Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Am J Physiol Lung Cell Mol Physiol ; 319(1): L82-L90, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32401676

RESUMEN

Goblet cell metaplasia (GCM) and mucin overproduction are a hallmark of chronic rhinosinusitis (CRS) and chronic obstructive pulmonary disease (COPD). In the airways, cigarette smoke (CS) induces activation of the epidermal growth factor receptor (EGFR) leading to GCM and overexpression of the gel-forming mucin MUC5AC. Although previous studies have demonstrated that a membrane-bound mucin, MUC1, modulates the activation of CS-induced EGFR, the role of MUC1 in CS-induced GCM and mucin overproduction has not been explored. In response to CS exposure, wild-type (WT) rats displayed Muc1 translocation from the apical surface of airway epithelium to the intracellular compartment of hyperplastic intermediate cells, EGFR phosphorylation, GCM, and Muc5ac overproduction. Similarly, human CRS sinonasal tissues demonstrated hyperplasia of intermediate cells enriched with MUC1 in the intracellular compartment, which was accompanied by GCM and increased MUC5AC expression. To further evaluate the role of Muc1 in vivo, a Muc1 knockout (KO) rat (MUC in humans and Muc in animals) was developed. In contrast to WT littermates, Muc1-KO rats exhibited no activation of EGFR, and were protected from GCM and Muc5ac overproduction. Genetic knockdown of MUC1 in human lung or Muc1 knockout in primary rat airway epithelial cells led to significantly diminished EGF-induced MUC5AC production. Together, these findings suggest that MUC1-dependent EGFR activation mediates CS-induced GCM and mucin overproduction. Strategies designed to suppress MUC1-dependent EGFR activation may provide a novel therapeutic approach for treating mucin hypersecretion in CRS and COPD.


Asunto(s)
Células Caliciformes/metabolismo , Mucina 5AC/metabolismo , Mucina-1/metabolismo , Fumar/efectos adversos , Animales , Línea Celular Tumoral , Polaridad Celular , Células Epiteliales/metabolismo , Epitelio/metabolismo , Epitelio/patología , Receptores ErbB/metabolismo , Células Caliciformes/patología , Metaplasia , Fosforilación , Ratas Sprague-Dawley
2.
Int Forum Allergy Rhinol ; 9(10): 1159-1188, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31430424

RESUMEN

BACKGROUND: Rhinovirus (RV) infections are the most common cause of viral upper respiratory infections (URIs), and in the majority of persons they are self-limiting. However, in others, viral URIs can progress to bacterial sinusitis and induce chronic rhinosinusitis (CRS) exacerbations. METHODS: We conducted a comprehensive Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) review through April 2018 based on MEDLINE, EMBASE, Web of Science-Science Citation Index (SCI), and Conference Proceedings Citation Index- Science (CPCI-S) using keywords: RV, respiratory virus, sinusitis, and airway epithelial cells. The goal of this systematic review was to: (1) determine the prevalence between RV and CRS, (2) study the changes that occur after experimental RV inoculation, (3) investigate the pathophysiologic mechanisms by which RV induces sinonasal inflammation, and (4) explore the treatment options available for RV-associated sinusitis. Data regarding study design, research question, intervention, subjects, outcomes, and biases was extracted. RESULTS: The initial search yielded 2395 unique abstracts, of which 614 were selected for full-text review; 147 were included in the final review. We determined that (1) the prevalence of RV infections is increased in those with CRS, (2) humans challenged in vivo with RV secrete local inflammatory mediators with radiographic mucosal thickening, (3) RV species RV-A and RV-C challenges in vitro to sinonasal epithelia produce robust cytokine responses and differential gene changes, and (4) no current therapies have produced consistent and significant resolution of disease. CONCLUSION: RV infections are common in persons with CRS, and incite inflammatory reactions that may result in CRS exacerbations and progression of disease. Further studies assessing RV species, and the host-virome response are required to develop new strategies targeting RV-induced CRS.


Asunto(s)
Infecciones por Picornaviridae/epidemiología , Mucosa Respiratoria/inmunología , Rinitis/epidemiología , Rhinovirus/fisiología , Sinusitis/epidemiología , Animales , Enfermedad Crónica , Citocinas/metabolismo , Modelos Animales de Enfermedad , Glicoproteínas/uso terapéutico , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/metabolismo , Interferones/uso terapéutico , Infecciones por Picornaviridae/inmunología , Infecciones por Picornaviridae/terapia , Mucosa Respiratoria/virología , Rinitis/inmunología , Rinitis/terapia , Sinusitis/inmunología , Sinusitis/terapia
3.
BMC Cancer ; 18(1): 595, 2018 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-29801480

RESUMEN

BACKGROUND: Metastatic breast cancer carries a poor prognosis despite the success of newly targeted therapies. Treatment options remain especially limited for the subtype of triple negative breast cancer (TNBC). Several signaling pathways, including NF-κB, are altered in TNBC, and the complexity of this disease implies multi-faceted pathway interactions. Given that IKKε behaves as an oncogene in breast cancer, we hypothesized that IKKε regulates NF-κB signaling to control diverse oncogenic functions in TNBC. METHODS: Vector expression and RNA interference were used to investigate the functional role of IKKε in triple-negative breast cancer cells. Viability, protein expression, NF-κB binding activity, invasion, anoikis, and spheroid formation were examined in cells expressing high or low levels of IKKε, in conjunction with p52 RNA interference or MEK inhibition. RESULTS: This study found that non-canonical NF-κB p52 levels are inversely proportional to ΙΚΚε, and growth of TNBC cells in anchorage supportive, high-attachment conditions requires IKKε and activated MEK. Growth of these cells in anchorage resistant conditions requires IKKε and activated MEK or p52. In this model, IKKε and MEK cooperate to support overall viability whereas the p52 transcription factor is only required for viability in low attachment conditions, underscoring the contrasting roles of these proteins. CONCLUSIONS: This study illustrates the diverse functions of IKKε in TNBC and highlights the adaptability of NF-κB signaling in maintaining cancer cell survival under different growth conditions. A better understanding of the diversity of NF-κB signaling may ultimately improve the development of novel therapeutic regimens for TNBC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Quinasa I-kappa B/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular/genética , Femenino , Humanos , Quinasa I-kappa B/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Subunidad p52 de NF-kappa B/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/genética , Neoplasias de la Mama Triple Negativas/patología
4.
Cancer Res ; 77(24): 6927-6940, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29074539

RESUMEN

Understanding the mechanisms supporting tumor-initiating cells (TIC) is vital to combat advanced-stage recurrent cancers. Here, we show that in advanced ovarian cancers NFκB signaling via the RelB transcription factor supports TIC populations by directly regulating the cancer stem-like associated enzyme aldehyde dehydrogenase (ALDH). Loss of RelB significantly inhibited spheroid formation, ALDH expression and activity, chemoresistance, and tumorigenesis in subcutaneous and intrabursal mouse xenograft models of human ovarian cancer. RelB also affected expression of the ALDH gene ALDH1A2 Interestingly, classical NFκB signaling through the RelA transcription factor was equally important for tumorigenesis in the intrabursal model, but had no effect on ALDH. In this case, classical signaling via RelA was essential for proliferating cells, whereas the alternative signaling pathway was not. Our results show how NFκB sustains diverse cancer phenotypes via distinct classical and alternative signaling pathways, with implications for improved understanding of disease recurrence and therapeutic response. Cancer Res; 77(24); 6927-40. ©2017 AACR.


Asunto(s)
Carcinogénesis/genética , Proliferación Celular/genética , Isoenzimas/metabolismo , Células Madre Neoplásicas/fisiología , Neoplasias Ováricas/genética , Retinal-Deshidrogenasa/metabolismo , Factor de Transcripción ReIA/fisiología , Familia de Aldehído Deshidrogenasa 1 , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral , Femenino , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , FN-kappa B/genética , FN-kappa B/fisiología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Transducción de Señal/genética , Factor de Transcripción ReIA/genética
5.
Biochem Biophys Res Commun ; 493(3): 1230-1235, 2017 11 25.
Artículo en Inglés | MEDLINE | ID: mdl-28916165

RESUMEN

BACKGROUND: MUC1 (MUC in human and Muc in animals) is a membrane-tethered mucin expressed on the apical surface of lung epithelial cells. However, in the lungs of patients with interstitial lung disease, MUC1 is aberrantly expressed in hyperplastic alveolar type II epithelial (ATII) cells and alveolar macrophages (AM), and elevated levels of extracellular MUC1 are found in bronchoalveolar lavage (BAL) fluid and the serum of these patients. While pro-fibrotic effects of extracellular MUC1 have recently been described in cultured fibroblasts, the contribution of MUC1 to the pathobiology of pulmonary fibrosis is unknown. In this study, we hypothesized that MUC1 deficiency would reduce susceptibility to pulmonary fibrosis in a mouse model of silicosis. METHODS: We employed human MUC1 transgenic mice, Muc1 deficient mice and wild-type mice on C57BL/6 background in these studies. Some mice received a one-time dose of crystalline silica instilled into their oropharynx in order to induce pulmonary fibrosis and assess the effects of Muc1 deficiency on fibrotic and inflammatory responses in the lung. RESULTS: As previously described in other mouse models of pulmonary fibrosis, we found that extracellular MUC1 levels were markedly increased in whole lung tissues, BALF and serum of human MUC1 transgenic mice after silica. We also detected an increase in total MUC1 levels in the lungs of these mice, indicating that production as well as release contributed to elevated levels after lung injury. Immunohistochemical staining revealed that increased MUC1 expression was mostly confined to ATII cells and AMs in areas of fibrotic remodeling, illustrating a pattern similar to the expression of MUC1 in human fibrotic lung tissues. However, contrary to our hypothesis, we found that Muc1 deficiency resulted in a worsening of fibrotic remodeling in the mouse lung as judged by an increase in number of silicotic nodules, an increase in lung collagen deposition and an increase in the severity of pulmonary inflammation. CONCLUSIONS: Altogether, our results indicate that Muc1 has anti-fibrotic properties in the mouse lung and suggest that elevated levels of MUC1 in patients with interstitial lung disease may serve a protective role, which aims to limit the severity of tissue remodeling in the lung.


Asunto(s)
Mucina-1/metabolismo , Fibrosis Pulmonar/fisiopatología , Silicosis/fisiopatología , Animales , Modelos Animales de Enfermedad , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mucina-1/genética , Neumonía/genética , Neumonía/patología , Fibrosis Pulmonar/inducido químicamente , Dióxido de Silicio/toxicidad
6.
Oncotarget ; 8(31): 51402-51415, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28881656

RESUMEN

Ovarian cancer (OC) is a heterogeneous disease characterized by defective DNA repair. Very few targets are universally expressed in the high grade serous (HGS) subtype. We previously identified that CHK1 was overexpressed in most of HGSOC. Here, we sought to understand the DNA damage response (DDR) to CHK1 inhibition and increase the anti-tumor activity of this pathway. We found BRD4 suppression either by siRNA or BRD4 inhibitor JQ1 enhanced the cytotoxicity of CHK1 inhibition. Interestingly, BRD4 was amplified and/or upregulated in a subset of HGSOC with statistical correlation to overall survival. BRD4 inhibition increased CBX5 (HP1α) level. CHK1 inhibitor induced DDR marker, γ-H2AX, but BRD4 suppression did not. Furthermore, nuclear localization of CBX5 and γ-H2AX was mutually exclusive in BRD4-and CHK1-inhibited cells, suggesting BRD4 facilitates DDR by repressing CBX5. Our results provide a strong rationale for clinical investigation of CHK1 and BRD4 co-inhibition, especially for HGSOC patients with BRD4 overexpression.

7.
Biochem Biophys Res Commun ; 492(2): 231-235, 2017 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-28822766

RESUMEN

Alveolar macrophages (AMs) play a critical role in the clearance of Pseudomonas aeruginosa (Pa) from the airways. However, hyper-activation of macrophages can impair bacterial clearance and contribute to morbidity and mortality. MUC1 mucin is a membrane-tethered, high molecular mass glycoprotein expressed on the apical surface of mucosal epithelial cells and some hematopoietic cells, including macrophages, where it counter-regulates inflammation. We recently reported that Pa up-regulates the expression of MUC1 in primary human AMs and THP-1 macrophages, and that increased MUC1 expression in these cells prevents hyper-activation of macrophages that appears to be important for host defense against severe pathology of Pa lung infection. The aims of this study were to elucidate the mechanism by which Pa increases MUC1 expression in macrophages. The results showed that: (a) Pa stimulation of THP-1 macrophages increased MUC1 expression both at transcriptional and protein levels in a dose-dependent manner; (b) Both Pa- and LPS-induced MUC1 expression in THP-1 cells were significantly diminished by an inhibitory peptide of TLR4; and (c) LPS-stimulated MUC1 expression was diminished at both the mRNA and protein levels by an inhibitor of the p38 mitogen-activated protein kinase, but not by inhibitors of ERK1/2, JNK, or IKK. We conclude that Pa-stimulated MUC1 expression in THP-1 macrophages is regulated mainly through the TLR4-p38 signaling pathway.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Macrófagos/microbiología , Mucina-1/genética , Infecciones por Pseudomonas/genética , Pseudomonas aeruginosa/fisiología , Regulación hacia Arriba , Línea Celular , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Mucina-1/inmunología , Infecciones por Pseudomonas/inmunología , Infecciones por Pseudomonas/patología , Transducción de Señal , Receptor Toll-Like 4/inmunología
8.
BMC Cancer ; 16: 678, 2016 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-27558154

RESUMEN

BACKGROUND: shRNA-mediated lethality screening is a useful tool to identify essential targets in cancer biology. Ovarian cancer (OC) is extremely heterogeneous and most cases are advanced stages at diagnosis. OC has a high response rate initially, but becomes resistant to standard chemotherapy. We previously employed high throughput global shRNA sensitization screens to identify NF-kB related pathways. Here, we re-analyzed our previous shRNA screens in an unbiased manner to identify clinically applicable molecular targets. METHODS: We proceeded with siRNA lethality screening using the top 55 genes in an expanded set of 6 OC cell lines. We investigated clinical relevance of candidate targets in The Cancer Genome Atlas OC dataset. To move these findings towards the clinic, we chose four pharmacological inhibitors to recapitulate the top siRNA effects: Oxozeaenol (for MAP3K7/TAK1), BI6727 (PLK1), MK1775 (WEE1), and Lapatinib (ERBB2). Cytotoxic effects were measured by cellular viability assay, as single agents and in 2-way combinations. Co-treatments were evaluated in either sequential or simultaneous exposure to drug for short term and extended periods to simulate different treatment strategies. RESULTS: Loss-of-function shRNA screens followed by short-term siRNA validation screens identified therapeutic targets in OC cells. Candidate genes were dysregulated in a subset of TCGA OCs although the alterations of these genes showed no statistical significance to overall survival. Pharmacological inhibitors such as Oxozeaenol, BI6727, and MK1775 showed cytotoxic effects in OC cells regardless of cisplatin responsiveness, while all OC cells tested were cytostatic to Lapatinib. Co-treatment with BI6727 and MK1775 at sub-lethal concentrations was equally potent to BI6727 alone at lethal concentrations without cellular re-growth after the drugs were washed off, suggesting the co-inhibition at reduced dosages may be more efficacious than maximal single-agent cytotoxic concentrations. CONCLUSIONS: Loss-of-function screen followed by in vitro target validation using chemical inhibitors identified clinically relevant targets. This approach has the potential to systematically refine therapeutic strategies in OC. These molecular target-driven strategies may provide additional therapeutic options for women whose tumors have become refractory to standard chemotherapy.


Asunto(s)
Neoplasias Ováricas/genética , Transcriptoma , Western Blotting , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , FN-kappa B/metabolismo , Neoplasias Ováricas/metabolismo , ARN Interferente Pequeño/genética
9.
Gynecol Oncol ; 142(2): 332-40, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27235858

RESUMEN

OBJECTIVE: The value of cell lines for pre-clinical work lies in choosing those with similar characteristics. Selection of cell lines is typically based on patient history, histological subtype at diagnosis, mutation patterns, or signaling pathways. Although recent studies established consensus regarding molecular characteristics of ovarian cancer cell lines, data on in vivo tumorigenicity remains only sporadically available, impeding translation of in vitro work to xenograft models. METHODS: We introduced 18 ovarian cancer cell lines into athymic nude mice through subcutaneous, intraperitoneal, and ovary intrabursal routes, and observed tumor development over 6weeks. We also profiled cell line gene expression and identified differentially expressed gene sets based on their ability to form tumors in the subcutaneous or intraperitoneal locations. Representative cell lines were further subjected to proteomic analyses. RESULTS: Ovarian cancer cell lines showed variable ability to grow in mice when implanted subcutaneous, intraperitoneal, or intrabursal. While some cell lines grew well in both SC and IP locations, others showed a strong propensity to grow in one location only. Gene expression profiles suggested that cell lines showing preference for IP growth had gene expression patterns more similar to primary tumors. CONCLUSIONS: We report the tumorigenicity of 17 human ovarian cancer cell lines and one mouse cell line in three distinct anatomical locations, and associated gene networks. Growth patterns and histopathology, linked to molecular characteristics, provide a valuable resource to the research community, and better guide the choice of cell lines for in vitro studies to translate efficiently into xenograft testing.


Asunto(s)
Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales/métodos , Neoplasias Ováricas/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Femenino , Xenoinjertos , Humanos , Ratones , Trasplante de Neoplasias , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo
11.
Clin Imaging ; 39(5): 920-2, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25982495

RESUMEN

We present a case of vesical varices manifesting as hematuria in a patient with severe portal hypertension. Vesical varices are an uncommon type of ectopic varices in patients with portal hypertension and are usually seen in patients who have undergone prior interventions that alter portal pressure dynamics. Our case demonstrates that this unusual entity was present in a patient without prior intervention and serves as reminder to assess for ectopic varices in order to mitigate or prevent associated morbidity.


Asunto(s)
Hematuria/etiología , Hipertensión Portal/etiología , Vejiga Urinaria/irrigación sanguínea , Várices/complicaciones , Diagnóstico Diferencial , Hematuria/diagnóstico , Humanos , Hipertensión Portal/diagnóstico , Masculino , Persona de Mediana Edad , Tomografía Computarizada por Rayos X , Ultrasonografía Doppler en Color/métodos , Vejiga Urinaria/diagnóstico por imagen , Várices/diagnóstico
12.
BMC Cancer ; 15: 196, 2015 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-25884494

RESUMEN

BACKGROUND: Topotecan (TPT) is a therapeutic option for women with platinum-resistant or -refractory ovarian cancer. However, the dose-limiting toxicity of TPT is myelosuppression. This led us to seek a combination treatment to augment TPT anti-cancer activity in a cancer-targeted manner. Ovarian serous cancers, a major subtype, show dysregulated DNA repair pathway and often display a high level of CHEK1 (CHK1), a cell cycle regulator and DNA damage sensor. CHEK1 inhibitors are a novel approach to treatment, and have been used as single agents or in combination chemotherapy in many cancers. METHODS: We evaluated the cellular effects of TPT in a panel of high grade serous (HGS) and non-HGS ovarian cancer cells. We then determined IC50s of TPT in the absence and presence of CHEK1 inhibitor, PF477736. Synergism between TPT and PF477736 was calculated based on cellular viability assays. Cytotoxic effect of the combined treatment was compared with apoptotic activities by Caspase3/7 activity assay and Western blotting of cleaved-PARP1 and γH2AX. RESULTS: Non-HGS ovarian cancer cells were generally more sensitive to TPT treatment compared to HGS ovarian cancer cells. When combined with CHEK1 inhibitor, TPT potently and synergistically inhibited the proliferation of HGS ovarian cancer cells. This dramatic synergism in cellular toxicity was consistent with increases in markers of apoptosis. CONCLUSIONS: Our findings suggest that the addition of CHEK1 inhibitor increases the response of ovarian cancer cells to TPT. Furthermore, reduced dosages of both drugs achieved maximal cytotoxic effects by combining TPT with CHEK1 inhibitor. This strategy would potentially minimize side effects of the drugs for extended clinical benefit.


Asunto(s)
Benzodiazepinonas/administración & dosificación , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Proteínas Quinasas/genética , Pirazoles/administración & dosificación , Inhibidores de Topoisomerasa I/administración & dosificación , Topotecan/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptosis/efectos de los fármacos , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Cisplatino/administración & dosificación , Cisplatino/efectos adversos , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Humanos , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Inhibidores de Topoisomerasa I/efectos adversos , Topotecan/efectos adversos
13.
Oncotarget ; 5(24): 12788-802, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25474241

RESUMEN

Ovarian cancer (OC) is extremely heterogeneous, implying that therapeutic strategies should be specifically designed based on molecular characteristics of an individual's tumor. Previously, we showed that IKKε promotes invasion and metastasis in a subset of OCs. Here, we identified CHEK1 as an IKKε-dependent lethal gene from shRNA kinome library screen. In subsequent pharmacological intervention studies, the co-inhibition of IKKε and CHEK1 was more effective in killing OC cells than single treatment. At the molecular level, co-inhibition dramatically decreased pro-survival proteins, but increased proteins involved in DNA damage and apoptosis. IKKε-knockdown increased p21 levels, while overexpression of wild-type IKKε, but not a kinase dead IKKε mutant decreased p21 levels. We further demonstrated that the depletion of p21 rendered OC cells more resistant to cell death induced by co-inhibition of IKKε and CHEK1. In conclusion, we revealed a novel interplay between IKKε, CHEK1 and p21 signaling in survival of OC. Our study provides a rationale for the clinical development of specific IKKε inhibitor and for usage of IKKε as an exploratory marker for resistance to CHEK1 inhibitors in the clinic. The interplay provides one potential explanation as to why very few clinical responses were achieved in patients treated with single-agent CHEK1 inhibitors.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Quinasa I-kappa B/metabolismo , Neoplasias Ováricas/metabolismo , Proteínas Quinasas/metabolismo , Apoptosis/fisiología , Línea Celular Tumoral , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Daño del ADN , Femenino , Humanos , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Proteínas Quinasas/deficiencia , Proteínas Quinasas/genética , ARN Interferente Pequeño , Transducción de Señal
14.
Proc Natl Acad Sci U S A ; 109(52): E3659-67, 2012 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-23236152

RESUMEN

Cancers with Ras mutations represent a major therapeutic problem. Recent RNAi screens have uncovered multiple nononcogene addiction pathways that are necessary for the survival of Ras mutant cells. Here, we identify the evolutionarily conserved gene enhancer of rudimentary homolog (ERH), in which depletion causes greater toxicity in cancer cells with mutations in the small GTPase KRAS compared with KRAS WT cells. ERH interacts with the spliceosome protein SNRPD3 and is required for the mRNA splicing of the mitotic motor protein CENP-E. Loss of ERH leads to loss of CENP-E and consequently, chromosome congression defects. Gene expression profiling indicates that ERH is required for the expression of multiple cell cycle genes, and the gene expression signature resulting from ERH down-regulation inversely correlates with KRAS signatures. Clinically, tumor ERH expression is inversely associated with survival of colorectal cancer patients whose tumors harbor KRAS mutations. Together, these findings identify a role of ERH in mRNA splicing and mitosis, and they provide evidence that KRAS mutant cancer cells are dependent on ERH for their survival.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/genética , Secuencia Conservada , Evolución Molecular , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Empalme del ARN/genética , Factores de Transcripción/metabolismo , Proteínas ras/genética , Ciclo Celular/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas Humanos/metabolismo , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Oncogenes , Unión Proteica , Proteínas Proto-Oncogénicas p21(ras) , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia , Proteínas Nucleares snRNP/metabolismo
15.
Cancer Res ; 72(21): 5494-504, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22942254

RESUMEN

Inhibitor of IκB kinases (IKK) are key regulators of NF-κB signaling. Three IKK isoforms-α, ß, and ε-have been linked to oncogenesis, yet the precise components of NF-κB signaling in ovarian cancer have not yet been dissected. We surveyed 120 ovarian cancer specimens for IKK-ε expression. Notably, cytoplasmic expression was elevated in metastatic lesions relative to primary tumors (P = 0.03). Therefore, we hypothesized that IKK-ε drives ovarian cancer metastasis. IKK-ε was identified previously as a breast cancer oncogene and was associated with poor clinical outcome in ovarian cancer. We now define an ovarian cancer-specific IKK-ε-regulated gene expression signature using stably expressed short hairpin RNA targeting IKK-ε. Pathway analysis of the signature indicated that IKK-ε regulates expression of genes involved in cell motility and inflammation. We further showed that IKK-ε depletion in metastatic ovarian cancer cell lines decreased growth, adhesion, and invasion. Consistently, human xenografts depleted of IKK-ε in mice showed decreased aggressiveness, whereas overexpression of IKK-ε in a less invasive ovarian cancer cell line increased metastasis in vivo. Taken together, these data provide evidence that IKK-ε is a key coordinator of invasion and metastasis programs in ovarian cancer. Inhibition of IKK-ε signaling thus emerges as a viable therapeutic strategy in women whose ovarian cancer shows aberrant activation of this pathway.


Asunto(s)
Quinasa I-kappa B/metabolismo , Neoplasias Glandulares y Epiteliales/enzimología , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Animales , Western Blotting , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica , Metástasis de la Neoplasia , Transducción de Señal/fisiología , Análisis de Matrices Tisulares , Transcriptoma , Trasplante Heterólogo
16.
Mol Biol Cell ; 21(19): 3487-96, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20719962

RESUMEN

Sprouty (Spry) proteins are negative regulators of receptor tyrosine kinase signaling; however, their exact mechanism of action remains incompletely understood. We identified phosphatidylinositol-specific phospholipase C (PLC)-γ as a partner of the Spry1 and Spry2 proteins. Spry-PLCγ interaction was dependent on the Src homology 2 domain of PLCγ and a conserved N-terminal tyrosine residue in Spry1 and Spry2. Overexpression of Spry1 and Spry2 was associated with decreased PLCγ phosphorylation and decreased PLCγ activity as measured by production of inositol (1,4,5)-triphosphate (IP(3)) and diacylglycerol, whereas cells deficient for Spry1 or Spry1, -2, and -4 showed increased production of IP(3) at baseline and further increased in response to growth factor signals. Overexpression of Spry 1 or Spry2 or small-interfering RNA-mediated knockdown of PLCγ1 or PLCγ2 abrogated the activity of a calcium-dependent reporter gene, suggesting that Spry inhibited calcium-mediated signaling downstream of PLCγ. Furthermore, Spry overexpression in T-cells, which are highly dependent on PLCγ activity and calcium signaling, blocked T-cell receptor-mediated calcium release. Accordingly, cultured T-cells from Spry1 gene knockout mice showed increased proliferation in response to T-cell receptor stimulation. These data highlight an important action of Spry, which may allow these proteins to influence signaling through multiple receptors.


Asunto(s)
Proteínas de la Membrana/metabolismo , Fosfolipasa C gamma/metabolismo , Fosfoproteínas/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación de Linfocitos T/metabolismo , Biomarcadores/metabolismo , Calcio/metabolismo , Diglicéridos/metabolismo , Activación Enzimática , Inmunoprecipitación , Inositol 1,4,5-Trifosfato/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Espacio Intracelular/metabolismo , Lectinas Tipo C/metabolismo , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Células 3T3 NIH , Unión Proteica , Proteínas Serina-Treonina Quinasas , Linfocitos T/metabolismo , Transcripción Genética , Proteínas ras/metabolismo
17.
Clin Cancer Res ; 16(17): 4339-52, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20601445

RESUMEN

PURPOSE: To understand the changes in gene expression in polycythemia vera (PV) progenitor cells and their relationship to JAK2V617F. EXPERIMENTAL DESIGN: Messenger RNA isolated from CD34(+) cells from nine PV patients and normal controls was profiled using Affymetrix arrays. Gene expression change mediated by JAK2V617F was determined by profiling CD34(+) cells transduced with the kinase and by analysis of leukemia cell lines harboring JAK2V617F, treated with an inhibitor. RESULTS: A PV expression signature was enriched for genes involved in hematopoietic development, inflammatory responses, and cell proliferation. By quantitative reverse transcription-PCR, 23 genes were consistently deregulated in all patient samples. Several of these genes such as WT1 and KLF4 were regulated by JAK2, whereas others such as NFIB and EVI1 seemed to be deregulated in PV by a JAK2-independent mechanism. Using cell line models and comparing gene expression profiles of cell lines and PV CD34(+) PV specimens, we have identified panels of 14 JAK2-dependent genes and 12 JAK2-independent genes. These two 14- and 12-gene sets could separate not only PV from normal CD34(+) specimens, but also other MPN such as essential thrombocytosis and primary myelofibrosis from their normal counterparts. CONCLUSIONS: A subset of the aberrant gene expression in PV progenitor cells can be attributed to the action of the mutant kinase, but there remain a significant number of genes characteristic of the disease but deregulated by as yet unknown mechanisms. Genes deregulated in PV as a result of the action of JAK2V617F or independent of the kinase may represent other targets for therapy.


Asunto(s)
Perfilación de la Expresión Génica , Janus Quinasa 2/genética , Mutación , Policitemia Vera/genética , Sustitución de Aminoácidos , Antígenos CD34/genética , Antígenos CD34/metabolismo , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Análisis por Conglomerados , Células Eritroides/citología , Células Eritroides/metabolismo , Eritropoyetina/farmacología , Humanos , Janus Quinasa 2/metabolismo , Factor 4 Similar a Kruppel , Análisis de Secuencia por Matrices de Oligonucleótidos , Policitemia Vera/sangre , Policitemia Vera/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
18.
Blood ; 114(13): 2764-73, 2009 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-19546476

RESUMEN

Sequential administration of DNA methyltransferase (DNMT) inhibitors and histone deacetylase (HDAC) inhibitors has demonstrated clinical efficacy in patients with hematologic malignancies. However, the mechanism behind their clinical efficacy remains controversial. In this study, the methylation dynamics of 4 TSGs (p15(INK4B), CDH-1, DAPK-1, and SOCS-1) were studied in sequential bone marrow samples from 30 patients with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) who completed a minimum of 4 cycles of therapy with 5-azacytidine and entinostat. Reversal of promoter methylation after therapy was observed in both clinical responders and nonresponders across all genes. There was no association between clinical response and either baseline methylation or methylation reversal in the bone marrow or purified CD34(+) population, nor was there an association with change in gene expression. Transient global hypomethylation was observed in samples after treatment but was not associated with clinical response. Induction of histone H3/H4 acetylation and the DNA damage-associated variant histone gamma-H2AX was observed in peripheral blood samples across all dose cohorts. In conclusion, methylation reversal of candidate TSGs during cycle 1 of therapy was not predictive of clinical response to combination "epigenetic" therapy. This trial is registered with http://www.clinicaltrials.gov under NCT00101179.


Asunto(s)
Azacitidina/administración & dosificación , Benzamidas/administración & dosificación , Daño del ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/tratamiento farmacológico , Piridinas/administración & dosificación , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Análisis Citogenético , Daño del ADN/fisiología , Esquema de Medicación , Epigénesis Genética/fisiología , Femenino , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Tiempo
19.
Proc Natl Acad Sci U S A ; 106(27): 11154-9, 2009 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-19549856

RESUMEN

WT1, a critical regulator of kidney development, is a tumor suppressor for nephroblastoma but in some contexts functions as an oncogene. A limited number of direct transcriptional targets of WT1 have been identified to explain its complex roles in tumorigenesis and organogenesis. In this study we performed genome-wide screening for direct WT1 targets, using a combination of ChIP-ChIP and expression arrays. Promoter regions bound by WT1 were highly G-rich and resembled the sites for a number of other widely expressed transcription factors such as SP1, MAZ, and ZNF219. Genes directly regulated by WT1 were implicated in MAPK signaling, axon guidance, and Wnt pathways. Among directly bound and regulated genes by WT1, nine were identified in the Wnt signaling pathway, suggesting that WT1 modulates a subset of Wnt components and responsive genes by direct binding. To prove the biological importance of the interplay between WT1 and Wnt signaling, we showed that WT1 blocked the ability of Wnt8 to induce a secondary body axis during Xenopus embryonic development. WT1 inhibited TCF-mediated transcription activated by Wnt ligand, wild type and mutant, stabilized beta-catenin by preventing TCF4 loading onto a promoter. This was neither due to direct binding of WT1 to the TCF binding site nor to interaction between WT1 and TCF4, but by competition of WT1 and TCF4 for CBP. WT1 interference with Wnt signaling represents an important mode of its action relevant to the suppression of tumor growth and guidance of development.


Asunto(s)
Pruebas Genéticas , Genoma/genética , Transducción de Señal/genética , Proteínas WT1/metabolismo , Proteínas Wnt/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Proteína de Unión a CREB/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , ADN/metabolismo , Embrión no Mamífero/metabolismo , Desarrollo Embrionario , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas/genética , Unión Proteica , Factores de Transcripción TCF/metabolismo , Transcripción Genética , Xenopus/embriología
20.
Mol Cancer Res ; 6(7): 1225-31, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18644985

RESUMEN

In its role as a tumor suppressor, WT1 transactivates several genes that are regulators of cell growth and differentiation pathways. For instance, WT1 induces the expression of the cell cycle regulator p21, the growth-regulating glycoprotein amphiregulin, the proapoptotic gene Bak, and the Ras/mitogen-activated protein kinase (MAPK) inhibitor Sprouty1. Here, we show that WT1 transactivates another important negative regulator of the Ras/MAPK pathway, MAPK phosphatase 3 (MKP3). In a WT1-inducible cell line that exhibits decreased cell growth and increased apoptosis on expression of WT1, microarray analysis showed that MKP3 is the most highly induced gene. This was confirmed by real-time PCR where MKP3 and other members of the fibroblast growth factor 8 syn expression group, which includes Sprouty 1 and the Ets family of transcription factors, were induced rapidly following WT1 expression. WT1 induction was associated with a block in the phosphorylation of extracellular signal-regulated kinase in response to epidermal growth factor stimulation, an effect mediated by MKP3. In the presence of a dominant-negative MKP3, WT1 could no longer block phosphorylation of extracellular signal-regulated kinase. Lastly, when MKP3 expression is down-regulated by short hairpin RNA, WT1 is less able to block Ras-mediated transformation of 3T3 cells.


Asunto(s)
Fosfatasa 6 de Especificidad Dual/biosíntesis , Proteínas WT1/metabolismo , Animales , Proliferación Celular , Fosfatasa 6 de Especificidad Dual/genética , Activación Enzimática , Inducción Enzimática , Factor 8 de Crecimiento de Fibroblastos/metabolismo , Regulación Enzimológica de la Expresión Génica , Humanos , Ratones , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Células 3T3 NIH , Proteína Oncogénica p21(ras) , Fosforilación , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA