Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Anticancer Res ; 31(1): 1-10, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21273574

ABSTRACT

AIMS: The molecular mechanisms of triptolide responsible for its antitumor properties are not yet fully understood. The ubiquitin/proteasome system is an important pathway of protein degradation in cells. This study investigated whether triptolide may inhibit proteasomal activity and induce apoptosis in human cancer cells. MATERIALS AND METHODS: In vitro proteasome inhibition was measured by incubation of a purified 20S proteasome with triptolide. Human breast and prostate cancer cell lines were also treated with different doses of triptolide for different times, followed by measurement of proteasome inhibition (levels of the chymotrypsin-like activity, ubiquitinated proteins and three well-known proteasome target proteins, p27, IκB-α and Bax) and apoptosis induction (caspase-3 activity and PARP cleavage). RESULTS: Triptolide did not inhibit the chymotrypsin-like activity of purified 20S proteasome. However, treatment of triptolide was able to cause decreased levels of cellular proteasomal chymotrypsin-like activity and accumulation of ubiquitinated proteins and three well-known proteasome target proteins in human breast and prostate cancer cells, associated with apoptosis induction. CONCLUSION: It is possible that at least one of metabolites of triptolide has proteasome-inhibitory activity.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Diterpenes/pharmacology , Phenanthrenes/pharmacology , Plants, Medicinal/chemistry , Prostatic Neoplasms/drug therapy , Proteasome Inhibitors , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 3/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Epoxy Compounds/pharmacology , Female , Humans , I-kappa B Proteins/metabolism , Male , NF-KappaB Inhibitor alpha , Poly(ADP-ribose) Polymerases/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tumor Cells, Cultured , Ubiquitin/metabolism
2.
Int J Mol Med ; 26(4): 447-55, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20818481

ABSTRACT

Curcumin (diferuloylmethane) is the main active ingredient of turmeric, a traditional herbal medicine and food of south Asia. Curcumin has been found to have a wide range of biological activities, including antioxidant, anti-inflammatory, chemopreventive and chemotherapeutic activities. Curcumin is currently being tested in clinical trials for treatment of various types of cancers, including multiple myeloma, pancreatic cancer and colon cancer. Although no toxicity associated with curcumin (even at very high doses) has been observed, the effects of curcumin in other solid tumors have been modest, primarily due to poor water solubility and poor bioavailability in tissues remote from the gastrointestinal tract. Therefore, there is a need for the discovery of curcumin analogs with better water solubility or greater bioavailability for the treatment of solid tumors such as prostate cancer. In this study, curcumin acetates and amino acid conjugates of curcumin were studied in terms of their proteasome inhibitory and antiproliferative effects against several human cancer cell lines. It was found that the water soluble amino acid conjugates of curcumin showed a potent antiproliferative effect and are potent proteasome inhibitors. Docking studies of the curcumin amino acid conjugates for proteasome inhibition were carried out to explain their biological activities. It is suggested that they may serve as the water soluble analogs of curcumin.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Curcumin/chemistry , Curcumin/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Acetates/chemistry , Acetates/pharmacology , Amino Acids/chemistry , Amino Acids/pharmacology , Cell Line, Tumor , Curcuma/chemistry , Humans , Models, Molecular , Neoplasms/drug therapy , Proteasome Endopeptidase Complex/chemistry , Protein Binding , Solubility
3.
Cancer Res ; 70(10): 3996-4004, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20424113

ABSTRACT

Frequent genetic alterations of the components in the phosphoinositide 3-kinase (PI3K)/PTEN/AKT signaling pathway contribute greatly to breast cancer initiation and progression, which makes targeting this signaling pathway a promising therapeutic strategy for breast cancer treatment. In this study, we showed that in the presence of copper (Cu), disulfiram (DSF), a clinically used antialcoholism drug, could potently inhibit breast cancer cell growth regardless of the PIK3CA status. Surprisingly, the treatment with a mixture of DSF and copper (DSF-Cu) led to the decreased expression of PTEN protein and the activation of AKT in a dose- and time-dependent manner in different cell lines with or without PIK3CA mutations. Treatment of breast cancer cell lines with a combination of DSF-Cu and LY294002, a pan-PI3K inhibitor, resulted in the significant inhibition of cell growth when compared with either drug alone. In addition, the combined treatment of DSF and LY294002 significantly inhibited the growth of the breast tumor xenograft in nude mice induced by MDA-MB-231 cells expressing mutant PIK3CA-H1047R and PIK3CA-E545K, whereas neither DSF nor LY294002 alone could significantly retard tumor growth. Finally, the observed in vivo inhibitory effects are found associated with aberrant signaling alterations and apoptosis-inducing activities in tumor samples. Thus, our finding shows for the first time that treatment of breast cancer with DSF results in a novel feedback mechanism that activates AKT signaling. Our study also suggests that the combination of DSF and a PI3K inhibitor may offer a new combinational treatment model for breast cancer, particularly for those with PIK3CA mutations.


Subject(s)
Breast Neoplasms/drug therapy , Disulfiram/pharmacology , Enzyme Inhibitors/pharmacology , Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Chromones/pharmacology , Class I Phosphatidylinositol 3-Kinases , Copper/pharmacology , Drug Therapy, Combination , Female , Humans , In Vitro Techniques , Mice , Mice, Nude , Morpholines/pharmacology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Xenograft Model Antitumor Assays
5.
Cancer Lett ; 292(1): 48-53, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-19962231

ABSTRACT

Epidemiological studies support the cancer-preventive effects of green tea and its main constituent (-)-epigallocatechin gallate [(-)-EGCG], however, (-)-EGCG is unstable under physiological conditions. Here we report that two novel fluoro-substituted (-)-EGCG analogs inhibited tumor growth with similar potency to that of Pro-EGCG (1) which has improved potency over parental compound (-)-EGCG in human breast cancer MDA-MB-231 xenografts. MDA-MB-231 tumors treated with each fluoro-substituted (-)-EGCG analog showed proteasome inhibition and apoptotic cell death, suggesting that the proteasome might be one of the cellular targets of fluoro-(-)-EGCGs and that proteasome inhibition is partially responsible for the observed antitumor activity.


Subject(s)
Antineoplastic Agents/therapeutic use , Catechin/analogs & derivatives , Fluorine , Mammary Neoplasms, Experimental/drug therapy , Proteasome Inhibitors , Animals , Apoptosis/drug effects , Catechin/chemistry , Catechin/pharmacology , Catechin/therapeutic use , Cell Line, Tumor , Female , Humans , Hydrocarbons, Fluorinated/therapeutic use , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Nude , Xenograft Model Antitumor Assays
6.
J Biol Inorg Chem ; 15(2): 259-69, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19809836

ABSTRACT

We have previously reported that when mixed with copper, 8-hydroxyquinoline (8-OHQ) and its analog clioquinol (CQ) inhibited the proteasomal activity and proliferation in cultured human cancer cells. CQ treatment of high-copper-containing human tumor xenografts also caused cancer suppression, associated with proteasome inhibition in vivo. However, the nature of the copper dependence of these events has not been elucidated experimentally. In the current study, using chemical probe molecules that mimic the structures of 8-OHQ and CQ, but have no copper-binding capability, we dissected the complex cellular processes elicited by 8-OHQ-Cu and CQ-Cu mixtures and revealed that copper binding to 8-OHQ or CQ is required for transportation of the copper complex into human breast cancer cells and the consequent proteasome-inhibitory, growth-suppressive, and apoptosis-inducing activities. In contrast, the non-copper-binding analogs of 8-OHQ or CQ blocked the very first step-copper binding-in this chain of events mediated by 8-OHQ-Cu or CQ-Cu.


Subject(s)
Antineoplastic Agents/pharmacology , Clioquinol/pharmacology , Copper/metabolism , Neoplasms/pathology , Oxyquinoline/pharmacology , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Biological Transport/drug effects , Cell Proliferation/drug effects , Clioquinol/metabolism , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Neoplasms/drug therapy , Neoplasms/enzymology , Organometallic Compounds/chemical synthesis , Organometallic Compounds/chemistry , Organometallic Compounds/pharmacology , Oxyquinoline/metabolism , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Structure-Activity Relationship , Tumor Cells, Cultured
7.
Inorg Chem ; 48(13): 5928-37, 2009 Jul 06.
Article in English | MEDLINE | ID: mdl-19496541

ABSTRACT

In this study, we compare the proteasome inhibition capabilities of two anticancer candidates, [Ni(L(IA))(2)] (1) and [Zn(L(IA))(2)] (2), where L(IA-) is the deprotonated form of the ligand 2,4-diiodo-6-(((2-pyridinylmethyl)amino)methyl)phenol. Species 1 contains nickel(II), a considerably inert ion that favors covalency, whereas 2 contains zinc(II), a labile transition metal ion that favors predominantly ionic bonds. We report on the synthesis and characterization of 1 and 2 using various spectroscopic, spectrometric, and structural methods. Furthermore, the pharmacological effects of 1 and 2, along with those of the salts NiCl(2) and ZnCl(2), were evaluated in vitro and in cultured human cancer cells in terms of their proteasome-inhibitory and apoptotic cell-death-inducing capabilities. It is shown that neither NiCl(2) nor 1 have the ability to inhibit the proteasome activity at any sustained levels. However, ZnCl(2) and 2 showed superior inhibitory activity versus the chymotrypsin-like activity of both the 26S proteasome (IC(50) = 5.7 and 4.4 micromol/L, respectively) and the purified 20S proteasome (IC(50) = 16.6 and 11.7 micromol/L, respectively) under cell-free conditions. Additionally, inhibition of proteasomal activity in cultured prostate cancer cells by 2 was associated with higher levels of ubiquitinated proteins and apoptosis. Treatment with either the metal complex or the salt was relatively nontoxic toward human normal cells. These results strengthen the current working hypothesis that fast ligand dissociation is required to generate an [ML(IA)](+) pharmacophore, capable of interaction with the proteasome. This interaction, possibly via N-terminal threonine amino acids present in the active sites, renders the proteasome inactive. Our results present a compelling rationale for 2 along with its gallium(III) and copper(II) congeners to be further investigated as potential anticancer drugs that act as proteasome inhibitiors.


Subject(s)
Nickel/chemistry , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Zinc/chemistry , Cell Line, Tumor , Cell Proliferation , Humans , Ligands , Magnetic Resonance Spectroscopy , Male , Models, Molecular , Molecular Structure , Prostatic Neoplasms/pathology , Protease Inhibitors/chemistry , Proteasome Endopeptidase Complex , Spectrometry, Mass, Electrospray Ionization , Ubiquitination
8.
Pharm Res ; 26(8): 1874-80, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19421843

ABSTRACT

PURPOSE: Emerging evidence clearly suggests the potential chemopreventive and anti-tumor activity of a well known "natural agent" curcumin. However, studies have shown that curcumin is not readily bioavailable, and thus the tissue bioavailability of curcumin is also poor except for gastrointestinal track. Because of the potential biological activity of curcumin, many studies have attempted for making a better analog of curcumin that is equally effective or better with increased bioavailability, which was the purpose of our current study. METHODS: We have designed and synthesized new difluoro Knoevenagel condensates of curcumin and Schiff bases along with their copper (II) complexes and evaluated their biological activities with respect to the inhibitory effects on purified rabbit 26S proteasome, and growth inhibition and induction of apoptosis in colon and pancreatic cancer cell lines. RESULTS: All copper complexes possess distorted square planar geometries with 1:1 metal to ligand stoichiometry with reversible copper redox couple. The difluoro compound CDF exhibited inhibitory effects on purified rabbit 20S proteasome or cellular 26S proteasome, and caused both growth inhibition of cancer cell lines and induced apoptotic cell death in our preliminary assessment. CONCLUSION: Our results suggest that our newly synthesized classes of curcumin analogs could be useful as chemopreventive and/or therapeutic agents against cancers.


Subject(s)
Apoptosis/drug effects , Copper/chemistry , Curcumin/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Proteasome Inhibitors , Schiff Bases/chemistry , Animals , Cell Line, Tumor , Curcumin/chemistry , Humans , Magnetic Resonance Spectroscopy , Rabbits
9.
Environ Health Perspect ; 117(3): 379-86, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19337512

ABSTRACT

BACKGROUND: Because of the vital importance of the proteasome pathway, chemicals affecting proteasome activity could disrupt essential cellular processes. Although the toxicity of organotins to both invertebrates and vertebrates is well known, the essential cellular target of organotins has not been well identified. We hypothesize that the proteasome is a molecular target of environmental toxic organotins. OBJECTIVES: Our goal was to test the above hypothesis by investigating whether organotins could inhibit the activity of purified and cellular proteasomes and, if so, the involved molecular mechanisms and downstream events. RESULTS: We found that some toxic organotins [e.g., triphenyltin (TPT)] can potently and preferentially inhibit the chymotrypsin-like activity of purified 20S proteasomes and human breast cancer cellular 26S proteasomes. Direct binding of tin atoms to cellular proteasomes is responsible for the observed irreversible inhibition. Inhibition of cellular proteasomes by TPT in several human cell lines results in the accumulation of ubiquitinated proteins and natural proteasome target proteins, accompanied by induction of cell death. CONCLUSIONS: The proteasome is one of the molecular targets of environmental toxic organotins in human cells, and proteasome inhibition by organotins contributes to their cellular toxicity.


Subject(s)
Environmental Pollutants/toxicity , Organotin Compounds/toxicity , Proteasome Endopeptidase Complex/drug effects , Blotting, Western , Cell Death/drug effects , Cell Line, Tumor , Humans , In Situ Nick-End Labeling , Jurkat Cells , Mass Spectrometry , Organotin Compounds/metabolism , Proteasome Endopeptidase Complex/metabolism
10.
Int J Cancer ; 124(10): 2450-9, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19165859

ABSTRACT

Dysregulation of the ubiquitin-proteasome pathway plays an essential role in tumor growth and development. Shikonin, a natural naphthoquinone isolated from the traditional Chinese medicine Zi Cao (gromwell), has been reported to possess tumor cell-killing activity, and results from a clinical study using a shikonin-containing mixture demonstrated its safety and efficacy for the treatment of late-stage lung cancer. In this study, we reported that shikonin is an inhibitor of tumor proteasome activity in vitro and in vivo. Our computational modeling predicts that the carbonyl carbons C(1) and C(4) of shikonin potentially interact with the catalytic site of beta 5 chymotryptic subunit of the proteasome. Indeed, shikonin potently inhibits the chymotrypsin-like activity of purified 20S proteasome (IC(50) 12.5 micromol/L) and tumor cellular 26S proteasome (IC(50) between 2-16 micromol/L). Inhibition of the proteasome by shikonin in murine hepatoma H22, leukemia P388 and human prostate cancer PC-3 cultures resulted in accumulation of ubiquitinated proteins and several proteasome target proapoptotic proteins (I kappaB-alpha, Bax and p27), followed by induction of cell death. Shikonin treatment resulted in tumor growth inhibition in both H22 allografts and PC-3 xenografts, associated with suppression of the proteasomal activity and induction of cell death in vivo. Finally, shikonin treatment significantly prolonged the survival period of mice bearing P388 leukemia. Our results indicate that the tumor proteasome is one of the cellular targets of shikonin and inhibition of the proteasome activity by shikonin contributes to its antitumor property.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Death/drug effects , Naphthoquinones/pharmacology , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Flow Cytometry , Humans , Male , Mice , Mice, Nude , Models, Molecular , Naphthoquinones/chemistry , Neoplasms, Experimental/pathology , Survival Rate
11.
Cancer Lett ; 268(1): 63-9, 2008 Sep 08.
Article in English | MEDLINE | ID: mdl-18468785

ABSTRACT

An ongoing strategy for cancer treatment is selective induction of apoptosis in cancer over normal cells. N-thiolated beta-lactams were found to induce DNA damage, growth arrest and apoptosis in cultured human cancer cells. However, whether these compounds have a similar effect in vivo has not been studied. We report here that treatment with the beta-lactam L-1 caused a significant inhibition of tumor growth in a breast cancer xenograft mouse model, associated with induction of DNA damage and apoptosis in vivo. These results suggest that the synthetic antibiotic N-thiolated beta-lactams hold great potential to be developed as novel anti-cancer drugs.


Subject(s)
Breast Neoplasms/drug therapy , Monobactams/therapeutic use , Animals , Anti-Bacterial Agents/pharmacology , Antigens, Differentiation/metabolism , Apoptosis , DNA Damage , Female , HSP70 Heat-Shock Proteins/metabolism , Humans , Jurkat Cells , Mice , Mice, Nude , Monobactams/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
12.
Int J Mol Sci ; 9(6): 951-961, 2008 Jun.
Article in English | MEDLINE | ID: mdl-19325839

ABSTRACT

The most potent catechin in green tea is (-)-epigallocatechin-3-gallate [(-)-EGCG], which, however, is unstable under physiological conditions. To discover more stable and more potent polyphenol proteasome inhibitors, we synthesized several novel fluoro-substituted (-)-EGCG analogs, named F-EGCG analogs, as well as their prodrug forms with all of -OH groups protected by acetate. We report that the prodrug form of one F-EGCG analog exhibited greater potency than the previously reported peracetate of (-)-EGCG to inhibit proteasomal activity, suppress cell proliferation, and induce apoptosis in human leukemia Jurkat T cells, demonstrating the potential of these compounds to be developed into novel anti-cancer and cancer-preventive agents.

13.
Int J Mol Med ; 20(6): 919-25, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17982703

ABSTRACT

Apoptosis has a central role in the pathogenesis of many human diseases, one of which is cancer. One of the most important strategies to regulate apoptosis is via the ubiquitin-proteasome pathway. It has been shown that inhibition of proteasomal chymotrypsin-like activity is a strong apoptosis-inducing stimulus and that actively proliferating cancer cells are more sensitive to proteasome inhibitors than normal or untransformed cells. Dithioscarbamates are a class of metal-chelating compounds with various applications in medicine. We reported previously that certain members of dithiocarbamates, such as pyrrolidine dithiocarbamate (PDTC), diethyldithiocarbamate and disulfiram, are able to bind with tumor cellular copper, forming an active complex with proteasome-inhibitory, apoptosis-inducing and anti-cancer activities. In the current study, we synthesized eight PDTC analogues with substitutions made to the pyrrolidine ring and studied their structure-activity relationships. We found that substitution of the pyrrolidine ring with piperidine had almost no effect on their proteasome-inhibitory and anti-proliferative potencies in human breast cancer cells. However, after the pyrrolidine ring was substituted with morpholine, the activity of the mixtures slightly decreased but was completely lost when piperazine with the attached ethyl group was used for the substitution. This structure-activity relationship was confirmed by the results generated with the corresponding copper complexes. Our data further support the novel concept of using accumulated copper in human cancer cells as a selective approach for chemotherapy.


Subject(s)
Apoptosis/physiology , Copper/metabolism , Proteasome Endopeptidase Complex , Pyrrolidines/chemistry , Thiocarbamates/chemistry , Animals , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Molecular Structure , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Pyrrolidines/metabolism , Pyrrolidines/pharmacology , Structure-Activity Relationship , Thiocarbamates/metabolism , Thiocarbamates/pharmacology
14.
Cancer Res ; 67(19): 9258-65, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17909033

ABSTRACT

The investigation of metal-based complexes with potential antitumor activity has been of paramount importance in recent years due to the successful use of cisplatin against various cancers. Gallium(III) and subsequently developed gallium(III)-containing complexes have shown promising antineoplastic effects when tested in a host of malignancies, specifically in lymphomas and bladder cancer. However, the molecular mechanism responsible for their anticancer effect is yet to be fully understood. We report here for the first time that the proteasome is a molecular target for gallium complexes in a variety of prostate cancer cell lines and in human prostate cancer xenografts. We tested five gallium complexes (1-5) in which the gallium ion is bound to an NN'O asymmetrical ligand containing pyridine and substituted phenolate moieties in a 1:2 (M/L) ratio. We found that complex 5 showed superior proteasome inhibitory activity against both 26S proteasome (IC50, 17 micromol/L) and purified 20S (IC50, 16 micromol/L) proteasome. Consistently, this effect was associated with apoptosis induction in prostate cancer cells. Additionally, complex 5 was able to exert the same effect in vivo by inhibiting growth of PC-3 xenografts in mice (66%), which was associated with proteasome inhibition and apoptosis induction. Our results strongly suggest that gallium complexes, acting as potent proteasome inhibitors, have a great potential to be developed into novel anticancer drugs.


Subject(s)
Gallium/pharmacology , Organometallic Compounds/pharmacology , Prostatic Neoplasms/drug therapy , Proteasome Inhibitors , Androgens/physiology , Animals , Apoptosis/drug effects , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Biomimetic Materials/pharmacology , Cell Line, Tumor , Chymotrypsin/antagonists & inhibitors , Chymotrypsin/chemistry , Chymotrypsin/metabolism , Gallium/chemistry , Gallium/metabolism , Humans , Male , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/enzymology , Organometallic Compounds/chemistry , Organometallic Compounds/metabolism , Prostatic Neoplasms/enzymology , Proteasome Endopeptidase Complex/metabolism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
15.
Int J Mol Med ; 19(5): 809-16, 2007 May.
Article in English | MEDLINE | ID: mdl-17390087

ABSTRACT

Diethyldithiocarbamate (DDTC) is a member of the dithiocarbamate family and a potent copper-chelating agent. DDTC was used in a clinical trial for patients with HIV-1 infection and showed a significant delay in progression to AIDS. In this study, we investigated the effects of DDTC-copper complex in human prostate and breast cancer cells. We found that DDTC was capable of binding copper and forming a new complex that potently inhibited the proteasomal chemotrypsin-like activity, decreased expression of androgen receptor (AR), estrogen receptor (ER) alpha and ERbeta proteins, and induced apoptosis in both prostate and breast cancer cells. Our data support the concept of using accumulated copper in cancer cells and tissues as a novel target for chemotherapy. This study provides a mechanistic interpretation for utilization of copper chelators in cancer treatment.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Apoptosis/drug effects , Breast Neoplasms/pathology , Copper/metabolism , Ditiocarb/pharmacology , Prostatic Neoplasms/pathology , Proteasome Inhibitors , Cell Line, Tumor , Cell Nucleus/metabolism , Disease Progression , Ditiocarb/chemistry , Female , Humans , Male , Solutions
16.
Cancer Res ; 67(4): 1636-44, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17308104

ABSTRACT

Tumor growth and metastasis depend on angiogenesis that requires the cofactor copper. Consistently, high levels of copper have been found in many types of human cancers, including prostate, breast, colon, and lung. Recent studies suggest that copper could be used as a novel selective target for cancer therapies. Clioquinol is capable of forming stable complexes with copper and currently used in clinics for treatment of Alzheimer's disease. Most recently, it has been reported that clioquinol possesses antitumor effects. However, the underlying molecular mechanism is unclear. We report here that after binding to copper, clioquinol can inhibit the proteasomal chymotrypsin-like activity, repress androgen receptor (AR) protein expression, and induce apoptotic cell death in human prostate cancer LNCaP and C4-2B cells. In addition, clioquinol alone exhibits similar effects in prostate cancer cell lines with elevated copper at concentrations similar to those found in patients. Addition of dihydrotestosterone did not affect clioquinol-mediated proteasome inhibition in both prostate cancer cell lines. However, dihydrotestosterone partially inhibited clioquinol-induced AR suppression and apoptosis only in androgen-dependent LNCaP cells. Animal studies show that clioquinol treatment significantly inhibits the growth of human prostate tumor C4-2B xenografts (by 66%), associated with in vivo proteasome inhibition, AR protein repression, angiogenesis suppression, and apoptosis induction. Our study provides strong evidence that clioquinol is able to target tumor proteasome in vivo in a copper-dependent manner, resulting in formation of an active AR inhibitor and apoptosis inducer that is responsible for its observed antiprostate tumor effect.


Subject(s)
Androgen Receptor Antagonists , Apoptosis/drug effects , Clioquinol/pharmacology , Copper/metabolism , Prostatic Neoplasms/drug therapy , Protease Inhibitors/pharmacology , Animals , Cell Line, Tumor , Clioquinol/metabolism , Humans , Male , Mice , Mice, Nude , Neoplasms, Hormone-Dependent/blood supply , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protease Inhibitors/metabolism , Proteasome Inhibitors , Xenograft Model Antitumor Assays
17.
Front Biosci ; 12: 1935-45, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17127432

ABSTRACT

Diet high in vegetables and fruits has been associated with reduced cancer risk. However, the involved mechanisms are unknown. Previously, we reported that the dietary flavonoid apigenin could inhibit the proteasome activity and induce apoptosis in tumor cells. To further investigate the structure-proteasome-inhibitory activity relationships, we chose and tested five dietary flavonoids, including luteolin, apigenin, chrysin, naringenin and eriodictyol. We found that the order of inhibitory potencies and apoptosis-inducing potencies of these five compounds in 20S purified proteasome and tumor cells was: (1) luteolin > apigenin > chrysin, and (2) apigenin >> naringenin, and luteolin >> eriodictyol. Therefore, flavonoids with hydroxylized B ring and/or unsaturated C ring are natural potent proteasome inhibitors and tumor cell apoptosis inducers. Furthermore, neither apigenin nor luteolin could inhibit the proteasome and induce apoptosis in non-transformed human natural killer cells. This finding may provide a molecular basis for the clinically observed cancer-preventive effects of fruits and vegetables.


Subject(s)
Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/pharmacology , Flavonoids/chemistry , Flavonoids/pharmacology , Proteasome Inhibitors , Apigenin/pharmacology , Apoptosis , Caspase 3/metabolism , Cell Line, Tumor , Diet , Humans , Jurkat Cells , Killer Cells, Natural/enzymology , Luteolin/pharmacology , Male , Prostatic Neoplasms/enzymology , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Structure-Activity Relationship
18.
Cancer Res ; 66(21): 10425-33, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17079463

ABSTRACT

Disulfiram (DSF), a member of the dithiocarbamate family capable of binding copper and an inhibitor of aldehyde dehydrogenase, is currently being used clinically for the treatment of alcoholism. Recent studies have suggested that DSF may have antitumor and chemosensitizing activities, although the detailed molecular mechanisms remain unclear. Copper has been shown to be essential for tumor angiogenesis processes. Consistently, high serum and tissue levels of copper have been found in many types of human cancers, including breast, prostate, and brain, supporting the idea that copper could be used as a potential tumor-specific target. Here we report that the DSF-copper complex potently inhibits the proteasomal activity in cultured breast cancer MDA-MB-231 and MCF10DCIS.com cells, but not normal, immortalized MCF-10A cells, before induction of apoptotic cancer cell death. Furthermore, MDA-MB-231 cells that contain copper at concentrations similar to those found in patients, when treated with just DSF, undergo proteasome inhibition and apoptosis. In addition, when administered to mice bearing MDA-MB-231 tumor xenografts, DSF significantly inhibited the tumor growth (by 74%), associated with in vivo proteasome inhibition (as measured by decreased levels of tumor tissue proteasome activity and accumulation of ubiquitinated proteins and natural proteasome substrates p27 and Bax) and apoptosis induction (as shown by caspase activation and apoptotic nuclei formation). Our study shows that inhibition of the proteasomal activity can be achieved by targeting tumor cellular copper with the nontoxic compound DSF, resulting in selective apoptosis induction within tumor cells.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Copper/metabolism , Disulfiram/pharmacology , Proteasome Inhibitors , Animals , Breast Neoplasms/pathology , Chymotrypsin/antagonists & inhibitors , Female , Humans , Mice , Neoplasm Transplantation , Transplantation, Heterologous
19.
Cancer Res ; 66(9): 4758-65, 2006 May 01.
Article in English | MEDLINE | ID: mdl-16651429

ABSTRACT

Interest in the use of traditional medicines for cancer prevention and treatment is increasing. In vitro, in vivo, and clinical studies suggest the potential use of proteasome inhibitors as novel anticancer drugs. Celastrol, an active compound extracted from the root bark of the Chinese medicine "Thunder of God Vine" (Tripterygium wilfordii Hook F.), was used for years as a natural remedy for inflammatory conditions. Although Celastrol has been shown to induce leukemia cell apoptosis, the molecular target involved has not been identified. Furthermore, whether Celastrol has antitumor activity in vivo has never been conclusively shown. Here, we report, for the first time, that Celastrol potently and preferentially inhibits the chymotrypsin-like activity of a purified 20S proteasome (IC(50) = 2.5 micromol/L) and human prostate cancer cellular 26S proteasome (at 1-5 micromol/L). Inhibition of the proteasome activity by Celastrol in PC-3 (androgen receptor- or AR-negative) or LNCaP (AR-positive) cells results in the accumulation of ubiquitinated proteins and three natural proteasome substrates (IkappaB-alpha, Bax, and p27), accompanied by suppression of AR protein expression (in LNCaP cells) and induction of apoptosis. Treatment of PC-3 tumor-bearing nude mice with Celastrol (1-3 mg/kg/d, i.p., 1-31 days) resulted in significant inhibition (65-93%) of the tumor growth. Multiple assays using the animal tumor tissue samples from both early and end time points showed in vivo inhibition of the proteasomal activity and induction of apoptosis after Celastrol treatment. Our results show that Celastrol is a natural proteasome inhibitor that has a great potential for cancer prevention and treatment.


Subject(s)
Prostatic Neoplasms/drug therapy , Proteasome Inhibitors , Triterpenes/pharmacology , Androgen Receptor Antagonists , Animals , Apoptosis/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Chymotrypsin/antagonists & inhibitors , Chymotrypsin/metabolism , Diterpenes/pharmacology , Diterpenes, Kaurane , Humans , Male , Mice , Mice, Nude , Pentacyclic Triterpenes , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Proteasome Endopeptidase Complex/metabolism , Rabbits , Receptors, Androgen/biosynthesis , Tripterygium/chemistry , Xenograft Model Antitumor Assays
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...