Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Cancer ; 18(1): 185, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29439667

RESUMO

BACKGROUND: Several efforts have been focused on identification of pathways involved in malignancy, progression, and response to treatment in Glioblastoma (GB). Overexpression of PKCε was detected in histological samples from GB, anaplastic astrocytoma, and gliosarcoma and is considered an important marker of negative disease outcome. In multiple studies on GB, autophagy has been shown as a survival mechanism during cellular stress, contributing to resistance against anti-cancer agents. The main object of this research was to determine the influence of PKCε downregulation on the expression of genes involved in autophagy pathways in glioblastoma cell lines U-138 MG and U-118 MG with high PKCε level. METHODS: We conducted siRNA-mediated knockdown of PKCε in glioblastoma cell lines and studied the effects of autophagy pathway. The expression of autophagy-related genes was analyzed using qPCR and Western blot analysis was carried out to assess protein levels. Immunostaining was used to detect functional autophagic maturation process. RESULTS: We found that these cell lines exhibited a high basal expression of autophagy-related genes. Our results suggest that the loss of PKCε contributes to the downregulation of genes involved in autophagy pathways. Moreover, most of the changes we observed in Western blot analysis and endogenous immunofluorescence experiments confirmed dysfunction of autophagy programs. We found that knockdown of PKCε induced a decrease in the expression of Beclin1, Atg5, PI3K, whereas the expression of other autophagy-related proteins mTOR and Bcl2 was increased. Treatment of control siRNA glioma cells with rapamycin-induced autophagosome formation and increase in LC3-II level and caused a decrease in the expression of p62. Additionally, PKCε siRNA caused a diminution in the Akt phosphorylation at Ser473 and in the protein level in both cell lines. Moreover, we observed reduction in the adhesion of glioblastoma cells, accompanied by the decrease in total FAK protein level and phosphorylation. CONCLUSIONS: Effects of down-regulation of PKCε in glioma cells raised the possibility that the expression of PKCε is essential for the autophagic signal transduction pathways in these cells. Thus, our results identify an important role of PKCε in autophagy and may, more importantly, identifyit as a novel therapeutic target.


Assuntos
Autofagia/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Proteína Quinase C-épsilon/genética , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Linhagem Celular Tumoral , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteína Quinase C-épsilon/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Interferência de RNA , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
2.
Nutr Cancer ; 68(2): 290-304, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26847268

RESUMO

Autophagy is important in the regulation of survival and death signaling pathways in cancer. PKCϵ revealed high transforming potential and the ability to increase cell migration, invasion, and metastasis. Zapotin (5,6,2',6'-tetramethoxyflavone), a natural flavonoid, showed chemopreventive and anticancer properties. Previously, we reported that downmodulation of induced PKCϵ level by zapotin was associated with decreased migration and increased apoptosis in HeLa cell line containing doxycycline-inducible constitutively active PKCϵ (PKCϵA/E, Ala(159) → Glu). Depending on the genetic and environmental content of cells, autophagy may either precede apoptosis or occur simultaneously. The purpose of this study was to assess the effect of zapotin on autophagy. Increasing concentration of zapotin (from 7.5 µM to 30 µM) caused an inhibition of the formation of autophagosomes and a decline in microtubule-associated protein 1 light chain 3 (LC3) protein levels. The gene expression level of major negative regulator of autophagy was noticeably increased. Moreover, the expression of the pivotal autophagy genes was decreased. These changes were accompanied by alternation in autophagy-related protein levels. In conclusion, our results implied that both the antiautophagic and the proapoptosis effect of zapotin in HeLaPKCϵA/E cells are associated with the protein kinase C epsilon signaling pathway and lead to programmed cell death.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Flavonas/farmacologia , Proteína Quinase C-épsilon/metabolismo , Autofagia/genética , Sobrevivência Celular/efeitos dos fármacos , Doxiciclina/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa/efeitos dos fármacos , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Fosforilação/efeitos dos fármacos , Proteína Quinase C-épsilon/genética , Transdução de Sinais/efeitos dos fármacos
3.
Arch Pharm (Weinheim) ; 347(8): 599-608, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24917378

RESUMO

A series of 2-pyridylhydrazones derived from phenyl-pyridazin-3-yl-methanones were prepared in search for potential novel antitumor agents. The stereochemistry of these compounds was established by means of NMR spectroscopy. Whereas hydrazones derived from 3-benzoylpyridazines (IC50 = 0.99-8.74 µM) inhibited the proliferation of the tumor cell lines tested, the non-fully aromatic 3-benzoylpyridazinone hydrazones (IC50 >10 µM) turned out to be inactive. Compounds E-1b (IC50 = 0.12 µM) and E-1d (IC50 = 0.18 µM) exert high cytotoxic activities in clonogenic assays involving human tumor cells of different tissue origins. In vivo application of compound E-1b (300 mg/kg/day) resulted in a 66% reduction in tumor burden.


Assuntos
Antineoplásicos/síntese química , Proliferação de Células/efeitos dos fármacos , Hidrazonas/síntese química , Piridazinas/química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Hidrazonas/química , Hidrazonas/farmacologia , Hidrazonas/uso terapêutico , Concentração Inibidora 50 , Camundongos Nus , Estrutura Molecular , Especificidade de Órgãos , Ensaio Tumoral de Célula-Tronco , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Med Chem ; 57(8): 3235-46, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24712764

RESUMO

Ten protein kinase C (PKC) isozymes play divergent roles in signal transduction. Because of sequence similarities, it is particularly difficult to generate isozyme-selective small molecule inhibitors. In order to identify such a selective binder, we derived a pharmacophore model from the peptide EAVSLKPT, a fragment of PKCε that inhibits the interaction of PKCε and receptor for activated C-kinase 2 (RACK2). A database of 330 000 molecules was screened in silico, leading to the discovery of a series of thienoquinolines that disrupt the interaction of PKCε with RACK2 in vitro. The most active molecule, N-(3-acetylphenyl)-9-amino-2,3-dihydro-1,4-dioxino[2,3-g]thieno[2,3-b]quinoline-8-carboxamide (8), inhibited this interaction with a measured IC50 of 5.9 µM and the phosphorylation of downstream target Elk-1 in HeLa cells with an IC50 of 11.2 µM. Compound 8 interfered with MARCKS phosphorylation and TPA-induced translocation of PKCε (but not that of PKCδ) from the cytosol to the membrane. The compound reduced the migration of HeLa cells into a gap, reduced invasion through a reconstituted basement membrane matrix, and inhibited angiogenesis in a chicken egg assay.


Assuntos
Proteína Quinase C-épsilon/antagonistas & inibidores , Quinolinas/farmacologia , Receptores de Superfície Celular/antagonistas & inibidores , Animais , Proliferação de Células/efeitos dos fármacos , Embrião de Galinha , Descoberta de Drogas , Células HeLa , Humanos , Modelos Moleculares , Fosforilação , Ligação Proteica , Proteína Quinase C-épsilon/química , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico/efeitos dos fármacos , Receptores de Quinase C Ativada , Receptores de Superfície Celular/química , Relação Estrutura-Atividade , Proteínas Elk-1 do Domínio ets/metabolismo
5.
Mol Biol Rep ; 40(8): 4995-5004, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23677713

RESUMO

The aim of the study was to analyze the consequence of silencing genes coding for the key subunits of the telomerase complex, i.e. TERT, TERC and TP1 in human breast cancer MCF7 and MDA-MB-231cells. The transfection was performed using Lipofectamine2000 and pooled siRNAs. The cytotoxic and/or antiproliferative effect of siRNA was measured by the SRB assay, the cell cycle was analysed by flow cytometry and DNA fragmentation by TUNEL analysis. Telomerase activity was assessed by TRAP, followed by PAGE and ELISA assays. Telomerase downregulation was also assessed using qPCR in order to estimate the changes in the expression profile of genes engaged in apoptosis. It was revealed that treatment of breast cancer cells with different siRNAs (100 nM) resulted in a cell type and time-dependent effects. The downregulation of telomerase subunits was followed by reduction of telomerase activity down to almost 60% compared to control cells. However, a significant effect was only observed when the TERT subunit was downregulated. Its silencing resulted in a significant (p<0.05) increase of apoptosis (over 10% in MCF7 and about 5% in MDA-MB-231 cells, corresponding to the Annexin V assay) and DNA fragmentation (almost 30% in MCF7 and over 25% in MDA-MB-231 cells). Interestingly, also several proapoptotic genes were induced after the downregulation of the key telomerase subunit, including Bax, Bik or caspase-1 and caspase-14, as well as NGFR and TNFSF10 which were upregulated twice and more.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/fisiologia , Neoplasias da Mama/enzimologia , Fragmentação do DNA , Regulação Enzimológica da Expressão Gênica/fisiologia , Telomerase/metabolismo , Eletroforese em Gel de Poliacrilamida , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica/genética , Humanos , Marcação In Situ das Extremidades Cortadas , Células MCF-7 , Subunidades Proteicas/metabolismo , RNA Interferente Pequeno/genética , Telomerase/genética , Transfecção
6.
Eur J Pharmacol ; 682(1-3): 21-8, 2012 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-22381066

RESUMO

Zapotin, a tetramethoxyflavone, is a natural compound with a wide spectrum of activities in neoplastic cells. Protein kinase C epsilon (PKCε) has been shown to be oncogenic, with the ability to increase cell migration, invasion and survival of tumor cells. Here we report that zapotin inhibits cell proliferation. In wild-type HeLa cells with basal endogenous expression of PKCε, the IC(50) was found to be 17.9 ± 1.6 µM. In HeLa cells overexpressing doxycycline-inducible constitutively active PKCε (HeLaPKCεA/E), the IC(50) was 7.6 ± 1.3 µM, suggesting that PKCε enhances the anti-proliferative effect of zapotin. Moreover, we found that zapotin selectively activated PKCε in comparison with other PKC family members, but attenuated doxycycline-induced PKCε expression. As a result of zapotin treatment for 6, 12 and 24h, the doxycycline-induced levels of the two differently phosphorylated PKCε forms (87 kDa and 95 kDa) were decreased. Migration assays revealed that increasing concentrations of zapotin (from 3.5 to 15 µM) decreased migration of HeLaPKCεA/E cells. Furthermore, zapotin significantly increased the fraction of apoptotic cells in doxycycline-induced (HeLaPKCεA/E) cells after 24h and decreased the levels of Bcl-2, c-Jun, c-Fos. This was accompanied by a degradation of PARP-1. In summary, activation of PKCε and down-modulation of the induced PKCε level by zapotin were associated with decreased migration and increased apoptosis. These observations are consistent with the previously reported chemopreventive and chemotherapeutic action of zapotin.


Assuntos
Antineoplásicos/farmacologia , Flavonas/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteína Quinase C-épsilon/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxiciclina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Isoenzimas/metabolismo
7.
Curr Top Med Chem ; 11(11): 1305-19, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21513500

RESUMO

In recent years the development of small organic molecules modulating protein-protein interactions (P-PIs) has drawn major attention in both academic and industrial research. Despite the appreciable progress being made, targeting such extensive interaction areas with comparatively small, drug-like agents has proven to be an ambitious objective. This review highlights the reasons rendering this task highly challenging and provides an overview on the latest developments in rational design approaches for P-PI modulators. The significance, scope and limitations of computational methods in this particular field of research are analyzed. Recent successfully identified and designed P-PI modulators are discussed. Thereby, particular focus is taken on small organic molecules disrupting protein-protein interfaces of protein kinases.


Assuntos
Desenho de Fármacos , Compostos Orgânicos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases/metabolismo , Biologia Computacional , Humanos , Simulação de Dinâmica Molecular , Terapia de Alvo Molecular , Compostos Orgânicos/síntese química , Compostos Orgânicos/química , Ligação Proteica/efeitos dos fármacos , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química
8.
J Biochem ; 149(3): 331-6, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21186251

RESUMO

Protein kinase C (PKC) is a family of at least 10 isozymes involved in the activation of different signal transduction pathways. The exact function of these isozymes is not known at present. Isozyme-selective inhibitors would be important to explain the function of the different PKCs and are anticipated to have pharmaceutical potential. Here we report that the small organic molecule BAS 02104951 [5-(1,3-benzodioxol-5-ylmethylene)-1-(phenylmethyl)-2,4,6(1H,3H,5H)-pyrimidinetrion], a barbituric acid derivative, inhibited PKCη and PKCε in vitro (IC(50) 18 and 36 µM, respectively). BAS 02104951 also inhibited the interaction of PKCε with its adaptor protein receptor for activated C-kinase 2 (RACK2) (IC(50) 28.5 µM). BAS 02104951 also inhibited 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced Elk-1 phosphorylation in HeLa cells, translocation of PKCε and PKCη to the membrane following treatment of PC3 cells with TPA. The compound did not inhibit the proliferation of PC3 and HeLa cells. BAS 02104951 can be used as selective inhibitor of PKCε in cells not expressing PKCη and may serve as a basis for the rational development of a selective inhibitor of PKCε or PKCη, or for an inhibitor of the PKCε/RACK2 interaction.


Assuntos
Barbitúricos/química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Proteína Quinase C-épsilon/metabolismo , Proteína Quinase C/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Animais , Linhagem Celular , Imunofluorescência , Células HeLa , Humanos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ratos , Proteínas Elk-1 do Domínio ets/metabolismo
9.
Pharmacol Rep ; 63(6): 1500-17, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22358098

RESUMO

Multidrug resistance (MDR) in leukemia patients is a great incentive to the development of new drugs. In a search for potential multidrug resistance modulators we tested a group of oleanolic acid (OA) analogues modified at C-3, C-11, C-12 and C-28 using an experimental model consisting of three human acute lymphoblastic leukemia cell lines (CCRF-CEM and the multidrug resistant sublines CCRF-VCR1000 and CCRF-ADR5000). The most effective compound, methyl 3,11-dioxoolean-12-en-28-olate (DIOXOL) was more potent in cell viability inhibition than its precursor - OA, and showed similar or even higher activity in the drug resistant than in the wild-type cells. Resistance factor (RF) values obtained for CCRF-VCR1000 and CCRF-ADR-5000 cells using MTT assay were 0.7 and 0.8 (24 h of treatment) and after 72 h of treatment 0.9 and 1.1, respectively. Moreover, 5 µM DIOXOL significantly reduced the expression of the ABCB1 gene in MDR cells by around 30%, and also decreased the level of P-gp protein. Compared to untreated control cells, DIOXOL treatment resulted in a significant P-gp decrease (30% in CCRF-ADR5000 and 50% in CCRF-VCR1000), that was detected by western blot and confirmed by flow cytometry analysis. Moreover, DIOXOL (at 10 µM) significantly inhibited P-gp transport function by more than twofold comparing to control, untreated cells that was demonstrated using rhodamine 123-based functional test. The compound exhibited synergistic activity with ABCB1 substrate - adriamycin in CCRF-VCR1000 cells, indicating partial but significant MDR reversing ability.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Dioxolanos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ácido Oleanólico/análogos & derivados , Ácido Oleanólico/administração & dosagem , Subfamília B de Transportador de Cassetes de Ligação de ATP , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Resistência a Múltiplos Medicamentos/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Humanos
10.
Chem Biol Interact ; 185(1): 25-32, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20188713

RESUMO

Protein kinase C (PKC) isoenzymes are expressed and activated in a cell type-specific manner, and play an essential role in tissue-specific signal transduction. The presence of butyrate at millimolar concentrations in the colon raises the question of whether it affects the expression of PKC isoenzymes in the different cell types of the colonic epithelium. We investigated the protein expression levels of PKCgamma, Thr(514)-phosphorylated PKCgamma (pPKCgamma-Thr(514)), and their subcellular distribution as affected by butyrate in a set of colon cancer cell lines. Thr(514)-phosphorylation of de novo synthesized PKCgamma is the first step in priming of the inactive PKCgamma before its release into the cytoplasm. For immunoblot analysis, we employed three antibodies, one against an unmodified sequence, mapping within 50 amino acids at its C-terminus, a second against pPKCgamma-Thr(514), and a third against pPKCgamma-pan-Thr(514). The antibody against an unmodified C-terminal peptide epitope did not recognize pPKCgamma-Thr(514), suggesting that phosphorylation at this site interferes with the binding of the antibody to the C-terminus. Marked butyrate-induced upregulation of PKCgamma occurred in HT29 cells (model for colonocyte stem cells) and HT29-derived cell lines. However, in Caco2 and IEC-18 cells (models for differentiated intestinal epithelial cells), PKCgamma was insensitive to upregulation, and present exclusively as pPKCgamma-Thr(514). Lovo and SW480 expressed higher levels of PKCgamma. In HT29 cells, butyrate-induced upregulation of the non-phosphorylated PKCgamma was observed in both the membrane and the cytosolic fraction. In Caco2 cells, the Thr(514)-phosphorylated form was present at high levels in both fractions. The presence of unphosphorylated PKCgamma in HT29 cells, and its complete absence in Caco2 cells demonstrates a cell type-dependent differential coupling of Thr(514)-phosphorylation with de novo synthesis of PKCgamma in colon cancer cells.


Assuntos
Butiratos/farmacologia , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Citosol/efeitos dos fármacos , Citosol/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Fosforilação , Proteína Quinase C/análise , Treonina/metabolismo
11.
Mol Endocrinol ; 24(4): 813-21, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20172962

RESUMO

Dehydroepiandrosterone sulfate (DHEAS) is the most abundant steroid in the human circulation and is secreted by the adrenals in an age-dependent fashion, with maximum levels during the third decade and very low levels in old age. DHEAS is considered an inactive metabolite, whereas cleavage of the sulfate group generates dehydroepiandrosterone (DHEA), a crucial sex steroid precursor. However, here we show that DHEAS, but not DHEA, increases superoxide generation in primed human neutrophils in a dose-dependent fashion, thereby impacting on a key bactericidal mechanism. This effect was not prevented by coincubation with androgen and estrogen receptor antagonists but was reversed by the protein kinase C inhibitor Bisindolylmaleimide 1. Moreover, we found that neutrophils are unique among leukocytes in expressing an organic anion-transporting polypeptide D, able to mediate active DHEAS influx transport whereas they did not express steroid sulfatase that activates DHEAS to DHEA. A specific receptor for DHEAS has not yet been identified, but we show that DHEAS directly activated recombinant protein kinase C-beta (PKC-beta) in a cell-free assay. Enhanced PKC-beta activation by DHEAS resulted in increased phosphorylation of p47(phox), a crucial component of the active reduced nicotinamide adenine dinucleotide phosphate complex responsible for neutrophil superoxide generation. Our results demonstrate that PKC-beta acts as an intracellular receptor for DHEAS in human neutrophils, a signaling mechanism entirely distinct from the role of DHEA as sex steroid precursor and with important implications for immunesenescence, which includes reduced neutrophil superoxide generation in response to pathogens.


Assuntos
Sulfato de Desidroepiandrosterona/farmacologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Proteína Quinase C/metabolismo , Superóxidos/metabolismo , Células Cultivadas , Humanos , Indóis/farmacologia , Masculino , Maleimidas/farmacologia , NADPH Oxidases/genética , Neutrófilos/enzimologia , Transportadores de Ânions Orgânicos/genética , Fosforilação/efeitos dos fármacos , Reação em Cadeia da Polimerase , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C beta
12.
Cell Signal ; 21(5): 745-52, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19168130

RESUMO

The protein kinase C (PKC) family is the most prominent target of tumor-promoting phorbol esters. For the PKCepsilon isozyme, different intracellular localizations and oncogenic potential in several but not all experimental systems have been reported. To obtain information about PKCepsilon-signaling, we investigated the effects of constitutively active rat PKCepsilon (PKCepsilonA/E, alanine 159 is replaced by glutamic acid) in HeLa cells in a doxycycline-inducible vector. Upon induction of PKCepsilonA/E expression by doxycycline, the major part of PKCepsilonA/E was localized to the Golgi. This led (i) to phosphorylations of PKCepsilon(S729), Elk-1(S383), PDK1(S241) and Rb(S807/S811), (ii) to elevated expression of receptor of activated C kinase 2 (RACK2) after 12 h, and (iii) increased colony formation in soft agar, increased cell migration and invasion, but not to decreased doubling time. Following induction of PKCepsilonA/E-expression by doxycycline for 24 h and additional short-term treatment with 12-O-tetradecanoylphorbol-13-acetate (TPA), PKCepsilonA/E translocated to the plasma membrane and increased phosphorylation of MARCKS(S152/156). Treatment with doxycycline/TPA or TPA alone increased phosphorylations of Elk-1(S383), PDK1(S241), Rb(S807/S811), PKCdelta(T505), p38MAPK(T180/Y182), MEK1/2(S217/S221) and ERK2(T185/T187). MARCKS was not phosphorylated after treatment with TPA alone, demonstrating that in this system it is phosphorylated only by PKCepsilon localized to the plasma membrane but not by PKCalpha or delta, the other TPA-responsive PKC isozymes in HeLa cells. These results demonstrate that PKCepsilon can induce distinctly different signaling from the Golgi and from the plasma membrane.


Assuntos
Proteína Quinase C-épsilon/metabolismo , Transdução de Sinais , Substituição de Aminoácidos , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Doxiciclina/farmacologia , Células HeLa , Humanos , Fosforilação , Ratos , Acetato de Tetradecanoilforbol/farmacologia
13.
Invest New Drugs ; 27(3): 189-202, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18612590

RESUMO

The novel compound N-benzoxazol-2-yl-N'-1-(isoquinolin-3-yl-ethylidene)-hydrazine (EPH136) has been shown to exhibit antitumor activity in vitro and in vivo. A COMPARE analysis showed that the patterns of cellular effects of EPH136 are not related to any of 175 standard antitumor agents with a known mechanism of action. In order to help identify the mechanism of action we employed a bioinformatics approach called partial least squares modelling in latent variables in which the expression levels of approximately 8,000 genes in each of 56 untreated NCI panel cell lines were correlated with the respective IC(50) values of each cell line following treatment with EPH136. The 60 genes found to be most important for the antiproliferative effect of EPH136 are involved in nucleoside, nucleotide, nucleic acid binding and metabolism, developmental processes, protein modification and metabolism. In addition, using a DNA microarray we measured the expression of approximately 5,000 known genes following treatment of HT-29 colon carcinoma cells with a two-fold IC(50) concentration of EPH136. The genes that were up-regulated more than two-fold compared to untreated controls belong to the same classes as found by the bioinformatic approach. Many of these proteins are regulated by oxidation/reduction and so we concluded that formation of radicals may be involved in the mechanism of action. We show here that EPH136 leads to generation of oxygen radicals, swelling of mitochondria and dissipation of the mitochondrial membrane potential. The antiproliferative activity of EPH136 was prevented by the radical scavenger N-acetylcysteine. Cells with elevated glutathione exhibited resistance to EPH136. In summary, the mechanism of the novel experimental anticancer drug EPH136 is generation of radicals and dissipation of the mitochondrial membrane potential.


Assuntos
Antineoplásicos/farmacologia , Benzoxazóis/farmacologia , Radicais Livres/metabolismo , Hidrazonas/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Antineoplásicos/química , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Benzoxazóis/química , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Biologia Computacional , DNA/biossíntese , Ensaios de Seleção de Medicamentos Antitumorais , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Hidrazonas/química , Análise de Sequência com Séries de Oligonucleotídeos , RNA/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Ribonucleotídeo Redutases/antagonistas & inibidores
14.
J Infect Dis ; 198(8): 1243-6, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18752432

RESUMO

Using laser scanning microscopy, we investigated whether platelets are capable of internalizing Aspergillus conidia and examined Aspergillus-platelet adherence. The influence of platelets on fungal growth was evaluated by assessing galactomannan (GM) release, hyphal elongation, and colony size. A secretion assay with [(3)H]-serotonin (5-hydroxytryptamine [5-HT]) was performed. Exposure to platelets resulted in significantly decreased GM release (p<.05), hyphal elongation (p<.001), colony size, pigmentation, and 5-HT release ( p<.05). A lack of antifungal effects was observed with the microfilament inhibitor cytochalasin D. Platelets attenuate the virulence of Aspergillus species in vitro on the basis of granule-dependent effects.


Assuntos
Aspergillus fumigatus/patogenicidade , Aspergillus/patogenicidade , Plaquetas/fisiologia , Plaquetas/ultraestrutura , Grânulos Citoplasmáticos/metabolismo , Aspergillus/classificação , Aspergillus/metabolismo , Aspergillus/fisiologia , Aspergillus fumigatus/metabolismo , Aspergillus fumigatus/fisiologia , Adesão Celular , Galactose/análogos & derivados , Humanos , Mananas/metabolismo , Microscopia Confocal , Microscopia de Fluorescência , Serotonina/metabolismo , Serotonina/farmacologia
15.
J Med Chem ; 49(21): 6343-50, 2006 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-17034140

RESUMO

Recently we have described the antitumor activities of 2-benzoxazolylhydrazones derived from 2-formyl and 2-acetylpyridines. In search of a more efficacious analogue, compounds in which the 2-acetylpyridine moiety has been replaced by 2-acylpyridine and alpha-(N)-acetyldiazine/quinoline groups have been synthesized. The 2-acylpyridyl hydrazones inhibited in vitro cell proliferation in the nM range, whereas the hydrazones derived from the alpha-(N)-acetyldiazines/quinolines inhibited cell growth in the muM range. Compounds tested in the NCI-60 cell assay were effective inhibitors of leukemia, colon, and ovarian cancer cells. E-13k [N-benzoxazol-2-yl-N'-(1-isoquinolin-3-yl-ethylidene)-hydrazine] inhibited the proliferation of MCF-7 breast carcinoma cells more efficiently than nontransformed MCF-10A cells. It is not transported by P-glycoprotein and a weak MRP substrate. Increased concentrations of serum or alpha(1)-acid glycoprotein did not reduce the antiproliferative activity of the compound. In the in vivo hollow fiber assay, E-13k achieved a score of 24, with a net cell kill of OVCAR-3 (ovarian) and SF2-95 (CNS) tumor cells.


Assuntos
Antineoplásicos/síntese química , Benzoxazóis/síntese química , Hidrazonas/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Benzoxazóis/química , Benzoxazóis/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Hidrazonas/química , Hidrazonas/farmacologia , Especificidade de Órgãos , Estereoisomerismo , Relação Estrutura-Atividade
16.
Bioorg Med Chem ; 14(1): 109-20, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16185886

RESUMO

We have developed the synthesis of boronated porphyrins for potential application in cancer treatment, based on the functional derivatives of 5,10,15,20-tetraphenylporphyrin. Boronated amide derivatives starting from 5,10,15,20-tetra(p-aminophenyl)porphyrin and 9-o- and 9-m-carborane carboxylic acid chlorides were prepared. Also, the reaction of 2-formyl-5,10,15,20-tetraphenylporphyrin with closo-C-lithium-o- and m-carboranes, as well as with closo-C-lithium monocarbon carborane, yielded neutral and anionic boronated hydroxy derivatives of 5,10,15,20-tetraphenylporphyrin, respectively. Water-soluble forms of neutral compounds were prepared by deboronation of closo-polyhedra with Bu4NF into nido-7,8- and nido-7,9-dicarbaundecaborate anions. Monocarbon carborane conjugated with copper (II) complex of 5,10,15,20-tetraphenylporphyrin was active for a variety of tumor cell lines (IC50 approximately 5 microM after 48-72 h of exposure) but was inert for non-malignant fibroblasts at up to 100 microM. At low micromolar concentrations, this compound caused the death of cells that express P-glycoprotein and other mechanisms of resistance to conventional anticancer drugs.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/toxicidade , Compostos de Boro/química , Resistencia a Medicamentos Antineoplásicos , Porfirinas/síntese química , Porfirinas/toxicidade , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/análise , Linhagem Celular Tumoral , Humanos , Porfirinas/química
17.
Res Microbiol ; 156(4): 499-505, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15862448

RESUMO

An extracellular protease from Pedobacter cryoconitis, isolated from alpine cryoconite on glacier ice, was purified and characterized. Despite high cell densities at a temperature range of 1-25 degrees C, the optimum temperature for protease production was 15 degrees C. Maximum enzyme production was achieved when the strain was grown in a pH-neutral medium containing soybean meal, wheat flour and citrate over 72 h. The 27-kDa enzyme was a metalloprotease (sensitive to EDTA, EGTA and phenanthroline) and showed maximal activity towards azocasein at 40 degrees C and pH 8. The protease was stable for 60 min at 20-30 degrees C, lost 50% of activity after 30 min at 40 degrees C, and was inactivated at 50 degrees C, but was resistant to repeated freezing and thawing. Calcium ions had no protective effect against thermal denaturation. More than 80% of the maximum activity were retained at a pH in the range of 7-10. No activity loss was detected after 1 h at pH 7-9 and 20 degrees C, nor after 1 h of incubation with 3 M urea or 0.1% perborate.


Assuntos
Bacteroidetes/enzimologia , Metaloproteases/metabolismo , Sequência de Aminoácidos , Bacteroidetes/crescimento & desenvolvimento , Sítios de Ligação , Caseínas/metabolismo , Temperatura Baixa , Meios de Cultura , Estabilidade Enzimática , Concentração de Íons de Hidrogênio , Metaloproteases/química , Metaloproteases/isolamento & purificação , Dados de Sequência Molecular , Peso Molecular , Temperatura , Fatores de Tempo
18.
Curr Cancer Drug Targets ; 4(2): 125-46, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15032665

RESUMO

Protein kinase C (PKC) comprises a family of isozymes (alpha, betaI, betaII, gamma, delta, epsilon, theta, eta, lambda/iota [mouse/human], and zeta) which are involved in signal transduction from membrane receptors to the nucleus. Activation of PKC by phorbol esters promotes tumor formation, and from that it was concluded that inhibitors of PKC might prevent carcinogenesis or inhibit tumor proliferation. However, the situation is more complicated because the exact function of the different PKC isozymes is not known at present. They have been shown to be involved in synaptic transmissions, the activation of ion fluxes, secretion, cell cycle control, differentiation, proliferation, tumorigenesis, metastasis and apoptosis. Modulators such as bryostatin-1, phospholipid analogues, PKC-activating adriamycin derivatives, CGP41251, UCN-01, and antisense oligonucleotides directed against PKCalpha, have shown antitumor activity in cancer patients. PKC inhibitors are not specific to PKC, but also interact with other signaling molecules, which may contribute to the antitumor effects. Modulators of PKC have also been shown to influence non-MDR1-mediated and MDR1-mediated antitumor drug resistance. This review is focussed on the role of PKC isozymes in human cell proliferation, apoptosis and antitumor drug resistance, and on the use of PKC modulators as antitumor agents.


Assuntos
Antineoplásicos/farmacologia , Proteína Quinase C/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Divisão Celular/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Humanos , Isoenzimas/antagonistas & inibidores , Dados de Sequência Molecular , Proteínas de Neoplasias/química , Proteínas de Neoplasias/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Neoplasias/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...