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










Base de datos
Intervalo de año de publicación
1.
J Appl Microbiol ; 132(1): 78-89, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34218487

RESUMEN

Plastics have become ubiquitous in both their adoption as materials and as environmental contaminants. Widespread pollution of these versatile, man-made and largely petroleum-derived polymers has resulted from their long-term mass production, inappropriate disposal and inadequate end of life management. Polyethylene (PE) is at the forefront of this problem, accounting for one-third of plastic demand in Europe in part due to its extensive use in packaging. Current recycling and incineration processes do not represent sustainable solutions to tackle plastic waste, especially once it becomes littered, and the development of new waste-management and remediation technologies are needed. Mycoremediation (fungal-based biodegradation) of PE has been the topic of several studies over the last two decades. The utility of these studies is limited by an inconclusive definition of biodegradation and a lack of knowledge regarding the biological systems responsible. This review highlights relevant features of fungi as potential bioremediation agents, before discussing the evidence for fungal biodegradation of both high- and low-density PE. An up-to-date perspective on mycoremediation as a future solution to PE waste is provided.


Asunto(s)
Plásticos , Polietileno , Biodegradación Ambiental , Hongos , Humanos , Reciclaje
2.
Molecules ; 26(14)2021 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-34299465

RESUMEN

Lytic transglycosylases such as Slt35 from E. coli are enzymes involved in bacterial cell wall remodelling and recycling, which represent potential targets for novel antibacterial agents. Here, we investigated a series of known glycosidase inhibitors for their ability to inhibit Slt35. While glycosidase inhibitors such as 1-deoxynojirimycin, castanospermine, thiamet G and miglitol had no effect, the phenothiazinium dye thionine acetate was found to be a weak inhibitor. IC50 values and binding constants for thionine acetate were similar for Slt35 and the hen egg white lysozyme. Molecular docking simulations suggest that thionine binds to the active site of both Slt35 and lysozyme, although it does not make direct interactions with the side-chain of the catalytic Asp and Glu residues as might be expected based on other inhibitors. Thionine acetate also increased the potency of the beta-lactam antibiotic ampicillin against a laboratory strain of E. coli.


Asunto(s)
Glicosiltransferasas/metabolismo , Fenotiazinas/farmacología , Acetatos/metabolismo , Secuencia de Aminoácidos/genética , Proteínas Bacterianas/química , Sitios de Unión/genética , Dominio Catalítico/genética , Pared Celular/metabolismo , Cristalografía por Rayos X/métodos , Escherichia coli/metabolismo , Proteínas de Escherichia coli/efectos de los fármacos , Proteínas de Escherichia coli/metabolismo , Glicosiltransferasas/antagonistas & inhibidores , Glicosiltransferasas/efectos de los fármacos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Muramidasa/antagonistas & inhibidores , Muramidasa/metabolismo , Peptidoglicano/metabolismo , Fenotiazinas/metabolismo , Conformación Proteica/efectos de los fármacos
3.
Int J Oncol ; 46(3): 1099-108, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25502497

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is often diagnosed when metastatic events have occurred. Cancer stem cells (CSCs) play an important role in tumor initiation, metastasis, chemoresistance and relapse. A growing number of studies have suggested that CSCs exist in a dynamic equilibrium with more differentiated cancer cells via a bi­directional regeneration that is dependent on the environmental stimuli. In this investigation, we obtain, by using a selective medium, PDAC CSCs from five out of nine PDAC cell lines, endowed with different tumorsphere­forming ability. PDAC CSCs were generally more resistant to the action of five anticancer drugs than parental cell lines and were characterized by an increased expression of EpCAM and CD44v6, typical stem cell surface markers, and a decreased expression of E­cadherin, the main marker of the epithelial state. PDAC CSCs were able to re­differentiate into parental cells once cultured in parental growth condition, as demonstrated by re­acquisition of the epithelial morphology, the decreased expression levels of EpCAM and CD44v6 and the increased sensitivity to anticancer drugs. Finally, PDAC CSCs injected into nude mice developed a larger subcutaneous tumor mass and showed a higher metastatic activity compared to parental cells. The present study demonstrates the ability to obtain CSCs from several PDAC cell lines and that these cells are differentially resistant to various anticancer agents. This variability renders them a model of great importance to deeply understand pancreatic adenocarcinoma biology, to discover new biomarkers and to screen new therapeutic compounds.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Desdiferenciación Celular , Diferenciación Celular , Células Madre Neoplásicas/fisiología , Neoplasias Pancreáticas/patología , Animales , Biomarcadores de Tumor/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/fisiopatología , Desdiferenciación Celular/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Células Cultivadas , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/fisiopatología
4.
Eur J Pharm Biopharm ; 85(3 Pt A): 373-80, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23791684

RESUMEN

The aim of this work was the preparation, characterization, and preliminary evaluation of the targeting ability toward pancreatic adenocarcinoma cells of liposomes containing the gemcitabine lipophilic prodrug [4-(N)-lauroyl-gemcitabine, C12GEM]. Hyaluronic acid (HA) was selected as targeting agent since it is biodegradable, biocompatible, and can be chemically modified and its cell surface receptor CD44 is overexpressed on various tumors. For this purpose, conjugates between a phospholipid, the 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine (DPPE), and HA of two different low molecular weights 4800 Da (12 disaccharidic units) and 12,000 Da (32 disaccharidic units), were prepared, characterized, and introduced in the liposomes during the preparation. Different liposomal formulations were prepared and their characteristics were analyzed: size, Z potential, and TEM analyses underline a difference in the HA-liposomes from the non-HA ones. In order to better understand the HA-liposome cellular localization and to evaluate their interaction with CD44 receptor, confocal microscopy studies were performed. The results demonstrate that HA facilitates the recognition of liposomes by MiaPaCa2 cells (CD44(+)) and that the uptake increases with increase in the polymer molecular weight. Finally, the cytotoxicity of the different preparations was evaluated and data show that incorporation of C12GEM increases their cytotoxic activity and that HA-liposomes inhibit cell growth more than plain liposomes. Altogether, the results demonstrate the specificity of C12GEM targeting toward CD44-overexpressing pancreatic adenocarcinoma cell line using HA as a ligand.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antimetabolitos Antineoplásicos/administración & dosificación , Desoxicitidina/análogos & derivados , Ácido Hialurónico/química , Neoplasias Pancreáticas/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patología , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Desoxicitidina/administración & dosificación , Desoxicitidina/química , Desoxicitidina/farmacología , Sistemas de Liberación de Medicamentos , Regulación Neoplásica de la Expresión Génica , Humanos , Receptores de Hialuranos/genética , Liposomas , Peso Molecular , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Fosfatidiletanolaminas/química , Profármacos
5.
Biochim Biophys Acta ; 1828(5): 1396-404, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23384419

RESUMEN

Pancreatic adenocarcinoma is often diagnosed when metastatic events have occurred. The early spread of circulating cancer cells expressing the CD44 receptor may play a crucial role in this process. In this study, we have investigated the cellular delivery ability and both in vitro and in vivo anti-tumoral activity of liposomes conjugated with two different low molecular weight hyaluronic acids (HA 4.8kDa and HA 12kDa), the primary ligand of CD44, and containing a lipophilic gemcitabine (GEM) pro-drug. By confocal microscopy and flow cytometry analyses, we demonstrate that the cellular uptake into a highly CD44-expressing pancreatic adenocarcinoma cell line is higher with HA-conjugated (12kDa>4.8kDa) than non-conjugated liposomes. Consistently, in vitro cytotoxic assays display an increased sensitivity towards GEM containing HA-liposomes, compared to non-conjugated liposomes. Conversely, CD44 non-expressing normal cells show a similar uptake and in vitro cytotoxicity with both HA-conjugated and non-conjugated liposomes. Furthermore, we demonstrate that the HA-liposomes are taken up into the cells via lipid raft-mediated endocytosis. All the liposome formulations containing GEM show a higher antitumoral activity than free GEM in a mouse xenograft tumor model of human pancreatic adenocarcinoma. The 12kDa HA-liposomes have the strongest efficiency, while non-conjugated liposomes and the 4.8kDa HA-liposomes are similarly active. Taken together, our results provide a strong rationale for further development of HA-conjugated liposomes to treat pancreatic adenocarcinoma.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Desoxicitidina/análogos & derivados , Liposomas/química , Neoplasias Pancreáticas/tratamiento farmacológico , 1,2-Dipalmitoilfosfatidilcolina/química , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Antimetabolitos Antineoplásicos/química , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Colesterol/química , Desoxicitidina/química , Desoxicitidina/farmacología , Citometría de Flujo , Humanos , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/química , Ácido Hialurónico/metabolismo , Liposomas/metabolismo , Ratones , Ratones Desnudos , Microscopía Confocal , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Fosfatidiletanolaminas/química , Profármacos/química , Profármacos/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
6.
Biochim Biophys Acta ; 1833(3): 672-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23124112

RESUMEN

Mitochondrial uncoupling protein 2 (UCP2) can moderate oxidative stress by favoring the influx of protons into the mitochondrial matrix, thus reducing electron leakage from respiratory chain and mitochondrial superoxide production. Here, we demonstrate that UCP2 inhibition by genipin or UCP2 siRNA strongly increases reactive oxygen species (ROS) production inhibiting pancreatic adenocarcinoma cell growth. We also show that UCP2 inhibition triggers ROS-dependent nuclear translocation of the glycolytic enzyme glyceraldehyde 3-phosphate dehydrogenase (GAPDH), formation of autophagosomes, and the expression of the autophagy marker LC3-II. Consistently, UCP2 over-expression significantly reduces basal autophagy confirming the anti-autophagic role of UCP2. Furthermore, we demonstrate that autophagy induced by UCP2 inhibition determines a ROS-dependent cell death, as indicated by the apoptosis decrease in the presence of the autophagy inhibitors chloroquine (CQ) or 3-methyladenine (3-MA), or the radical scavenger NAC. Intriguingly, the autophagy induced by genipin is able to potentiate the autophagic cell death triggered by gemcitabine, the standard chemotherapeutic drug for pancreatic adenocarcinoma, supporting the development of an anti-cancer therapy based on UCP2 inhibition associated to standard chemotherapy. Our results demonstrate for the first time that UCP2 plays a role in autophagy regulation bringing new insights into mitochondrial uncoupling protein field.


Asunto(s)
Adenocarcinoma/patología , Autofagia , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Canales Iónicos/antagonistas & inhibidores , Iridoides/farmacología , Proteínas Mitocondriales/antagonistas & inhibidores , Neoplasias Pancreáticas/patología , Especies Reactivas de Oxígeno/metabolismo , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colagogos y Coleréticos/farmacología , Técnica del Anticuerpo Fluorescente , Humanos , Canales Iónicos/genética , Canales Iónicos/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Estrés Oxidativo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Transporte de Proteínas , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Desacopladora 2
7.
Apoptosis ; 18(3): 337-46, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23238993

RESUMEN

TP53 mutations compromising p53 transcriptional function occur in more than 50 % of human cancers, including pancreatic adenocarcinoma, and render cancer cells more resistant to conventional therapy. In the last few years, many efforts have been addressed to identify p53-reactivating molecules able to restore the wild-type transcriptionally competent conformation of the mutated proteins. Here, we show that two of these compounds, CP-31398 and RITA, induce cell growth inhibition, apoptosis, and autophagy by activating p53/DNA binding and p53 phosphorylation (Ser15), without affecting the total p53 amount. These effects occur in both wild-type and mutant p53 pancreatic adenocarcinoma cell lines, whereas they are much less pronounced in normal human primary fibroblasts. Furthermore, CP-31398 and RITA regulate the axis SESN1-2/AMPK/mTOR by inducing AMPK phosphorylation on Thr172, which has a crucial role in the autophagic response. The protective role of autophagy in cell growth inhibition by CP-31398 and RITA is supported by the finding that the AMPK inhibitor compound C or the autophagy inhibitors chloroquine or 3-methyladenine sensitize both pancreatic adenocarcinoma cell lines to the apoptotic response induced by p53-reactivating molecules. Our results demonstrate for the first time a survival role for autophagy induced by p53-reactivating molecules, supporting the development of an anti-cancer therapy based on autophagy inhibition associated to p53 activation.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Pirimidinas/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Línea Celular Tumoral , Activación Enzimática , Furanos/farmacología , Humanos , Serina-Treonina Quinasas TOR/metabolismo , Proteína p53 Supresora de Tumor/genética
8.
Biochim Biophys Acta ; 1823(10): 1856-63, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22705884

RESUMEN

Cancer cells exhibit an endogenous constitutive oxidative stress higher than that of normal cells, which renders tumours vulnerable to further reactive oxygen species (ROS) production. Mitochondrial uncoupling protein 2 (UCP2) can mitigate oxidative stress by increasing the influx of protons into the mitochondrial matrix and reducing electron leakage and mitochondrial superoxide generation. Here, we demonstrate that chemical uncouplers or UCP2 over-expression strongly decrease mitochondrial superoxide induction by the anticancer drug gemcitabine (GEM) and protect cancer cells from GEM-induced apoptosis. Moreover, we show that GEM IC(50) values well correlate with the endogenous level of UCP2 mRNA, suggesting a critical role for mitochondrial uncoupling in GEM resistance. Interestingly, GEM treatment stimulates UCP2 mRNA expression suggesting that mitochondrial uncoupling could have a role also in the acquired resistance to GEM. Conversely, UCP2 inhibition by genipin or UCP2 mRNA silencing strongly enhances GEM-induced mitochondrial superoxide generation and apoptosis, synergistically inhibiting cancer cell proliferation. These events are significantly reduced by the addition of the radical scavenger N-acetyl-l-cysteine or MnSOD over-expression, demonstrating a critical role of the oxidative stress. Normal primary fibroblasts are much less sensitive to GEM/genipin combination. Our results demonstrate for the first time that UCP2 has a role in cancer cell resistance to GEM supporting the development of an anti-cancer therapy based on UCP2 inhibition associated to GEM treatment.


Asunto(s)
Desoxicitidina/análogos & derivados , Resistencia a Antineoplásicos/efectos de los fármacos , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Acetilcisteína/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/genética , Iridoides/farmacología , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/genética , Neoplasias/enzimología , Neoplasias/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Desacopladores , Proteína Desacopladora 2 , Gemcitabina
9.
J Feline Med Surg ; 13(9): 672-80, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21872794

RESUMEN

UNLABELLED: PRESENTATION AND LESION LOCALISATION: Seven adult domestic shorthair cats were presented with a 1- to 6-day history of progressive neurological signs. A focal skin puncture and subcutaneous swelling over the dorsal part of the head were detected on physical examination. Neurological examination indicated lesion(s) in the right forebrain in four cats, multifocal forebrain in one cat, left forebrain in one cat, and multifocal forebrain and brainstem in the remaining cat. In all cats, magnetic resonance imaging revealed a space-occupying forebrain lesion causing a severe mass effect on adjacent brain parenchyma. CLINICAL APPROACH AND OUTCOME: All cats were managed with a combination of medical and surgical treatment. At surgery a small penetrating calvarial fracture was detected in all cats, and a tooth fragment was found within the content of the abscess in two cats. The combination of surgical intervention, intensive care and intravenous antimicrobials led to a return to normal neurological function in five cats. PRACTICAL RELEVANCE: As this series of cases indicates, successful resolution of a brain abscess due to a bite injury depends on early recognition and combined used of antimicrobials and surgical intervention. A particular aim of surgery is to remove any skull and foreign body (tooth) fragments that may represent a continuing focus of infection.


Asunto(s)
Mordeduras y Picaduras/veterinaria , Absceso Encefálico/veterinaria , Enfermedades de los Gatos/diagnóstico , Enfermedades de los Gatos/terapia , Gatos/lesiones , Traumatismos Penetrantes de la Cabeza/veterinaria , Animales , Mordeduras y Picaduras/complicaciones , Absceso Encefálico/diagnóstico , Absceso Encefálico/etiología , Absceso Encefálico/terapia , Enfermedades de los Gatos/patología , Femenino , Traumatismos Penetrantes de la Cabeza/complicaciones , Imagen por Resonancia Magnética/veterinaria , Masculino , Examen Neurológico/veterinaria , Prosencéfalo , Resultado del Tratamiento
10.
Free Radic Biol Med ; 50(8): 926-33, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21236335

RESUMEN

Pancreatic adenocarcinoma is a common malignancy that remains refractory to all available therapies, including the gold standard drug gemcitabine (GEM). We investigated the effect of the combination of GEM and each of the ionophore compounds pyrrolidine dithiocarbamate (PDTC) and disulfiram [DSF; 1-(diethylthiocarbamoyldisulfanyl)-N,N-diethylmethanethioamide] on p53(-/-) pancreatic adenocarcinoma cell growth. PDTC or DSF synergistically inhibited cell proliferation when used in combination with GEM by inducing apoptotic cell death. This effect was associated with an increased mitochondrial O(2)(•-) production and was further enhanced by zinc ions. Basal levels of mitochondrial O(2)(•-) or manganese superoxide dismutase (MnSOD) strictly correlated with the IC(50) for GEM or the percentage of synergism. Thus, the most relevant values of the antiproliferative synergism were obtained in GEM-resistant pancreatic adenocarcinoma cell lines. Interestingly, the GEM-sensitive T3M4 cells transfected with MnSOD expression vector showed mitochondrial O(2)(•-) and IC(50) for GEM similar to those of resistant cell lines. In vivo experiments performed on nude mice xenotransplanted with the GEM-resistant PaCa44 cell line showed that only the combined treatment with GEM and DSF/Zn completely inhibited the growth of the tumoral masses. These results and the consideration that DSF is already used in clinics strongly support the GEM and DSF/Zn combination as a new approach to overcoming pancreatic cancer resistance to standard chemotherapy.


Asunto(s)
Adenocarcinoma/patología , Desoxicitidina/análogos & derivados , Neoplasias Pancreáticas/patología , Tiocarbamatos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/farmacología , Sinergismo Farmacológico , Humanos , Gemcitabina
11.
Bioconjug Chem ; 21(4): 635-45, 2010 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-20201569

RESUMEN

"Zero-length" dimers of ribonuclease A, a novel type of dimers formed by two RNase A molecules bound to each other through a zero-length amide bond [Simons, B. L., et al. (2007) Proteins 66, 183-195], were further characterized and tested for their possible in vitro cytotoxic activity. Results obtained are the following. Besides dimers, also trimers and higher oligomers could be identified among the products of the covalently linking reaction, and the "zero-length" dimers prepared by us appear not to be a unique species. The product was indeed heterogeneous, and results obtained with two RNase A mutants, E9A and K66A, indicated that amino and carboxyl groups others than those belonging to Lys66 and Glu9 are involved in the amide bond. As for their functional properties, the "zero-length" dimers degrade poly(A).poly(U) (dsRNA) with an activity that increases with the increase of the oligomer's basicity and yeast RNA (ssRNA) with an activity that instead decreases with the increase of oligomer's basicity, which is in agreement with previous data. No cytotoxicity of the RNase A "zero-length" dimers could be evidenced in assays performed with various tumor cells lines; the dimers, instead, become cytotoxic if cationized by conjugation with polyethylenimine (PEI) [Futami et al. (2005) J. Biosci. Bioengin. 99, 95-103]. However, PEI derivatives of RNase A "zero-length" dimers and PEI derivatives of native RNase A resulted to be equally cytotoxic. In other words, protein "dimericity" does not play any role in this case. Moreover, the acquired cytotoxicity does not seem to be specific for tumor cells: PEI-cationized native RNase A was also cytotoxic toward human monocytes.


Asunto(s)
Ribonucleasa Pancreática/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Dimerización , Relación Dosis-Respuesta a Droga , Células HeLa , Humanos , Monocitos/citología , Mutación , Polietileneimina/química , Polietileneimina/farmacología , Ribonucleasa Pancreática/metabolismo , Relación Estructura-Actividad
12.
Nucleic Acids Res ; 37(20): 6681-90, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19745053

RESUMEN

The aim of the present study was to analyse the molecular mechanisms involved in the Interleukin-6 (IL-6) silencing in pancreatic adenocarcinoma cell lines. Our results demonstrate that TNF-alpha, a major IL-6 inducer, is able to induce IL-6 only in three out of six cell lines examined. 5-aza-2'-deoxycytidine (DAC), but not trichostatin A (TSA), activates the expression of IL-6 in all cell lines, indicating that DNA methylation, but not histone deacetylation, plays an essential role in IL-6 silencing. Indeed, the IL-6 upstream region shows a methylation status that correlates with IL-6 expression and binds MeCP2 and H3meK9 only in the non-expressing cell lines. Our results suggest that critical methylations located from positions -666 to -426 relative to the transcription start site of IL-6 may act as binding sites for MeCP2.


Asunto(s)
Adenocarcinoma/genética , Azacitidina/análogos & derivados , Silenciador del Gen , Interleucina-6/antagonistas & inhibidores , Proteína 2 de Unión a Metil-CpG/metabolismo , Neoplasias Pancreáticas/genética , Azacitidina/farmacología , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , ADN/metabolismo , Metilación de ADN , Decitabina , Humanos , Ácidos Hidroxámicos/farmacología , Interleucina-6/genética , Interleucina-6/metabolismo , FN-kappa B/metabolismo
13.
BMC Mol Biol ; 9: 56, 2008 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-18549468

RESUMEN

BACKGROUND: Despite the importance of the BCL2L11 (BIM) protein in various apoptotic processes in development and disease, little is known of the promoter structure of the human BCL2L11 locus and of the cis-acting elements regulating expression of the human gene. RESULTS: In the search for novel promoter sequences in the human BCL2L11 locus, we have identified previously unrecognized genomic sequences displaying promoter activity and E2F responsiveness, and driving the expression of BCL2L11 coding transcripts. In man, transcripts originating from this novel putative promoter contribute significantly to total BCL2L11 mRNA expression in testis, heart and liver. In HEK293 cells, this novel candidate promoter originates BCL2L11 transcripts whose expression can be modulated by a known modulator of BCL2L11 expression (Trichostatin A) and by E2F, a characterized transcriptional regulator of BCL2L11 expression. CONCLUSION: The identification of a novel putative human BCL2L11 promoter provides new insights into the structure and regulation of the BCL2L11 locus.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Perfilación de la Expresión Génica , Proteínas de la Membrana/genética , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Secuencia de Bases , Proteína 11 Similar a Bcl2 , Línea Celular , Biología Computacional , Factores de Transcripción E2F/genética , Factores de Transcripción E2F/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Exones/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ácidos Hidroxámicos/farmacología , Hígado/metabolismo , Masculino , Modelos Biológicos , Datos de Secuencia Molecular , Miocardio/metabolismo , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Testículo/metabolismo , Transcripción Genética/efectos de los fármacos
14.
Bioconjug Chem ; 18(6): 1946-55, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17914869

RESUMEN

Polyspermine-ribonuclease A (PS-RNase A) and polyspermine-dimeric ribonuclease A (PS-dimeric RNase A) were prepared by cross-linking ribonuclease A or its covalently linked dimer to polyspermine (PS) using dimethyl suberimidate. The two RNase A derivatives were tested for a possible antitumor action. The in vitro and in vivo cytotoxic activity of PS-RNase A, although strong, is not higher than that known for free polyspermine. PS-dimeric RNase A, which was characterized by mass spectroscopy, titration of free amine groups, and enzymatic assays, proved instead to be a definitely more efficient antitumor agent, both in vitro and in vivo. This result could tentatively be explained in view of the importance of positive charges for ribonuclease activity, considering the higher basicity of PS-dimeric RNase A compared to that of PS-(monomeric)RNase A. It must be also taken into account that the dimeric RNase A moiety of PS-dimeric RNase A could evade the cytoplasmic ribonuclease inhibitor, which instead could trap the monomeric RNase A moiety of the other derivative. The two RNase A derivatives degrade poly(A).poly(U) under conditions where native RNase A is inactive. The results of this work demonstrate once again the importance of positive charges for the functions of mammalian pancreatic type ribonucleases in general, in particular for RNase A derivatives, and the potential therapeutic use of the ribonuclease A derivatives.


Asunto(s)
Ribonucleasa Pancreática/química , Ribonucleasa Pancreática/toxicidad , Espermina/química , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cromatografía en Gel , Reactivos de Enlaces Cruzados/química , Dimerización , Humanos , Estructura Molecular , ARN/metabolismo , Ribonucleasa Pancreática/aislamiento & purificación , Ribonucleasa Pancreática/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
15.
Biochim Biophys Acta ; 1773(7): 1095-106, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17555830

RESUMEN

We investigated the ability of the histone deacetylase (HDAC) inhibitor trichostatin A (TSA) to interact with gemcitabine (GEM) in inducing pancreatic cancer cell death. The combined treatment with TSA and GEM synergistically inhibited growth of four pancreatic adenocarcinoma cell lines and induced apoptosis. This effect was associated with the induction of reactive oxygen species (ROS) by GEM, increased expression of the pro-apoptotic BIM gene by both TSA and GEM and downregulation of the 5'-nucleotidase UMPH type II gene by TSA. The expression of other genes critical for GEM resistance (nucleoside transporters, deoxycytidine kinase, cytidine deaminase, and ribonucleotide reductase genes) was not affected by TSA. The functional role of ROS in cell growth inhibition by GEM was supported by (i) a significantly reduced GEM-associated growth inhibition by the free radical scavenger N-acetyl-L-cysteine, and (ii) a positive correlation between the basal level of ROS and sensitivity to GEM in 10 pancreatic cancer cell lines. The functional role of both Bim and 5'-nucleotidase UMPH type II in cell growth inhibition by TSA and GEM was assessed by RNA interference assays. In vivo studies on xenografts of pancreatic adenocarcinoma cells in nude mice showed that the association of TSA and GEM reduced to 50% the tumour mass and did not cause any apparent form of toxicity, while treatments with TSA or GEM alone were ineffective. In conclusion, the present study demonstrates a potent anti-tumour activity of TSA/GEM combination against pancreatic cancer cells in vitro and in vivo, strongly supporting the use of GEM in combination with an HDAC inhibitor for pancreatic cancer therapy.


Asunto(s)
Adenocarcinoma/metabolismo , Proliferación Celular/efectos de los fármacos , Desoxicitidina/análogos & derivados , Inhibidores Enzimáticos , Ácidos Hidroxámicos , Neoplasias Pancreáticas/metabolismo , 5'-Nucleotidasa/genética , 5'-Nucleotidasa/metabolismo , Adenocarcinoma/patología , Animales , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteína 11 Similar a Bcl2 , Ciclo Celular/fisiología , Línea Celular Tumoral , Desoxicitidina/metabolismo , Desoxicitidina/farmacología , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Glicoproteínas/genética , Glicoproteínas/metabolismo , Inhibidores de Histona Desacetilasas , Humanos , Ácidos Hidroxámicos/metabolismo , Ácidos Hidroxámicos/farmacología , Proteínas de la Membrana/metabolismo , Ratones , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Gemcitabina
16.
Biochim Biophys Acta ; 1763(9): 917-26, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16904205

RESUMEN

We present evidence that pyrrolidine dithiocarbamate (PDTC) inhibits growth of p53-negative pancreatic adenocarcinoma cell lines via cell cycle arrest in the S-phase, while it has no effect on primary fibroblast proliferation. Growth inhibition of cancer cells is dependent on ROS and ERK1/2 induction as indicated by a significantly reduced PDTC-associated growth inhibition by the free radical scavenger N-acetyl-L-cysteine (NAC) or the MEK/ERK1/2 inhibitor (PD98059). Moreover, ERK1/2 induction is dependent on ROS production as demonstrated by a complete removal of PDTC-mediated ERK1/2 phosphorylation by NAC. p21(WAF1/CIP1) activation has a central role in growth inhibition by PDTC, as revealed by P21(WAF1/CIP1) silencing experiments with antisense oligonucleotide, and occurs via increased mRNA stability largely mediated by ROS/ERK induction. Conversely, PDTC does not affect P21(WAF1/CIP1) gene expression in primary fibroblasts, although it is able to activate p53 and the p53-regulated antioxidant SESN2. These results suggest that the resistance of fibroblasts to the cytotoxic action of PDTC may be related to the up-regulation of p53-dependent antioxidant genes. Finally, in vivo studies on PaCa44 cells subcutaneously xenografted in nude mice show that treatment with 100 or 200 mg/kg PDTC reduces of 30% or 60% the tumour volume, respectively, and does not cause any apparent form of toxicity.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Pirrolidinas/farmacología , Estabilidad del ARN , Tiocarbamatos/farmacología , Acetilcisteína/farmacología , Animales , Línea Celular Tumoral/citología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Cartilla de ADN , Fibroblastos , Flavonoides/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Humanos , Immunoblotting , Ratones , Oligonucleótidos Antisentido , Pirrolidinas/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tiocarbamatos/uso terapéutico
17.
Virchows Arch ; 448(6): 797-804, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16568310

RESUMEN

Pancreatic cancer is an aggressive neoplasia, and standard chemotherapies are by and large ineffective. The purpose of this work was to get a comprehensive preclinical study on the ability of anticancer drug combinations that best inhibit growth of pancreatic adenocarcinoma cells. We evaluated the in vitro growth inhibition of ten pancreatic cancer cell lines to gemcitabine and 5-fluorouracil, newer generation cytotoxic agents (oxaliplatin, irinotecan), targeted therapy (gefitinib) and a histone deacetylase (HDAC) inhibitor (trichostatin A). Cells were treated with the single drug alone and all pairwise drug association. Our results demonstrate that TSA can effectively increase the drug sensitivity of all the cell lines studied. The association of TSA and irinotecan determines an increase in growth inhibition on the highest percentage of cell lines (80%). Our findings may represent an experimental basis for potential clinical application of HDAC inhibitors, in particular in association with drugs used in cancer clinical treatment, supporting the idea that HDAC inhibitors could act as sensitizers for chemotherapy.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de la Síntesis de la Proteína/farmacología , Adenocarcinoma/patología , Antineoplásicos Fitogénicos/farmacología , Camptotecina/análogos & derivados , Camptotecina/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Concentración 50 Inhibidora , Irinotecán , Neoplasias Pancreáticas/patología
18.
Biochim Biophys Acta ; 1693(3): 167-76, 2004 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-15363630

RESUMEN

The histone deacetylase inhibitor trichostatin A (TSA) has been previously shown to block cellular growth in G2 and induce apoptosis in human pancreatic cancer cell lines. In order to better understand this phenomenon, we have analyzed the gene expression profiles in PaCa44 cells after treatment with TSA using microarrays containing 22,283 probesets. TSA was found to cause both the induction and repression of a large number of genes, although the number whose expression was up-regulated was greater than the number of genes that were down-regulated. When a threshold value of 3 was used as a cutoff level, a total of 306 (3.4%) of the detectable genes had altered expression. When categorized according to cellular function, the differentially expressed genes were found to be involved in a wide variety of cellular processes, including cell proliferation, signaling, regulation of transcription, and apoptosis. Moreover, Sp1/Sp3 transcription factor binding sites were significantly more abundant among TSA-induced genes. One prominent feature was the increased ratio between the levels of expression of pro-apoptotic (BIM) and anti-apoptotic (Bcl-XL and Bcl-W) genes. This result was confirmed in eight additional pancreatic cancer cell lines after treatment with TSA, suggesting that this event may be a strong determinant for the induction of apoptosis by TSA.


Asunto(s)
Adenocarcinoma/genética , Apoptosis/genética , Perfilación de la Expresión Génica , Histona Desacetilasas/fisiología , Ácidos Hidroxámicos/farmacología , Neoplasias Pancreáticas/genética , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
19.
Biochim Biophys Acta ; 1692(1): 17-24, 2004 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-15158360

RESUMEN

The human IL-6 promoter contains multiple regulatory elements such as those binding transcription factors belonging to the NF-kappaB (-75/-63), C/EBP (-158/-145 and -87/-76) and AP-1 (-283/-277) families. Herein, we report that ectopic expression of c-Jun, C/EBPdelta, and the p65 subunit of NF-kappaB synergistically activates an IL-6 promoter construct containing only a TATA box and a kappaB binding site. These results suggest that interactions among NF-kappaB, C/EBP, and AP-1, which are all activated by the most powerful physiological inducers of the IL-6 gene, namely TNF-alpha and IL-1, may be crucial for maximal activation of the IL-6 promoter in response to the two cytokines. Furthermore, we show that a mutated form of c-Jun lacking the transactivation domain (TAM-67) was a much stronger activator of the IL-6 promoter than c-Jun. In combination with p65 and/or C/EBPdelta, TAM-67 also synergistically activated the IL-6 promoter, while it inhibited TNF-alpha induced AP-1 activity directing an AP-1-responsive reporter plasmid. Lastly, electrophoretic mobility shift assay (EMSA) results strongly suggest the formation of complexes between p65, C/EBPdelta, and/or c-Jun or TAM-67 on the kappaB site, supporting the idea that the functional synergism is determined by a physical interaction. These data provide new insight into the molecular mechanisms regulating the formation of the transcription complex responsible for IL-6 promoter activation.


Asunto(s)
Proteínas de Unión al Calcio , Interleucina-6/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-jun/genética , Factores de Transcripción , Sitios de Unión , Proteína delta de Unión al Potenciador CCAAT , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Genes Reporteros , Células HeLa , Humanos , Glicoproteínas de Membrana/metabolismo , Mutación , FN-kappa B/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Fragmentos de Péptidos/metabolismo , Plásmidos/metabolismo , Unión Proteica , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-jun/metabolismo , Sinaptotagmina I , Sinaptotagminas , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción ReIA , Transcripción Genética , Activación Transcripcional , Transfección
20.
Mol Carcinog ; 38(2): 59-69, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14502645

RESUMEN

In cells with an altered p53 gene, the expression of p21(WAF1/CIP1), a potent inhibitor of cyclin-dependent kinases, can be induced by histone deacetylase (HDAC) inhibitors via a p53-independent pathway, which may play a critical role in arrest of cell growth. Accordingly, HDAC inhibitors such as trichostatin A (TSA) have potential utility in pancreatic cancer, as most of these tumors possess mutations in p53, which in fact is the main cause of chemoresistance to 5-fluorouracil. We have analyzed the effect of TSA on the proliferation of nine pancreatic adenocarcinoma cell lines, all containing a mutated p53 gene. TSA strongly inhibited the cellular growth of all these cell lines at submicromolar concentrations. The cellular mechanisms underlying this effect consisted of cell cycle arrest at the G2 phase and apoptotic cell death. The expression of p21(WAF1/CIP1) normally induced at the transcriptional level by p53 was also strongly activated by TSA. These findings suggest that inhibitors of HDAC may represent a novel therapeutic strategy for treatment of pancreatic cancer.


Asunto(s)
Adenocarcinoma/patología , Inhibidores Enzimáticos/farmacología , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/farmacología , Neoplasias Pancreáticas/patología , Adenocarcinoma/prevención & control , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/metabolismo , Humanos , Neoplasias Pancreáticas/prevención & control , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...