Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Biochim Biophys Acta ; 1823(4): 789-99, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22289349

RESUMO

The tumor suppressor Pdcd4 is involved in multiple pathways. Considering its biological action conflicting data in the literature exist and, consequently, our own studies point to a cell type specific action of Pdcd4. In the present study, using several Pdcd4 knock down cell lines we succeeded to identify angiopoietin-2 (Ang-2) as a gene up-regulated on the mRNA and protein level. The subsequent enhanced peptide secretion forced wild type Bon-1 cells in a neoplastic direction demonstrated by increased proliferation and colony formation while cell adhesion was decreased. Most likely, the stimulation of Ang-2 is in part mediated by increased activation of AP-1 but different signal transduction pathways may also be involved since we found opposite activation of PI3K/Akt/mTOR and MAPK7ERK pathways (both known to regulate in Ang-2 expression). Ang-2 is a modulator of vascular remodeling. Therefore, we analyzed the effect of supernatants from Pdcd4 knock-down cell lines on endothelial cells. Again, we detected reduced cell adhesion and increased colony formation. Probably, the most impressive effect was described on tube formation in a model for angiogenesis. Tube length and junctions of endothelial cells treated with conditioned medium from Pdcd4 knock-down cells were considerably increased. Both, up-regulation of Ang-2 and down-regulation of Pdcd4 are described for many tumors. However, this is the first study showing a direct impact of Pdcd4 on Ang-2 levels and, thereby, angiogenesis. Our data suggest a completely new mechanism for Pdcd4 to act as a tumor suppressor rendering Pdcd4 an attractive target for new therapeutic strategies in cancer treatment.


Assuntos
Angiopoietina-2/genética , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/metabolismo , Técnicas de Silenciamento de Genes , Neovascularização Fisiológica/genética , Proteínas de Ligação a RNA/metabolismo , Regulação para Cima/genética , Angiopoietina-2/metabolismo , Proteínas Reguladoras de Apoptose/genética , Adesão Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Meios de Cultivo Condicionados/farmacologia , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , Neovascularização Fisiológica/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Transfecção , Regulação para Cima/efeitos dos fármacos
2.
Biochem Biophys Res Commun ; 417(1): 29-34, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22133680

RESUMO

BACKGROUND: We investigated the role of the programmed cell death 4 (PDCD4) tumor suppressor gene in specimens of transitional cell carcinoma and of healthy individuals. METHODS: PDCD4 immunohistochemical expression was investigated in 294 cases in histologically proven transitional cell carcinoma in different tumorous stages (28 controls, 122 non-muscle invasive urothelial carcinoma, stages Tis-T1, 119 invasive transitional cell carcinoma stages T2-T4 and 25 metastases). MiR-21 expression, an important PDCD4 regulator, was assessed with real-time PCR analysis and showed inverse correlation to tissue PDCD4 expression. RESULTS: Nuclear and cytoplasmatic PDCD4 immunostaining decreased significantly with histopathological progression of the tumor (p<0001). Controls showed strong nuclear and cytoplasmatic immunohistochemical staining. MiR-21 up regulation in tissue corresponded to PDCD4 suppression. CONCLUSIONS: These data support a decisive role for PDCD4 down regulation in transitional cell carcinoma and confirm miR-21 as a negative regulator for PDCD4. Additionally, PDCD4 immunohistochemical staining turns out to be a possible diagnostic marker for transitional cell carcinoma.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Carcinoma de Células de Transição/metabolismo , MicroRNAs/biossíntese , Proteínas de Ligação a RNA/biossíntese , Neoplasias Urológicas/metabolismo , Urotélio/metabolismo , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células de Transição/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Neoplasias Urológicas/patologia , Urotélio/patologia
3.
Biol Cell ; 101(6): 309-17, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19356152

RESUMO

Pdcd4 (programmed cell death 4) has been known as a tumour suppressor gene and potential target for anticancer therapies for several years. Initially, Pdcd4 was identified as a gene that is up-regulated during apoptosis, but its precise role still remains to be defined. However, there is increasing evidence that Pdcd4 levels influence transcription, as well as translation, modulate different signal transduction pathways and might act as a tumour suppressor. Interestingly, recent data suggest that Pdcd4 function may depend on cell type and/or genetic background. This review summarizes the current knowledge regarding the function and regulation of Pdcd4.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Genes Supressores de Tumor , Neoplasias/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Humanos , Neoplasias/tratamento farmacológico , Proteínas de Ligação a RNA/genética , Transdução de Sinais
4.
Sci Rep ; 10(1): 18924, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33144657

RESUMO

Inhibition of the kinase ATR, a central regulator of the DNA damage response, eliminates subsets of cancer cells in certain tumors. As previously shown, this is at least partly attributable to synthetic lethal interactions between ATR and POLD1, the catalytic subunit of the polymerase δ. Various POLD1 variants have been found in colorectal cancer, but their significance as therapeutic targets for ATR pathway inhibition remains unknown. Using CRISPR/Cas9 in the colorectal cancer cell line DLD-1, which harbors four POLD1 variants, we established heterozygous POLD1-knockout clones with exclusive expression of distinct variants to determine the functional relevance of these variants individually by assessing their impact on ATR pathway activation, DNA replication, and cellular sensitivity to inhibition of ATR or its effector kinase CHK1. Of the four variants analyzed, only POLD1R689W affected POLD1 function, as demonstrated by compensatory ATR pathway activation and impaired DNA replication. Upon treatment with ATR or CHK1 inhibitors, POLD1R689W strongly decreased cell survival in vitro, which was attributable at least partly to S phase impairment and apoptosis. Similarly, treatment with the ATR inhibitor AZD6738 inhibited growth of murine xenograft tumors, harboring the POLD1R689W variant, in vivo. Our POLD1-knockout model thus complements algorithm-based models to predict the pathogenicity of tumor-specific variants of unknown significance and illustrates a novel and potentially clinically relevant therapeutic approach using ATR/CHK1 inhibitors in POLD1-deficient tumors.


Assuntos
Substituição de Aminoácidos , Neoplasias Colorretais/tratamento farmacológico , DNA Polimerase III/genética , Pirimidinas/administração & dosagem , Sulfóxidos/administração & dosagem , Animais , Sistemas CRISPR-Cas , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Quinase 1 do Ponto de Checagem/antagonistas & inibidores , Neoplasias Colorretais/genética , Replicação do DNA/efeitos dos fármacos , Técnicas de Inativação de Genes , Humanos , Indóis , Camundongos , Morfolinas , Pirimidinas/farmacologia , Sulfonamidas , Sulfóxidos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Biol Cell ; 100(12): 703-15, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18549351

RESUMO

BACKGROUND INFORMATION: Pdcd4 (programmed cell death 4) is up-regulated during apoptosis and seems to play an important role as a tumour suppressor. To gain further insights into its biological functions, we suppressed Pdcd4 expression in the neuroendocrine cell line Bon-1 via siRNA (small interfering RNA) technology. RESULTS: Using this cell line, we found that suppression of Pdcd4 resulted in an increased release of CgA (chromogranin A) and Sg II (secretogranin II), and was accompanied by an up-regulation of intracellular PC1 (proprotein convertase 1/3). The enhanced secretion of CgA and Sg II seemed to be mediated by an activation of protein kinase Akt via PI3K (phosphoinositide 3-kinase). In accordance with this, inhibition of PI3K activity and, thereby, reduced phosphorylation of Akt was shown to enhance Pdcd4 expression. Neither the PKC (protein kinase C) signal transduction cascade nor the MAPK (mitogen-activated protein kinase) pathway seemed to play a role in the regulation of CgA and Sg II secretion by Pdcd4. CONCLUSIONS: CgA is considered to be a marker for neuroendocrine tumours, and up-regulation of PC1 has been reported in various types of cancers. The repression of PC1 by Pdcd4 may represent a novel mechanism for the function of Pdcd4 as a tumour suppressor. Our results are of particular interest, as we observed that pioglitazone, an oral medication used in the treatment of Type 2 diabetes, decreased Pdcd4 levels, activated Akt, increased CgA and Sg II secretion and augmented PC1 protein in Bon-1 cells. Enhanced PC1 levels, leading to improved processing of proinsulin and proglucagon, may contribute to the benefits of pioglitazone therapy. The in vivo relevance of our findings was highlighted by data indicating elevated CgA amounts in the sera of patients treated with pioglitazone. This is the first study connecting Pdcd4 levels, secretion behaviour of neuroendocrine cells and regulation of PI3K activity.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Cromogranina A/metabolismo , Células Neuroendócrinas/metabolismo , Pró-Proteína Convertase 1/genética , Proteínas de Ligação a RNA/genética , Secretogranina II/metabolismo , Regulação para Cima , Proteínas Reguladoras de Apoptose/metabolismo , Linhagem Celular Tumoral , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Ativação Enzimática , Técnicas de Silenciamento de Genes , Humanos , Masculino , Células Neuroendócrinas/efeitos dos fármacos , Pioglitazona , Pró-Proteína Convertase 1/metabolismo , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Tiazolidinedionas/farmacologia , Tiazolidinedionas/uso terapêutico
6.
Neoplasia ; 20(11): 1135-1143, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30257222

RESUMO

The phosphoinositide 3-kinase-related kinase ATR is a central regulator of the DNA damage response. Its chemical inhibition eliminates subsets of cancer cells in various tumor types. This effect is caused at least partly by the synthetically lethal relationship between ATR and certain DNA repair genes. In a previous screen using an siRNA library against DNA repair genes, we identified PRIM1, a part of the polymerase α-primase complex, as acting synthetically lethal with ATR. Applying a genetic ATR knock-in model of colorectal cancer cells, we confirmed that PRIM1 depletion inhibited proliferation of ATR-deficient cells and excluded artifacts due to clonal variation using an ATR reexpressing cell clone. We expanded these data by demonstrating in different cell lines that also chemical inhibition of ATR or its main effector kinase CHK1 reduces proliferation upon depletion of PRIM1. Mechanistically, PRIM1 depletion in ATR-deficient cells caused S-phase stasis in the absence of increased DNA damage followed by Wee1-mediated activation of caspase 8 and apoptosis. As PRIM1 inactivation sensitizes cancer cells to ATR and CHK1 inhibitors, mutations in PRIM1 or other components of the polymerase α-primase complex could represent novel targets for individualized tumor therapeutic approaches using ATR/CHK1 inhibitors, as has been previously demonstrated for POLD1, the catalytic subunit of polymerase δ.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/antagonistas & inibidores , Quinase 1 do Ponto de Checagem/antagonistas & inibidores , DNA Primase/antagonistas & inibidores , Resistencia a Medicamentos Antineoplásicos , Inibidores de Proteínas Quinases/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Expressão Gênica , Histonas/metabolismo , Humanos , Interferência de RNA , RNA Interferente Pequeno/genética , Mutações Sintéticas Letais
7.
Cancer Res ; 76(1): 96-107, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26669866

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) carries the most dismal prognosis of all solid tumors and is generally strongly resistant to currently available chemo- and/or radiotherapy regimens, including targeted molecular therapies. Therefore, unraveling the molecular mechanisms underlying the aggressive behavior of pancreatic cancer is a necessary prerequisite for the development of novel therapeutic approaches. We previously identified the protein placenta-specific 8 (PLAC8, onzin) in a genome-wide search for target genes associated with pancreatic tumor progression and demonstrated that PLAC8 is strongly ectopically expressed in advanced preneoplastic lesions and invasive human PDAC. However, the molecular function of PLAC8 remained unclear, and accumulating evidence suggested its role is highly dependent on cellular and physiologic context. Here, we demonstrate that in contrast to other cellular systems, PLAC8 protein localizes to the inner face of the plasma membrane in pancreatic cancer cells, where it interacts with specific membranous structures in a temporally and spatially stable manner. Inhibition of PLAC8 expression strongly inhibited pancreatic cancer cell growth by attenuating cell-cycle progression, which was associated with transcriptional and/or posttranslational modification of the central cell-cycle regulators CDKN1A, retinoblastoma protein, and cyclin D1 (CCND1), but did not impact autophagy. Moreover, Plac8 deficiency significantly inhibited tumor formation in genetically engineered mouse models of pancreatic cancer. Together, our findings establish PLAC8 as a central mediator of tumor progression in PDAC and as a promising candidate gene for diagnostic and therapeutic targeting.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas/metabolismo , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Progressão da Doença , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Prognóstico , Proteínas/genética , Análise Serial de Tecidos , Transfecção
8.
Anticancer Res ; 35(11): 5873-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26504010

RESUMO

BACKGROUND: Fibroblast growth factor receptors are expressed in diverse cell types. They play a critical role in tumor development. Their activation promotes cell-cycle progression, angiogenesis, and cell survival by induction/suppression of the expression of proteins involved. MATERIALS AND METHODS: Non-small cell lung cancer (NSCLC) cells (line H1581) were treated with NVP-BGJ398 to evaluate effects on growth by western blot, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide assay and cell-cycle analysis. RESULTS: NVP-BGJ398 induced cell death in H1581 cells by activating caspase-dependent mitochondrial and non-mitochondrial pathways. Caspase-independent apoptosis was also activated. Cells were found to be arrested in the G0/G1 phase. Furthermore, the expression of the tumor-suppressor gene programmed cell death 4 (PDCD4) was up-regulated with suppression of angiopoietin 2 (ANG2). This represents an additional mechanism by which NVP-BGJ389 inhibits tumor growth. CONCLUSION: Various pathways induce apoptosis in NSCLC cells by employing NVP-BGJ398. These data reflect the potential of cancer treatment utilizing small FGFR inhibitors.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/patologia , Caspases/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Neoplasias Pulmonares/patologia , Compostos de Fenilureia/farmacologia , Pirimidinas/farmacologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Antineoplásicos/farmacologia , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Células Tumorais Cultivadas
9.
Clin Cancer Res ; 21(19): 4356-64, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26015511

RESUMO

PURPOSE: FGFR1 copy-number gain (CNG) occurs in head and neck squamous cell cancers (HNSCC) and is used for patient selection in FGFR-specific inhibitor clinical trials. This study explores FGFR1 mRNA and protein levels in HNSCC cell lines, primary tumors, and patient-derived xenografts (PDX) as predictors of sensitivity to the FGFR inhibitor, NVP-BGJ398. EXPERIMENTAL DESIGN: FGFR1 status, expression levels, and BGJ398 sensitive growth were measured in 12 HNSCC cell lines. Primary HNSCCs (n = 353) were assessed for FGFR1 CNG and mRNA levels, and HNSCC TCGA data were interrogated as an independent sample set. HNSCC PDXs (n = 39) were submitted to FGFR1 copy-number detection and mRNA assays to identify putative FGFR1-dependent tumors. RESULTS: Cell line sensitivity to BGJ398 is associated with FGFR1 mRNA and protein levels, not FGFR1 CNG. Thirty-one percent of primary HNSCC tumors expressed FGFR1 mRNA, 18% exhibited FGFR1 CNG, 35% of amplified tumors were also positive for FGFR1 mRNA. This relationship was confirmed with the TCGA dataset. Using high FGFR1 mRNA for selection, 2 HNSCC PDXs were identified, one of which also exhibited FGFR1 CNG. The nonamplified tumor with high mRNA levels exhibited in vivo sensitivity to BGJ398. CONCLUSIONS: FGFR1 expression associates with BGJ398 sensitivity in HNSCC cell lines and predicts tyrosine kinase inhibitor sensitivity in PDXs. Our results support FGFR1 mRNA or protein expression, rather than FGFR1 CNG as a predictive biomarker for the response to FGFR inhibitors in a subset of patients suffering from HNSCC.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/genética , Resistencia a Medicamentos Antineoplásicos/genética , Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Compostos de Fenilureia/farmacologia , Pirimidinas/farmacologia , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Feminino , Dosagem de Genes , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Hibridização in Situ Fluorescente , Masculino , Compostos de Fenilureia/uso terapêutico , Prognóstico , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/uso terapêutico , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço
10.
FEBS Lett ; 533(1-3): 42-6, 2003 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-12505156

RESUMO

Within the adaptive immune system the transporter associated with antigen processing (TAP) plays a pivotal role in loading of peptides onto major histocompatibility (MHC) class I molecules. As a central tool to investigate the structure and function of the TAP complex, we created cysteine-less human TAP subunits by de novo gene synthesis, replacing all 19 cysteines in TAP1 and TAP2. After expression in TAP-deficient human fibroblasts, cysteine-less TAP1 and TAP2 are functional with respect to adenosine triphosphate (ATP)-dependent peptide transport and inhibition by ICP47 from herpes simplex virus. Cysteine-less TAP1 and TAP2 restore maturation and intracellular trafficking of MHC class I molecules to the cell surface.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Transportadores de Cassetes de Ligação de ATP/genética , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Membro 3 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/metabolismo , Substituição de Aminoácidos , Apresentação de Antígeno , Transporte Biológico Ativo , Linhagem Celular , Cisteína/química , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Modelos Moleculares , Subunidades Proteicas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfecção
11.
Ann N Y Acad Sci ; 1014: 220-1, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15153438

RESUMO

PDCD4 is a new tumor suppressor gene. In the current study, we show that overexpression of PDCD4 in carcinoid cells results in inhibition of cell proliferation. This is most likely caused by a PDCD4-induced downregulation of carbonic anhydrase type II which catalyzes the production of bicarbonate, a fundamental substrate for many cellular pathways.


Assuntos
Apoptose/fisiologia , Tumor Carcinoide , Genes Supressores de Tumor/fisiologia , Neoplasias Pancreáticas , Proteínas de Ligação a RNA/fisiologia , Proteínas Reguladoras de Apoptose , Linhagem Celular Tumoral , Humanos
12.
EJNMMI Res ; 4: 31, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25006548

RESUMO

BACKGROUND: Various diseases derive from pathologically altered ß-cells. Their function can be increased, leading to hyperinsulinism, or decreased, resulting in diabetes. Non-invasive imaging of the ß-cell-specific glucagon-like peptide receptor-1 (GLP-1R) would allow the assessment of both ß-cell mass and derived tumours, potentially improving the diagnosis of various conditions. We tested three new (67/68)Ga-labelled derivatives of exendin-4, an agonist of GLP-1R, in vitro and in vivo. We determined the influence of the chelator NODAGA conjugated to resident lysines either at positions 12 and 27 or the C-terminally attached lysine at position 40 on the binding and kinetics of the peptide. METHODS: Binding and internalisation of (67)Ga-labelled Ex4NOD12, Ex4NOD27 and Ex4NOD40 were tested on Chinese hamster lung (CHL) cells stably transfected to express the GLP-1 receptor (GLP-1R). In vivo biodistribution of (68)Ga-labelled peptides was investigated in CD1 nu/nu mice with subcutaneous CHL-GLP-1R positive tumours; the specificity of the binding to GLP-1R was determined by pre-injecting excess peptide. RESULTS: All peptides showed good in vitro binding affinities to GLP-1R in the range of 29 to 54 nM. (67/68)Ga-Ex4NOD40 and (67/68)Ga-Ex4NOD12 show excellent internalisation (>30%) and high specific uptake in GLP-1R positive tissue, but high activity was also found in the kidneys. CONCLUSIONS: We show that of the three peptides, Ga-Ex4NOD40 and Ga-Ex4NOD12 demonstrate the most favourable in vitro properties and in vivo binding to GLP-1R positive tissue. Therefore, we conclude that the lysines at positions 12 and 40 might preferentially be utilised for modifying exendin-4.

13.
Apoptosis ; 13(1): 157-64, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17955371

RESUMO

Pdcd4 (programmed cell death protein 4) was identified as a gene up-regulated during apoptosis and, additionally, seems to have a function as a tumor suppressor. However, there are conflicting data concerning its role in programmed cell death and most results for its action as an inhibitor for neoplastic transformation are derived from experiments with epidermal cells. Therefore, we were interested to investigate if the action of Pdcd4 might be cell type specific. For that purpose we examined the expression of Pdcd4 and several other proteins in various tumor cell lines. We could not find any correlation of Pdcd4 levels and expression of proteins associated with cell cycle and/or apoptosis in different cell lines. Furthermore, we stably transfected two cell lines (Bon-1 and HCT116) to over-express Pdcd4 and analyzed protein expression. Although we found several regulated proteins none of these proteins were affected in both cell lines in the same manner. For instance, dUTPase expression was reduced in Bon-1 cells but not changed in HCT116 cells. This regulation might be important for the sensitivity of cells to anti-cancer drugs like inhibitors of thymidilate synthase. Therefore, we conclude that the function of Pdcd4 might be cell type specific. A role for Pdcd4 in apoptosis or as a tumor suppressor might be limited to certain cell types.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/fisiologia , Pirofosfatases/metabolismo , Proteínas de Ligação a RNA/metabolismo , Antimetabólitos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Fluoruracila/farmacologia , Humanos , Transfecção
14.
Digestion ; 71(3): 131-40, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15785039

RESUMO

AIM: We investigated the effects of the tyrosine kinase inhibitor imatinib (Gleevec) on neuroendocrine tumor cells. METHODS: Neuroendocrine tumor cells were incubated without and with imatinib. The effects on growth were examined by methylthiazoletetrazolium (MTT) assay. The c-Kit expression in human endocrine tumor tissue and cell lines was determined by immunohistochemistry and Western blot analysis, respectively. Cytotoxicity assay was performed by fluorescence-activated cell sorting. The telomerase activity was determined using the telomeric repeat amplification protocol. RESULTS: 28% of the insulinomas, 100% of the gastrinomas, and 38% of the carcinoids expressed c-Kit. Imatinib at concentrations >5 microM inhibited cell proliferation and induced apoptosis in both c-Kit-positive and c-Kit-negative cell lines. The PI-3K inhibitor wortmannin did not enhance the effects of imatinib. Imatinib did not sensitize endocrine tumor cells to doxorubicin and 5-fluorouracil. Imatinib inhibited the telomerase activity. CONCLUSION: Imatinib inhibits neuroendocrine tumor cell growth independently of c-Kit by inhibition of other tyrosine kinases or through tyrosine kinase independent pathways.


Assuntos
Proliferação de Células/efeitos dos fármacos , Tumores Neuroendócrinos/tratamento farmacológico , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirimidinas/farmacologia , Apoptose/efeitos dos fármacos , Benzamidas , Western Blotting , Citometria de Fluxo , Humanos , Mesilato de Imatinib , Imuno-Histoquímica , Técnicas In Vitro , Tumores Neuroendócrinos/enzimologia , Tumores Neuroendócrinos/patologia , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-kit/biossíntese , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/patologia
15.
Biol Cell ; 95(8): 515-9, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14630388

RESUMO

Pdcd4 is a novel gene first identified as a differentially expressed protein during apoptosis. In the meantime not only the impact of Pdcd4 in programmed cell death but also an implication in transformation suppression by inhibition of protein translation is discussed. These features implicate a potential value of Pdcd4 as a molecular target in cancer therapy. This review summarizes the current knowledge about expression, structure and function of Pdcd4.


Assuntos
Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/fisiologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/fisiologia , Animais , Apoptose , Proteínas Reguladoras de Apoptose , Fator de Iniciação Eucariótico 4G/metabolismo , Camundongos , Neoplasias/terapia , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/química , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/química , Regulação para Cima
16.
Physiol Rev ; 82(1): 187-204, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11773612

RESUMO

The adaptive immune systems have evolved to protect the organism against pathogens encountering the host. Extracellular occurring viruses or bacteria are mainly bound by antibodies from the humoral branch of the immune response, whereas infected or malignant cells are identified and eliminated by the cellular immune system. To enable the recognition, proteins are cleaved into peptides in the cytosol and are presented on the cell surface by class I molecules of the major histocompatibility complex (MHC). The transport of the antigenic peptides into the lumen of the endoplasmic reticulum (ER) and loading onto the MHC class I molecules is an essential process for the presentation to cytotoxic T lymphocytes. The delivery of these peptides is performed by the transporter associated with antigen processing (TAP). TAP is a heterodimer of TAP1 and TAP2, each subunit containing transmembrane domains and an ATP-binding motif. Sequence homology analysis revealed that TAP belongs to the superfamily of ATP-binding cassette transporters. Loss of TAP function leads to a loss of cell surface expression of MHC class I molecules. This may be a strategy for tumors and virus-infected cells to escape immune surveillance. Structure and function of the TAP complex as well as the implications of loss or downregulation of TAP is the topic of this review.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Membro 2 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Membro 3 da Subfamília B de Transportadores de Cassetes de Ligação de ATP , Animais , Doenças Autoimunes/fisiopatologia , Regulação para Baixo , Humanos , Imunidade/fisiologia , Neoplasias/etiologia , Viroses/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA