isoform of RUNX1, parallels with lineage commitment and hematopoietic emergence from hPSCs, including both human embryonic stem cells and inducible pluripotent stem cells. In a defined hematopoietic differentiation system, ectopic expression of RUNX1a facilitates emergence of hematopoietic progenitor cells (HPCs) and positively regulates expression of mesoderm and hematopoietic differentiation-related factors, including Brachyury, KDR, SCL, GATA2, and PU.1. HPCs derived from RUNX1a hPSCs show enhanced expansion ability, and the ex vivo-expanded cells are capable of differentiating into multiple lineages. Expression of RUNX1a in embryoid bodies (EBs) promotes definitive hematopoiesis that generates erythrocytes with ß-globin production. Moreover, HPCs generated from RUNX1a EBs possess ≥9-week repopulation ability and show multilineage hematopoietic reconstitution in vivo. Together, our results suggest that RUNX1a facilitates the process of producing therapeutic HPCs from hPSCs.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Células-Tronco Embrionárias/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Western Blotting , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula/genética , Proliferação de Células , Células Cultivadas , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias/citologia , Proteínas Fetais/genética , Proteínas Fetais/metabolismo , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Microscopia Confocal , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Transativadores/genética , Transativadores/metabolismo
7.
J Biol Chem ; 288(8): 5381-8, 2013 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-23322776

RESUMO

SON is a DNA- and RNA-binding protein localized in nuclear speckles. Although its function in RNA splicing for effective cell cycle progression and genome stability was recently unveiled, other mechanisms of SON functions remain unexplored. Here, we report that SON regulates GATA-2, a key transcription factor involved in hematopoietic stem cell maintenance and differentiation. SON is highly expressed in undifferentiated hematopoietic stem/progenitor cells and leukemic blasts. SON knockdown leads to significant depletion of GATA-2 protein with marginal down-regulation of GATA-2 mRNA. We show that miR-27a is up-regulated upon SON knockdown and targets the 3'-UTR of GATA-2 mRNA in hematopoietic cells. Up-regulation of miR-27a was due to activation of the promoter of the miR-23a∼27a∼24-2 cluster, suggesting that SON suppresses this promoter to lower the microRNAs from this cluster. Our data revealed a previously unidentified role of SON in microRNA production via regulating the transcription process, thereby modulating GATA-2 at the protein level during hematopoietic differentiation.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Fator de Transcrição GATA2/metabolismo , Regulação da Expressão Gênica , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Animais , Diferenciação Celular , Proteínas de Ligação a DNA/genética , Hematopoese , Humanos , Células K562 , Leucemia Mieloide Aguda/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Antígenos de Histocompatibilidade Menor , Modelos Biológicos , Regiões Promotoras Genéticas , Splicing de RNA , RNA Mensageiro/metabolismo , Células U937 , Regulação para Cima
8.
Genome Res ; 21(5): 798-810, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21451113

RESUMO

Emerging evidence suggests that microRNAs (miRNAs), an abundant class of ∼22-nucleotide small regulatory RNAs, play key roles in controlling the post-transcriptional genetic programs in stem and progenitor cells. Here we systematically examined miRNA expression profiles in various adult tissue-specific stem cells and their differentiated counterparts. These analyses revealed miRNA programs that are common or unique to blood, muscle, and neural stem cell populations and miRNA signatures that mark the transitions from self-renewing and quiescent stem cells to proliferative and differentiating progenitor cells. Moreover, we identified a stem/progenitor transition miRNA (SPT-miRNA) signature that predicts the effects of genetic perturbations, such as loss of PTEN and the Rb family, AML1-ETO9a expression, and MLL-AF10 transformation, on self-renewal and proliferation potentials of mutant stem/progenitor cells. We showed that some of the SPT-miRNAs control the self-renewal of embryonic stem cells and the reconstitution potential of hematopoietic stem cells (HSCs). Finally, we demonstrated that SPT-miRNAs coordinately regulate genes that are known to play roles in controlling HSC self-renewal, such as Hoxb6 and Hoxa4. Together, these analyses reveal the miRNA programs that may control key processes in normal and aberrant stem and progenitor cells, setting the foundations for dissecting post-transcriptional regulatory networks in stem cells.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica/genética , MicroRNAs/metabolismo , Células-Tronco/metabolismo , Animais , Diferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , MicroRNAs/genética , Mutação , Mioblastos/citologia , Mioblastos/metabolismo , Células-Tronco Neurais , Especificidade de Órgãos , Células-Tronco/citologia
9.
Development ; 138(19): 4255-65, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21852394

RESUMO

Wnt target gene activation in C. elegans requires simultaneous elevation of ß-catenin/SYS-1 and reduction of TCF/POP-1 nuclear levels within the same signal-responsive cell. SYS-1 binds to the conserved N-terminal ß-catenin-binding domain (CBD) of POP-1 and functions as a transcriptional co-activator. Phosphorylation of POP-1 by LIT-1, the C. elegans Nemo-like kinase homolog, promotes POP-1 nuclear export and is the main mechanism by which POP-1 nuclear levels are lowered. We present a mechanism whereby SYS-1 and POP-1 nuclear levels are regulated in opposite directions, despite the fact that the two proteins physically interact. We show that the C terminus of POP-1 is essential for LIT-1 phosphorylation and is specifically bound by the diverged ß-catenin WRM-1. WRM-1 does not bind to the CBD of POP-1, nor does SYS-1 bind to the C-terminal domain. Furthermore, binding of WRM-1 to the POP-1 C terminus is mutually inhibitory with SYS-1 binding at the CBD. Computer modeling provides a structural explanation for the specificity in WRM-1 and SYS-1 binding to POP-1. Finally, WRM-1 exhibits two independent and distinct molecular functions that are novel for ß-catenins: WRM-1 serves both as the substrate-binding subunit and an obligate regulatory subunit for the LIT-1 kinase. Mutual inhibitory binding would result in two populations of POP-1: one bound by WRM-1 that is LIT-1 phosphorylated and exported from the nucleus, and another, bound by SYS-1, that remains in the nucleus and transcriptionally activates Wnt target genes. These studies could provide novel insights into cancers arising from aberrant Wnt activation.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Grupo de Alta Mobilidade/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/metabolismo , beta Catenina/biossíntese , Sequência de Aminoácidos , Animais , Padronização Corporal , Caenorhabditis elegans , Núcleo Celular/metabolismo , Proteínas de Fluorescência Verde/química , Células HeLa , Humanos , Modelos Biológicos , Dados de Sequência Molecular , Fosforilação , Ligação Proteica , Homologia de Sequência de Aminoácidos , beta Catenina/química
10.
Blood ; 119(13): 3155-63, 2012 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-22223820

RESUMO

The t(8;21)(q22;q22) is common in adult acute myeloid leukemia (AML). The RUNX1-ETO fusion protein that is expressed by this translocation is poorly leukemogenic and requires additional mutations for transformation. Loss of sex chromosome (LOS) is frequently observed in t(8;21) AML. In the present study, to evaluate whether LOS cooperates with t(8;21) in leukemogenesis, we first used a retroviral transduction/transplantation model to express RUNX1-ETO in hematopoietic cells from XO mice. The low frequency of leukemia in these mice suggests that the potentially critical gene for suppression of t(8;21) leukemia in humans is not conserved on mouse sex chromosomes. The gene encoding the GM-CSF receptor α subunit (CSF2RA) is located on X and Y chromosomes in humans but on chromosome 19 in mice. GM-CSF promotes myeloid cell survival, proliferation, and differentiation. To determine whether GM-CSF signaling affects RUNX1-ETO leukemogenesis, hematopoietic stem/progenitor cells that lack GM-CSF signaling were used to express RUNX1-ETO and transplanted into lethally irradiated mice, and a high penetrance of AML was observed in recipients. Furthermore, GM-CSF reduced the replating ability of RUNX1-ETO-expressing cells. These results suggest a possible tumor-suppressor role of GM-CSF in RUNX1-ETO leukemia. Loss of the CSF2RA gene may be a critical mutation explaining the high incidence of LOS associated with the t(8;21)(q22;q22) translocation.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Leucemia Mieloide Aguda/genética , Transdução de Sinais/fisiologia , Translocação Genética , Adulto , Animais , Células Cultivadas , Cromossomos Humanos Par 21/genética , Cromossomos Humanos Par 8/genética , Cromossomos de Mamíferos/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Proteínas de Ligação a DNA/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas de Fusão Oncogênica/genética , Proteínas Proto-Oncogênicas/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/fisiologia , Cromossomos Sexuais/genética , Cromossomos Sexuais/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Transcrição/genética
11.
Blood ; 120(19): 4028-37, 2012 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-22919028

RESUMO

Mutations of RUNX1 are detected in patients with myelodysplastic syndrome (MDS). In particular, C-terminal truncation mutations lack a transcription regulatory domain and have increased DNA binding through the runt homology domain. The expression of the runt homology domain, RUNX1(41-214), in mouse hematopoietic cells induced progression to MDS and acute myeloid leukemia. Analysis of premyelodysplastic animals found expansion of c-Kit(+)Sca-1(+)Lin(-) cells and skewed differentiation to myeloid at the expense of the lymphoid lineage. These abnormalities correlate with the phenotype of Runx1-deficient animals, as expected given the reported dominant-negative role of C-terminal mutations over the full-length RUNX1. However, MDS is not observed in Runx1-deficient animals. Gene expression profiling found that RUNX1(41-214) c-Kit(+)Sca-1(+)Lin(-) cells have an overlapping yet distinct gene expression profile from Runx1-deficient animals. Moreover, an unexpected parallel was observed between the hematopoietic phenotype of RUNX1(41-214) and aged animals. Genes deregulated in RUNX1(41-214), but not in Runx1-deficient animals, were inversely correlated with the aging gene signature of HSCs, suggesting that disruption of the expression of genes related to normal aging by RUNX1 mutations contributes to development of MDS. The data presented here provide insights into the mechanisms of development of MDS in HSCs by C-terminal mutations of RUNX1.


Assuntos
Transformação Celular Neoplásica/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Regulação da Expressão Gênica , Células-Tronco Hematopoéticas/metabolismo , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/metabolismo , Animais , Apoptose/genética , Medula Óssea/patologia , Ciclo Celular/genética , Linhagem Celular , Análise por Conglomerados , Perfilação da Expressão Gênica , Hematopoese/genética , Transplante de Células-Tronco Hematopoéticas , Homeostase/genética , Humanos , Leucemia Experimental , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Camundongos , Transdução Genética
12.
Blood ; 119(21): 4953-62, 2012 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-22498736

RESUMO

Fusion protein AML1-ETO, resulting from t(8;21) translocation, is highly related to leukemia development. It has been reported that full-length AML1-ETO blocks AML1 function and requires additional mutagenic events to promote leukemia. We have previously shown that the expression of AE9a, a splice isoform of AML1-ETO, can rapidly cause leukemia in mice. To understand how AML1-ETO is involved in leukemia development, we took advantage of our AE9a leukemia model and sought to identify its interacting proteins from primary leukemic cells. Here, we report the discovery of a novel AE9a binding partner PRMT1 (protein arginine methyltransferase 1). PRMT1 not only interacts with but also weakly methylates arginine 142 of AE9a. Knockdown of PRMT1 affects expression of a specific group of AE9a-activated genes. We also show that AE9a recruits PRMT1 to promoters of AE9a-activated genes, resulting in enrichment of H4 arginine 3 methylation, H3 Lys9/14 acetylation, and transcription activation. More importantly, knockdown of PRMT1 suppresses the self-renewal capability of AE9a, suggesting a potential role of PRMT1 in regulating leukemia development.


Assuntos
Proliferação de Células , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Proteínas Repressoras/metabolismo , Células-Tronco/fisiologia , Ativação Transcricional , Animais , Células Cultivadas , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Perfilação da Expressão Gênica , Regulação Leucêmica da Expressão Gênica , Células HEK293 , Humanos , Células K562 , Camundongos , Análise em Microsséries , Proteínas de Fusão Oncogênica/fisiologia , Ligação Proteica/fisiologia , Proteína 1 Parceira de Translocação de RUNX1 , Células-Tronco/metabolismo , Ativação Transcricional/genética , Regulação para Cima/genética , Regulação para Cima/fisiologia
13.
Blood ; 120(7): 1473-84, 2012 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-22740448

RESUMO

Chromosome translocation 8q22;21q22 [t(8;21)] is commonly associated with acute myeloid leukemia (AML), and the resulting AML1-ETO fusion proteins are involved in the pathogenesis of AML. To identify novel molecular and therapeutic targets, we performed combined gene expression microarray and promoter occupancy (ChIP-chip) profiling using Lin(-)/Sca1(-)/cKit(+) cells, the major leukemia cell population, from an AML mouse model induced by AML1-ETO9a (AE9a). Approximately 30% of the identified common targets of microarray and ChIP-chip assays overlap with the human t(8;21)-gene expression molecular signature. CD45, a protein tyrosine phosphatase and a negative regulator of cytokine/growth factor receptor and JAK/STAT signaling, is among those targets. Its expression is substantially down-regulated in leukemia cells. Consequently, JAK/STAT signaling is enhanced. Re-expression of CD45 suppresses JAK/STAT activation, delays leukemia development, and promotes apoptosis of t(8;21)-positive cells. This study demonstrates the benefit of combining gene expression and promoter occupancy profiling assays to identify molecular and potential therapeutic targets in human cancers and describes a previously unappreciated signaling pathway involving t(8;21) fusion proteins, CD45, and JAK/STAT, which could be a potential novel target for treating t(8;21) AML.


Assuntos
Cromossomos Humanos Par 21/genética , Cromossomos Humanos Par 8/genética , DNA de Neoplasias/metabolismo , Perfilação da Expressão Gênica , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Translocação Genética , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Imunoprecipitação da Cromatina , Ativação Enzimática , Regulação Leucêmica da Expressão Gênica , Redes Reguladoras de Genes/genética , Genes Neoplásicos/genética , Humanos , Janus Quinases/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas/genética , Reprodutibilidade dos Testes , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais/genética
14.
J Immunol ; 188(10): 4776-81, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22491252

RESUMO

Dendritic cells (DCs) represent the key cells linking innate and adaptive immune responses. It is critical to understand the molecular factors regulating DC differentiation. Usp18 is an IFN-inducible member of the ubiquitin-specific protease family, which deconjugates ubiquitin-like modifier ISG15 from target proteins and competitively inhibits IFN-α/ß-induced JAK/STAT activation. This study demonstrates that the frequency of conventional CD11b(+) DCs in the spleen of Usp18(-/-) mice was significantly reduced, whereas the frequencies of conventional CD8(+) DCs and plasmacytoid DCs remained normal. In addition, Usp18(-/-) bone marrow (BM) cells generate DCs less efficiently in GM-CSF-supplemented culture, demonstrating a fundamental defect throughout the DC differentiation pathway. Usp18(-/-) BM cells were rescued by exogenous expression of either wild-type or deconjugation-inactive Usp18, and superimposition of an IFN-α/ß receptor knockout returned in vivo DC populations to normal, clearly showing that the defect seen is due solely to Usp18's effect on IFN signaling. Finally, Usp18(-/-) BM-derived DCs expressed high levels of SOCS1/SOCS3, known inhibitors of GM-CSF signaling, providing a mechanistic explanation for the phenotype. In conclusion, we have identified a novel role of Usp18 in modulating conventional CD11b(+) DC development via its inhibitory effect on type I IFN signaling.


Assuntos
Antígeno CD11b/biossíntese , Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Endopeptidases/fisiologia , Animais , Antígenos CD8/biossíntese , Contagem de Células , Diferenciação Celular/genética , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/enzimologia , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Endopeptidases/deficiência , Endopeptidases/genética , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Fator Estimulador de Colônias de Granulócitos e Macrófagos/fisiologia , Substâncias de Crescimento/deficiência , Substâncias de Crescimento/genética , Substâncias de Crescimento/fisiologia , Masculino , Camundongos , Camundongos Knockout , Proteína 1 Supressora da Sinalização de Citocina , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/biossíntese , Proteínas Supressoras da Sinalização de Citocina/fisiologia , Ubiquitina Tiolesterase
15.
Crit Rev Eukaryot Gene Expr ; 23(2): 103-13, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23582033

RESUMO

Microarray technology has contributed valuable information to gene expression signatures of leukemia and other types of cancers and helped to identify biological markers and potential therapeutic targets for treating these diseases. Acute myeloid leukemia (AML) is often caused by aberrant fusion transcription factors resulting from chromosomal translocations, and the dysregulated genes detected by microarray include both direct and indirect targets of the oncogenic transcription factors. The ChIP-chip technology enables the identification of direct targets of a transcription factor based on its promoter occupancy and cellular context. Using AML1-ETO9a-induced AML as a cancer model and using a combined gene expression and promoter occupancy profiling approach, we recently identified CD45 as a direct down-regulated target of t(8;21) fusion proteins. This finding subsequently led us to discover the enhanced Janus activated kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathway, which is negatively regulated by CD45, in t(8;21) AML. This review summarizes the background of t(8;21) leukemia, structural features of the translocation fusion proteins, and the merits of combining gene discovery technologies for the identification of therapeutic targets in t(8;21) leukemia.


Assuntos
Perfilação da Expressão Gênica/métodos , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/genética , Translocação Genética , Animais , Cromossomos Humanos Par 21 , Cromossomos Humanos Par 8 , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Impressões Digitais de DNA , Humanos , Janus Quinases/metabolismo , Leucemia Mieloide Aguda/metabolismo , Antígenos Comuns de Leucócito/genética , Camundongos , Terapia de Alvo Molecular/métodos , Proteínas de Fusão Oncogênica/genética , Regiões Promotoras Genéticas , Proteína 1 Parceira de Translocação de RUNX1 , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo
16.
Dev Biol ; 355(1): 115-23, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21539828

RESUMO

In the canonical Wnt pathway, signaling results in the stabilization and increased levels of ß-catenin in responding cells. ß-catenin then enters the nucleus, functioning as a coactivator for the Wnt effector, TCF/LEF protein. In the absence of Wnt signaling, TCF is complexed with corepressors, together repressing Wnt target genes. In C. elegans, Wnt signaling specifies the E blastomere to become the endoderm precursor. Activation of endoderm genes in E requires not only an increase in ß-catenin level, but a concomitant decrease in the nuclear level of POP-1, the sole C. elegans TCF. A decrease in nuclear POP-1 levels requires Wnt-induced phosphorylation of POP-1 and 14-3-3 protein-mediated nuclear export. Nuclear POP-1 levels remain high in the sister cell of E, MS, where POP-1 represses the expression of endoderm genes. Here we express three vertebrate TCF proteins (human TCF4, mouse LEF1 and Xenopus TCF3) in C. elegans embryos and compare their localization, repression and activation functions to POP-1. All three TCFs are localized to the nucleus in C. elegans embryos, but none undergoes Wnt-induced nuclear export. Although unable to undergo Wnt-induced nuclear export, human TCF4, but not mouse LEF1 or Xenopus TCF3, can repress endoderm genes in MS, in a manner very similar to POP-1. This repressive activity requires that human TCF4 recognizes specific promoter sequences upstream of endoderm genes and interacts with C. elegans corepressors. Domain swapping identified two regions of POP-1 that are sufficient to confer nuclear asymmetry to human TCF4 when swapped with its corresponding domains. Despite undergoing Wnt-induced nuclear export, the human TCF4/POP-1 chimeric protein continues to function as a repressor for endoderm genes in E, a result we attribute to the inability of hTCF4 to bind to C. elegans ß-catenin. Our results reveal a higher degree of species specificity among TCF proteins for coactivator interactions than for corepressor interactions, and uncover a basic difference between how POP-1 and human TCF4 steady state nuclear levels are regulated.


Assuntos
Caenorhabditis elegans/embriologia , Fatores de Transcrição TCF/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Caenorhabditis elegans/metabolismo , Proteínas Correpressoras/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Ligação Proteica/fisiologia , Proteínas Wnt/fisiologia , Proteínas de Xenopus/metabolismo , beta Catenina/fisiologia
17.
Proc Natl Acad Sci U S A ; 105(44): 17103-8, 2008 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-18952841

RESUMO

AML1-ETO is generated from t(8;21)(q22;q22), which is a common form of chromosomal translocation associated with development of acute myeloid leukemia (AML). Although full-length AML1-ETO alone fails to promote leukemia because of its detrimental effects on cell proliferation, an alternatively spliced isoform, AML1-ETO9a, without its C-terminal NHR3/NHR4 domains, strongly induces leukemia. However, full-length AML1-ETO is a major form of fusion product in many t(8;21) AML patients, suggesting additional molecular mechanisms of t(8;21)-related leukemogenesis. Here, we report that disruption of the zinc-chelating structure in the NHR4 domain of AML1-ETO by replacing only one critical amino acid leads to rapid onset of leukemia, demonstrating that the NHR4 domain with the intact structure generates inhibitory effects on leukemogenesis. Furthermore, we identified SON, a DNA/RNA-binding domain containing protein, as a novel NHR4-interacting protein. Knock-down of SON by siRNA resulted in significant growth arrest, and disruption of the interaction between AML1-ETO and endogenous SON rescued cells from AML1-ETO-induced growth arrest, suggesting that SON is an indispensable factor for cell growth, and AML1-ETO binding to SON may trigger signals inhibiting leukemogenesis. In t(8;21) AML patient-derived primary leukemic cells and cell lines, abnormal cytoplasmic localization of SON was detected, which may keep cells proliferating in the presence of full-length AML1-ETO. These results uncovered the crucial role of the NHR4 domain in determination of cellular fate during AML1-ETO-associated leukemogenesis.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/química , Proteínas de Ligação a DNA/metabolismo , Leucemia Mieloide Aguda/metabolismo , Proteínas de Fusão Oncogênica/química , Animais , Sítios de Ligação , Proliferação de Células , Cromossomos Humanos Par 21/genética , Cromossomos Humanos Par 8/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/genética , Células HeLa , Humanos , Células K562 , Leucemia Mieloide Aguda/genética , Camundongos , Antígenos de Histocompatibilidade Menor , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Estrutura Terciária de Proteína , Proteína 1 Parceira de Translocação de RUNX1 , Transfecção , Células U937
19.
Exp Hematol ; 35(6): 978-88, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17533052

RESUMO

Runx1/AML1 plays important roles in hematopoiesis, including the commitment of cells to hematopoiesis during embryonic development, and in the maintenance of hematopoietic cell populations. It is also one of the most common genes involved in chromosomal translocations related to leukemia. One such translocation is t(8;21), which fuses the Runx1 gene to the MTG8/ETO gene and generates the Runx1-MTG8 (AML1-ETO) fusion gene. Both Runx1 and MTG8 have two additional family members that are much less studied in hematopoiesis. Here we report the expression of every member of the Runx and MTG families as well as the Runx heterodimerization partner CBFbeta during hematopoietic differentiation of murine embryonic stem cells. We observed substantially increased expression of Runx1, Runx2, and MTG16 during hematopoietic differentiation. Furthermore, the increase in Runx2 expression is delayed relative to Runx1 expression, suggesting their possible sequential contribution to hematopoiesis.


Assuntos
Diferenciação Celular/fisiologia , Subunidades alfa de Fatores de Ligação ao Core/biossíntese , Proteínas de Ligação a DNA/biossíntese , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica/fisiologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Nucleares/biossíntese , Proteínas Proto-Oncogênicas/biossíntese , Fatores de Transcrição/biossíntese , Animais , Fator de Ligação a CCAAT/metabolismo , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas/citologia , Leucemia/metabolismo , Camundongos , Proteínas de Fusão Oncogênica , Proteínas Repressoras , Translocação Genética
20.
Oncol Rep ; 39(6): 3041-3047, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29620216

RESUMO

Targeting cancer stem cells (CSCs) is a key strategy to prevent cancers from developing drug resistance and metastasis. Mitochondria have been reported to be a vulnerability of CSCs by multiple studies. Here, we report that doxycycline, functioning as an inhibitor of mitochondrial biogenesis, can effectively target breast cancer stem cells (BCSCs). Our results revealed that doxycycline significantly decreased the frequency of aldehyde dehydrogenase­positive (ALDH+) BCSCs as well as mammosphere formation efficiency in HER2+ and triple­negative breast cancer (TNBC) subtypes. Doxycycline also ameliorated paclitaxel­induced enrichment of ALDH+ BCSCs in TNBC. Mechanistically, we showed that doxycycline decreased the level of reactive oxygen species and their downstream p38 MAPK pathway. In agreement with the key role for p38 in maintaining BCSCs, a specific inhibitor targeting this MAPK pathway significantly decreased the number of ALDH+ cells. Doxycycline is a FDA­approved drug with minor and limited side­effects. Given doxycycline's low toxicity and strong effect on BCSC inhibition, we report that doxycycline should be safe to be used concomitantly with chemotherapy drugs to eradicate both CSCs and bulk tumor cells.


Assuntos
Aldeído Desidrogenase/metabolismo , Neoplasias da Mama/metabolismo , Doxiciclina/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células MCF-7 , Células-Tronco Neoplásicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA