Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Eur J Med Chem ; 226: 113795, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34597896

ABSTRACT

P-glycoprotein (P-gp; ABCB1)-mediated drug efflux causes multidrug resistance in cancer. Previous synthetic methylated epigallocatechin (EGC) possessed promising P-gp modulating activity. In order to further improve the potency, we have synthesized some novel stereoisomers of methylated epigallocatechin (EGC) and gallocatechin (GC) as well as epicatechin (EC) and catechin (C). The (2R, 3S)-trans-methylated C derivative 25 and the (2R, 3R)-cis-methylated EC derivative 31, both containing dimethyoxylation at ring B, tri-methoxylation at ring D and oxycarbonylphenylcarbamoyl linker between ring D and C3, are the most potent in reversing P-gp mediated drug resistance with EC50 ranged from 32 nM to 93 nM. They are non-toxic to fibroblast with IC50 > 100 µM. They can inhibit the P-gp mediated drug efflux and restore the intracellular drug concentration to a cytotoxic level. They do not downregulate surface P-gp protein level to enhance drug retention. They are specific for P-gp with no or low modulating activity towards MRP1- or BCRP-mediated drug resistance. In summary, methylated C 25 and EC 31 derivatives represent a new class of potent, specific and non-toxic P-gp modulator.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Catechin/analogs & derivatives , Catechin/pharmacology , Doxorubicin/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Catechin/chemical synthesis , Catechin/chemistry , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Humans , Methylation , Molecular Structure , Stereoisomerism , Structure-Activity Relationship
2.
Acta Pharmacol Sin ; 42(3): 460-469, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32647340

ABSTRACT

Sphingosine-1-phosphate (S1P), the backbone of most sphingolipids, activating S1P receptors (S1PRs) and the downstream G protein signaling has been implicated in chemoresistance. In this study we investigated the role of S1PR2 internalization in 5-fluorouracil (5-FU) resistance in human colorectal cancer (CRC). Clinical data of randomly selected 60 CRC specimens showed the correlation between S1PR2 internalization and increased intracellular uracil (P < 0.001). Then we explored the regulatory mechanisms in CRC model of villin-S1PR2-/- mice and CRC cell lines. We showed that co-administration of S1P promoted S1PR2 internalization from plasma membrane (PM) to endoplasmic reticulum (ER), thus blunted 5-FU efficacy against colorectal tumors in WT mice, compared to that in S1PR2-/- mice. In HCT116 and HT-29 cells, application of S1P (10 µM) empowered S1PR2 to internalize from PM to ER, thus inducing 5-FU resistance, whereas the specific S1PR2 inhibitor JTE-013 (10 µM) effectively inhibited S1P-induced S1PR2 internalization. Using Mag-Fluo-AM-labeling [Ca2+]ER and LC-ESI-MS/MS, we revealed that internalized S1PR2 triggered elevating [Ca2+]ER levels to activate PERK-eLF2α-ATF4 signaling in HCT116 cells. The activated ATF4 upregulated RNASET2-mediated uracil generation, which impaired exogenous 5-FU uptake to blunt 5-FU therapy. Overall, this study reveals a previously unrecognized mechanism of 5-FU resistance resulted from S1PR2 internalization-upregulated uracil generation in colorectal cancer, and provides the novel insight into the significance of S1PR2 localization in predicting the benefit of CRC patients from 5-FU-based chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Fluorouracil/therapeutic use , Lysophospholipids/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Sphingosine/analogs & derivatives , Uracil/metabolism , Activating Transcription Factor 4/metabolism , Animals , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Endoplasmic Reticulum/metabolism , Female , HCT116 Cells , Humans , Male , Mice, Inbred C57BL , Middle Aged , Ribonucleases/metabolism , Signal Transduction/physiology , Sphingosine/metabolism , Tumor Suppressor Proteins/metabolism
4.
Oncogene ; 39(29): 5214-5227, 2020 07.
Article in English | MEDLINE | ID: mdl-32546724

ABSTRACT

Aberrant sphingolipid metabolism has been implicated in chemoresistance, but the underlying mechanisms are still poorly understood. Herein we revealed a previously unrecognized mechanism of 5-fluorouracil (5-FU) resistance contributed by high SphK2-upregulated dihydropyrimidine dehydrogenase (DPD) in colorectal cancer (CRC), which is evidenced from human CRC specimens, animal models, and cancer cell lines. TMA samples from randomly selected 60 CRC specimens firstly identified the clinical correlation between high SphK2 and increased DPD (p < 0.001). Then the regulatory mechanism was explored in CRC models of villin-SphK2 Tg mice, SphK2-/-mice, and human CRC cells xenografted nude mice. Assays of ChIP-Seq and luciferase reporter gene demonstrated that high SphK2 upregulated DPD through promoting the HDAC1-mediated H3K56ac, leading to the degradation of intracellular 5-FU into inactive α-fluoro-ß-alanine (FBAL). Lastly, inhibition of SphK2 by SLR080811 exhibited excellent inhibition on DPD expression and potently reversed 5-FU resistance in colorectal tumors of villin-SphK2 Tg mice. Overall, this study manifests that SphK2high conferred 5-FU resistance through upregulating tumoral DPD, which highlights the strategies of blocking SphK2 to overcome 5-FU resistance in CRC.


Subject(s)
Colorectal Neoplasms/genetics , Dihydrouracil Dehydrogenase (NADP)/metabolism , Drug Resistance, Neoplasm/genetics , Fluorouracil/therapeutic use , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Animals , Colorectal Neoplasms/pathology , Fluorouracil/pharmacology , Humans , Mice , Up-Regulation
5.
ACS Chem Neurosci ; 11(11): 1634-1642, 2020 06 03.
Article in English | MEDLINE | ID: mdl-32374999

ABSTRACT

Neurodegenerative diseases are irreversible conditions that result in progressive degeneration and death of nerve cells. Although the underlying mechanisms may vary, oxidative stress is considered to be one of the major causes of neuronal loss. Importantly, there are still no comprehensive treatments to completely cure these diseases. Therefore, protecting neurons from oxidative damage may be the most effective therapeutic strategy. Here we report a neuroprotective effects of a novel hybrid compound (dlx-23), obtained by conjugating α-lipoic acid (ALA), a natural antioxidant agent, and 3-n-butylphthalide (NBP), a clinical anti-ischemic drug. Dlx-23 protected against neuronal death induced by both H2O2 induced oxidative stress in Cath.-a-differentiated (CAD) cells and 6-OHDA, a toxin model of Parkinson's disease (PD) in SH-SY5Y cells. These activities proved to be more potent than the parent compound (ALA) alone. Dlx-23 scavenged free radicals, increased glutathione levels, and prevented mitochondria damage. In addition, live imaging of primary cortical neurons demonstrated that dlx-23 protected against neuronal growth cone damage induced by H2O2. Taken together these results suggest that dlx-23 has substantial potential to be further developed into a novel neuroprotective agent against oxidative damage and toxin induced neurodegeneration.


Subject(s)
Neuroprotective Agents , Thioctic Acid , Benzofurans , Cell Line, Tumor , Hydrogen Peroxide/toxicity , Neurons , Neuroprotective Agents/pharmacology , Oxidative Stress , Oxidopamine/toxicity , Reactive Oxygen Species , Thioctic Acid/pharmacology
6.
Pharmacol Res ; 155: 104717, 2020 05.
Article in English | MEDLINE | ID: mdl-32088343

ABSTRACT

In this study, S1PR2 was reckoned as a brand-new GPCR target for designing inhibitors to reverse 5-FU resistance. Herein a series of pyrrolidine pyrazoles as the S1PR2 inhibitors were designed, synthesized and evaluated for their activities of anti-FU-resistance. Among them, the most promising compound JTE-013, exhibited excellent inhibition on DPD expression and potent anti-FU-resistance activity in various human cancer cell lines, along with the in vivo HCT116DPD cells xenograft model, in which the inhibition rate of 5-FU was greatly increased from 13.01%-75.87%. The underlying mechanism was uncovered that JTE-013 demonstrated an anti-FU-resistance activity by blocking S1PR2 internalization to the endoplasmic reticulum (ER), which inhibited the degradation of 5-FU into α-fluoro-ß-alanine (FBAL) by downregulating tumoral DPD expression. Overall, JTE-013 could serve as the lead compound for the discovery of new anti-FU-resistance drugs. SIGNIFICANCE: This study provides novel insights that S1PR2 inhibitors could sensitize 5-FU therapy in colorectal cancer.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Fluorouracil/therapeutic use , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Sphingosine-1-Phosphate Receptors/antagonists & inhibitors , Animals , Cell Line, Tumor , Dihydrouracil Dehydrogenase (NADP)/genetics , Down-Regulation/drug effects , Humans , Mice, Nude , Molecular Docking Simulation , Pyrazoles/pharmacology , Pyridines/pharmacology , Sphingosine-1-Phosphate Receptors/metabolism
7.
RSC Adv ; 8(52): 29548-29554, 2018 Aug 20.
Article in English | MEDLINE | ID: mdl-35547324

ABSTRACT

A simple and efficient protocol was developed for the syntheses of oridonin analogues, i.e. 6,20-epoxy ent-kaurane diterpenoid analogues from oridonin via diethylaminosulfur trifluoride (DAST) promoted rearrangement, most of which exhibited superior anticancer activities compared with their precursor.

8.
Eur J Med Chem ; 125: 795-806, 2017 Jan 05.
Article in English | MEDLINE | ID: mdl-27750197

ABSTRACT

In the present study, a total of 25 novel ningalin B analogues were synthesized and evaluated for their P-gp modulating activity in a P-gp overexpressed breast cancer cell line LCC6MDR. Preliminary structure-activity study shows that A ring and its two methoxy groups are important pharmacophores for P-gp inhibiting activity. Among all derivatives, 23 is the most potent P-gp modulator with EC50 of 120-165 nM in reversing paclitaxel, DOX, vinblastine and vincristine resistance. It is relatively safe to use with selective index at least greater than 606 compared to verapamil. Mechanistic study demonstrates that compound 23 reverses P-gp mediated drug resistance by inhibiting transport activity of P-gp, thereby restoring intracellular drug accumulation. In summary, our study demonstrates that ningalin B analogue 23 is a non-cytotoxic and effective P-gp chemosensitizer that can be used in the future for reversing P-gp mediated clinical cancer drug resistance.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Heterocyclic Compounds, 3-Ring/chemistry , ATP Binding Cassette Transporter, Subfamily B/drug effects , ATP-Binding Cassette Transporters/drug effects , Aquatic Organisms/chemistry , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Structure-Activity Relationship
9.
Bioorg Med Chem ; 23(17): 5566-73, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26233798

ABSTRACT

In the present study, a total of 9 novel permethyl ningalin B analogs have been synthesized and evaluated for their P-gp modulating activity in a P-gp overexpressed breast cancer cell line LCC6MDR. Among these derivatives, compound 12 with dimethoxy groups at rings A and B and tri-substitution at ring C with ortho-methoxyethylmorpholine, meta-bromo and para-benzyloxy groups displays the most potent P-gp modulating activity with EC50 of 423 nM to reverse paclitaxel resistance. It is non-toxic towards L929 fibroblast with IC50 greater than 100 µM and with selective index greater than 236. Its mechanism to reverse P-gp mediated drug resistance is by virtue of inhibiting transport activity of P-gp, restoring intracellular drug accumulation and eventually chemosensitizing the cancer cells to anticancer drug again. Moreover, compound 12 showed better solubility (405 ng/mL) than hit compound 1 in phosphate buffer (pH 4.0). In summary, our study demonstrates that permethyl ningalin B derivative 12 is non-toxic and efficient P-gp inhibitor that is a potential candidate to be used clinically to reverse P-gp mediated cancer drug resistance.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Heterocyclic Compounds, 3-Ring/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Cell Line, Tumor , Drug Resistance, Multiple , Humans , Molecular Structure , Structure-Activity Relationship
10.
J Med Chem ; 58(11): 4529-49, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25985195

ABSTRACT

We are interested in developing novel natural product-derived P-gp inhibitors to reverse cancer drug resistance. Here, we have synthesized 55 novel derivatives of methylated epigallocatechin (EGC), gallocatechin (GC), and dihydromyricetin (DHM). Three EGC derivatives (23, 35, and 36) and three GC derivatives (50, 51, and 53) are significantly better than epigallocatechin gallate (EGCG) with a relative fold (RF) ranging from 31.4 to 53.6. The effective concentration (EC50) of 23 and 51 ranges from 102 to 195 nM. Compounds 23 and 51 are noncytotoxic to fibroblasts with IC50 > 100 µM. Compound 23 is specific for P-gp without modulating activity toward MRP1 or BCRP. Compounds 23 and 51 are non-P-gp substrates. Important pharmacophores for P-gp modulation were identified. In summary, methylated EGC and GC derivatives represent a new class of potent, specific, noncytotoxic, and nonsubstrate P-gp modulators.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Agents/pharmacology , Catechin/analogs & derivatives , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Flavonols/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents/chemistry , Catechin/chemistry , Catechin/pharmacology , Cell Proliferation/drug effects , Flavonols/chemistry , Humans , Methylation , Models, Molecular , Molecular Structure , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Structure-Activity Relationship , Tumor Cells, Cultured
11.
Mar Drugs ; 12(10): 5209-21, 2014 Oct 17.
Article in English | MEDLINE | ID: mdl-25329704

ABSTRACT

In this study, new marine ningalin B analogues containing a piperazine or a benzoloxy group at ring C have been synthesized and evaluated on their P-gp modulating activity in human breast cancer and leukemia cell lines. Their structure-activity relationship was preliminarily studied. Compounds 19 and 20 are potent P-gp inhibitors. These two synthetic analogues of permethyl ningalin B may be potentially used as effective modulators of P-gp-mediated drug resistance in cancer cells.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Biological Products/chemistry , Biological Products/pharmacology , Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Humans , K562 Cells , Leukemia/drug therapy , Piperazine , Piperazines/chemistry , Piperazines/pharmacology , Structure-Activity Relationship
12.
Shanghai Kou Qiang Yi Xue ; 21(3): 283-6, 2012 Jun.
Article in Chinese | MEDLINE | ID: mdl-22885488

ABSTRACT

PURPOSE: To explore the application value of the cervical lymphatic imaging in interstitial magnetic resonance lymphography using submucosal injection of Dextran-DTPA-Gd. METHODS: 0.2 mL Dextran-DTPA-Gd (3.96 mmol/L) was injected into the submucosa of the bilateral lingual margins in 12 New Zealand rabbits,and then massaged the injection site for 30 seconds. MR images were obtained before injection and 10, 15, 20, 25, 30, 35, 40, 50, 90 minutes after injection by 3D TOF CE-MRA sequence.The signal intensities of cervical lymph node were measured, the enhancing rates(E%) were calculated and the signal enhancing rates -time curve was drawn. The data was analysed using SPSS11.5 software package. RESULTS: The cervical lymph nodes,the first and second lymphatics were strengthened significantly after injecting Dextran-DTPA-Gd, but the blood vessels were not enhanced at the same time. The enhancing rates of cervical lymph node reached the peak(344%) at 30-min,and the best strengthening effect was achieved between 20-min and 50-min. CONCLUSIONS: As IMRLG contrast agent,the Dextran-DTPA-Gd could image lymphatic drainage lines of the neck and the cervical lymph nodes efficiently.


Subject(s)
Gadolinium DTPA , Lymphography , Animals , Contrast Media , Dextrans , Lymph Nodes , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Neck , Rabbits
13.
Eur J Med Chem ; 54: 413-22, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22743241

ABSTRACT

Three methylated quercetins and a series of O-3 substituted 5,7,3',4'-tetra-O-methylated quercetin derivatives have been synthesized and evaluated on the modulating activity of P-gp, BCRP and MRP1 in cancer cell lines. Compound 17 (with a 2-((4-methoxybenzoyl)oxy)ethyl at O-3) is the most potent P-gp modulator. Three derivatives, compound 9 (3,7,3',4'-tetra-O-methylated quercetin), compound 14 (with a 2-((3-oxo-3-(3,4,5trimethoxyphenyl)prop-1-en-1-yl)oxy)ethyl at O-3) and compound 17, consistently exhibited promising BCRP-modulating activity. Interestingly, compound 17 was found to be equipotent against both P-gp and BCRP. Importantly, these synthetic quercetin derivatives did not exhibit any inherent cytotoxicity to cancer cell lines or normal mouse fibroblast cell lines. These quercetin derivatives can be employed as safe and effective modulators of P-gp- or BCRP-mediated drug resistance in cancer.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP-Binding Cassette Transporters/metabolism , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Drug Resistance, Multiple/drug effects , Neoplasm Proteins/metabolism , Quercetin/chemical synthesis , Quercetin/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Chemistry Techniques, Synthetic , Humans , Methylation , Mice , Multidrug Resistance-Associated Proteins/metabolism , Quercetin/chemistry
14.
Int J Mol Med ; 29(1): 102-6, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22012631

ABSTRACT

The proteasome plays a vital role in the degradation of proteins involved in several pathways including the cell cycle, cellular proliferation and apoptosis and is a validated target in cancer treatment. Bortezomib (Velcade®, PS-341) is the first US FDA approved proteasome inhibitor anticancer drug used in the treatment of refractory multiple myeloma. In spite of its improved efficacy compared to alternative therapies, about 60% of patients do not respond to bortezomib due to the emergence of resistance. We hypothesized that novel small molecules could enhance the proteasome-inhibitory and anticancer activities of bortezomib in resistant multiple myeloma cells in vitro and in vivo. The dietary polyphenol curcumin has been shown to exert anti-cancer activity in several cancer cell lines, but the effects of curcumin in solid tumors have been modest primarily due to poor water solubility and poor bioavailability in tissues remote from the gastrointestinal tract. Here we show that the water-soluble analog of curcumin #12, but not curcumin, in combination with bortezomib could enhance the proteasome-inhibitory effect in multiple myeloma cells. Furthermore, the sensitivity of the myeloma cells to cytotoxic killing in the presence of otherwise sublethal concentrations of bortezomib was enhanced by incubation with the curcumin analog #12. These findings justify further investigation into those combinations that may yield potential therapeutic benefit.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Boronic Acids/pharmacology , Curcumin/analogs & derivatives , Multiple Myeloma/drug therapy , Proteasome Inhibitors , Pyrazines/pharmacology , Apoptosis/drug effects , Boronic Acids/administration & dosage , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/administration & dosage , Curcumin/pharmacology , Drug Synergism , Humans , Immunoblotting , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Proteasome Endopeptidase Complex/metabolism , Pyrazines/administration & dosage
15.
Adv Clin Chem ; 53: 155-77, 2011.
Article in English | MEDLINE | ID: mdl-21404918

ABSTRACT

Cancer-preventive effects of tea polyphenols, especially epigallocatechin-3-gallate (EGCG), have been demonstrated by epidemiological, preclinical, and clinical studies. Green tea polyphenols such as EGCG have the potential to affect multiple biological pathways, including gene expression, growth factor-mediated pathways, the mitogen-activated protein kinase-dependent pathway, and the ubiquitin/proteasome degradation pathway. Therefore, identification of the molecular targets of EGCG should greatly facilitate a better understanding of the mechanisms underlying its anticancer and cancer-preventive activities. Performing structure-activity relationship (SAR) studies could also greatly enhance the discovery of novel tea polyphenol analogs as potential anticancer and cancer-preventive agents. In this chapter, we review the relevant literature as it relates to the effects of natural and synthetic green tea polyphenols and EGCG analogs on human cancer cells and their potential molecular targets as well as their antitumor effects. We also discuss the implications of green tea polyphenols in cancer prevention.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Catechin/analogs & derivatives , Flavonoids/therapeutic use , Neoplasms/drug therapy , Neoplasms/prevention & control , Phenols/therapeutic use , Prodrugs/pharmacology , Tea/chemistry , Anticarcinogenic Agents/chemistry , Catechin/chemistry , Catechin/therapeutic use , DNA Damage , DNA Methylation , Flavonoids/chemistry , Humans , Phenols/chemistry , Polyphenols , Prodrugs/chemistry , Proteasome Inhibitors , Structure-Activity Relationship
16.
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
17.
J Med Chem ; 53(14): 5108-20, 2010 Jul 22.
Article in English | MEDLINE | ID: mdl-20560605

ABSTRACT

A series of novel N-arylalkyl-3,4-diaryl-substituted pyrrole-2,5-diones were synthesized. They exhibited promising P-gp modulating activity in a P-gp overexpressing breast cancer cell line (LCC6MDR). Compound 6 (with three methoxy groups at D-ring) displayed the highest P-gp modulating activity. 6 at 1 microM can sensitize LCC6MDR cells toward paclitaxel by 18.2-fold. Interestingly, a synergy on modulating P-gp was noted when 6 and 25 were used together (fractional inhibitory concentration index FICI = 0.42). Combination of 6 (0.5 microM) and 25 (0.5 microM) resulted in a 66-fold sensitization of LCC6MDR cells toward paclitaxel. They also reversed P-gp mediated doxorubicin (DOX) and vincristine resistance. Kinetic characterization suggests that permethyl ningalin B analogues likely act as a noncompetitive inhibitor of P-gp-mediated DOX transport (K(i) = 5.4-5.8 microM). The present study demonstrates that synthetic analogues of permethyl ningalin B can be employed as effective and safe modulators of P-gp-mediated drug resistance in cancer cells.


Subject(s)
Antineoplastic Agents/chemical synthesis , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Heterocyclic Compounds, 3-Ring/chemical synthesis , Pyrroles/chemical synthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Design , Heterocyclic Compounds, 3-Ring/chemistry , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Paclitaxel/pharmacology , Pyrroles/chemistry , Pyrroles/pharmacology , Stereoisomerism , Structure-Activity Relationship , Vincristine/pharmacology
18.
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
19.
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.

20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...