Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Anal Biochem ; 543: 37-42, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29175137

RESUMO

Digestion techniques for ICP analysis have been poorly studied for biological samples. This report describes an optimized method for analysis of trace metals that can be used across a variety of sample types. Digestion methods were tested and optimized with the analysis of trace metals in cancerous as compared to normal tissue as the end goal. Anthropological, forensic, oncological and environmental research groups can employ this method reasonably cheaply and safely whilst still being able to compare between laboratories. We examined combined HNO3 and H2O2 digestion at 170 °C for human, porcine and bovine samples whether they are frozen, fresh or lyophilized powder. Little discrepancy is found between microwave digestion and PFA Teflon pressure vessels. The elements of interest (Cu, Zn, Fe and Ni) yielded consistently higher and more accurate values on standard reference material than samples heated to 75 °C or samples that utilized HNO3 alone. Use of H2SO4 does not improve homogeneity of the sample and lowers precision during ICP analysis. High temperature digestions (>165 °C) using a combination of HNO3 and H2O2 as outlined are proposed as a standard technique for all mammalian tissues, specifically, human tissues and yield greater than 300% higher values than samples digested at 75 °C regardless of the acid or acid combinations used. The proposed standardized technique is designed to accurately quantify potential discrepancies in metal loads between cancerous and healthy tissues and applies to numerous tissue studies requiring quick, effective and safe digestions.


Assuntos
Rim/química , Fígado/química , Pulmão/química , Pâncreas/química , Oligoelementos/análise , Animais , Rim/metabolismo , Fígado/metabolismo , Pulmão/metabolismo , Espectrometria de Massas , Pâncreas/metabolismo , Padrões de Referência , Suínos , Oligoelementos/metabolismo
2.
Bioorg Med Chem Lett ; 21(2): 730-3, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21193311

RESUMO

Shp2 protein tyrosine phosphate (PTP) is a novel target for anticancer drug discovery. We identified estramustine phosphate as a Shp2 PTP inhibitor from the National Cancer Institute Approved Oncology Drug set. A focused structure-activity relationship study indicated that the 17-phosphate group is required for the Shp2 PTP inhibitor activity of estramustine phosphate. A search for estramustine phosphate analogs led to identification of two triterpenoids, enoxolone, and celastrol, having Shp2 PTP inhibitor activity. With the previously reported PTP1B inhibitor trodusquemine, our study reveals steroids and triterpenoids with negatively charged phosphate, carboxylate, or sulfonate groups as novel pharmacophores of selective PTP inhibitors.


Assuntos
Antineoplásicos Hormonais/química , Antineoplásicos Hormonais/farmacologia , Estramustina/análogos & derivados , Estramustina/farmacologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Humanos , Modelos Moleculares , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/química , Relação Estrutura-Atividade , Triterpenos/química , Triterpenos/farmacologia
3.
Biochem J ; 432(2): 255-65, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20868368

RESUMO

Recent work has highlighted roles for JAK (Janus kinase) family members in haemopoietic diseases. Although sequencing efforts have uncovered transforming JAK1 mutations in acute leukaemia, they have also identified non-transforming JAK1 mutations. Thus with limited knowledge of the mechanisms of JAK1 activation by mutation, sequencing may not readily identify transforming mutations. Therefore we sought to further understand the repertoire of transforming mutations of JAK1. We identified seven randomly generated transforming JAK1 mutations, including V658L and a deletion of amino acids 629-630 in the pseudokinase domain, as well as L910P, F938S, P960S, K1026E and Y1035C within the kinase domain. These mutations led to differential signalling activation, but exhibited similar transforming abilities, in BaF3 cells. Interestingly, these properties did not always correlate with JAK1 activation-loop phosphorylation. We also identified a JAK1 mutant that did not require a functional FERM (4.1/ezrin/radixin/moesin) domain for transformation. Although we isolated a mutation of JAK1 at residue Val658, which is found mutated in acute leukaemia patients, most of the mutations we identified are within the kinase domain and have yet to be identified in patients. Interestingly, compared with cells expressing JAK1-V658F, cells expressing these mutants had higher STAT1 (signal transducer and activator of transcription 1) phosphorylation and were more sensitive to interferon-γ-mediated growth inhibition. The differential STAT1 activation and interferon-sensitivity of JAK1 mutants may contribute to the determination of which specific JAK1 mutations ultimately contribute to disease and thus are identified in patients. Our characterization of these novel mutations contributes to a better understanding of mutational activation of JAK1.


Assuntos
Janus Quinase 1/genética , Antígenos Transformantes de Poliomavirus/genética , Sítios de Ligação , Clonagem Molecular , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Ativação Enzimática , Células HEK293 , Humanos , Immunoblotting , Interferon gama/farmacologia , Janus Quinase 1/química , Janus Quinase 1/metabolismo , Mutagênese , Mutagênese Sítio-Dirigida , Mutação , Plasmídeos/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Transdução de Sinais
4.
Front Biosci ; 12: 135-44, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17127289

RESUMO

The trace element copper is vital to the healthy functioning of organisms. Copper is used in a multitude of cellular activities including respiration, angiogenesis, and immune responses. Recently, copper has become a focus in medical research ranging from Alzheimer's disease to cancer. Copper modulation has been suggested to be a potential modality for therapy in these diseases. Several copper-binding compounds have been found to spontaneously complex with copper and form active proteasome inhibitors and apoptosis inducers. This review examines compounds in the quinoline and dithiocarbamate families and from the National Cancer Institute (NCI) Diversity Set that bind with copper and act as anticancer agents. In each case, it is shown that these compounds can bind with copper, inhibit the proteasome activity, and induce apoptosis in cancer cells. These activities are absent when copper is not present. Compounds alone, clioquinol and pyrrolidinedithiocarbamate as examples, are shown to have no effects in normal breast cells. Current research suggests that a possible therapeutic modality for cancer may be developed using the difference of high copper load in tumors versus low copper load in normal cells. This strategy would convert tumor cellular copper into a potent, specific proteasome inhibitor and apoptosis inducer. Thus, this approach could pave the way for the development of nontoxic anticancer therapy.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Cobre/metabolismo , Inibidores de Proteassoma , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Clioquinol/farmacologia , Humanos , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neovascularização Patológica , Oxiquinolina/farmacologia , Inibidores de Proteases/química , Inibidores de Proteases/farmacologia , Inibidores de Proteases/uso terapêutico , Pirrolidinas/farmacologia , Tiocarbamatos/farmacologia
5.
Int J Mol Med ; 18(4): 625-32, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16964415

RESUMO

Previously, we showed that ester carbon-containing tea polyphenols, including (-)-epigallocatechin gallate [(-)-EGCG] and (-)-epicatechin-3-gallate [(-)-ECG], potently inhibit proteasomal chymotrypsin-like activity. In addition, our in silico docking study suggested that a particular pose of (-)-EGCG could lead to potential covalent modification of the N-terminal threonine (Thr 1) of the proteasome beta5 subunit in the chymotrypsin-like active site. It has been suggested that some major biotransformation reactions, such as methylation, could result in reduced biological activity of (-)-EGCG in vivo. We hypothesize that methylation reduces binding of (-)-EGCG to the beta5 subunit of the proteasome and, therefore, decreases its proteasomal chymotrypsin-like-inhibitory potency. Here, we report that, while methylation has no effect on nucleophilic susceptibility of (-)-EGCG and (-)-ECG, it may disrupt the ability of these polyphenols to interact with Thr 1 of the proteasome beta5 subunit. In silico docking shows that methylation results in the tea polyphenols' ester carbon being moved away or blocked entirely from Thr 1. Additionally, methylation impairs the ability of (-)-EGCG and (-)-ECG to dock in a consistent low energy pose. These observations, no change in nucleophilic susceptibility, moving or blocking the ester carbon from Thr 1, and lack of a consistent docking pose, suggest that methylation disrupts the ability of (-)-EGCG and (-)-ECG to bind to the proteasome beta5 subunit, which may then diminish their proteasomal chymotrypsin-inhibitory and, therefore, other biological activities.


Assuntos
Flavonoides/metabolismo , Fenóis/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Chá/química , Catequina/análogos & derivados , Catequina/química , Catequina/metabolismo , Catequina/farmacologia , Quimotripsina/antagonistas & inibidores , Quimotripsina/metabolismo , Relação Dose-Resposta a Droga , Flavonoides/química , Flavonoides/farmacologia , Metilação , Modelos Moleculares , Conformação Molecular , Fenóis/química , Fenóis/farmacologia , Polifenóis , Inibidores de Proteassoma , Ligação Proteica , Subunidades Proteicas/antagonistas & inibidores , Subunidades Proteicas/metabolismo
6.
Curr Cancer Drug Targets ; 5(7): 529-41, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16305349

RESUMO

Tumor growth and metastasis depend on the formation of blood vessels, angiogenesis, to supply the developing mass with nutrients, oxygen, and waste removal. The proteasome, a massive multisubunit catabolic body, exerts a regulatory influence on angiogenesis. Inhibition of the proteasome activity has been found to inhibit angiogenesis and induce apoptosis in human cancer cells with limited toxicity to normal cells. Therefore, the dual action of angiogenesis inhibition and cell death induction makes proteasome inhibition an attractive modality for chemotherapy. A variety of proteasome inhibitors have been studied including: antibiotics such as lactacystin, the green tea polyphenols, and the boronic acid Velcade (MLN-341). Most recently, certain classes of copper compounds have been found to act as potent proteasome inhibitors. The potential of particular organic compounds, such as 8-hydroxyquinoline, to spontaneously bind with tumor cellular copper and form proteasome inhibitors provides a new modality of anti-proteasome and anti-angiogenesis chemotherapy. This review examines angiogenesis, the proteasome, representative proteasome inhibitors, and the emerging role of copper. The formation of new blood vessels, or angiogenesis, is an important and necessary function in both embryonic development and wound repair. Therefore, the ability to regenerate or form new vessels for blood flow is essential. The control of angiogenic pathways is tightly regulated in normal differentiated adult cells, which generally do not stimulate blood vessel growth unless injury occurs. However, cancerous tissues stimulate angiogenesis that in turn leads to increased tumor formation and possible metastases. Many of the factors involved in angiogenesis are regulated by the proteasome, which recently has become a focus in anti-cancer therapies due to its involvement in cell cycle and apoptosis control. Here we discuss angiogenesis and its relation to the proteasome. Additionally, current modalities of anti-angiogenic treatment, mainly proteasome inhibitory strategies, are reviewed. Furthermore, proteasome inhibitors, both natural and synthetic, and their anti-angiogenic effects as well as future approaches to anti-angiogenic chemotherapies are also discussed.


Assuntos
Inibidores da Angiogênese/farmacologia , Antineoplásicos/farmacologia , Inibidores de Proteassoma , Acetilcisteína/análogos & derivados , Acetilcisteína/farmacologia , Ácidos Borônicos/farmacologia , Bortezomib , Humanos , Neoplasias/irrigação sanguínea , Neoplasias/enzimologia , Neoplasias/prevenção & controle , Neovascularização Patológica/prevenção & controle , Inibidores de Proteases/farmacologia , Pirazinas/farmacologia
7.
Breast Cancer Res ; 7(6): R897-908, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16280039

RESUMO

INTRODUCTION: A physiological feature of many tumor tissues and cells is the tendency to accumulate high concentrations of copper. While the precise role of copper in tumors is cryptic, copper, but not other trace metals, is required for angiogenesis. We have recently reported that organic copper-containing compounds, including 8-hydroxyquinoline-copper(II) and 5,7-dichloro-8-hydroxyquinoline-copper(II), comprise a novel class of proteasome inhibitors and tumor cell apoptosis inducers. In the current study, we investigate whether clioquinol (CQ), an analog of 8-hydroxyquinoline and an Alzheimer's disease drug, and pyrrolidine dithiocarbamate (PDTC), a known copper-binding compound and antioxidant, can interact with copper to form cancer-specific proteasome inhibitors and apoptosis inducers in human breast cancer cells. Tetrathiomolybdate (TM), a strong copper chelator currently being tested in clinical trials, is used as a comparison. METHODS: Breast cell lines, normal, immortalized MCF-10A, premalignant MCF10AT1K.cl2, and malignant MCF10DCIS.com and MDA-MB-231, were treated with CQ or PDTC with or without prior interaction with copper, followed by measurement of proteasome inhibition and cell death. Inhibition of the proteasome was determined by levels of the proteasomal chymotrypsin-like activity and ubiquitinated proteins in protein extracts of the treated cells. Apoptotic cell death was measured by morphological changes, Hoechst staining, and poly(ADP-ribose) polymerase cleavage. RESULTS: When in complex with copper, both CQ and PDTC, but not TM, can inhibit the proteasome chymotrypsin-like activity, block proliferation, and induce apoptotic cell death preferentially in breast cancer cells, less in premalignant breast cells, but are non-toxic to normal/non-transformed breast cells at the concentrations tested. In contrast, CQ, PDTC, TM or copper alone had no effects on any of the cells. Breast premalignant or cancer cells that contain copper at concentrations similar to those found in patients, when treated with just CQ or PDTC alone, but not TM, undergo proteasome inhibition and apoptosis. CONCLUSION: The feature of breast cancer cells and tissues to accumulate copper can be used as a targeting method for anticancer therapy through treatment with novel compounds such as CQ and PDTC that become active proteasome inhibitors and breast cancer cell killers in the presence of copper.


Assuntos
Anti-Infecciosos Locais/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/patologia , Clioquinol/farmacologia , Cobre/metabolismo , Inibidores de Proteassoma , Pirrolidinas/farmacologia , Tiocarbamatos/farmacologia , Antioxidantes/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Interações Medicamentosas , Feminino , Humanos , Lesões Pré-Cancerosas/patologia
8.
Front Biosci ; 10: 2932-9, 2005 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15970547

RESUMO

Recent research suggests that copper could be used as a novel selective target for cancer therapies. Copper is a co-factor essential for tumor angiogenesis processes and high levels of copper have been found in many types of human cancers, including prostate, breast and brain. We have reported that organic copper-containing compounds, such as 8-hydroxyquinoline-copper(II), are a novel class of proteasome inhibitors and tumor cell apoptosis inducers (Daniel et al., Biochem Pharmacol. 2004;67:1139-51). Most recently, we have found that when complexed with copper, the known antioxidant pyrrolidine dithiocarbamate (PDTC) forms a potent proteasome inhibitor in human breast cancer, but not normal cells (Daniel, Chen, et al., submitted). In the current study, we investigate whether the PDTC-copper complex can play similar roles in inhibiting the proteasomal activity and consequently inducing apoptosis in human prostate cancer cells. We used tetrathiomolybdate (TM), a strong copper chelator currently being tested in clinical trials, as a control. We report here that after binding to copper, PDTC, but not TM, can inhibit the proteasomal chymotrypsin-like activity, suppress proliferation, induce apoptotic cell death, and inhibit uptake of radiopharmaceutical 2-[18F]Fluoro-2-deoxy-D-glucose in cultured human prostate cancer cells. In contrast, PDTC, TM or copper alone or a TM-copper mixture had no such effects. Our study suggests that high copper levels in human prostate cancer in vivo can be targeted by a ligand such as PDTC, resulting in formation of an active proteasome inhibitor and apoptosis inducer specifically in prostate tumor, but not normal cells.


Assuntos
Apoptose/efeitos dos fármacos , Cobre/química , Neoplasias da Próstata/patologia , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Pirrolidinas/farmacologia , Tiocarbamatos/farmacologia , Animais , Cobre/farmacologia , Fluordesoxiglucose F18/metabolismo , Humanos , Masculino , Complexo de Endopeptidases do Proteassoma/metabolismo , Pirrolidinas/química , Tiocarbamatos/química , Células Tumorais Cultivadas
9.
Front Biosci ; 10: 1010-23, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15769601

RESUMO

It has been suggested that proteasome activity is essential for tumor cell proliferation and drug resistance development. We have previously shown that natural and synthetic ester bond-containing tea polyphenols are selective inhibitors of the chymotrypsin-like activity of the proteasome. The most abundant catechin in green tea is (-)-epigallocatechin-3-gallate [(-)-EGCG], which has been found by many laboratories to exhibit the most potent anticancer activity. We have reported that (-)-EGCG is also the most effective proteasome inhibitor among all the natural green tea catechins tested. Unfortunately, (-)-EGCG is very unstable in neutral and alkaline conditions. In an attempt to increase the stability and thus the efficacy, we synthesized several (-)-EGCG analogs with acetyl protected -OH groups as prodrugs. Here we report, for the first time, that these acetylated synthetic tea analogs are much more potent than natural (-)-EGCG in inhibiting the proteasome in cultured tumor cells. Consistently, these protected analogs showed much higher potency than (-)-EGCG to inhibit proliferation and transforming activity and to induce apoptosis in human leukemic, prostate, breast, and simian virus 40-transformed cells. Additionally, these protected analogs had greatly reduced effects on human normal and non-transformed cells. Therefore, these peracetate protected tea polyphenols are more efficacious than (-)-EGCG and possess great potential to be developed into novel anticancer drugs. Identification of the cytosolic metabolite(s) of peracetate-protected polyphenols in cultured tumor cells and examination of their in vivo tumor growth-inhibitory activity are currently underway in our laboratory.


Assuntos
Antineoplásicos/síntese química , Apoptose/efeitos dos fármacos , Flavonoides/síntese química , Fenóis/síntese química , Inibidores de Proteassoma , Chá/química , Antineoplásicos/farmacologia , Apoptose/fisiologia , Catequina/análogos & derivados , Catequina/farmacologia , Flavonoides/farmacologia , Humanos , Peróxido de Hidrogênio/metabolismo , Células Jurkat , Fenóis/farmacologia , Polifenóis , Células Tumorais Cultivadas
10.
Front Biosci ; 10: 1183-90, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15769617

RESUMO

The development of novel anti-cancer drugs that induce apoptosis has long been a focus of drug discovery. Beta-lactam antibiotics have been used for over 60 years to fight bacterial infectious diseases with little or no side effects observed. Recently a new class of N-methylthiolated beta-lactams has been discovered that have potent activity against methicillin resistant Staphylococcus aureas. Most recently, we determined the potential effects of these N-thiolated beta-lactams on tumorigenic cell growth and found that they are apoptosis-inducers in human cancer cell lines. In the current study, we further determined the effects of the substitution of the O-methyl moiety on C3 and stereochemistry of the beta-lactams on the anti-proliferative and apoptosis-inducing abilities. We have found that lactam 18, in which C3 is substituted with an acrylate ester group, is a very effective proliferation inhibitor against human premalignant and malignant breast, leukemic, and simian virus 40-transformed fibroblast cells. Generally speaking, increasing the size of the moiety on C3 decreases its anti-proliferation potency, possibly indicating steric hindrance with the cellular target or decreased permeability through the cell membrane. We also found that the stereochemistry of the beta-lactams plays an important role in their potency. The 3S,4R isomers are more effective than their enantiomers (3R,4S), suggesting that 3S,4R configuration is more favorable for target interaction.


Assuntos
Antibacterianos/farmacologia , Apoptose/efeitos dos fármacos , beta-Lactamas/farmacologia , Antibacterianos/química , Humanos , Relação Estrutura-Atividade , Células Tumorais Cultivadas , beta-Lactamas/química
11.
Biochem Pharmacol ; 69(10): 1421-32, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15857606

RESUMO

It has been shown that proteasome activity is required for cancer cell survival and consumption of fruits and vegetables is associated with decreased cancer risk. Previously, we reported that grape extract could inhibit proteasome activity and induce apoptosis in tumor cells. In this study, we examined the flavonoids apigenin, quercetin, kaempferol and myricetin for their proteasome-inhibitory and apoptosis-inducing abilities in human tumor cells. We report that apigenin and quercetin are much more potent than kaempferol and myricetin at: (i) inhibiting chymotrypsin-like activity of purified 20S proteasome and of 26S proteasome in intact leukemia Jurkat T cells; (ii) accumulating putative ubiquitinated forms of two proteasome target proteins, Bax and Inhibitor of nuclear factor kappabeta-alpha in Jurkat T cells and (iii) inducing activation of caspase-3 and cleavage of poly(ADP-ribose) polymerase in Jurkat T cells. The proteasome-inhibitory abilities of these compounds correlated with their apoptosis-inducing potencies. Results from computational modeling of the potential interactions of these flavonoids to the chymotrypsin site (beta5 subunit) of the proteasome were consistent with the obtained proteasome-inhibitory activities. We found that the C(4) carbon may be a site of nucleophilic attack by the OH group of N-terminal threonine of proteasomal beta5 subunit and that the C(3) hydroxyl may alter the ability of these flavonoids to inhibit the proteasome. Finally, apigenin neither effectively inhibited the proteasome activity nor induced apoptosis in non-transformed human natural killer cells. Our results suggested that the proteasome may be a target of these dietary flavonoids in human tumor cells and that inhibition of the proteasome by flavonoids may be one of the mechanisms responsible for their cancer-preventive effects.


Assuntos
Anticarcinógenos/farmacologia , Apoptose/efeitos dos fármacos , Flavonoides/farmacologia , Inibidores de Proteassoma , Apigenina/farmacologia , Simulação por Computador , Inibidores de Cisteína Proteinase , Dieta , Humanos , Células Jurkat , Quempferóis/farmacologia , Células Matadoras Naturais/efeitos dos fármacos , Quercetina/farmacologia , Relação Estrutura-Atividade
12.
Eur J Med Chem ; 90: 315-331, 2015 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-25437618

RESUMO

Inhibition of anti-apoptotic Mcl-1 is a promising anticancer strategy to overcome the survival and chemoresistance of a broad spectrum of human cancers. We previously reported on the identification of a natural product marinopyrrole A (1) that induces apoptosis in Mcl-1-dependent cells through Mcl-1 degradation. Here, we report the design and synthesis of novel marinopyrrole-based analogs and their evaluation as selective inhibitors of Mcl-1 as well as dual Mcl-1/Bcl-xL inhibitors. The most selective Mcl-1 antagonists were 34, 36 and 37 with 16-, 13- and 9-fold more selectivity for disrupting Mcl-1/Bim over Bcl-xL/Bim binding, respectively. Among the most potent dual inhibitors is 42 which inhibited Mcl-1/Bim and Bcl-xL/Bim binding 15-fold (IC50 = 600 nM) and 33-fold (500 nM) more potently than (±)-marinopyrrole A (1), respectively. Fluorescence quenching, NMR analysis and molecular docking indicated binding of marinopyrroles to the BH3 binding site of Mcl-1. Several marinopyrroles potently decreased Mcl-1 cellular levels and induced caspase 3 activation in human breast cancer cells. Our studies provide novel "lead" marinopyrroles for further optimization as selective Mcl-1 inhibitors and dual Mcl-1 and Bcl-xL inhibitors.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Desenho de Fármacos , Proteína de Sequência 1 de Leucemia de Células Mieloides/antagonistas & inibidores , Pirróis/farmacologia , Proteína bcl-X/antagonistas & inibidores , Sítios de Ligação/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Pirróis/síntese química , Pirróis/química , Relação Estrutura-Atividade , Proteína bcl-X/metabolismo
13.
Proteins ; 54(1): 58-70, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14705024

RESUMO

Previously, we demonstrated that natural and synthetic ester bond-containing green tea polyphenols were potent and specific non-peptide proteasome inhibitors. However, the molecular mechanism of inhibition is currently unknown. Here, we report that inhibition of the chymotrypsin activity of the 20S proteasome by (-)-epigallocatechin-3-gallate (EGCG) is time-dependent and irreversible, implicating acylation of the beta5-subunit's catalytic N-terminal threonine (Thr 1). This knowledge is used, along with in silico docking experiments, to aid in the understanding of binding and inhibition. On the basis of these docking experiments, we propose that (-)-EGCG binds the chymotrypsin site in an orientation and conformation that is suitable for a nucleophilic attack by Thr 1. Consistently, the distance from the electrophilic carbonyl carbon of (-)-EGCG to the hydroxyl group of Thr 1 was measured as 3.18 A. Furthermore, the A ring of (-)-EGCG acts as a tyrosine mimic, binding to the hydrophobic S1 pocket of the beta5-subunit. In the process, the (-)-EGCG scissile bond may become strained, which could lower the activation energy for attack by the hydroxyl group of Thr 1. This model is validated by comparison of predicted and actual activities of several EGCG analogs, either naturally occurring, previously synthesized, or rationally synthesized.


Assuntos
Catequina/análogos & derivados , Catequina/química , Cisteína Endopeptidases/efeitos dos fármacos , Inibidores de Cisteína Proteinase/química , Modelos Moleculares , Complexos Multienzimáticos/efeitos dos fármacos , Amidas/química , Sítios de Ligação , Camellia sinensis/química , Catequina/metabolismo , Catequina/farmacologia , Quimotripsina/metabolismo , Cisteína Endopeptidases/química , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Desenho de Fármacos , Flavonoides/química , Flavonoides/metabolismo , Flavonoides/farmacologia , Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Fenóis/química , Fenóis/metabolismo , Fenóis/farmacologia , Polifenóis , Complexo de Endopeptidases do Proteassoma , Ligação Proteica , Subunidades Proteicas/química , Subunidades Proteicas/efeitos dos fármacos , Subunidades Proteicas/metabolismo
14.
Front Biosci ; 9: 2652-62, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15358588

RESUMO

The trace element copper is vital to the healthy functioning of organisms. Copper is used in a multitude of cellular activities including respiration, angiogenesis, and immune responses. Like other metals, copper homeostasis is a tightly regulated process. Copper is transported from dietary intake through the serum and into cells via a variety of transporters. There are a variety of copper chaperones designed to insure that copper is sequestered from interaction with cellular membranes, proteins, or DNA where its properties can result in oxidative damage. However, there are disease states in which copper transporters crucial to homeostasis are impaired resulting in potentially toxic copper accumulation. Wilsons and Menkes diseases are two such cases. Wilsons disease (hepatolenticular degeneration) is an autosomal recessive disorder resulting in extreme accumulation of copper in the liver with deposits elsewhere in the body. Menkes is characterized by a systemic copper deficiency (different from the liver specificity of Wilsons disease) and is the result of an X-linked recessive mutation in a copper transporter. Uptake of copper is impaired due to inability to remove existing copper from cells primarily in the small intestine. Though the causes are dramatically different, cancer also shares a similar diagnostic in the accumulation of copper in effected tissues. Studies have shown greatly elevated levels of copper in cancer tissues, and some diagnostics and treatments from Wilsons and Menkes diseases, such as copper chelation therapy, have been used in the treatment of cancer. Given the commonality of copper accumulation in these diseases and that common therapies exist between them, it may prove beneficial to study all three diseases in light of copper homeostasis. This review will examine the chemical nature and biological roles of copper, Wilsons and Menkes disease and their therapies, and the use of copper related therapies in cancer.


Assuntos
Cobre/metabolismo , Degeneração Hepatolenticular/diagnóstico , Síndrome dos Cabelos Torcidos/diagnóstico , Neoplasias/diagnóstico , Animais , Quelantes/farmacologia , Desenho de Fármacos , Degeneração Hepatolenticular/patologia , Humanos , Ligantes , Síndrome dos Cabelos Torcidos/patologia , Mutação , Neoplasias/patologia , Estresse Oxidativo , Oxigênio/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo
15.
Front Biosci ; 9: 2618-31, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15358585

RESUMO

The cancer-preventive effects of green tea and its main constituent (-)-epigallocatechin gallate [(-)-EGCG] are widely supported by results from epidemiological, cell culture, animal and clinical studies in the recent decade. In vitro cell culture studies show that tea polyphenols potently induce apoptotic cell death and cell cycle arrest in tumor cells but not in their normal cell counterparts. Green tea polyphenols affect several signal transduction pathways, including growth factor-mediated, the mitogen-activated protein kinase (MAPK)-dependent, and ubiquitin/proteasome degradation pathways. Epidemiological studies have suggested that the consumption of green tea lowers the risk of cancer. Various animal studies have revealed that treatment by green tea inhibits tumor incidence and multiplicity in different organ sites such as skin, lung, liver, stomach, mammary gland and colon. Phase I and II clinical trials were carried out recently to explore the anticancer effects of green tea in patients with cancer. At this time, more mechanistic research, animal studies, and clinical trials are necessary to further evaluate the role of green tea in cancer prevention.


Assuntos
Anticarcinógenos/farmacologia , Flavonoides/farmacologia , Neoplasias/prevenção & controle , Fenóis/farmacologia , Chá , Apoptose , Ensaios Clínicos como Assunto , Flavonoides/química , Humanos , Sistema de Sinalização das MAP Quinases , Neoplasias/epidemiologia , Neoplasias/metabolismo , Fenóis/química , Polifenóis , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais , Ubiquitina/metabolismo
16.
Biochem Pharmacol ; 66(6): 965-76, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12963483

RESUMO

Epidemiological studies have suggested that increased soy consumption is associated with reduced cancer occurrence. Genistein, a soy isoflavone, has been reported to inhibit the growth of human tumor cells although the involved molecular mechanisms are not clearly defined. Here we report that genistein inhibits the proteasomal chymotrypsin-like activity in vitro and in vivo. Computational docking studies suggest that the interaction of genistein with the proteasomal beta 5 subunit is responsible for inhibition of the chymotrypsin-like activity. Inhibition of the proteasome by genistein in prostate cancer LNCaP and breast cancer MCF-7 cells is associated with accumulation of ubiquitinated proteins and three known proteasome target proteins, the cyclin-dependent kinase inhibitor p27(Kip1), inhibitor of nuclear factor-kappa B (I kappa B-alpha), and the pro-apoptotic protein Bax. Genistein-mediated proteasome inhibition was accompanied by induction of apoptosis in these solid tumor cells. Finally, genistein induced proteasome inhibition and apoptosis selectively in simian virus 40-transformed human fibroblasts, but not in their parental normal counterpart. Our results suggest that the proteasome is a potential target of genistein in human tumor cells and that inhibition of the proteasome activity by genistein might contribute to its cancer-preventive properties.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Cisteína Endopeptidases/efeitos dos fármacos , Genisteína/farmacologia , Complexos Multienzimáticos/efeitos dos fármacos , Neoplasias da Mama/patologia , Transformação Celular Viral/efeitos dos fármacos , Quimotripsina/metabolismo , Cisteína Endopeptidases/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Humanos , Isoflavonas/farmacologia , Masculino , Complexos Multienzimáticos/metabolismo , Neoplasias da Próstata/patologia , Complexo de Endopeptidases do Proteassoma , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/biossíntese
17.
Biochem Pharmacol ; 67(6): 1139-51, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15006550

RESUMO

Here we report that organic copper complexes can potently and selectively inhibit the chymotrypsin-like activity of the proteasome in vitro and in vivo. Several copper compounds, such as NCI-109268 and bis-8-hydroxyquinoline copper(II) [Cu(8-OHQ)(2)], can inhibit the chymotrypsin-like activity of purified 20S proteasome. In human leukemia cells, proteasome inhibition occurs within 15min after treatment, followed by apoptosis. Neither proteasome inhibition nor apoptosis occurs in non-transformed, immortalized human natural killer cells under the same treatment. Furthermore, proteasome inhibition and apoptosis induction were detected in prostate cancer cells treated with the ligand 8-OHQ alone following pre-treatment with copper(II) chloride. None of these events occurred in cells treated with copper(II) chloride alone, 8-OHQ alone (without growth in copper-enriched media), or nickel(II) chloride pre-treatment followed by 8-OHQ. Furthermore, we found that copper-mediated inhibition of purified 20S proteasome cannot be blocked by a reducing agent and that organic copper compounds do not generate hydrogen peroxide in the cells, suggesting that proteasome inhibition and apoptosis induction are not due to copper-mediated oxidative damage of proteins. Our results suggest that certain types of organic ligands could bind to tumor cellular copper, forming potent proteasome inhibitors and apoptosis inducers at copper concentrations found in tumor tissues.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Cobre/química , Complexos Multienzimáticos/antagonistas & inibidores , Antineoplásicos/química , Linhagem Celular Transformada , Quimotripsina/metabolismo , Cobre/farmacologia , Cisteína Endopeptidases , Humanos , Células Jurkat , Oxiquinolina/farmacologia , Complexo de Endopeptidases do Proteassoma , Células Tumorais Cultivadas
18.
Int J Mol Med ; 12(2): 247-52, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12851726

RESUMO

Various stimuli including anticancer drugs are capable of initiating the apoptotic death program in human tumor cells via activation of caspases. Mitochondria play an essential role for cell apoptotic commitment. Previous studies have shown a potential role of calpain activation in apoptosis, however, the involved molecular mechanisms remain to be defined. In the current study, we have examined the expression and activation of mitochondrial calpain in Jurkat T leukemia cells, MCF-7 breast carcinoma and LNCaP prostate cancer cells during apoptosis induced by an anticancer drug (VP-16, tamoxifen) or the specific p38 kinase inhibitor PD-169316. Our results suggest that increased expression and autolysis of the mitochondrial calpain small subunit are tightly associated with calpain activation in an early stage of apoptosis. In contrast, there were no correlations observed between the early calpain activation and changes in levels of mitochondrial calpain large subunit and the endogenous calpain inhibitor calpastatin. Furthermore, pretreatment with the specific pharmacological calpain inhibitor calpeptin blocked the drug-induced calpain small subunit autolysis and calpain activation in mitochondria and inhibited apoptosis-associated caspase-3 activation, demonstrating that mitochondrial calpain activation through small subunit cleavage is an essential step for inducing tumor cell apoptosis by various anticancer drugs.


Assuntos
Apoptose/fisiologia , Calpaína/metabolismo , Mitocôndrias/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autólise , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Grupo dos Citocromos c/efeitos dos fármacos , Grupo dos Citocromos c/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Etoposídeo/farmacologia , Feminino , Humanos , Imidazóis/farmacologia , Células Jurkat/efeitos dos fármacos , Masculino , Mitocôndrias/enzimologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Subunidades Proteicas , Tamoxifeno/farmacologia , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno
19.
Expert Opin Ther Pat ; 24(4): 369-82, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24450483

RESUMO

INTRODUCTION: Over the past 3 years, numerous patents and patent applications have been submitted and published involving compounds designed to inhibit the proteasome. Proteasome inhibition has been of great interest in cancer research since disruption of proteolysis leads to a significant buildup of cytotoxic proteins and activation of apoptotic pathways, particularly in rapidly proliferating cells. The current standards in proteasome inhibition are the only FDA-approved inhibitors, bortezomib and carfilzomib. Although these drugs are quite effective in treating multiple myeloma and other blood tumors, there are shortcomings, including toxicities and resistance. Most of the current patents attempt to improve on existing compounds, by increasing bioavailability and selectivity, while attempting to reduce toxicity. A general categorization of similar compounds was employed to evaluate and compare drug design strategies. AREAS COVERED: This review focuses on novel compounds and subsequent analogs developed for proteasome inhibition, used in preventing and treating human cancers. A comprehensive description and categorization of patents related to each type of compound and its derivatives, as well as their uses and efficacies as anticancer agents is included. A review of combination therapy patents has also been included. EXPERT OPINION: Although there are many diverse chemical scaffolds being published, there are few patented proteasome inhibitors whose method of inhibition is genuinely novel. Most patents utilize a destructive chemical warhead to attack the catalytic threonine residue of the proteasome active sites. Few patents try to depart from this, emphasizing the need for developing new mechanisms of action and specific targeting.


Assuntos
Neoplasias/tratamento farmacológico , Patentes como Assunto , Inibidores de Proteassoma/uso terapêutico , Animais , Desenho de Fármacos , Quimioterapia Combinada , Humanos , Inibidores de Proteassoma/química , Inibidores de Proteassoma/farmacologia
20.
PLoS One ; 9(12): e114131, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25501935

RESUMO

Lung cancer is the second most common cancer and the leading cause of cancer-related deaths. Despite recent advances in the development of targeted therapies, patients with advanced disease remain incurable, mostly because metastatic non-small cell lung carcinomas (NSCLC) eventually become resistant to tyrosine kinase inhibitors (TKIs). Kinase inhibitors have the potential for target promiscuity because the kinase super family is the largest family of druggable genes that binds to a common substrate (ATP). As a result, TKIs often developed for a specific purpose have been found to act on other targets. Drug affinity chromatography has been used to show that dasatinib interacts with the TGFß type I receptor (TßR-I), a serine-threonine kinase. To determine the potential biological relevance of this association, we studied the combined effects of dasatinib and TGFß on lung cancer cell lines. We found that dasatinib treatment alone had very little effect; however, when NSCLC cell lines were treated with a combination of TGFß and dasatinib, apoptosis was induced. Combined TGFß-1 + dasatinib treatment had no effect on the activity of Smad2 or other non-canonical TGFß intracellular mediators. Interestingly, combined TGFß and dasatinib treatment resulted in a transient increase in p-Smad3 (seen after 3 hours). In addition, when NSCLC cells were treated with this combination, the pro-apoptotic protein BIM was up-regulated. Knockdown of the expression of Smad3 using Smad3 siRNA also resulted in a decrease in BIM protein, suggesting that TGFß-1 + dasatinib-induced apoptosis is mediated by Smad3 regulation of BIM. Dasatinib is only effective in killing EGFR mutant cells, which is shown in only 10% of NSCLCs. Therefore, the observation that wild-type EGFR lung cancers can be manipulated to render them sensitive to killing by dasatinib could have important implications for devising innovative and potentially more efficacious treatment strategies for this disease.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pulmonares/patologia , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tiazóis/farmacologia , Fator de Crescimento Transformador beta/metabolismo , Compostos de Anilina/metabolismo , Compostos de Anilina/farmacologia , Antineoplásicos/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Dasatinibe , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Simulação de Acoplamento Molecular , Nitrilas/metabolismo , Nitrilas/farmacologia , Conformação Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Pirimidinas/metabolismo , Quinolinas/metabolismo , Quinolinas/farmacologia , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/química , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteína Smad3/metabolismo , Tiazóis/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA