Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Cancer Growth Metastasis ; 7: 19-25, 2014.
Article in English | MEDLINE | ID: mdl-24987270

ABSTRACT

The aim of cancer chemoprevention is disruption or delay of the molecular pathways that lead to carcinogenesis. Chemopreventive blocking and/or suppressing agents disrupt the molecular mechanisms that drive carcinogenesis such as DNA damage by reactive oxygen species, increased signal transduction to NF-κB, epigenomic deregulation, and the epithelial mesenchymal transition that leads to metastatic progression. Numerous dietary phytochemicals have been observed to inhibit the initiation phase of carcinogenesis, and therefore are useful in primary chemoprevention. Moreover, phytochemicals are capable of interfering with the molecular mechanisms of metastasis. Likewise, numerous synthetic compounds are relevant and clinically viable as chemopreventive agents during the fundamental stages of carcinogenesis. While molecularly targeted anti-cancer therapies are in constant stages of development, superior patient outcomes are observed if carcinogenic processes are prevented altogether. This article reviews the role of chemopreventive compounds in inhibition of cancer initiation and their ability to reduce cancer progression.

3.
Clin Lab Sci ; 25(1): 38-44, 2012.
Article in English | MEDLINE | ID: mdl-22458048

ABSTRACT

The cellular proteasome is an important molecular target in cancer therapy and drug resistance research. Proteasome inhibitors are effective agents against multiple myeloma and mantle cell lymphoma and display great potential as treatment for a variety of other malignancies. The proteasome is a large multicatalytic, proteinase complex located in the cytosol and the nucleus of eukaryotic cells. The ubiquitin proteasome system is responsible for the degradation of most intracellular proteins and therefore plays an essential regulatory role in critical cellular processes including cell cycle progression, proliferation, differentiation, angiogenesis, and apoptosis. Cancer cells are particularly sensitive to proteasome inhibitors, indicating the utility for inhibition of the ubiquitin-proteasome pathway as an approach for cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/enzymology , Proteasome Inhibitors , Humans , Ubiquitin/metabolism
4.
Cancer Res ; 68(18): 7283-92, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794115

ABSTRACT

Curcumin (diferuloylmethane) is the major active ingredient of turmeric (Curcuma longa) used in South Asian cuisine for centuries. Curcumin has been shown to inhibit the growth of transformed cells and to have a number of potential molecular targets. However, the essential molecular targets of curcumin under physiologic conditions have not been completely defined. Herein, we report that the tumor cellular proteasome is most likely an important target of curcumin. Nucleophilic susceptibility and in silico docking studies show that both carbonyl carbons of the curcumin molecule are highly susceptible to a nucleophilic attack by the hydroxyl group of the NH(2)-terminal threonine of the proteasomal chymotrypsin-like (CT-like) subunit. Consistently, curcumin potently inhibits the CT-like activity of a purified rabbit 20S proteasome (IC(50) = 1.85 micromol/L) and cellular 26S proteasome. Furthermore, inhibition of proteasome activity by curcumin in human colon cancer HCT-116 and SW480 cell lines leads to accumulation of ubiquitinated proteins and several proteasome target proteins, and subsequent induction of apoptosis. Furthermore, treatment of HCT-116 colon tumor-bearing ICR SCID mice with curcumin resulted in decreased tumor growth, associated with proteasome inhibition, proliferation suppression, and apoptosis induction in tumor tissues. Our study shows that proteasome inhibition could be one of the mechanisms for the chemopreventive and/or therapeutic roles of curcumin in human colon cancer. Based on its ability to inhibit the proteasome and induce apoptosis in both HCT-116 and metastatic SW480 colon cancer cell lines, our study suggests that curcumin could potentially be used for treatment of both early-stage and late-stage/refractory colon cancer.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Curcumin/pharmacology , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Growth Processes/drug effects , Dose-Response Relationship, Drug , Female , HCT116 Cells , Humans , Leupeptins/pharmacology , Mice , Models, Molecular , Poly(ADP-ribose) Polymerases/metabolism , Proteasome Endopeptidase Complex/metabolism , Xenograft Model Antitumor Assays
5.
J Cell Biochem ; 105(2): 514-23, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18636546

ABSTRACT

Flavonoids are polyphenolic compounds widely distributed in the plant kingdom. Compelling research indicates that flavonoids have important roles in cancer chemoprevention and chemotherapy possibly due to biological activities that include action through anti-inflammation, free radical scavenging, modulation of survival/proliferation pathways, and inhibition of the ubiquitin-proteasome pathway. Plant polyphenols including the green tea polyphenol (-)-epigallocatechin gallate or (-)-EGCG, and the flavonoids apigenin, luteolin, quercetin, and chrysin have been shown to inhibit proteasome activity and induce apoptosis in human leukemia cells. However, biotransformation reactions to the reactive hydroxyl groups on polyphenols could reduce their biological activities. Although methylated polyphenols have been suggested to be metabolically more stable than unmethylated polyphenols, the practical use of methylated polyphenols as cancer preventative agents warrants further investigation. In the current study, methylated and unmethylated flavonoids were studied for their proteasome-inhibitory and apoptosis-inducing abilities in human leukemia HL60 cells. Methylated flavonoids displayed sustained bioavailability and inhibited cellular proliferation by arresting cells in the G(1) phase. However, they did not act as proteasome inhibitors in either an in vitro system or an in silico model and only weakly induced apoptosis. In contrast, unmethylated flavonoids exhibited inhibition of the proteasomal activity in intact HL60 cells, accumulating proteasome target proteins and inducing caspase activation and poly(ADP-ribose) polymerase cleavage. We conclude that methylated flavonoids lack potent cytotoxicity against human leukemia cells and most likely have limited ability as chemopreventive agents.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Flavonoids/pharmacology , Neoplasms/pathology , Caspases/metabolism , Cell Line, Tumor , Diet , Flavonoids/chemistry , G1 Phase , Humans , Methylation , Poly(ADP-ribose) Polymerases/metabolism , Structure-Activity Relationship
6.
J Cell Biochem ; 103(1): 234-44, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17541980

ABSTRACT

Pristimerin is a natural product derived from the Celastraceae and Hippocrateaceae families that were used as folk medicines for anti inflammation in ancient times. Although it has been shown that pristimerin induces apoptosis in breast cancer cells, the involved mechanism of action is unknown. The purpose of the current study is to investigate the primary target of pristimerin in human cancer cells, using prostate cancer cells as a working model. Nucleophilic susceptibility and in silico docking studies show that C6 of pristimerin is highly susceptible towards a nucleophilic attack by the hydroxyl group of N-terminal threonine of the proteasomal chymotrypsin subunit. Consistently, pristimerin potently inhibits the chymotrypsin-like activity of a purified rabbit 20S proteasome (IC50 2.2 micromol/L) and human prostate cancer 26S proteasome (IC50 3.0 micromol/L). The accumulation of ubiquitinated proteins and three proteasome target proteins, Bax, p27 and I kappa B-alpha, in androgen receptor (AR)-negative PC-3 prostate cancer cells supports the conclusion that proteasome inhibition by pristimerin is physiologically functional. This observed proteasome inhibition subsequently led to the induction of apoptotic cell death in a dose- and kinetic-dependent manner. Furthermore, in AR-positive, androgen-dependent LNCaP and AR-positive, androgen-independent C4-2B prostate cancer cells, proteasome inhibition by pristimerin results in suppression of AR protein prior to apoptosis. Our data demonstrate, for the first time, that the proteasome is a primary target of pristimerin in prostate cancer cells and inhibition of the proteasomal chymotrypsin-like activity by pristimerin is responsible for its cancer cell death-inducing property.


Subject(s)
Apoptosis/drug effects , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Triterpenes/pharmacology , Androgens/pharmacology , Animals , Cell Extracts , Cell Line, Tumor , Computational Biology , Electrons , Humans , Kinetics , Male , Models, Molecular , Molecular Structure , Pentacyclic Triterpenes , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Proteasome Endopeptidase Complex/chemistry , Protein Subunits/chemistry , Protein Subunits/metabolism , Rabbits , Receptors, Androgen/metabolism , Serine Endopeptidases/metabolism , Triterpenes/chemistry
7.
Bioorg Med Chem ; 15(15): 5076-82, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17544279

ABSTRACT

Analogs of (-)-EGCG containing a para-amino group on the D-ring in place of the hydroxyl groups have been synthesized and their proteasome inhibitory activities were studied. We found that, the O-acetylated (-)-EGCG analogs possessing a p-NH(2) or p-NHBoc (Boc; tert-butoxycarbonyl) D-ring (5 and 7) act as novel tumor cellular proteasome inhibitors and apoptosis inducers with potency similar to natural (-)-EGCG and similar to (-)-EGCG peracetate. These data suggest that the acetylated amino-GTP analogs have the potential to be developed into novel anticancer agents.


Subject(s)
Catechin/analogs & derivatives , Tea/chemistry , Apoptosis/drug effects , B-Lymphocytes/drug effects , Caspase 3/metabolism , Catechin/chemistry , Catechin/pharmacology , Cell Line, Tumor , Humans , Leukemia/drug therapy , Molecular Structure , Proteasome Endopeptidase Complex/metabolism , Structure-Activity Relationship
8.
J Cell Physiol ; 213(1): 252-60, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17477351

ABSTRACT

Under physiological conditions, biotransformation reactions, such as methylation, can modify green tea polyphenols (GTPs) and therefore limit their in vivo cancer-preventive activity. Although a recent study suggested that methylated polyphenols are less cancer-protective, the molecular basis is unknown. We previously reported that ester bond-containing GTPs, for example (-)-epigallocatechin-3-gallate [(-)-EGCG] or (-)-epicatechin-3-gallate [(-)-ECG], potently and specifically inhibit the proteasomal chymotrypsin-like activity. In this study, we hypothesize that methylated GTPs have decreased proteasome-inhibitory abilities. To test this hypothesis, methylated (-)-EGCG and (-)-ECG analogs that can be found in vivo were synthesized and studied for their structure-activity relationships (SARs) using a purified 20S proteasome. The addition of a single methyl group on (-)-EGCG or (-)-ECG led to decreased proteasome inhibition and, as the number of methyl groups increased, the inhibitory potencies further decreased. These SARs were supported by our findings from in silico docking analysis published recently. Previously, we synthesized a peracetate-protected (-)-EGCG molecule, Pro-EGCG (1), to enhance its cellular permeability and stability, and current HPLC analysis confirms conversion of Pro-EGCG (1) to (-)-EGCG in cultured human leukemic Jurkat T cells. Furthermore, in this study, peracetate-protected forms of methylated GTPs were added in intact Jurkat T cells to observe the intracellular effects of methylation. Peracetate-protected, monomethylated (-)-EGCG induced greater cellular proteasome inhibition and apoptosis than did peracetate-protected, trimethylated (-)-EGCG, consistent with the potencies of the parent methylated analogs against a purified 20S proteasome. Therefore, methylation on GTPs, under physiological conditions, could decrease their proteasome-inhibitory activity, contributing to decreased cancer-preventive effects of tea consumption.


Subject(s)
Flavonoids/pharmacology , Phenols/pharmacology , Proteasome Inhibitors , Tea/chemistry , Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/pharmacology , Cell Death/drug effects , Flavonoids/chemistry , Flavonoids/isolation & purification , Humans , Jurkat Cells , Methylation , Phenols/chemistry , Phenols/isolation & purification , Polyphenols , Protease Inhibitors/pharmacology
9.
Cancer Res ; 67(9): 4303-10, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17483343

ABSTRACT

The most abundant and biologically active green tea catechin, (-)-epigallocatechin-3-gallate or (-)-EGCG, has been shown to act as a proteasome inhibitor and tumor cell death inducer. However, (-)-EGCG is unstable under physiologic conditions and has poor bioavailability. Previously, in an attempt to increase the stability of (-)-EGCG, we introduced peracetate protections to its reactive hydroxyl groups and showed that this peracetate-protected (-)-EGCG [Pro-EGCG (1); formerly named compound 1] could be converted into (-)-EGCG under cell-free conditions. In the current study, we provide evidence that when cultured human breast cancer MDA-MB-231 cells were treated with Pro-EGCG (1), (-)-EGCG was not only converted but also accumulated, accompanied by enhanced levels of proteasome inhibition, growth suppression, and apoptosis induction, compared with cells treated with natural (-)-EGCG. To investigate the potential use of Pro-EGCG (1) as a novel prodrug that converts to a cellular proteasome inhibitor and anticancer agent in vivo, MDA-MB-231 tumors were induced in nude mice, followed by treatment with Pro-EGCG (1) or (-)-EGCG for 31 days. Results of this in vivo study showed a significant inhibition of breast tumor growth by Pro-EGCG (1), compared with (-)-EGCG, associated with increased proteasome inhibition and apoptosis induction in tumor tissues. In conclusion, we have shown that Pro-EGCG (1) increases the bioavailability, stability, and proteasome-inhibitory and anticancer activities of (-)-EGCG in human breast cancer cells and tumors, suggesting its potential use for cancer prevention and treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Catechin/analogs & derivatives , Peracetic Acid/pharmacology , Prodrugs/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Catechin/pharmacokinetics , Catechin/pharmacology , Cell Growth Processes/drug effects , Cell Line, Tumor , Chromatography, High Pressure Liquid , Chymotrypsin/antagonists & inhibitors , Chymotrypsin/metabolism , Female , Humans , Mice , Mice, Nude , Peracetic Acid/pharmacokinetics , Prodrugs/pharmacokinetics , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/pharmacology , Xenograft Model Antitumor Assays
10.
J Cell Physiol ; 212(2): 298-306, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17458898

ABSTRACT

Proteasome inhibitors are known to induce apoptosis in a variety of cancer cells. On the other hand, maspin, a non-inhibitory serine protease inhibitor, is shown to sensitize cancer cells to therapeutic agents that induce apoptosis. We examined the consequence of maspin expression in prostate cancer cells targeted for treatment with various proteasome inhibitors. We observed that proteasome inhibitors induced apoptosis more effectively in maspin transfected human prostate cancer DU145 cells than in control cells. Interestingly, increased apoptosis in these cells was associated with a significant induction of maspin expression. MG-132, a proteasome inhibitor, induced endogenous and ectopic [cytomegalovirus promoter (CMV)-driven] maspin expression, and maspin siRNA attenuated MG-132-induced apoptosis. Proteasome inhibitor-induced maspin expression was inhibited by actinomycin D (Act D) and cyclohexamide (CHX), and by the inhibitors of p38MAPK, but not ERK1/2 or NF-kappaB. Electrophoretic mobility-shift assay (EMSA) and promoter-reporter activity analyses suggested that p38MAPK activated transcription factor AP-1 is responsible for proteasome inhibitor-induced maspin expression. Taken together, these observations demonstrate that proteasome inhibitors induce maspin expression by activating p38MAPK pathway, and that maspin thus expressed, in turn, augments proteasome inhibitor-induced apoptosis in prostate cancer cells. Our results suggest that gene therapy involving ectopic maspin expression may dramatically improve the efficacy of proteasome inhibitors for the treatment of prostate cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Prostatic Neoplasms/drug therapy , Proteasome Inhibitors , Serpins/metabolism , Transcription, Genetic/drug effects , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cysteine Proteinase Inhibitors/therapeutic use , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Dose-Response Relationship, Drug , Genetic Vectors/drug effects , Humans , Imidazoles/pharmacology , Leupeptins/pharmacology , Luteolin/pharmacology , Male , Phosphorylation , Promoter Regions, Genetic/drug effects , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Serpins/genetics , Transcription Factor AP-1/metabolism , Transfection , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Tetrahedron ; 63(32): 7565-7570, 2007 Aug 06.
Article in English | MEDLINE | ID: mdl-21152270

ABSTRACT

The total and semi syntheses of (2R, 3R)-epigallocatechin-3-O-(4-hydroxybenzoate), a novel catechin from Cistus salvifolius, was accomplished. The proteasome inhibition and cytotoxic activities of the synthetic compound and its acetyl derivative were studied and compared with (2R, 3R)-epigallocatechin-3-gallate (EGCG), the active component from green tea.

12.
Breast Cancer Res ; 9(6): R80, 2007.
Article in English | MEDLINE | ID: mdl-18300387

ABSTRACT

INTRODUCTION: Proteasome inhibition is an attractive approach to anticancer therapy and may have relevancy in breast cancer treatment. Natural products, such as dietary flavonoids, have been suggested as natural proteasome inhibitors with potential use for cancer prevention and therapeutics. We previously reported that apigenin, a flavonoid widely distributed in many fruits and vegetables, can inhibit proteasome activity and can induce apoptosis in cultured leukemia Jurkat T cells. Whether apigenin has proteasome-inhibitory activity in the highly metastatic human breast MDA-MB-231 cells and xenografts,however, is unknown. METHODS: MDA-MB-231 breast cancer cell cultures and xenografts were treated with apigenin, followed by measurement of reduced cellular viability/proliferation,proteasome inhibition, and apoptosis induction. Inhibition of the proteasome was determined by levels of the proteasomal chymotrypsin-like activity, by ubiquitinated proteins, and by accumulation of proteasome target proteins in extracts of the treated cells or tumors. Apoptotic cell death was measured by caspase-3/caspase-7 activation, poly(ADP-ribose) polymerase cleavage, and immunohistochemistry for terminal nucleotidyltransferase-mediated nick end labeling positivity. RESULTS: We report for the first time that apigenin inhibits the proteasomal chymotrypsin-like activity and induces apoptosis not only in cultured MDA-MB-231 cells but also in MDA-MB-231 xenografts. Furthermore, while apigenin has antibreast tumor activity, no apparent toxicity to the tested animals was observed. CONCLUSION: We have shown that apigenin is an effective proteasome inhibitor in cultured breast cancer cells and in breast cancer xenografts. Furthermore, apigenin induces apoptotic cell death in human breast cancer cells and exhibits anticancer activities in tumors. The results suggest its potential benefits in breast cancer prevention and treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Apigenin/pharmacology , Breast Neoplasms/diet therapy , Breast Neoplasms/enzymology , Enzyme Inhibitors/pharmacology , Proteasome Inhibitors , Animals , Apoptosis/drug effects , Blotting, Western , Breast Neoplasms/physiopathology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Humans , In Situ Nick-End Labeling , Mice , Mice, Nude , Proteasome Endopeptidase Complex/metabolism , Random Allocation , Transplantation, Heterologous
13.
Cancer Res ; 66(21): 10478-86, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17079469

ABSTRACT

Although cisplatin has been used for decades to treat human cancer, some toxic side effects and resistance are observed. It has been suggested that gold(III) complexes, containing metal centers isoelectronic and isostructural to cisplatin, are promising anticancer drugs. Gold(III) dithiocarbamate complexes were shown to exhibit in vitro cytotoxicity, comparable with and even greater than cisplatin; however, the involved mechanism of action remained unknown. Because we previously reported that copper(II) dithiocarbamates are potent proteasome inhibitors, we hypothesized that gold(III) dithiocarbamate complexes could suppress tumor growth via direct inhibition of the proteasome activity. Here, for the first time, we report that a synthetic gold(III) dithiocarbamate (compound 2) potently inhibits the activity of a purified rabbit 20S proteasome and 26S proteasome in intact highly metastatic MDA-MB-231 breast cancer cells, resulting in the accumulation of ubiquitinated proteins and the proteasome target protein p27 and induction of apoptosis. The compound 2-mediated proteasome inhibition and apoptosis induction were completely blocked by addition of a reducing agent DTT or N-acetyl-L-cysteine, showing that process of oxidation is required for proteasome inhibition by compound 2. Treatment of MDA-MB-231 breast tumor-bearing nude mice with compound 2 resulted in significant inhibition of tumor growth, associated with proteasome inhibition and massive apoptosis induction in vivo. Our findings reveal the proteasome as a primary target for gold(III) dithiocarbamates and support the idea for their potential use as anticancer therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Organogold Compounds/pharmacology , Proteasome Inhibitors , Thiocarbamates/pharmacology , Acetylcysteine/pharmacology , Animals , Apoptosis/drug effects , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Mice , Neoplasm Transplantation , Proteasome Endopeptidase Complex , Transplantation, Heterologous
14.
Int J Mol Med ; 18(4): 625-32, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16964415

ABSTRACT

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.


Subject(s)
Flavonoids/metabolism , Phenols/metabolism , Proteasome Endopeptidase Complex/metabolism , Tea/chemistry , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/metabolism , Catechin/pharmacology , Chymotrypsin/antagonists & inhibitors , Chymotrypsin/metabolism , Dose-Response Relationship, Drug , Flavonoids/chemistry , Flavonoids/pharmacology , Methylation , Models, Molecular , Molecular Conformation , Phenols/chemistry , Phenols/pharmacology , Polyphenols , Proteasome Inhibitors , Protein Binding , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism
15.
Int J Mol Med ; 17(5): 931-5, 2006 May.
Article in English | MEDLINE | ID: mdl-16596283

ABSTRACT

Persistent but relatively limited research has been devoted to the use of compounds related to polycyclic aromatic hydrocarbons (PAH) as anticancer agents. In previous reports, we have described the cytotoxicity of a number of new and novel PAH against human cancer cell lines. However, the involved molecular mechanisms of inducing cell death were not elucidated. In the current study, we describe the apoptotic pathway as apparently playing a crucial role in induced cell death in human leukemia Jurkat T cells by several diamide and diamine PAH that contain chrysene as their core aromatic ring system. Structure-activity relationships were analyzed. Importantly, no effect was demonstrated in a normal, non-transformed line of human natural killer cells. These results provide additional evidence for the potential chemotherapeutic use of PAH.


Subject(s)
Apoptosis/drug effects , Polycyclic Aromatic Hydrocarbons/pharmacology , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Chrysenes/chemistry , Chrysenes/pharmacology , Enzyme Activation/drug effects , Flow Cytometry , Humans , In Situ Nick-End Labeling , Jurkat Cells , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Leukemia/metabolism , Leukemia/pathology , Molecular Structure , Piperazines/chemistry , Piperazines/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Polycyclic Aromatic Hydrocarbons/chemistry , Structure-Activity Relationship
16.
Drug Resist Updat ; 9(6): 263-73, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17197231

ABSTRACT

A major challenge in cancer therapy is tumor drug resistance. To overcome it, it is essential to understand the mechanisms and identify the molecules involved, so that they can be specifically targeted in combination therapies. The proteasome is such a validated target: it plays a key role in cancer cell proliferation, inhibition of chemotherapy-induced apoptosis and drug resistance development. Bortezomib (Velcade, PS-341) was the first proteasome inhibitor to receive regulatory approval from the US Food and Drug Administration for the treatment of multiple myeloma. Clinical combination trials have demonstrated a chemo-sensitizing effect of bortezomib on conventional agents in hematological malignancies and some solid tumors such as androgen-independent prostate and ovarian cancer. Although generally well-tolerated, bortezomib still generates toxicity which underscores the need for less toxic proteasome inhibitors. Several naturally occurring products, such as green tea polyphenols and the antibiotic lactacystin, have been shown to be potent proteasome inhibitors. Significantly, green tea polyphenols, as well as several flavonoids such as genistein, curcumin and resveratrol, have also been shown to have chemo-sensitizing properties in prostate, breast, hepatic, and lung tumors. Further studies on natural proteasome inhibitors as chemo-sensitizers could lead to identification of more potent and less toxic compounds that could be used in combination therapies for drug-resistant tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Proteasome Inhibitors , Pyrazines/pharmacology , Antineoplastic Agents/therapeutic use , Boronic Acids/adverse effects , Boronic Acids/therapeutic use , Bortezomib , Catechin/analogs & derivatives , Catechin/pharmacology , Catechin/therapeutic use , Clinical Trials as Topic , Curcumin/pharmacology , Curcumin/therapeutic use , Drug Therapy, Combination , Genistein/pharmacology , Genistein/therapeutic use , Humans , Proteasome Endopeptidase Complex/physiology , Pyrazines/adverse effects , Pyrazines/therapeutic use , Radiation-Sensitizing Agents/adverse effects , Radiation-Sensitizing Agents/pharmacology , Radiation-Sensitizing Agents/therapeutic use , Resveratrol , Stilbenes/pharmacology , Stilbenes/therapeutic use , Structure-Activity Relationship , Ubiquitin/physiology
17.
J Invest Dermatol ; 125(2): 207-12, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16098028

ABSTRACT

Proteasome inhibitors have emerged as a clinically important therapy for neoplastic disease, with velcade, an organoboron compound used extensively in multiple myeloma. Recently, (-)-epigallocatechin gallate has been found to be a potent inhibitor of the proteasomal chymotrypsin-like activity. Other compounds that inhibit angiogenesis and are active as chemopreventive agents, such as curcumin, also inhibit proteasome activity. We have screened natural product extracts using ras-transformed endothelial cells (SVR cells) as a bioassay, and found that extracts of mate tea (Ilex paraguayensis) inhibit the growth of these endothelial cells. The extract was fractionated and found to have novel cinnamate esters that inhibit proteasome activity. Based upon the structures of the compounds isolated from mate tea, we examined synthetic analogs of these compounds for proteasome activity. Cinnamic acid amides had no inhibitory activity against proteasomes, whereas cinnamate esters displayed the activity. Based upon these findings, preclinical and clinical trials of topical cinnamate esters as proteasome inhibitors are warranted for psoriasis and other inflammatory disorders.


Subject(s)
Cinnamates/chemistry , Endothelial Cells/drug effects , Ilex paraguariensis/chemistry , Plant Extracts/chemistry , Proteasome Inhibitors , Cell Division/drug effects , Cell Line, Transformed , Chlorogenic Acid/chemistry , Chlorogenic Acid/pharmacology , Chromatography, High Pressure Liquid , Cinnamates/pharmacology , Endothelial Cells/cytology , Esters/chemistry , Esters/pharmacology , G2 Phase/drug effects , Humans , Jurkat Cells , Nuclear Magnetic Resonance, Biomolecular , Plant Extracts/pharmacology
18.
Biochem Pharmacol ; 69(10): 1421-32, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15857606

ABSTRACT

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.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Flavonoids/pharmacology , Proteasome Inhibitors , Apigenin/pharmacology , Computer Simulation , Cysteine Proteinase Inhibitors , Diet , Humans , Jurkat Cells , Kaempferols/pharmacology , Killer Cells, Natural/drug effects , Quercetin/pharmacology , Structure-Activity Relationship
19.
Front Biosci ; 10: 1183-90, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15769617

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Apoptosis/drug effects , beta-Lactams/pharmacology , Anti-Bacterial Agents/chemistry , Humans , Structure-Activity Relationship , Tumor Cells, Cultured , beta-Lactams/chemistry
20.
Int J Mol Med ; 15(4): 735-42, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15754040

ABSTRACT

The anti-cancer and cancer-preventive effects of green tea and its main constituent (-)-epigallocatechin gallate [(-)-EGCG] are well documented by a variety of studies, including epidemiological, cell culture, animal, and clinical studies. While (-)-EGCG remains the most potent polyphenol in green tea, it is very unstable in neutral or alkaline conditions (i.e. physiologic pH). In an effort to discover more stable polyphenol proteasome inhibitors, we synthesized several novel (-)-EGCG analogs with -OH groups eliminated from the B- and/or D-rings. In addition, we also synthesized their putative prodrugs with -OH groups protected by peracetate that can be removed by cellular cytosolic esterases. We first examined the structure-activity relationship of these unprotected and protected compounds to their proteasome-inhibitory potentials. We found that decreasing -OH groups from either the B- or D-ring leads to diminished proteasome-inhibitory activity in vitro. However, in cultured tumor cells only the protected analogs were capable of potently inhibiting the proteasome activity. Furthermore, these protected analogs induced apoptotic cell death in a tumor cell-specific manner. The superior efficacy of the protected (-)-EGCG analogs indicates the formation of an entirely new compound(s) in intact tumor cells. These data suggest that the B-ring/D-ring peracetate-protected EGCG analogs have great potential to be developed into novel anti-cancer and cancer-preventive agents.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Catechin/analogs & derivatives , Catechin/pharmacology , Prodrugs/pharmacology , Proteasome Inhibitors , Flavonoids/pharmacology , Humans , Jurkat Cells , Phenols/pharmacology , Polyphenols , Tea/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...