Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 48(20): 11434-11451, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33080033

RESUMEN

Core regulatory circuitry (CRC)-dependent transcriptional network is critical for developmental tumors in children and adolescents carrying few gene mutations. However, whether and how CRC contributes to transcription regulation in Ewing sarcoma is unknown. Here, we identify and functionally validate a CRC 'trio' constituted by three transcription factors (TFs): KLF15, TCF4 and NKX2-2, in Ewing sarcoma cells. Epigenomic analyses demonstrate that EWS-FLI1, the primary fusion driver for this cancer, directly establishes super-enhancers of each of these three TFs to activate their transcription. In turn, KLF15, TCF4 and NKX2-2 co-bind to their own and each other's super-enhancers and promoters, forming an inter-connected auto-regulatory loop. Functionally, CRC factors contribute significantly to cell proliferation of Ewing sarcoma both in vitro and in vivo. Mechanistically, CRC factors exhibit prominent capacity of co-regulating the epigenome in cooperation with EWS-FLI1, occupying 77.2% of promoters and 55.6% of enhancers genome-wide. Downstream, CRC TFs coordinately regulate gene expression networks in Ewing sarcoma, controlling important signaling pathways for cancer, such as lipid metabolism pathway, PI3K/AKT and MAPK signaling pathways. Together, molecular characterization of the oncogenic CRC model advances our understanding of the biology of Ewing sarcoma. Moreover, CRC-downstream genes and signaling pathways may contain potential therapeutic targets for this malignancy.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Sarcoma de Ewing/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Metabolismo de los Lípidos/genética , Ratones Desnudos , Proteínas Nucleares , Proteínas de Fusión Oncogénica/fisiología , Proteína Proto-Oncogénica c-fli-1/fisiología , Proteína EWS de Unión a ARN/fisiología , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología , Transducción de Señal , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Factores de Transcripción , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
2.
Nucleic Acids Res ; 47(3): 1255-1267, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30496486

RESUMEN

As the second most common malignant bone tumor in children and adolescents, Ewing sarcoma is initiated and exacerbated by a chimeric oncoprotein, most commonly, EWS-FLI1. In this study, we apply epigenomic analysis to characterize the transcription dysregulation in this cancer, focusing on the investigation of super-enhancer and its associated transcriptional regulatory mechanisms. We demonstrate that super-enhancer-associated transcripts are significantly enriched in EWS-FLI1 target genes, contribute to the aberrant transcriptional network of the disease, and mediate the exceptional sensitivity of Ewing sarcoma to transcriptional inhibition. Through integrative analysis, we identify MEIS1 as a super-enhancer-driven oncogene, which co-operates with EWS-FLI1 in transcriptional regulation, and plays a key pro-survival role in Ewing sarcoma. Moreover, APCDD1, another super-enhancer-associated gene, acting as a downstream target of both MEIS1 and EWS-FLI1, is also characterized as a novel tumor-promoting factor in this malignancy. These data delineate super-enhancer-mediated transcriptional deregulation in Ewing sarcoma, and uncover numerous candidate oncogenes which can be exploited for further understanding of the molecular pathogenesis for this disease.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Sarcoma de Ewing/genética , Transcripción Genética , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Elementos de Facilitación Genéticos , Regulación Neoplásica de la Expresión Génica , Humanos , Motivos de Nucleótidos/genética , Proteínas de Fusión Oncogénica/genética , Proteína Proto-Oncogénica c-fli-1/genética , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/patología , Transducción de Señal/genética
3.
Proc Natl Acad Sci U S A ; 114(47): 12548-12553, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29109286

RESUMEN

The circadian system regulates numerous physiological processes including immune responses. Here, we show that mice deficient of the circadian clock genes Cry1 and Cry2 [Cry double knockout (DKO)] develop an autoimmune phenotype including high serum IgG concentrations, serum antinuclear antibodies, and precipitation of IgG, IgM, and complement 3 in glomeruli and massive infiltration of leukocytes into the lungs and kidneys. Flow cytometry of lymphoid organs revealed decreased pre-B cell numbers and a higher percentage of mature recirculating B cells in the bone marrow, as well as increased numbers of B2 B cells in the peritoneal cavity of Cry DKO mice. The B cell receptor (BCR) proximal signaling pathway plays a critical role in autoimmunity regulation. Activation of Cry DKO splenic B cells elicited markedly enhanced tyrosine phosphorylation of cellular proteins compared with cells from control mice, suggesting that overactivation of the BCR-signaling pathway may contribute to the autoimmunity phenotype in the Cry DKO mice. In addition, the expression of C1q, the deficiency of which contributes to the pathogenesis of systemic lupus erythematosus, was significantly down-regulated in Cry DKO B cells. Our results suggest that B cell development, the BCR-signaling pathway, and C1q expression are regulated by circadian clock CRY proteins and that their dysregulation through loss of CRY contributes to autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Autoinmunidad/genética , Linfocitos B/inmunología , Relojes Circadianos/inmunología , Criptocromos/inmunología , Animales , Anticuerpos Antinucleares/biosíntesis , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/metabolismo , Enfermedades Autoinmunes/patología , Linfocitos B/metabolismo , Linfocitos B/patología , Relojes Circadianos/genética , Complemento C1q/genética , Criptocromos/deficiencia , Criptocromos/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , Riñón/inmunología , Riñón/patología , Pulmón/inmunología , Pulmón/patología , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal , Bazo/inmunología , Bazo/metabolismo , Bazo/patología
4.
Int J Cancer ; 134(1): 207-17, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23775785

RESUMEN

We have synthesized 39 1,25-dihydroxyvitamin D3 [1,25(OH)2D3] analogs having two side chains attached to carbon-20 (Gemini) with various modifications and compared their anticancer activities. Five structure-function rules emerged to identify analogs with enhanced anticancer activity. One of these active analogs, BXL-01-0126, was more potent than 1,25(OH)2D3 in mediating 50% clonal inhibition of cancer cell growth. Murine studies found that BXL-01-0126 and 1,25(OH)2D3 had nearly the same potency to raise serum calcium levels. Taken together, BXL-01-0126 when compared to 1,25(OH)2D3 has greater anticancer potency, but similar toxicity causing hypercalcemia. We focused on the effect of these compounds on the stimulation of expression of human cathelicidin antimicrobial peptide (CAMP) whose gene has a vitamin D response element in its promoter. Expression of CAMP mRNA and protein increased in a dose-response fashion after exposure of acute myeloid leukemia (AML) cells to the Gemini analog, BXL-01-126, in vitro. A xenograft model of AML was developed using U937 AML cells injected into NSG-immunodeficient mice. Administration of vitamin D3 compounds to these mice resulted in substantial levels of CAMP in the systemic circulation. This suggests a unique prophylactic treatment at diagnosis or during induction chemotherapy for AML patients to provide them with protection against various microbial infections through CAMP induction.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Calcitriol/análogos & derivados , Catelicidinas/biosíntesis , Colecalciferol/farmacología , Animales , Péptidos Catiónicos Antimicrobianos , Antineoplásicos/química , Calcitriol/síntesis química , Calcitriol/química , Calcitriol/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colecalciferol/análogos & derivados , Colecalciferol/síntesis química , Citometría de Flujo , Xenoinjertos , Humanos , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Relación Estructura-Actividad
5.
Blood ; 120(16): 3310-7, 2012 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-22942183

RESUMEN

Fms-like tyrosine kinase 3 (FLT3) is a receptor tyrosine kinase with important roles in hematopoietic progenitor cell survival and proliferation. It is mutated in approximately one-third of AML patients, mostly by internal tandem duplications (ITDs). Adaptor protein Lnk is a negative regulator of hematopoietic cytokine signaling. In the present study, we show that Lnk interacts physically with both wild-type FLT3 (FLT3-WT) and FLT3-ITD through the SH2 domains. We have identified the tyrosine residues 572, 591, and 919 of FLT3 as phosphorylation sites involved in direct binding to Lnk. Lnk itself was tyrosine phosphorylated by both FLT3 ligand (FL)-activated FLT3-WT and constitutively activated FLT3-ITD. Both shRNA-mediated depletion and forced overexpression of Lnk demonstrated that activation signals emanating from both forms of FLT3 are under negative regulation by Lnk. Moreover, Lnk inhibited 32D cell proliferation driven by different FLT3 variants. Analysis of primary BM cells from Lnk-knockout mice showed that Lnk suppresses the expansion of FL-stimulated hematopoietic progenitors, including lymphoid-primed multipotent progenitors. The results of the present study show that through direct binding to FLT3, Lnk suppresses FLT3-WT/ITD-dependent signaling pathways involved in the proliferation of hematopoietic cells. Therefore, modulation of Lnk expression levels may provide a unique therapeutic approach for FLT3-ITD-associated hematopoietic disease.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Células Madre Hematopoyéticas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Leucemia Mieloide Aguda/metabolismo , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales , Animales , Proliferación Celular , Transformación Celular Neoplásica/patología , Ensayo de Unidades Formadoras de Colonias , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Leucemia Mieloide Aguda/patología , Proteínas de la Membrana , Ratones , Ratones Noqueados , Mutación/genética , Fosforilación , ARN Interferente Pequeño/genética , Transducción de Señal , Secuencias Repetidas en Tándem/genética , Tirosina/metabolismo , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/metabolismo , Dominios Homologos src
6.
Nat Commun ; 15(1): 2484, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38509096

RESUMEN

Squamous cell carcinomas (SCCs) are common and aggressive malignancies. Immune check point blockade (ICB) therapy using PD-1/PD-L1 antibodies has been approved in several types of advanced SCCs. However, low response rate and treatment resistance are common. Improving the efficacy of ICB therapy requires better understanding of the mechanism of immune evasion. Here, we identify that the SCC-master transcription factor TP63 suppresses interferon-γ (IFNγ) signaling. TP63 inhibition leads to increased CD8+ T cell infiltration and heighten tumor killing in in vivo syngeneic mouse model and ex vivo co-culture system, respectively. Moreover, expression of TP63 is negatively correlated with CD8+ T cell infiltration and activation in patients with SCC. Silencing of TP63 enhances the anti-tumor efficacy of PD-1 blockade by promoting CD8+ T cell infiltration and functionality. Mechanistically, TP63 and STAT1 mutually suppress each other to regulate the IFNγ signaling by co-occupying and co-regulating their own promoters and enhancers. Together, our findings elucidate a tumor-extrinsic function of TP63 in promoting immune evasion of SCC cells. Over-expression of TP63 may serve as a biomarker predicting the outcome of SCC patients treated with ICB therapy, and targeting TP63/STAT/IFNγ axis may enhance the efficacy of ICB therapy for this deadly cancer.


Asunto(s)
Carcinoma de Células Escamosas , Interferón gamma , Animales , Humanos , Ratones , Antígeno B7-H1/metabolismo , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Linfocitos T CD8-positivos , Línea Celular Tumoral , Inmunidad , Interferón gamma/metabolismo , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/metabolismo , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factores de Transcripción/metabolismo , Microambiente Tumoral , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
7.
Int J Cancer ; 132(12): 2730-7, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23165325

RESUMEN

Despite recent advances in therapy, breast cancer remains the second most common cause of death from malignancy in women. Chemotherapy plays a major role in breast cancer management, and combining chemotherapeutic agents with nonchemotherapeutic agents is of considerable clinical interest. Cucurbitacins are triterpenes compounds found in plants of the Cucurbitaceae family, reported to have anticancer and anti-inflammatory activities. Previously, we have shown antiproliferative activity of cucurbitacin B (CuB) in breast cancer, and we hypothesized that combining CuB with chemotherapeutic agents can augment their antitumor effect. Here, we show that a combination of CuB with either docetaxel (DOC) or gemcitabine (GEM) synergistically inhibited the proliferation of MDA-MB-231 breast cancer cells in vitro. This antiproliferative effect was accompanied by an increase in apoptosis rates. Furthermore, in vivo treatment of human breast cancer orthotopic xenografts in immunodeficient mice with CuB at either low (0.5 mg/kg) or high (1 mg/kg) doses in combination with either DOC (20 mg/kg) or GEM (12.5mg/kg) significantly reduced tumor volume as compared with monotherapy of each drug. Importantly, no significant toxicity was noted with low-dose CuB in combination with either DOC or GEM. In conclusion, combination of CuB at a relatively low concentration with either of the chemotherapeutic agents, DOC or GEM, shows prominent antiproliferative activity against breast cancer cells without increased toxicity. This promising combination should be examined in therapeutic trials of breast cancer.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama/patología , Triterpenos/farmacología , Animales , Antineoplásicos Fitogénicos/toxicidad , Apoptosis/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Docetaxel , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Taxoides/farmacología , Triterpenos/toxicidad , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
8.
Br J Haematol ; 161(6): 811-20, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23590807

RESUMEN

Somatic point mutations in the PH domain of SH2B3 (LNK), an adaptor protein that is highly expressed in haematopoietic cells, were recently described in patients with myeloproliferative neoplasms. We studied the effect of these mutations on the JAK2 signalling pathway in cells expressing either wild type JAK2 or the JAK2 V617F mutation. Compared to wild type SH2B3, PH domain mutants have mild loss of function, with no evidence for a dominant-negative effect. Mutants retain binding capacity for JAK2, an established SH2B3 target, as well as for the adaptor proteins 14-3-3 and CBL. Our data suggest that the loss of SH2B3 inhibitory function conferred by the PH domain mutations is mild and may collaborate with JAK2 V617F and CBL mutations in order to promote either the development or the progression of myeloproliferative neoplasms.


Asunto(s)
Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Mutación , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/metabolismo , Proteínas/genética , Proteínas 14-3-3/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Proteínas/química , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Transducción de Señal
9.
J Ovarian Res ; 16(1): 80, 2023 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-37087441

RESUMEN

BACKGROUND: Standard platinum-based therapy for ovarian cancer is inefficient against ovarian clear cell carcinoma (OCCC). OCCC is a distinct subtype of epithelial ovarian cancer. OCCC constitutes 25% of ovarian cancers in East Asia (Japan, Korea, China, Singapore) and 6-10% in Europe and North America. The cancer is characterized by frequent inactivation of ARID1A and 10% of cases of endometriosis progression to OCCC. The aim of this study was to identify drugs that are either FDA-approved or in clinical trials for the treatment of OCCC. RESULTS: High throughput screening of 166 compounds that are either FDA-approved, in clinical trials or are in pre-clinical studies identified several cytotoxic compounds against OCCC. ARID1A knockdown cells were more sensitive to inhibitors of either mTOR (PP242), dual mTOR/PI3K (GDC0941), ATR (AZD6738) or MDM2 (RG7388) compared to control cells. Also, compounds targeting BH3 domain (AZD4320) and SRC (AZD0530) displayed preferential cytotoxicity against ARID1A mutant cell lines. In addition, WEE1 inhibitor (AZD1775) showed broad cytotoxicity toward OCCC cell lines, irrespective of ARID1A status. CONCLUSIONS: In a selection of 166 compounds we showed that inhibitors of ATR and WEE1 were cytotoxic against a panel of OCCC cell lines. These two drugs are already in other clinical trials, making them ideal candidates for treatment of OCCC.


Asunto(s)
Adenocarcinoma de Células Claras , Proteínas de la Ataxia Telangiectasia Mutada , Neoplasias Ováricas , Proteínas Tirosina Quinasas , Femenino , Humanos , Adenocarcinoma de Células Claras/tratamiento farmacológico , Adenocarcinoma de Células Claras/patología , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Carcinoma Epitelial de Ovario , Proteínas de Ciclo Celular/metabolismo , China , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
10.
Curr Opin Genet Dev ; 17(1): 78-83, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17210248

RESUMEN

Transcription factors comprise a high proportion of the proteins with dysregulated expression or altered activity in cancer cells. In recent years, great progress has been made in elucidating crucial pathways in leukemogenesis, partially by identifying key transcription factors that are normally indispensable for hematopoiesis but which are often associated with malignant transformation when functioning improperly. The significance of transcription factors is highlighted by the multiple mechanisms used by leukemic cells to abrogate their normal activity. Delineating these mechanisms can benefit the diagnosis and treatment of hematological malignancies and might also be applicable to certain solid tumors.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Hematológicas/genética , Transducción de Señal/genética , Factores de Transcripción/genética , Translocación Genética/genética , Humanos , Modelos Biológicos , Mutación Puntual/genética
11.
Oncotarget ; 12(18): 1749-1762, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34504648

RESUMEN

Triple negative breast cancer (TNBC) is a deadly disease with limited treatment options. Selinexor is a selective inhibitor of nuclear export that binds covalently to exportin 1 thereby reactivating tumor suppressor proteins and downregulating expression of oncogenes and DNA damage repair (DDR) proteins. Olaparib is a poly (ADP-ribose) polymerase (PARP) inhibitor approved for the treatment of patients with breast cancer harboring BRCA mutations. We examined the effects of co-treatment with selinexor and olaparib in TNBC cell lines. BRCA1 wildtype (BRCA1-wt) and BRCA1 mutant (BRCA1-mut) TNBC cell lines were treated with selinexor and/or olaparib and effects on cell viability and cell cycle were evaluated. The effects of treatment were also evaluated in mouse xenograft models generated with BRCA1-wt and BRCA1-mut TNBC cell lines. Treatment with selinexor inhibited cell proliferation and survival of all TNBC cell lines tested in vitro. This effect was enhanced following treatment of the cells with the combination of selinexor and olaparib, which showed synergistic effects on tumor growth inhibition in MDA-MB-468-derived (BRCA1-wt) and MDA-MB-436-derived (BRCA1-mut) xenografts. As co-treatment with selinexor and olaparib exhibits anti-tumor activity regardless of BRCA1 mutation status, the clinical implications of the combination warrant further investigation.

12.
Oncogene ; 40(10): 1851-1867, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33564073

RESUMEN

Soft tissue sarcoma (STS) is a heterogeneous disease that arises from connective tissues. Clinical outcome of patients with advanced tumors especially de-differentiated liposarcoma and uterine leiomyosarcoma remains unsatisfactory, despite intensive treatment regimens including maximal surgical resection, radiation, and chemotherapy. MAP kinase-interacting serine/threonine-protein kinase 1 and 2 (MNK1/2) have been shown to contribute to oncogenic translation via phosphorylation of eukaryotic translation initiation factor 4E (eIF4E). However, little is known about the role of MNK1/2 and their downstream targets in STS. In this study, we show that depletion of either MNK1 or MNK2 suppresses cell viability, anchorage-independent growth, and tumorigenicity of STS cells. We also identify a compelling antiproliferative efficacy of a novel, selective MNK inhibitor ETC-168. Cellular responsiveness of STS cells to ETC-168 correlates positively with that of phosphorylated ribosomal protein S6 (RPS6). Mirroring MNK1/2 silencing, ETC-168 treatment strongly blocks eIF4E phosphorylation and represses expression of sarcoma-driving onco-proteins including E2F1, FOXM1, and WEE1. Moreover, combination of ETC-168 and MCL1 inhibitor S63845 exerts a synergistic antiproliferative activity against STS cells. In summary, our study reveals crucial roles of MNK1/2 and their downstream targets in STS tumorigenesis. Our data encourage further clinical translation of MNK inhibitors for STS treatment.


Asunto(s)
Proteínas de Ciclo Celular/genética , Factor de Transcripción E2F1/genética , Proteína Forkhead Box M1/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Sarcoma/tratamiento farmacológico , Carcinogénesis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Sarcoma/genética , Sarcoma/patología , Tiofenos/farmacología
13.
Nat Commun ; 12(1): 4362, 2021 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-34272396

RESUMEN

Squamous cell carcinomas (SCCs) comprise one of the most common histologic types of human cancer. Transcriptional dysregulation of SCC cells is orchestrated by tumor protein p63 (TP63), a master transcription factor (TF) and a well-researched SCC-specific oncogene. In the present study, both Gene Set Enrichment Analysis (GSEA) of SCC patient samples and in vitro loss-of-function assays establish fatty-acid metabolism as a key pathway downstream of TP63. Further studies identify sterol regulatory element binding transcription factor 1 (SREBF1) as a central mediator linking TP63 with fatty-acid metabolism, which regulates the biosynthesis of fatty-acids, sphingolipids (SL), and glycerophospholipids (GPL), as revealed by liquid chromatography tandem mass spectrometry (LC-MS/MS)-based lipidomics. Moreover, a feedback co-regulatory loop consisting of SREBF1/TP63/Kruppel like factor 5 (KLF5) is identified, which promotes overexpression of all three TFs in SCCs. Downstream of SREBF1, a non-canonical, SCC-specific function is elucidated: SREBF1 cooperates with TP63/KLF5 to regulate hundreds of cis-regulatory elements across the SCC epigenome, which converge on activating cancer-promoting pathways. Indeed, SREBF1 is essential for SCC viability and migration, and its overexpression is associated with poor survival in SCC patients. Taken together, these data shed light on mechanisms of transcriptional dysregulation in cancer, identify specific epigenetic regulators of lipid metabolism, and uncover SREBF1 as a potential therapeutic target and prognostic marker in SCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Metabolismo de los Lípidos/genética , Neoplasias Pulmonares/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Acetilación , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Secuenciación de Inmunoprecipitación de Cromatina , Cromatografía Liquida , Epigenómica , Receptores ErbB/genética , Receptores ErbB/metabolismo , Neoplasias Esofágicas/genética , Ácidos Grasos/biosíntesis , Ácidos Grasos/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Histonas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Neoplasias Pulmonares/genética , Elementos Reguladores de la Transcripción , Transducción de Señal/genética , Esfingolípidos/biosíntesis , Esfingolípidos/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Espectrometría de Masas en Tándem , Factores de Transcripción/genética , Transcriptoma/genética , Proteínas Supresoras de Tumor/genética
14.
Sci Adv ; 7(18)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33931443

RESUMEN

Molecular profiling of the most aggressive brain tumor glioblastoma (GBM) on the basis of gene expression, DNA methylation, and genomic variations advances both cancer research and clinical diagnosis. The enhancer architectures and regulatory circuitries governing tumor-intrinsic transcriptional diversity and subtype identity are still elusive. Here, by mapping H3K27ac deposition, we analyze the active regulatory landscapes across 95 GBM biopsies, 12 normal brain tissues, and 38 cell line counterparts. Analyses of differentially regulated enhancers and super-enhancers uncovered previously unrecognized layers of intertumor heterogeneity. Integrative analysis of variant enhancer loci and transcriptome identified topographies of transcriptional enhancers and core regulatory circuitries in four molecular subtypes of primary tumors: AC1-mesenchymal, AC1-classical, AC2-proneural, and AC3-proneural. Moreover, this study reveals core oncogenic dependency on super-enhancer-driven transcriptional factors, long noncoding RNAs, and druggable targets in GBM. Through profiling of transcriptional enhancers, we provide clinically relevant insights into molecular classification, pathogenesis, and therapeutic intervention of GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Cromatina/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/patología , Humanos
15.
Cancer Res ; 80(13): 2722-2736, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32332020

RESUMEN

Gastrointestinal adenocarcinomas (GIAC) of the tubular gastrointestinal (GI) tract including esophagus, stomach, colon, and rectum comprise most GI cancers and share a spectrum of genomic features. However, the unified epigenomic changes specific to GIAC are poorly characterized. Using 907 GIAC samples from The Cancer Genome Atlas, we applied mathematical algorithms to large-scale DNA methylome and transcriptome profiles to reconstruct transcription factor (TF) networks and identify a list of functionally hyperactive master regulator (MR) TF shared across different GIAC. The top candidate HNF4A exhibited prominent genomic and epigenomic activation in a GIAC-specific manner. A complex interplay between the HNF4A promoter and three distal enhancer elements was coordinated by GIAC-specific MRTF including ELF3, GATA4, GATA6, and KLF5. HNF4A also self-regulated its own promoter and enhancers. Functionally, HNF4A promoted cancer proliferation and survival by transcriptional activation of many downstream targets, including HNF1A and factors of interleukin signaling, in a lineage-specific manner. Overall, our study provides new insights into the GIAC-specific gene regulatory networks and identifies potential therapeutic strategies against these common cancers. SIGNIFICANCE: These findings show that GIAC-specific master regulatory transcription factors control HNF4A via three distal enhancers to promote GIAC cell proliferation and survival. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/13/2722/F1.large.jpg.


Asunto(s)
Adenocarcinoma/patología , Biomarcadores de Tumor/metabolismo , Epigenómica , Neoplasias Gastrointestinales/patología , Regulación Neoplásica de la Expresión Génica , Factor Nuclear 4 del Hepatocito/metabolismo , Factores de Transcripción/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/metabolismo , Redes Reguladoras de Genes , Genómica , Factor Nuclear 4 del Hepatocito/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Regiones Promotoras Genéticas , Tasa de Supervivencia , Factores de Transcripción/genética , Transcriptoma , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Int J Cancer ; 125(7): 1710-20, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19507250

RESUMEN

Our study explored the drug interaction of all-trans retinoic acid (ATRA) and RAD001 (everolimus), the inhibitor of mammalian target of rapamycin complex 1 (mTORC1), in acute myelogenous leukemia (AML) NB4 and HL60 cells. RAD001 (10 nM) significantly enhanced the ATRA-induced growth arrest and differentiation of these cells, as measured by colony-forming assay and cell cycle analysis, and expression of CD11b cell surface antigen and nitroblue tetrazolium reduction, respectively. ATRA (0.1-1 microM) upregulated levels of RTP801, a negative regulator of mTORC1, and inhibited mTORC1 signaling as assessed by measurement of the levels of p-p70S6K and p-4E-BP1 in HL60 and NB4 cells. ATRA (0.1-1 microM) in combination with RAD001 (10 nM) strikingly downregulated the levels of p-70S6K and p-4E-BP1 without affecting the total amount of these proteins. Notably, RAD001 (10 nM) significantly augmented ATRA-induced expression of CCAAT/enhancer-binding protein epsilon (C/EBPepsilon) and p27(kip1) and downregulated levels of c-Myc in these cells. Furthermore, RAD001 (5 mg/kg) enhanced the ability of ATRA (10 mg/kg) to inhibit the proliferation of HL60 cells growing as tumor xenografts in immune-deficient nude mice. Taken together, concomitant blockade of the RA and mTORC1 signaling may be a promising treatment strategy for individuals with AML.


Asunto(s)
Antineoplásicos/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Receptores de Ácido Retinoico/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Sirolimus/análogos & derivados , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Tretinoina/farmacología , Animales , Biomarcadores de Tumor/metabolismo , Western Blotting , Proteínas Potenciadoras de Unión a CCAAT , Antígeno CD11b/metabolismo , Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación hacia Abajo , Sinergismo Farmacológico , Everolimus , Femenino , Regulación Neoplásica de la Expresión Génica , Células HL-60/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Complejos Multiproteicos , Proteínas de Neoplasias/efectos de los fármacos , Proteínas de Neoplasias/metabolismo , Nitroazul de Tetrazolio/metabolismo , Proteínas , Proteínas Proto-Oncogénicas c-myc/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Factores de Transcripción/efectos de los fármacos , Trasplante Heterólogo , Ensayo de Tumor de Célula Madre , Regulación hacia Arriba
17.
Int J Cancer ; 124(4): 827-33, 2009 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-19035457

RESUMEN

Molecular-targeted therapy is a hopeful approach for pancreatic cancer. Silencing of tumor suppressor genes can occur by histone deacetylation and/or DNA methylation in the promoter. Here, we identified epigenetically silenced genes in pancreatic cancer cells. Pancreatic cancer cell line, PANC-1 cells were treated either with or without 5Aza-dC (a DNA methyltransferase inhibitor) and suberoylanilide hydroxamic acid (SAHA, a histone deacetylase inhibitor), and mRNA was isolated from these cells. Oligonucleotide microarray analysis revealed that 30 genes including UCHL1, C/EBPalpha, TIMP2 and IRF7 were up-regulated after treatment with 5Aza-dC and SAHA in PANC-1. The induction of these 4 genes was validated by real-time PCR in several pancreatic cancer cell lines. Interestingly, expression of C/EBPalpha was significantly restored in 6 of 6 pancreatic cancer cell lines. Chromatin immunoprecipitation assay revealed that histone H3 of the promoter region of C/EBPalpha was acetylated in PANC-1 treated with SAHA; and bisulfate sequencing showed methylation of its promoter region in several pancreatic cancer cell lines. Forced expression of C/EBPalpha markedly suppressed clonal proliferation of PANC-1 cells. Co-immunoprecipitation assay showed the interaction of C/EBPalpha and E2F1; and the interaction caused the inhibition of E2F1 transcriptional activity. Immunohistochemical analysis revealed that C/EBPalpha localized in the cytoplasm in pancreatic adenocarcinoma cells, whereas it localized predominantly in the nucleus in normal pancreatic cells. Our data demonstrated that aberrant silencing, as well as, inappropriate cytoplasmic localization of C/EBPalpha causes dysregulation of its function, suggesting that C/EBPalpha is a novel candidate tumor suppressor gene in pancreatic cancer cells.


Asunto(s)
Proteína alfa Potenciadora de Unión a CCAAT/genética , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Antígenos de Neoplasias/biosíntesis , Antineoplásicos/farmacología , Línea Celular Tumoral , Metilación de ADN , Factor de Transcripción E2F1/metabolismo , Silenciador del Gen , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Modelos Biológicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vorinostat
18.
Biochem J ; 415(2): 241-5, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18588518

RESUMEN

The adaptor protein Lnk is expressed in haemopoietic cells and plays a critical role in haemopoiesis. Animal model studies demonstrated that Lnk acts as a broad inhibitor of signalling pathways in haemopoietic lineages. Lnk belongs to a family of proteins sharing several structural motifs, including an SH2 (Src homology 2) domain which binds phosphotyrosine residues in various signal-transducing proteins. The SH2 domain is essential for Lnk-mediated negative regulation of several cytokine receptors [e.g. Mpl, EpoR (erythropoietin receptor), c-Kit]. Therefore inhibition of the binding of Lnk to cytokine receptors might lead to enhanced downstream signalling of the receptor and thereby to improved haemopoiesis in response to exposure to cytokines (e.g. erythropoietin in anaemic patients). This hypothesis led us to define the exact binding site of Lnk to the stem cell factor receptor c-Kit. Pull-down experiments using GST (glutathione transferase)-fusion proteins of the different domains of c-Kit showed that Lnk almost exclusively binds to the phosphorylated juxtamembrane domain. Binding of Lnk to the juxtamembrane domain was abolished by point mutation of Tyr(568) and was competed by peptides with a phosphotyrosine residue at position 568. Co-immunoprecipitation with full-length wild-type or Y568F mutant c-Kit and Lnk confirmed these results, thus showing the importance of this phosphorylated tyrosine residue. Lnk bound directly to c-Kit without requiring other interacting partners. The identification of the binding site of Lnk to c-Kit will be useful to discover inhibitory molecules that prevent the binding of these two proteins, thus making haemopoietic cells more sensitive to growth factors.


Asunto(s)
Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Tirosina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Línea Celular , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular , Mutación , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-kit/química , Tirosina/genética , Dominios Homologos src/genética
19.
Nat Commun ; 10(1): 1353, 2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30903020

RESUMEN

Liposarcomas (LPSs) are a group of malignant mesenchymal tumors showing adipocytic differentiation. Here, to gain insight into the enhancer dysregulation and transcriptional addiction in this disease, we chart super-enhancer structures in both LPS tissues and cell lines. We identify a bromodomain and extraterminal (BET) protein-cooperated FUS-DDIT3 function in myxoid LPS and a BET protein-dependent core transcriptional regulatory circuitry consisting of FOSL2, MYC, and RUNX1 in de-differentiated LPS. Additionally, SNAI2 is identified as a crucial downstream target that enforces both proliferative and metastatic potentials to de-differentiated LPS cells. Genetic depletion of BET genes, core transcriptional factors, or SNAI2 mitigates consistently LPS malignancy. We also reveal a compelling susceptibility of LPS cells to BET protein degrader ARV-825. BET protein depletion confers additional advantages to circumvent acquired resistance to Trabectedin, a chemotherapy drug for LPS. Moreover, this study provides a framework for discovering and targeting of core oncogenic transcriptional programs in human cancers.


Asunto(s)
Liposarcoma/genética , Proteínas de Neoplasias/metabolismo , Transcripción Genética , Animales , Azepinas/farmacología , Secuencia de Bases , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Elementos de Facilitación Genéticos/genética , Genoma Humano , Humanos , Ratones Endogámicos NOD , Ratones SCID , Proteínas de Fusión Oncogénica/metabolismo , Talidomida/análogos & derivados , Talidomida/farmacología , Transcripción Genética/efectos de los fármacos
20.
Int J Cancer ; 122(7): 1567-72, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18059030

RESUMEN

HPP1 is a recently discovered gene that is epigenetically silenced in a number of tumor types, suggesting a potential role as a tumor suppressor. However, whether HPP1 has tumor suppressor activity is not clearly known. We have sought to investigate the effects of HPP1 on tumor growth and survival and to identify signaling pathways that mediate HPP1's mechanism of action. Forced expression of HPP1 into HCT116 colon cancer cell lines blocked the ability of HCT116 tumors to grown in vivo in nude mice. In cell culture, ectopic expression of HPP1 induces apoptosis and potently inhibits soft agar colony formation. HPP1 overexpression was also associated with a moderate reduction in in vitro proliferation characterized by an accumulation of cells in the G0/G1 phase of the cell cycle. Microarray analysis revealed that ectopic expression of HPP1 resulted in a dramatic upregulation of STAT1 as well as a large number of associated interferon-inducible genes. RNA interference-mediated abrogation of STAT1 reversed HPP1's antiproliferative effects. We conclude that HPP1 demonstrates tumor suppressive and pro-apoptotic activity, both in vitro and in vivo. Coupled with its inactivation in a number of tumor types, our data provides evidence to support the role of HPP1 as a tumor suppressor gene. Moreover, activation of the STAT1 pathway likely represents the principal mediator of HPP1's tumor suppressive properties.


Asunto(s)
Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Genes Supresores de Tumor , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Factor de Transcripción STAT1/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis , Proliferación Celular , Regulación hacia Abajo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Ratones , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Interferencia de ARN , Transducción de Señal , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA