Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Nat Commun ; 15(1): 3415, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38649367

RESUMEN

An important epigenetic component of tyrosine kinase signaling is the phosphorylation of histones, and epigenetic readers, writers, and erasers. Phosphorylation of protein arginine methyltransferases (PRMTs), have been shown to enhance and impair their enzymatic activity. In this study, we show that the hyperactivation of Janus kinase 2 (JAK2) by the V617F mutation phosphorylates tyrosine residues (Y149 and Y334) in coactivator-associated arginine methyltransferase 1 (CARM1), an important target in hematologic malignancies, increasing its methyltransferase activity and altering its target specificity. While non-phosphorylatable CARM1 methylates some established substrates (e.g. BAF155 and PABP1), only phospho-CARM1 methylates the RUNX1 transcription factor, on R223 and R319. Furthermore, cells expressing non-phosphorylatable CARM1 have impaired cell-cycle progression and increased apoptosis, compared to cells expressing phosphorylatable, wild-type CARM1, with reduced expression of genes associated with G2/M cell cycle progression and anti-apoptosis. The presence of the JAK2-V617F mutant kinase renders acute myeloid leukemia (AML) cells less sensitive to CARM1 inhibition, and we show that the dual targeting of JAK2 and CARM1 is more effective than monotherapy in AML cells expressing phospho-CARM1. Thus, the phosphorylation of CARM1 by hyperactivated JAK2 regulates its methyltransferase activity, helps select its substrates, and is required for the maximal proliferation of malignant myeloid cells.


Asunto(s)
Apoptosis , Subunidad alfa 2 del Factor de Unión al Sitio Principal , Janus Quinasa 2 , Proteína-Arginina N-Metiltransferasas , Tirosina , Humanos , Fosforilación , Janus Quinasa 2/metabolismo , Janus Quinasa 2/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Tirosina/metabolismo , Línea Celular Tumoral , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Metilación , Especificidad por Sustrato , Células HEK293 , Ciclo Celular , Mutación
2.
J Clin Invest ; 133(13)2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37200093

RESUMEN

During emergency hematopoiesis, hematopoietic stem cells (HSCs) rapidly proliferate to produce myeloid and lymphoid effector cells, a response that is critical against infection or tissue injury. If unresolved, this process leads to sustained inflammation, which can cause life-threatening diseases and cancer. Here, we identify a role of double PHD fingers 2 (DPF2) in modulating inflammation. DPF2 is a defining subunit of the hematopoiesis-specific BAF (SWI/SNF) chromatin-remodeling complex, and it is mutated in multiple cancers and neurological disorders. We uncovered that hematopoiesis-specific Dpf2-KO mice developed leukopenia, severe anemia, and lethal systemic inflammation characterized by histiocytic and fibrotic tissue infiltration resembling a clinical hyperinflammatory state. Dpf2 loss impaired the polarization of macrophages responsible for tissue repair, induced the unrestrained activation of Th cells, and generated an emergency-like state of HSC hyperproliferation and myeloid cell-biased differentiation. Mechanistically, Dpf2 deficiency resulted in the loss of the BAF catalytic subunit BRG1 from nuclear factor erythroid 2-like 2-controlled (NRF2-controlled) enhancers, impairing the antioxidant and antiinflammatory transcriptional response needed to modulate inflammation. Finally, pharmacological reactivation of NRF2 suppressed the inflammation-mediated phenotypes and lethality of Dpf2Δ/Δ mice. Our work establishes an essential role of the DPF2-BAF complex in licensing NRF2-dependent gene expression in HSCs and immune effector cells to prevent chronic inflammation.


Asunto(s)
Cromatina , Neoplasias , Ratones , Animales , Antioxidantes , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Ensamble y Desensamble de Cromatina , Inflamación/genética , Expresión Génica , Proteínas de Unión al ADN/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Biochem Biophys Res Commun ; 625: 181-187, 2022 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-35970078

RESUMEN

Pathological vascular remodeling and cell proliferation play vital roles in many proliferative vascular diseases. Estrogen can protect the cardiovascular system, but its exact molecular mechanism is unknown. Here we report that 17ß-estradiol (E2) suppressed vascular smooth muscle cells (VSMCs) proliferation and inflammation. qRT-PCR and Western blot demonstrated that E2 decreased NF-κB p50 expression and reduced VSMCs proliferation and inflammation. Mechanistically, a dual luciferase reporter assay and chromatin immunoprecipitation suggested that KLF5 promoted NF-κB p50 expression by binding to the NF-κB p50 promoter, whereas E2 reduced the effect of KLF5 binding to the NF-κB p50 promoter and inhibited NF-κB p50 expression. Furthermore, a coimmunoprecipitation assay and immunofluorescence staining showed that the interaction between KLF5 and ERα increased in VSMCs treated with E2, which in turn decreased NF-κB p50 expression levels. Altogether, we reveal that E2 inhibits VSMCs proliferation and inflammation by reducing NF-κB expression induced by an increased interaction between KLF5 and ERα. These data provide further insights into how E2 inhibits vascular proliferation and inflammation.


Asunto(s)
Músculo Liso Vascular , FN-kappa B , Animales , Células Cultivadas , Estradiol/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Humanos , Inflamación/patología , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Ratones , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo
4.
Cancer Res ; 82(11): 2047-2056, 2022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35320354

RESUMEN

Innate immune cells participate in the detection of tumor cells via complex signaling pathways mediated by pattern-recognition receptors, such as Toll-like receptors and nucleotide-binding and oligomerization domain-like receptors. These pathways are finely tuned via multiple mechanisms, including epigenetic regulation. It is well established that hematopoietic progenitors generate innate immune cells that can regulate cancer cell behavior, and the disruption of normal hematopoiesis in pathologic states may lead to altered immunity and the development of cancer. In this review, we discuss the epigenetic and transcriptional mechanisms that underlie the initiation and amplification of innate immune signaling in cancer. We also discuss new targeting possibilities for cancer control that exploit innate immune cells and signaling molecules, potentially heralding the next generation of immunotherapy.


Asunto(s)
Epigénesis Genética , Inmunidad Innata , Neoplasias , Receptores Toll-Like/metabolismo , Humanos , Inmunidad Innata/genética , Neoplasias/inmunología , Transducción de Señal , Receptores Toll-Like/genética , Transcripción Genética
5.
JCI Insight ; 6(19)2021 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-34622806

RESUMEN

Myelodysplastic syndromes (MDS) are hematopoietic stem and progenitor cell (HSPC) malignancies characterized by ineffective hematopoiesis and an increased risk of leukemia transformation. Epigenetic regulators are recurrently mutated in MDS, directly implicating epigenetic dysregulation in MDS pathogenesis. Here, we identified a tumor suppressor role of the acetyltransferase p300 in clinically relevant MDS models driven by mutations in the epigenetic regulators TET2, ASXL1, and SRSF2. The loss of p300 enhanced the proliferation and self-renewal capacity of Tet2-deficient HSPCs, resulting in an increased HSPC pool and leukemogenicity in primary and transplantation mouse models. Mechanistically, the loss of p300 in Tet2-deficient HSPCs altered enhancer accessibility and the expression of genes associated with differentiation, proliferation, and leukemia development. Particularly, p300 loss led to an increased expression of Myb, and the depletion of Myb attenuated the proliferation of HSPCs and improved the survival of leukemia-bearing mice. Additionally, we show that chemical inhibition of p300 acetyltransferase activity phenocopied Ep300 deletion in Tet2-deficient HSPCs, whereas activation of p300 activity with a small molecule impaired the self-renewal and leukemogenicity of Tet2-deficient cells. This suggests a potential therapeutic application of p300 activators in the treatment of MDS with TET2 inactivating mutations.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular/genética , Leucemia Mieloide Aguda/genética , Síndromes Mielodisplásicos/genética , Factores de Transcripción p300-CBP/genética , Animales , Proteínas de Unión al ADN/genética , Dioxigenasas/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Epigénesis Genética , Células Madre Hematopoyéticas , Leucemia Mieloide Aguda/metabolismo , Ratones , Mutación , Síndromes Mielodisplásicos/metabolismo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Proteínas Represoras/genética , Factores de Empalme Serina-Arginina/genética , Tasa de Supervivencia
6.
Oncogene ; 40(40): 5950-5962, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34373577

RESUMEN

The activity of Rho family GTPase protein, RAC1, which plays important normal physiological functions, is dysregulated in multiple cancers. RAC1 is expressed in both estrogen receptor alpha (ER)-positive and ER-negative breast cancer (BC) cells. However, ER-positive BC is more sensitive to RAC1 inhibition. We have determined that reducing RAC1 activity, using siRNA or EHT 1864 (a small molecule Rac inhibitor), leads to rapid ER protein degradation. RAC1 interacts with ER within the ER complex and RAC1 localizes to chromatin binding sites for ER upon estrogen treatment. RAC1 activity is important for RNA Pol II function at both promoters and enhancers of ER target genes and ER-regulated gene transcription is blocked by EHT 1864, in a dose-dependent manner. Having identified that RAC1 is an essential ER cofactor for ER protein stability and ER transcriptional activity, we report that RAC1 inhibition could be an effective therapeutic approach for ER-positive BC.


Asunto(s)
Receptor alfa de Estrógeno/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Femenino , Humanos , Transfección
7.
Mediators Inflamm ; 2021: 9921897, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34220338

RESUMEN

Excessive release of cytokines such as IL-1ß and other inflammatory mediators synthesized and secreted by macrophages is the fundamental link of uncontrolled inflammatory response in sepsis. 17ß-Estradiol (E2) plays anti-inflammatory and vascular protective effects by regulating leukocyte infiltration and the expression of chemokines or cytokines induced by injury. However, the role of E2 in the inflammatory response of macrophages in sepsis and its mechanism are still not fully understood. In the present study, we show that E2 alleviates vascular inflammation in sepsis mice induced by cecal ligation puncture (CLP). E2 significantly decreases RAW 264.7 cell inflammation response by downregulating the expression of NLRP3. Furthermore, we found that miR-29a-5p was significantly downregulated in LPS-treated macrophages. Treating RAW 264.7 cells with E2 markedly upregulated the miR-29a-5p expression level. More importantly, we demonstrated that miR-29a-5p repressed NLRP3 expression by directly targeting its 3'-UTR. Loss- and gain-of-function experiments revealed that transfection of the miR-29a-5p mimic abrogates LPS-induced macrophage inflammation. Moreover, depletion of miR-29a-5p by its inhibitor largely promotes LPS-induced macrophage inflammation. In summary, miR-29a-5p upregulation induced by E2 alleviated RAW 264.7 cell inflammation response by aggravating miR-29a-5p repression of NLRP3 expression. E2 exerts significant anti-inflammatory efficacy in macrophages by regulating the miR-29a-5p/NLRP3 axis. Targeting miR-29a-5p may be a novel therapeutic strategy to suppress sepsis-induced vascular inflammation.


Asunto(s)
Estradiol/metabolismo , Regulación de la Expresión Génica , Lipopolisacáridos/metabolismo , Macrófagos/metabolismo , MicroARNs/metabolismo , Sepsis/metabolismo , Regiones no Traducidas 3' , Animales , Antiinflamatorios/uso terapéutico , Células HEK293 , Humanos , Técnicas In Vitro , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Células RAW 264.7 , Sepsis/fisiopatología , Regulación hacia Arriba
8.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 36(1): 62-66, 2020 Jan 28.
Artículo en Chino | MEDLINE | ID: mdl-32476374

RESUMEN

OBJECTIVE: To investigate the effects of cerium oxide (CeO2) nanoparticles on the viabilities of nerve cells PC12 and SH-SY5Y. METHODS: CeO2 nanoparticles were synthesized, structures were characterized and properties were evaluated. PC12 cells and SH-SY5Y cells were treated with CeO2 nanoparticles at different concentrations (1, 2.5, 5, 10, 25, 50, 75, 100, 150 µg/ml) for 24 h and the cell viability was measured by MTT assay. Then PC12 cells and SH-SY5Y cells were co-treated with CeO2 and active oxygen scavenger NAC and the cells were stained with DCFH-DA probe for ROS. The number of cells and the fluorescence intensity were observed under a fluorescent inverted microscope. Differences were assessed by one-way ANOVA. RESULTS: After treatment with CeO2 nanoparticles, the viabilities of both PC12 cells (P<0.01) and SH-SY5Y cells (P<0.01) were decreased comparing with the control group. After staining with DCFH-DA probe, the fluorescence intensity of the nerve cells was enhanced depending on the concentration of CeO2 nanoparticles suggesting that CeO2 induced the generation of reactive oxygen species (ROS). The fluorescence intensity of PC12 cells was decreased after CeO2 nanoparticles (100 µg/ml) co-treatment with active oxygen scavenger NAC. Compared with CeO2 nanoparticles alone at 25 µg/ml (P<0.01), 50 µg/ml (P<0.01), 75 µg/ml (P<0.01), 100 µg/ml (P<0.01), the cell viability was significantly increased after co-treatment with NAC. CONCLUSION: CeO2 nanoparticles has a negative effect on the viabilities of nerve cells PC12 and SH-SY5Y, and the effect might be depend on ROS.


Asunto(s)
Supervivencia Celular , Cerio/farmacología , Nanopartículas , Neuronas/efectos de los fármacos , Animales , Línea Celular Tumoral , Humanos , Células PC12 , Ratas , Especies Reactivas de Oxígeno/metabolismo
9.
Nucleic Acids Res ; 48(6): 2956-2968, 2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32025719

RESUMEN

Protein arginine methyltransferase 5 (PRMT5) catalyzes the symmetric di-methylation of arginine residues in histones H3 and H4, marks that are generally associated with transcriptional repression. However, we found that PRMT5 inhibition or depletion led to more genes being downregulated than upregulated, indicating that PRMT5 can also act as a transcriptional activator. Indeed, the global level of histone H3K27me3 increases in PRMT5 deficient cells. Although PRMT5 does not directly affect PRC2 enzymatic activity, methylation of histone H3 by PRMT5 abrogates its subsequent methylation by PRC2. Treating AML cells with an EZH2 inhibitor partially restored the expression of approximately 50% of the genes that are initially downregulated by PRMT5 inhibition, suggesting that the increased H3K27me3 could directly or indirectly contribute to the transcription repression of these genes. Indeed, ChIP-sequencing analysis confirmed an increase in the H3K27me3 level at the promoter region of a quarter of these genes in PRMT5-inhibited cells. Interestingly, the anti-proliferative effect of PRMT5 inhibition was also partially rescued by treatment with an EZH2 inhibitor in several leukemia cell lines. Thus, PRMT5-mediated crosstalk between histone marks contributes to its functional effects.


Asunto(s)
Arginina/metabolismo , Histonas/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Transcripción Genética , Animales , Ciclo Celular/genética , Línea Celular Tumoral , Eliminación de Gen , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Metilación , Ratones Noqueados , Modelos Biológicos , Nucleosomas/metabolismo , Proteína-Arginina N-Metiltransferasas/antagonistas & inhibidores
11.
Nat Commun ; 10(1): 4925, 2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31664040

RESUMEN

AML1-ETO (AE) is a fusion transcription factor, generated by the t(8;21) translocation, that functions as a leukemia promoting oncogene. Here, we demonstrate that TATA-Box Binding Protein Associated Factor 1 (TAF1) associates with K43 acetylated AE and this association plays a pivotal role in the proliferation of AE-expressing acute myeloid leukemia (AML) cells. ChIP-sequencing indicates significant overlap of the TAF1 and AE binding sites. Knockdown of TAF1 alters the association of AE with chromatin, affecting of the expression of genes that are activated or repressed by AE. Furthermore, TAF1 is required for leukemic cell self-renewal and its reduction promotes the differentiation and apoptosis of AE+ AML cells, thereby impairing AE driven leukemogenesis. Together, our findings reveal a role of TAF1 in leukemogenesis and identify TAF1 as a potential therapeutic target for AE-expressing leukemia.


Asunto(s)
Carcinogénesis/patología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Histona Acetiltransferasas/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factor de Transcripción TFIID/metabolismo , Acetilación , Animales , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Autorrenovación de las Células , Cromatina/metabolismo , Regulación Leucémica de la Expresión Génica , Histona Acetiltransferasas/química , Humanos , Lisina/metabolismo , Ratones Endogámicos C57BL , Células Mieloides/patología , Unión Proteica , Dominios Proteicos , Factores Asociados con la Proteína de Unión a TATA/química , Factor de Transcripción TFIID/química
13.
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
15.
J Clin Invest ; 128(12): 5383-5398, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30226831

RESUMEN

ASXL1 is frequently mutated in myeloid malignancies and is known to co-occur with other gene mutations. However, the molecular mechanisms underlying the leukemogenesis associated with ASXL1 and cooperating mutations remain to be elucidated. Here, we report that Asxl1 loss cooperated with haploinsufficiency of Nf1, a negative regulator of the RAS signaling pathway, to accelerate the development of myeloid leukemia in mice. Loss of Asxl1 and Nf1 in hematopoietic stem and progenitor cells resulted in a gain-of-function transcriptional activation of multiple pathways such as MYC, NRAS, and BRD4 that are critical for leukemogenesis. The hyperactive MYC and BRD9 transcription programs were correlated with elevated H3K4 trimethylation at the promoter regions of genes involving these pathways. Furthermore, pharmacological inhibition of both the MAPK pathway and BET bromodomain prevented leukemia initiation and inhibited disease progression in Asxl1Δ/Δ Nf1Δ/Δ mice. Concomitant mutations of ASXL1 and RAS pathway genes were associated with aggressive progression of myeloid malignancies in patients. This study sheds light on the effect of cooperation between epigenetic alterations and signaling pathways on accelerating the progression of myeloid malignancies and provides a rational therapeutic strategy for the treatment of myeloid malignancies with ASXL1 and RAS pathway gene mutations.


Asunto(s)
Epigénesis Genética , Regulación Leucémica de la Expresión Génica , Haploinsuficiencia , Células Madre Hematopoyéticas , Leucemia Mieloide , Mutación , Neurofibromina 1 , Proteínas Represoras , Transducción de Señal , Animales , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Histonas/genética , Histonas/metabolismo , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Metilación , Ratones , Ratones Transgénicos , Neurofibromina 1/genética , Neurofibromina 1/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
16.
Cell Rep ; 24(10): 2643-2657, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30184499

RESUMEN

Protein arginine methyltransferase 5 (PRMT5) is overexpressed in many cancer types and is a promising therapeutic target for several of them, including leukemia and lymphoma. However, we and others have reported that PRMT5 is essential for normal physiology. This dependence may become dose limiting in a therapeutic setting, warranting the search for combinatorial approaches. Here, we report that PRMT5 depletion or inhibition impairs homologous recombination (HR) DNA repair, leading to DNA-damage accumulation, p53 activation, cell-cycle arrest, and cell death. PRMT5 symmetrically dimethylates histone and non-histone substrates, including several components of the RNA splicing machinery. We find that PRMT5 depletion or inhibition induces aberrant splicing of the multifunctional histone-modifying and DNA-repair factor TIP60/KAT5, which selectively affects its lysine acetyltransferase activity and leads to impaired HR. As HR deficiency sensitizes cells to PARP inhibitors, we demonstrate here that PRMT5 and PARP inhibitors have synergistic effects on acute myeloid leukemia cells.


Asunto(s)
Proteína-Arginina N-Metiltransferasas/metabolismo , Empalme Alternativo/genética , Empalme Alternativo/fisiología , Puntos de Control del Ciclo Celular/genética , Puntos de Control del Ciclo Celular/fisiología , Muerte Celular , Línea Celular Tumoral , Reparación del ADN/genética , Reparación del ADN/fisiología , Código de Histonas/genética , Código de Histonas/fisiología , Histonas/metabolismo , Humanos , Lisina Acetiltransferasa 5/genética , Lisina Acetiltransferasa 5/metabolismo , Lisina Acetiltransferasas/genética , Lisina Acetiltransferasas/metabolismo , Proteína-Arginina N-Metiltransferasas/genética
17.
Cancer Cell ; 33(6): 1111-1127.e5, 2018 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-29894694

RESUMEN

Chromatin-modifying enzymes, and specifically the protein arginine methyltransferases (PRMTs), have emerged as important targets in cancer. Here, we investigated the role of CARM1 in normal and malignant hematopoiesis. Using conditional knockout mice, we show that loss of CARM1 has little effect on normal hematopoiesis. Strikingly, knockout of Carm1 abrogates both the initiation and maintenance of acute myeloid leukemia (AML) driven by oncogenic transcription factors. We show that CARM1 knockdown impairs cell-cycle progression, promotes myeloid differentiation, and ultimately induces apoptosis. Finally, we utilize a selective, small-molecule inhibitor of CARM1 to validate the efficacy of CARM1 inhibition in leukemia cells in vitro and in vivo. Collectively, this work suggests that targeting CARM1 may be an effective therapeutic strategy for AML.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Hematopoyesis/genética , Leucemia Mieloide/genética , Proteína-Arginina N-Metiltransferasas/genética , Enfermedad Aguda , Animales , Apoptosis/genética , Ciclo Celular/genética , Línea Celular Tumoral , Perfilación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos , Proteína-Arginina N-Metiltransferasas/metabolismo
18.
Oncotarget ; 9(42): 26603-26604, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29928471
19.
Int J Med Sci ; 15(3): 228-237, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29483814

RESUMEN

Aims: Whether pioglitazone (PIO), a peroxisome proliferator-activated receptor-gamma agonist, increases the risk of developing bladder cancer has been debated for several years. The aim of this study was to investigate the in vitro effects of PIO on normal urothelial transitional epithelium (NUTE) cells and bladder cancer (J82) cells to further evaluate the risk. Methods: NUTE cells were obtained from Sprague-Dawley rats. NUTE and J82 cells were treated with different concentrations of PIO for various time periods. Cell proliferation was tested by the MTT assay. Cell apoptosis was evaluated by flow cytometry. The expressions of p53, cyclin D1, Bcl-2, and Bax were determined by qRT-PCR and western blots. Results: After 24 hours, the treatment of NUTE cells with 10 µmol/L PIO led to morphological changes, without changes in J82 cells. Moreover, PIO inhibited the proliferation and induced apoptosis of NUTE cells, but not J82 cells, in a time- and dose-dependent manner. However, PIO did not alter the growth of cells from other tissues. In addition, treatment with PIO for up to 72 hours did not result in changes in the expressions of p53, cyclin D1, Bcl-2, and Bax in NUTE cells and J82 cells. Interestingly, PIO significantly downregulated the protein levels of p53 and cyclin D1 in J82 cells, but not NUTE cells after more than 192 hours of treatment. Conclusions: PIO did not promote malignant alterations of NUTE cells or stimulate proliferation of J82 cells. PIO decreased the expression of p53 and cyclin D1 in J82 cells after long-term culture, which suggested that PIO may be helpful for diabetic patients with bladder cancer.


Asunto(s)
Complicaciones de la Diabetes/tratamiento farmacológico , Hipoglucemiantes/administración & dosificación , Tiazolidinedionas/administración & dosificación , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclina D1/genética , Complicaciones de la Diabetes/genética , Complicaciones de la Diabetes/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , PPAR gamma/agonistas , Pioglitazona , Proteínas Proto-Oncogénicas c-bcl-2/genética , Ratas , Factores de Riesgo , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética , Neoplasias de la Vejiga Urinaria/complicaciones , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología , Urotelio/efectos de los fármacos , Urotelio/patología
20.
Nat Commun ; 8: 15456, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28593990

RESUMEN

ASXL2 is frequently mutated in acute myeloid leukaemia patients with t(8;21). However, the roles of ASXL2 in normal haematopoiesis and the pathogenesis of myeloid malignancies remain unknown. Here we show that deletion of Asxl2 in mice leads to the development of myelodysplastic syndrome (MDS)-like disease. Asxl2-/- mice have an increased bone marrow (BM) long-term haematopoietic stem cells (HSCs) and granulocyte-macrophage progenitors compared with wild-type controls. Recipients transplanted with Asxl2-/- and Asxl2+/- BM cells have shortened lifespan due to the development of MDS-like disease or myeloid leukaemia. Paired daughter cell assays demonstrate that Asxl2 loss enhances the self-renewal of HSCs. Deletion of Asxl2 alters the expression of genes critical for HSC self-renewal, differentiation and apoptosis in Lin-cKit+ cells. The altered gene expression is associated with dysregulated H3K27ac and H3K4me1/2. Our study demonstrates that ASXL2 functions as a tumour suppressor to maintain normal HSC function.


Asunto(s)
Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Síndromes Mielodisplásicos/patología , Células Mieloides/metabolismo , Proteínas Represoras/deficiencia , Animales , Linaje de la Célula , Autorrenovación de las Células , Progresión de la Enfermedad , Eliminación de Gen , Regulación Leucémica de la Expresión Génica , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Histonas/metabolismo , Leucemia Mieloide Aguda/genética , Lisina/metabolismo , Ratones , Síndromes Mielodisplásicos/genética , Células Mieloides/patología , Procesamiento Proteico-Postraduccional , Proteínas Represoras/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA