Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Oncotarget ; 6(32): 33675-88, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26418896

RESUMEN

Innate immune cells strongly influence cancer growth and progression via multiple mechanisms including regulation of epithelial to mesenchymal transition (EMT). In this study, we investigated whether expression of the metabolic gene, heme oxygenase-1 (HO-1) in tumor microenvironment imparts significant effects on prostate cancer progression.We showed that HO-1 is expressed in MARCO-positive macrophages in prostate cancer (PCa) xenografts and human prostate cancers. We demonstrated that macrophage specific (LyzM-Cre) conditional deletion of HO-1 suppressed growth of PC3 xenografts in vivo and delayed progression of prostate intraepithelial neoplasia (PIN) in TRAMP mice. However, initiation and progression of cancer xenografts in the presence of macrophages lacking HO-1 resulted in loss of E-cadherin, a known marker of poor prognosis as well as EMT. Application of CO, a product of HO-1 catalysis, increased levels of E-cadherin in the adherens junctions between cancer cells. We further showed that HO-1-driven expression of E-cadherin in cancer cells cultured in the presence of macrophages is dependent on mitochondrial activity of cancer cells.In summary, these data suggest that HO-1-derived CO from tumor-associated macrophages influences, in part, E-cadherin expression and thus tumor initiation and progression.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Macrófagos/enzimología , Proteínas de la Membrana/metabolismo , Neoplasias de la Próstata/enzimología , Animales , Cadherinas/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Progresión de la Enfermedad , Hemo-Oxigenasa 1/genética , Xenoinjertos , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Microambiente Tumoral
2.
Cancer Res ; 73(23): 7009-21, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24121491

RESUMEN

One classical feature of cancer cells is their metabolic acquisition of a highly glycolytic phenotype. Carbon monoxide (CO), one of the products of the cytoprotective molecule heme oxygenase-1 (HO-1) in cancer cells, has been implicated in carcinogenesis and therapeutic resistance. However, the functional contributions of CO and HO-1 to these processes are poorly defined. In human prostate cancers, we found that HO-1 was nuclear localized in malignant cells, with low enzymatic activity in moderately differentiated tumors correlating with relatively worse clinical outcomes. Exposure to CO sensitized prostate cancer cells but not normal cells to chemotherapy, with growth arrest and apoptosis induced in vivo in part through mitotic catastrophe. CO targeted mitochondria activity in cancer cells as evidenced by higher oxygen consumption, free radical generation, and mitochondrial collapse. Collectively, our findings indicated that CO transiently induces an anti-Warburg effect by rapidly fueling cancer cell bioenergetics, ultimately resulting in metabolic exhaustion.


Asunto(s)
Monóxido de Carbono/farmacología , Proliferación Celular/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Neoplasias/metabolismo , Neoplasias/patología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Monóxido de Carbono/administración & dosificación , Células Cultivadas , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Células HeLa , Células Hep G2 , Humanos , Ratones , Ratones Desnudos , Ratones Transgénicos , Células 3T3 NIH , Neoplasias/tratamiento farmacológico
3.
APMIS ; 121(11): 1017-9, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24106870

RESUMEN

In this mini-review, progress in our understanding of the link between miRNAs and cytokines in inflammatory skin diseases is highlighted with focus on miR-155 in mycosis fungoides (MF). MF is the most common variant of cutaneous T-cell lymphoma (CTCL) and is characterized by malignant T-cell proliferation in a chronic inflammatory environment in affected skin. Recent data show that miR-155 is expressed in situ by both malignant and non-malignant T cells, induced via the STAT5 signal pathway and IL-2Rbeta/gamma cytokines, and thus suggest the importance of miR-155 in malignant proliferation, clinical diagnosis, and prognosis in CTCL.


Asunto(s)
Dermatitis/genética , MicroARNs/fisiología , Micosis Fungoide/genética , Neoplasias Cutáneas/genética , Dermatitis/inmunología , Humanos , Interleucina-15/fisiología , Interleucina-2/fisiología , Micosis Fungoide/inmunología , Factor de Transcripción STAT5/fisiología , Neoplasias Cutáneas/inmunología
4.
Drug Des Devel Ther ; 7: 875-81, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24009414

RESUMEN

Prostate cancer (PCa) is the most common malignancy, and the third leading cancer-related cause of death among men of the Western world. Upon PCa progression into metastatic disease, androgen deprivation therapy is applied as the first-line treatment, and has been shown to be effective in most patients, leading to a decrease in serum prostate-specific antigen and relief of disease-related symptoms. However, advanced PCa almost inevitably progresses to a castration-resistant state, and is currently regarded as incurable. The large body of evidence indicates that PCa cells remain dependent on androgen receptor (AR) signaling even in an androgen-deprived environment. As such, development of drugs that target AR and AR signaling pathways have become one of the major milestones in treatment of castration-resistant PCa (CRPC). Nevertheless, currently available therapies that target AR signaling are still regarded as palliative and more potent therapies are in great need. Over the past few years, a wide range of novel therapies has entered clinical trial for treatment of CRPC, including androgen synthesis inhibitors (abiraterone acetate), chemotherapeutic agents (docetaxel and cabazitaxel), and immunotherapies (sipuleucel-T). In this context, enzalutamide (previously referred to as MDV3100) is a novel second generation antiandrogen that has been demonstrated to significantly improve survival in men with metastatic CRPC in several clinical trials. In this paper we summarize recently completed and ongoing clinical trials of enzalutamide, and briefly discuss the efficacy of the novel antiandrogen therapy and its limitations for treatment of CRPC.


Asunto(s)
Antineoplásicos/uso terapéutico , Feniltiohidantoína/análogos & derivados , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Antagonistas de Andrógenos/farmacología , Antagonistas de Andrógenos/uso terapéutico , Animales , Antineoplásicos/farmacología , Benzamidas , Ensayos Clínicos como Asunto , Progresión de la Enfermedad , Humanos , Masculino , Nitrilos , Feniltiohidantoína/farmacología , Feniltiohidantoína/uso terapéutico , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata Resistentes a la Castración/patología , Transducción de Señal , Tasa de Supervivencia
5.
PLoS One ; 8(8): e72210, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23991063

RESUMEN

Alterations in cellular pathways related to both endocrine and vascular endothelial growth factors (VEGF) may contribute to breast cancer progression. Inhibition of the elevated levels of these pathways is associated with clinical benefits. However, molecular mechanisms by which endocrine-related pathways and VEGF signalling cooperatively promote breast cancer progression remain poorly understood. In the present study, we show that the A-type cyclin, cyclin A1, known for its important role in the initiation of leukemia and prostate cancer metastasis, is highly expressed in primary breast cancer specimens and metastatic lesions, in contrasting to its barely detectable expression in normal human breast tissues. There is a statistically significant correlation between cyclin A1 and VEGF expression in breast cancer specimens from two patient cohorts (p<0.01). Induction of cyclin A1 overexpression in breast cancer cell line MCF-7 results in an enhanced invasiveness and a concomitant increase in VEGF expression. In addition, there is a formation of protein-protein complexes between cyclin A1 and estrogen receptor ER-α cyclin A1 overexpression increases ER-α expression in MCF-7 and T47D cells. In mouse tumor xenograft models in which mice were implanted with MCF-7 cells that overexpressed cyclin A1 or control vector, cyclin A1 overexpression results in an increase in tumor growth and angiogenesis, which is coincident with an enhanced expression of VEGF, VEGFR1 and ER-α Our findings unravel a novel role for cyclin A1 in growth and progression of breast cancer, and suggest that multiple cellular pathways, including cell cycle regulators, angiogenesis and estrogen receptor signalling, may cooperatively contribute to breast cancer progression.


Asunto(s)
Neoplasias de la Mama/genética , Ciclina A1/genética , Perfilación de la Expresión Génica , Neovascularización Patológica/genética , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclina A1/metabolismo , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Immunoblotting , Inmunohistoquímica , Metástasis Linfática , Células MCF-7 , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos , Ratones Desnudos , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
7.
Mol Oncol ; 7(3): 555-66, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23384557

RESUMEN

Recurrent prostate cancer remains a major clinical challenge. The lysine specific demethylase-1 (LSD1/KDM1A), together with the JmjC domain-containing JMJD2A and JMJD2C proteins, have emerged as critical regulators of histone lysine methylation. The LSD1-JMJD2 complex functions as a transcriptional co-regulator of hormone activated androgen and estrogen receptors at specific gene promoters. LSD1 also regulates DNA methylation and p53 function. LSD1 is overexpressed in numerous cancers including prostate cancer through an unknown mechanism. We investigated expression of the LSD1 and JMJD2A in malignant human prostate specimens. We correlated LSD1 and JMJD2A expression with known mediators of prostate cancer progression: VEGF-A and cyclin A1. We show that elevated expression of LSD1, but not JMJD2A, correlates with prostate cancer recurrence and with increased VEGF-A expression. We show that functional depletion of LSD1 expression using siRNA in prostate cancer cells decreases VEGF-A and blocks androgen induced VEGF-A, PSA and Tmprss2 expression. We demonstrate that pharmacological inhibition of LSD1 reduces proliferation of both androgen dependent (LnCaP) and independent cell lines (LnCaP: C42, PC3). We show a direct mechanistic link between LSD1 over-expression and increased activity of pro-angiogenic pathways. New therapies targeting LSD1 activity should be useful in the treatment of hormone dependent and independent prostate cancer.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Histona Demetilasas/metabolismo , Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Factor A de Crecimiento Endotelial Vascular/genética , Andrógenos/metabolismo , Línea Celular Tumoral , Proliferación Celular , Ciclina A1/genética , Histona Demetilasas/análisis , Histona Demetilasas/genética , Humanos , Masculino , Persona de Mediana Edad , Próstata/enzimología , Próstata/metabolismo , Neoplasias de la Próstata/enzimología , Interferencia de ARN , Activación Transcripcional , Factor A de Crecimiento Endotelial Vascular/metabolismo
8.
Anticancer Res ; 33(2): 363-70, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23393325

RESUMEN

The aim of this study was to examine the expression of serotonin receptors in patients with breast cancer and to explore their utility as diagnostic and prognostic markers. Immunohistochemical analysis was performed to examine the expression of serotonin (5-HT) receptor subtypes 1A, 1B, 2B and 4 in a tissue microarray containing tumor specimens from 102 patients. Statistical analysis was performed to correlate the expression of these proteins with regard to clinical parameters. We found that all four serotonin receptors (5-HTRs) exhibited different expression patterns in breast cancer specimens. In general strong staining for 5-HTR1A was observed on the membrane of cancer cells but it was detected only in the cytoplasm of non-malignant cells. 5-HTR1B and 5-HTR2B were predominantly expressed in the cytoplasm of breast cancer cells, while 5-HTR4 was exclusively found in the nucleus of malignant and non-malignant cells. Correlation analysis revealed a significant correlation of 5-HTR2B with estrogen receptor-α (ER-α) and 5-HTR4 with ER-α and progesterone (PR). In conclusion, the different expression patterns and subcellular localization of 5-HTRs in breast cancer may reflect their role in breast cancer progression.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Mama/metabolismo , Receptores de Serotonina/biosíntesis , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Femenino , Humanos , Inmunohistoquímica , Receptores de Serotonina/análisis , Análisis de Matrices Tisulares
9.
Curr Drug Targets ; 13(10): 1308-23, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22746994

RESUMEN

Most of the prostate cancers (PCa) in advanced stage will progress to castration-resistant prostate cancer (CRPC). Within CRPC group, 50-70% of the patients will develop bone metastasis in axial and other regions of the skeleton. Once PCa cells spread to the bone, currently, no treatment regimens are available to eradicate the metastasis, and cancer-related death becomes inevitable. In 2012, it is estimated that there will be 28,170 PCa deaths in the United States. Thus, PCa bone metastasis-associated clinical complications and treatment resistance pose major clinical challenges. In this review, we will present recent findings on the molecular and cellular pathways that are responsible for bone metastasis of PCa. We will address several novel mechanisms with a focus on the role of bone and bone marrow microenvironment in promoting PCa metastasis, and will further discuss why prostate cancer cells preferentially metastasize to the bone. Additionally, we will discuss novel roles of several key pathways, including angiogenesis and extracellular matrix remodeling in bone marrow and stem cell niches with their relationship to PCa bone metastasis and poor treatment response. We will evaluate how various chemotherapeutic drugs and radiation therapies may allow aggressive PCa cells to gain advantageous mutations leading to increased survival and rendering the cancer cells to become resistant to treatment. The novel concept relating several key survival and invasion signaling pathways to stem cell niches and treatment resistance will be reviewed. Lastly, we will provide an update of several recently developed novel drug candidates that target metastatic cancer microenvironments or niches, and discuss the advantages and significance provided by such therapeutic approaches in pursuit of overcoming drug resistance and treating advanced PCa.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Resistencia a Antineoplásicos , Humanos , Masculino , Factores de Riesgo
10.
Int J Oncol ; 40(1): 5-12, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21956088

RESUMEN

Bladder cancer is three times more common in men than in women. However, the physiological basis of the male predominance of bladder cancer remains poorly understood. A higher than expected association of prostate and bladder cancers has also been reported which may indicate a common mechanism of carcinogenesis. Consistent with this, androgens and the androgen receptor (AR) play essential roles in prostate carcinogenesis and are believed to play a role in bladder carcinogenesis. There is also evidence implicating cancer stem cells in prostate and bladder cancers. Indeed putative prostate and bladder cancer stem cells share some common molecular features. We highlight key proteins (CD49f, CD133, PTEN, CD44) which are implicated in both prostate and bladder cancers and are enriched in putative prostate and bladder cancer stem cells. We examine published chromatin immuno-precipitation studies analyzing the genome-wide distribution of the AR to identify AR association with, and by inference potential AR-regulation of, these loci. We discuss recent evidence indicating a role for the AR in the splicing of the key urological stem cell protein CD44. We propose a model whereby aberrant AR regulation of these putative stem cell proteins contributes to malignant transformation of prostate and bladder cells. For these reasons we propose that the relationship between androgens and cancer stem cell associated proteins warrants further investigation.


Asunto(s)
Células Madre Neoplásicas/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Femenino , Humanos , Masculino , Transducción de Señal
11.
Int J Cancer ; 128(12): 2843-52, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21128282

RESUMEN

The course of prostate cancer varies greatly, and additional prognostic markers are needed. Leucine-rich repeats and immunoglobulin-like domains protein 1 (LRIG1) is an endogenous inhibitor of growth factor signaling and a proposed tumor suppressor. Publicly available gene expression datasets indicate that LRIG1 may be overexpressed in prostate cancer. In our study, the expression of LRIG1 protein in prostate cancer was evaluated for the first time. Immunohistochemistry was performed on tissue microarrays from two different patient series: 355 Swedish patients diagnosed by transurethral resection and 293 American patients who underwent radical prostatectomy. In the Swedish series, high expression of LRIG1 correlated with Gleason score, T-stage, tumor cell proliferation, vascular density and epidermal growth factor receptor (EGFR) phosphorylation. Among the 256 Swedish patients, followed by watchful waiting, high LRIG1 expression was significantly associated with short overall and prostate cancer-specific survival. In contrast, in the US series, high LRIG1 expression was significantly associated with long overall survival. In vitro cell experiments showed that LRIG1 was induced by androgen stimulation, and its expression inhibited prostate cancer cell proliferation. Thus, LRIG1 expression was an independent marker for poor survival in the untreated patient series, perhaps as a secondary marker of androgen receptor and/or EGFR activation. On the contrary, LRIG1 was a marker for good prognosis after prostatectomy, which might be due to its growth inhibiting properties. We propose that LRIG1 is an important determinant of prostate cancer growth, and the implications of its expression on patient outcome depend on the clinical and biological circumstances.


Asunto(s)
Glicoproteínas de Membrana/fisiología , Neoplasias de la Próstata/metabolismo , Anciano , Anciano de 80 o más Años , Proliferación Celular , Humanos , Inmunohistoquímica , Masculino , Glicoproteínas de Membrana/genética , Persona de Mediana Edad , Prostatectomía , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/cirugía , Análisis de Matrices Tisulares
12.
PLoS One ; 4(11): e7953, 2009 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-19956729

RESUMEN

Cystatin C is believed to prevent tumor progression by inhibiting the activities of a family of lysosomal cysteine proteases. However, little is known about the precise mechanism of cystatin C function in prostate cancer. In the present study, we examined the expression of cystatin C and its association with matrix metalloproteinases 2 (MMP2) and androgen receptor (AR) in a tissue microarray comparing benign and malignant specimens from 448 patients who underwent radical prostatectomy for localized prostate cancer. Cystatin C expression was significantly lower in cancer specimens than in benign tissues (p<0.001) and there was a statistically significant inverse correlation between expression of cystatin C and MMP2 (r(s) (2) = -0.056, p = 0.05). There was a clear trend that patients with decreased level of cystatin C had lower overall survival. Targeted inhibition of cystatin C using specific siRNA resulted in an increased invasiveness of PC3 cells, whereas induction of cystatin C overexpression greatly reduced invasion rate of PC3 in vitro. The effect of cystatin C on modulating the PC3 cell invasion was provoked by Erk2 inhibitor that specifically inhibited MAPK/Erk2 activity. This suggests that cystatin C may mediate tumor cell invasion by modulating the activity of MAPK/Erk cascades. Consistent with our immunohistochemical findings that patients with low expression of cystatin C and high expression of androgen receptor (AR) tend to have worse overall survival than patients with high expression of cystatin C and high AR expression, induced overexpression of AR in PC3 cells expressing cystatin C siRNA greatly enhanced the invasiveness of PC3 cells. This suggests that there may be a crosstalk between cystatin C and AR-mediated pathways. Our study uncovers a novel role for cystatin C and its associated cellular pathways in prostate cancer invasion and metastasis.


Asunto(s)
Cistatina C/metabolismo , Regulación Neoplásica de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Supervivencia sin Enfermedad , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , ARN Interferente Pequeño/metabolismo , Factores de Tiempo , Resultado del Tratamiento
13.
J Pediatr Hematol Oncol ; 31(9): 696-701, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19707156

RESUMEN

Perturbation in the expression and signaling pathways of vascular endothelial growth factor (VEGF) has been linked to pathogenesis of hematologic malignancies. We investigated the expression and clinical importance of VEGF and two of its receptors, VEGFR-1 and VEGFR-2, in childhood precursor B-cell acute lymphoblastic leukemia (pre-B ALL) by using immunohistochemistry. These angiogenic proteins were expressed in the majority of leukemic bone marrow samples. Notably, pre-B ALL patients had significantly increased expression of VEGFR-1 compared with no expression in the nonmalignant group, indicating a link between VEGFR-1 protein expression and pre-B ALL. These novel findings suggest that VEGFR-1 may have clinical importance in childhood pre-B ALL.


Asunto(s)
Técnicas para Inmunoenzimas , Proteínas de Neoplasias/análisis , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Factor A de Crecimiento Endotelial Vascular/análisis , Receptor 1 de Factores de Crecimiento Endotelial Vascular/análisis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis , Adolescente , Médula Ósea/metabolismo , Médula Ósea/patología , Niño , Preescolar , Femenino , Estudios de Seguimiento , Regulación Leucémica de la Expresión Génica , Humanos , Lactante , Masculino , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neovascularización Patológica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Pronóstico , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/biosíntesis , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética
14.
Stem Cells Dev ; 18(7): 1093-108, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19480567

RESUMEN

Coordinated transcription factor networks have emerged as the master regulatory mechanisms of stem cell pluripotency and differentiation. Many stem cell-specific transcription factors, including the pluripotency transcription factors, OCT4, NANOG, and SOX2 function in combinatorial complexes to regulate the expression of loci, which are involved in embryonic stem (ES) cell pluripotency and cellular differentiation. This review will address how these pathways form a reciprocal regulatory circuit whereby the equilibrium between stem cell self-renewal, proliferation, and differentiation is in perpetual balance. We will discuss how distinct epigenetic repressive pathways involving polycomb complexes, DNA methylation, and microRNAs cooperate to reduce transcriptional noise and to prevent stochastic and aberrant induction of differentiation. We will provide a brief overview of how these networks cooperate to modulate differentiation along hematopoietic and neuronal lineages. Finally, we will describe how aberrant functioning of components of the stem cell regulatory network may contribute to malignant transformation of adult stem cells and the establishment of a "cancer stem cell" phenotype and thereby underlie multiple types of human malignancies.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas de Homeodominio/metabolismo , MicroARNs/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/metabolismo , Factores de Transcripción SOXB1/metabolismo , Transcripción Genética/fisiología , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Metilación de ADN/fisiología , Epigénesis Genética/fisiología , Proteínas de Homeodominio/genética , Humanos , MicroARNs/genética , Proteína Homeótica Nanog , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/citología , Factores de Transcripción SOXB1/genética
15.
Pediatr Hematol Oncol ; 26(1): 48-56, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19206008

RESUMEN

PTEN and SHP1 are tumor suppressor genes involved in the regulation of cell cycle control and apoptosis. The authors investigated the protein expression of PTEN and SHP1, by immunohistochemistry in tissue microarrays from bone marrow samples in children, diagnosed with acute lymphoblastic leukaemia and nonmalignant controls. PTEN was overexpressed in diagnostic ALL samples, while SHP1 showed a low expression. Both proteins showed a significant difference in expression compared to nonmalignant controls. The roles of PTEN and SHP1 are not well investigated in pediatric leukemia and could in the future play a role as prognostic factors.


Asunto(s)
Fosfohidrolasa PTEN/análisis , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 6/análisis , Adolescente , Biomarcadores de Tumor , Médula Ósea/patología , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Regulación Leucémica de la Expresión Génica , Humanos , Inmunohistoquímica , Lactante , Masculino , Análisis de Matrices Tisulares
16.
Mol Cell Biochem ; 320(1-2): 115-24, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18787932

RESUMEN

Understanding of molecular mechanisms underlying the effects of cell cycle proteins in response to the chemotherapeutic agents is of great importance for improving the efficacy of targeted therapeutics and overcoming resistance to chemotherapeutic agents. Staurosporine and tumor necrosis factor alpha (TNFalpha) are the therapeutic agents that inhibit tumor cell growth by inducing cell death. Staurosporine induces apoptosis through the intrinsic pathway, while TNFalpha trigger the cell death via the extrinsic apoptotic pathway. We have previously demonstrated that the cell cycle regulatory protein, cyclin A1 played an important role in the development of acute myeloid leukemia (AML), and cyclin A1 expression correlated with disease characteristics and patient outcome in leukemia. However, it remains unknown how cyclin A1 expression is regulated in leukemic cells treated with the therapeutic agents. Here, we demonstrate that cyclin A1 protein is regulated by proteasome-mediated ubiquitination and degradation in untreated U-937 cells. Interestingly, ubiquitination- and proteasomal-mediated degradation of cyclin A1 is prevented in cells treated with staurosporine or TNFalpha. Induction of apoptosis in U-937 cells by staurosporine or TNFalpha resulted in an increase in cyclin A1 protein expression, which correlated well with cyclin A1 protein modification and the activation of caspase-3. Blocking caspases activity by Z-VAD-FMK had no effect on the increased cyclin A1 expression, suggesting that cyclin A1 might be regulated by caspase-3 independent pathways. We further propose that CDC25C may be associated with cyclin A1 protein modification in response to staurosporine or TNFalpha treatment. Our results suggest that cyclin A1 protein is stabilized via post-transcriptional modification in response to apoptosis induced by staurosporine or TNFalpha.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclina A1/metabolismo , Inhibidores Enzimáticos/farmacología , Leucemia/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estaurosporina/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Caspasa 3/metabolismo , Inhibidores de Caspasas , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Ciclina A1/química , Ciclina A1/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Inhibidores de Cisteína Proteinasa/farmacología , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Ubiquitinación , Fosfatasas cdc25/metabolismo
17.
Anticancer Agents Med Chem ; 8(7): 723-31, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18855574

RESUMEN

Cyclins and CDKs play critical roles in DNA synthesis and cell division. Alterations in their function may lead to the disruption of normal cell growth and apoptosis, and subsequently, result in carcinogenesis. Elevated levels of cyclins and CDKs are frequently observed in a wide range of different types of human cancers. Understanding of molecular mechanisms underlying the cell cycle effects in response to the chemotherapeutic agents is of great importance for improving the efficacy of targeted therapeutics and overcoming resistance to chemotherapeutic agents. Despite the clinical applications of cell cycle specific chemotherapeutic agents, there is still an urgent need to develop novel drugs that can target multiple sites and pathways of the cell cycle while avoiding drug induced cytotoxicity. In this review article, we will summarize the development of novel agents that specifically target cell cycle pathways in human cancer. We will discuss drugs that can directly interfere with the mitotic process of tumor cells. Moreover, we tend to address the significance of using small molecule CDK inhibitors that are derived from natural products.


Asunto(s)
Antineoplásicos , Ciclo Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Neoplasias/tratamiento farmacológico , Antineoplásicos/aislamiento & purificación , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Estructura Molecular , Neoplasias/metabolismo , Neoplasias/patología
18.
J Natl Cancer Inst ; 100(14): 1022-36, 2008 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-18612129

RESUMEN

BACKGROUND: Cyclin A1 is a cell cycle regulator that has been implicated in the progression of prostate cancer. Its role in invasion and metastasis of this disease has not been characterized. METHODS: Immunohistochemistry and cDNA microarray analyses were used to assess protein and mRNA expression of cyclin A1 and proteins with roles in metastasis, including vascular endothelial growth factor (VEGF), metalloproteinase 2 (MMP2), and MMP9, in human prostate cancer. Transient transfection and infection with viral vectors expressing cyclin A1 and short hairpin RNA (shRNA) targeting cyclin A1 were used to study the effects of altered cyclin A1 expression in PC3 prostate cancer cells. The BrdU assay, annexin V staining, and invasion chambers were used to examine cyclin A1 effects on proliferation, apoptosis, and invasion, respectively. The role of cyclin A1 and androgen receptor (AR) in transcription of VEGF and MMP2 was assessed by promoter mutation and chromatin immunoprecipitation. The effect of cyclin A1 expression on tumor growth and metastasis was analyzed in a mouse model of metastasis. All statistical tests were two-sided. RESULTS: Cyclin A1 protein and mRNA expression were statistically significantly higher in prostate cancers than in adjacent benign tissues. A statistically significant correlation between expression of cyclin A1 and of MMP2, MMP9, and VEGF was observed in prostate tumors from 482 patients (P values from Spearman rank correlation tests < .001). PC3 cells that overexpressed cyclin A1 showed increased invasiveness, and inhibition of cyclin A1 expression via shRNA expression reduced invasiveness of these cells. Eight of 10 mice (80%) bearing PC3 cells overexpressing cyclin A1 had infiltration of tumor cells in lymph node, liver, and lung, but all 10 mice bearing tumors expressing control vector were free of liver and lung metastases and only one mouse from this group had lymph node metastasis (P values from Fisher exact tests < .001). Cyclin A1, in concert with AR, bound to and increased expression from the VEGF and MMP2 promoters. CONCLUSIONS: Cyclin A1 contributes to prostate cancer invasion by modulating the expression of MMPs and VEGF and by interacting with AR.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Ciclina A/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Anciano , Anciano de 80 o más Años , Animales , Inmunoprecipitación de Cromatina , Ciclina A/genética , Ciclina A1 , Ensayo de Inmunoadsorción Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Immunoblotting , Inmunohistoquímica , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Metribolona/farmacología , Ratones , Invasividad Neoplásica , Neovascularización Patológica/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias de la Próstata/irrigación sanguínea , ARN Mensajero/metabolismo , Receptores Androgénicos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Int J Cancer ; 122(7): 1521-9, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18027847

RESUMEN

Interleukin-6 (IL6) is a growth and survival factor in human prostate cancer (PCa) cells with aggressive phenotypes and has been implicated in the progression of hormone refractory PCas. In the present study, we characterized the IL6-triggered PI3K/Akt and MAPK/Erk signaling. We identified the A-type cyclin, cyclin A1 as an important downstream target of PI3K/Akt. Treatment of cells with PI3K inhibitor or cotransfection with a vector expressing wild-type PTEN decreased cyclin A1 promoter activity. Cyclin A1 promoter activity and its expression were upregulated by constitutively active myristoylated Akt and were downregulated by dominant negative Akt in response to IL6 stimulation. LNCaP cells overexpressing cyclin A1 are resistant to camptothecin-induced apoptosis. Conversely, targeted knockdown of cyclin A1 via shRNA in LNCaP IL6+ cells resulted in decreased survival after treatment with camptothecin. This suggests that cyclin A1 is an important downstream target of PI3K/Akt that transduces survival signals in response to IL6 stimulation. Xenograft tumors generated from LNCaP-IL6+ cells expressing IL6 had higher levels of cyclin A1 and had rapid tumor growth compared to LNCaP xenograft tumors. Taken together, IL6 might utilize PI3K/Akt and cyclin A1 to promote tumor cell survival in PCa.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Camptotecina/farmacología , Ciclina A/metabolismo , Interleucina-6/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Apoptosis , Supervivencia Celular , Ciclina A1 , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Masculino , Fosfohidrolasa PTEN/metabolismo , Plásmidos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/fisiopatología , Transducción de Señal/efectos de los fármacos , Transfección , Trasplante Heterólogo , Regulación hacia Arriba/efectos de los fármacos
20.
Int J Biochem Cell Biol ; 38(8): 1330-9, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16517207

RESUMEN

Deregulated cell growth and inhibition of apoptosis are hallmarks of cancer. All-trans retinoic acid induces clinical remission in patients with acute promyelocytic leukemia by inhibiting cell growth and inducing differentiation and apoptosis of the leukemic blasts. An important role of the cell cycle regulatory protein, cyclin A1, in the development of acute myeloid leukemia has previously been demonstrated in a transgenic mouse model. We have recently shown that there was a direct interaction between cyclin A1 and a major all-trans retinoic acid receptor, RAR alpha, following all-trans retinoic acid treatment of leukemic cells. In the present study, we investigated whether cyclin A1 might be involved in all-trans retinoic acid-induced apoptosis in U-937 leukemic cells. We found that all-trans retinoic acid-induced apoptosis was associated with concomitant increase in cyclin A1 expression. However, there was no induction of cyclin A1 mRNA expression following the all-trans retinoic acid-induced apoptosis. Treatment of cells with a caspase inhibitor was not able to prevent all-trans retinoic acid-induced up-regulation of cyclin A1 expression. Interestingly, induced cyclin A1 expression in U-937 cells led to a significant increase in the proportion of apoptotic cells. Further, U-937 cells overexpressing cyclin A1 appeared to be more sensitive to all-trans retinoic acid-induced apoptosis indicating the ability of cyclin A1 to mediate all-trans retinoic acid-induced apoptosis. Induced cyclin E expression was not able to initiate cell death in U-937 cells. Our results indicate that cyclin A1 might have a role in apoptosis by mediating all-trans retinoic acid-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclina A/metabolismo , Tretinoina/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ciclina A/genética , Ciclina A1 , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Immunoblotting , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Transcripción Genética/efectos de los fármacos , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...