Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
ACS Med Chem Lett ; 14(10): 1351-1357, 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37849557

ABSTRACT

KRAS G12D mutation has been found in approximately 45% of pancreatic ductal adenocarcinoma (PDAC) cases, making it an attractive therapeutic target. Through structure-based drug design, a series of potent and selective KRAS G12D inhibitors were designed. The lead compound, ERAS-5024, inhibited ERK1/2 phosphorylation and cell proliferation in three-dimensional Cell-Titer Glo assays in AsPC-1 PDAC cells with single-digit nanomolar potency and caused tumor regression in the in vivo efficacy studies. We describe here the details of the design and synthesis program that led to the discovery of ERAS-5024.

2.
Cancer Res ; 75(10): 2049-60, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25795708

ABSTRACT

Retinoid X receptor alpha (RXRα) and its N-terminally truncated version, tRXRα, are widely implicated in cancer development and represent intriguing targets for cancer prevention and treatment. Successful manipulation of RXRα and tRXRα requires the identification of their modulators that could produce therapeutic effects. Here, we report that a class of nitrostyrene derivatives bind to RXRα by a unique mechanism, of which the nitro group of nitrostyrene derivatives and Cys432 of RXRα are required for binding. The binding results in the potent activation of Gal4-DBD-RXRα-LBD transactivation. However, the binding inhibits the transactivation of RXRα homodimer, which might be due to the distinct conformation of RXRα homodimer induced by these nitrostyrene derivatives. Two RXRα point mutants with Cys432 substituted with Tyr and Trp, respectively, could mimic the bindings of two nitrostyrene derivatives and have the ability of autotransactivation. In studying the functional consequences of the binding, we show that these nitrostyrene derivatives could potently inhibit the TNFα/NFκB signaling pathway in a tRXRα-dependent manner. tRXRα promotes TNFα-induced NF-κB activation through its interaction with TRAF2 and enhances TNFα-induced ubiquitination of RIP1, which is strongly inhibited by nitrostyrene derivatives. The inhibition of TNFα-induced NF-κB activation results in the synergistic effect of the combination of nitrostyrene derivatives and TNFα on the induction of cancer cell apoptosis. Together, our results show a new class of RXRα modulators that induce apoptosis of cancer cells through their unique binding mode and new mechanism of action.


Subject(s)
Anthracenes/pharmacology , Antineoplastic Agents/pharmacology , NF-kappa B/metabolism , Naphthalenes/pharmacology , Retinoid X Receptor alpha/metabolism , Styrenes/pharmacology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis , HEK293 Cells , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Nude , NF-kappa B/genetics , Protein Binding , Retinoid X Receptor alpha/agonists , Signal Transduction , TNF Receptor-Associated Factor 2/metabolism , Transcriptional Activation , Ubiquitination , Xenograft Model Antitumor Assays
3.
Carcinogenesis ; 34(6): 1208-15, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23389291

ABSTRACT

We recently reported that an N-terminally truncated retinoid X receptor-α (tRXRα) produced in cancer cells acts to promote cancer cell growth and survival through AKT activation. However, how RXRα is cleaved and how the cleavage is regulated in cancer cells remain undefined. In this study, we demonstrated that calpain II could cleave RXRα protein in vitro, generating two truncated RXRα products. The cleavage sites in RXRα were mapped by Edman N-terminal sequencing to Gly(90)↓Ser(91) and Lys(118)↓Val(119). Transfection of the resulting cleavage product RXRα/90, but not RXRα/118, resulted in activation of AKT in cancer cells, similar to the effect of tRXRα. In support of the role of calpain II in cancer cells, transfection of calpain II expression vector or its activation by ionomycin enhanced the production of tRXRα, whereas treatment of cells with calpain inhibitors reduced the levels of tRXRα. Co-immunoprecipitation assays also showed an interaction between calpain II and RXRα. In studying the regulation of tRXRα production, we observed that treatment of cells with lithium chloride or knockdown of glycogen synthase kinase-3ß (GSK-3ß) significantly increased the production of tRXRα. Conversely, overexpression of GSK-3ß reduced tRXRα expression. Furthermore, we found that the inhibitory effect of GSK-3ß on tRXRα production was due to its suppression of calpain II expression. Taken together, our data demonstrate that GSK-3ß plays an important role in regulating tRXRα production by calpain II in cancer cells, providing new insights into the development of new strategies and agents for the prevention and treatment of tRXRα-related cancers.


Subject(s)
Calpain/metabolism , Glycogen Synthase Kinase 3/metabolism , Retinoid X Receptor alpha/metabolism , Amino Acid Sequence , Animals , Calcium Ionophores/pharmacology , Calpain/antagonists & inhibitors , Cell Line, Tumor , Down-Regulation , Enzyme Activation , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Hep G2 Cells , Humans , Ionomycin/pharmacology , Lithium Chloride/pharmacology , MCF-7 Cells , Mice , Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering , Sequence Analysis, Protein , Signal Transduction
4.
PLoS One ; 7(4): e35722, 2012.
Article in English | MEDLINE | ID: mdl-22545132

ABSTRACT

BACKGROUND: Retinoid X receptor-alpha (RXRα) is a key member of the nuclear receptor superfamily. We recently demonstrated that proteolytic cleavage of RXRα resulted in production of a truncated product, tRXRα, which promotes cancer cell survival by activating phosphatidylinositol-3-OH kinase (PI3K)/AKT pathway. However, how the tRXRα-mediated signaling pathway in cancer cells is regulated remains elusive. METHODOLOGY/PRINCIPAL FINDINGS: We screened a natural product library for tRXRα targeting leads and identified that triptolide, an active component isolated from traditional Chinese herb Trypterygium wilfordii Hook F, could modulate tRXRα-mediated cancer cell survival pathway in vitro and in animals. Our results reveal that triptolide strongly induces cancer cell apoptosis dependent on intracellular tRXRα expression levels, demonstrating that tRXRα serves as an important intracellular target of triptolide. We show that triptolide selectively induces tRXRα degradation and inhibits tRXRα-dependent AKT activity without affecting the full-length RXRα. Interestingly, such effects of triptolide are due to its activation of p38. Although triptolide also activates Erk1/2 and MAPK pathways, the effects of triptolide on tRXRα degradation and AKT activity are only reversed by p38 siRNA and p38 inhibitor. In addition, the p38 inhibitor potently inhibits tRXRα interaction with p85α leading to AKT inactivation. Our results demonstrate an interesting novel signaling interplay between p38 and AKT through tRXRα mediation. We finally show that targeting tRXRα by triptolide strongly activates TNFα death signaling and enhances the anticancer activity of other chemotherapies. CONCLUSIONS/SIGNIFICANCE: Our results identify triptolide as a new xenobiotic regulator of the tRXRα-dependent survival pathway and provide new insight into the mechanism by which triptolide acts to induce apoptosis of cancer cells. Triptolide represents one of the most promising therapeutic leads of natural products of traditional Chinese medicine with unfortunate side-effects. Our findings will offer new strategies to develop improved triptolide analogs for cancer therapy.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Diterpenes/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Neoplasms/drug therapy , Phenanthrenes/therapeutic use , Proto-Oncogene Proteins c-akt/metabolism , Retinoid X Receptor alpha/metabolism , Animals , Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line , Cell Line, Tumor , Diterpenes/pharmacology , Drugs, Chinese Herbal/pharmacology , Enzyme Activation/drug effects , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/metabolism , Phenanthrenes/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Retinoid X Receptor alpha/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism
5.
FASEB J ; 25(1): 192-205, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20847229

ABSTRACT

The orphan nuclear receptor Nur77 is an immediate-early response gene whose expression is rapidly induced by various extracellular stimuli. The aims of this study were to study the role of Nur77 expression in the growth and survival of colon cancer cells and the mechanism by which Nur77 expression was regulated. We showed that levels of Nur77 were elevated in a majority of human colon tumors (9/12) compared to their nontumorous tissues and that Nur77 expression could be strongly induced by different colonic carcinogens including deoxycholic acid (DCA). DCA-induced Nur77 expression resulted in up-regulation of antiapoptotic BRE and angiogenic VEGF, and it enhanced the growth, colony formation, and migration of colon cancer cells. In studying the mechanism by which Nur77 was regulated in colon cancer cells, we found that ß-catenin was involved in induction of Nur77 expression through its activation of the transcriptional activity of AP-1 (c-Fos/c-Jun) that bound to and transactivated the Nur77 promoter. Together, our results demonstrate that Nur77 acts to promote the growth and survival of colon cancer cells and serves as an important mediator of the Wnt/ß-catenin and AP-1 signaling pathways.


Subject(s)
Cell Proliferation , Colonic Neoplasms/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , beta Catenin/metabolism , Animals , Blotting, Western , Cell Survival , Colon/drug effects , Colon/metabolism , Colon/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Deoxycholic Acid/toxicity , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Immunohistochemistry , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transcription Factor AP-1/metabolism , beta Catenin/genetics
6.
Cancer Cell ; 17(6): 560-73, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20541701

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAIDs) exert their anticancer effects through cyclooxygenase-2 (COX-2)-dependent and independent mechanisms. Here, we report that Sulindac, an NSAID, induces apoptosis by binding to retinoid X receptor-alpha (RXRalpha). We identified an N-terminally truncated RXRalpha (tRXRalpha) in several cancer cell lines and primary tumors, which interacted with the p85alpha subunit of phosphatidylinositol-3-OH kinase (PI3K). Tumor necrosis factor-alpha (TNFalpha) promoted tRXRalpha interaction with the p85alpha, activating PI3K/AKT signaling. When combined with TNFalpha, Sulindac inhibited TNFalpha-induced tRXRalpha/p85alpha interaction, leading to activation of the death receptor-mediated apoptotic pathway. We designed and synthesized a Sulindac analog K-80003, which has increased affinity to RXRalpha but lacks COX inhibitory activity. K-80003 displayed enhanced efficacy in inhibiting tRXRalpha-dependent AKT activation and tRXRalpha tumor growth in animals.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Retinoid X Receptor alpha/antagonists & inhibitors , Signal Transduction/drug effects , Sulindac/analogs & derivatives , Sulindac/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cytoplasm/metabolism , Dinoprostone/metabolism , Drug Design , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Molecular , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Prostaglandin-Endoperoxide Synthases/drug effects , Prostaglandin-Endoperoxide Synthases/metabolism , Protein Binding/drug effects , Protein Binding/physiology , Protein Processing, Post-Translational/physiology , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Sequence Deletion/physiology , Signal Transduction/physiology , Sulindac/metabolism , Sulindac/therapeutic use , Transcriptional Activation/drug effects , Transfection , Tretinoin/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
7.
Cancer Res ; 68(21): 8871-80, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18974131

ABSTRACT

Shikonin derivatives, which are the active components of the medicinal plant Lithospermum erythrorhizon, exhibit many biological effects including apoptosis induction through undefined mechanisms. We recently discovered that orphan nuclear receptor Nur77 migrates from the nucleus to the mitochondria, where it binds to Bcl-2 to induce apoptosis. Here, we report that certain shikonin derivatives could modulate the Nur77/Bcl-2 apoptotic pathway by increasing levels of Nur77 protein and promoting its mitochondrial targeting in cancer cells. Structural modification of acetylshikonin resulted in the identification of a derivative 5,8-diacetoxyl-6-(1'-acetoxyl-4'-methyl-3'-pentenyl)-1,4-naphthaquinones (SK07) that exhibited improved efficacy and specificity in activating the pathway. Unlike other Nur77 modulators, shikonins increased the levels of Nur77 protein through their posttranscriptional regulation. The apoptotic effect of SK07 was impaired in Nur77 knockout cells and suppressed by cotreatment with leptomycin B that inhibited Nur77 cytoplasmic localization. Furthermore, SK07 induced apoptosis in cells expressing the COOH-terminal half of Nur77 protein but not its NH(2)-terminal region. Our data also showed that SK07-induced apoptosis was associated with a Bcl-2 conformational change and Bax activation. Together, our results show that certain shikonin derivatives act as modulators of the Nur77-mediated apoptotic pathway and identify a new shikonin-based lead that targets Nur77 for apoptosis induction.


Subject(s)
Anthraquinones/pharmacology , Apoptosis/drug effects , DNA-Binding Proteins/physiology , Receptors, Steroid/physiology , Anthraquinones/chemistry , Apoptosis/physiology , Base Sequence , Blotting, Western , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA Primers , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Humans , Microscopy, Fluorescence , Mitochondria/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1 , Protein Transport , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Processing, Post-Transcriptional/drug effects , Receptors, Steroid/drug effects , Receptors, Steroid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , bcl-2-Associated X Protein/metabolism
8.
Cancer Cell ; 14(4): 285-98, 2008 Oct 07.
Article in English | MEDLINE | ID: mdl-18835031

ABSTRACT

Bcl-2 can be converted into a proapoptotic molecule by nuclear receptor Nur77. However, the development of Bcl-2 converters as anticancer therapeutics has not been explored. Here we report the identification of a Nur77-derived Bcl-2-converting peptide with 9 amino acids (NuBCP-9) and its enantiomer, which induce apoptosis of cancer cells in vitro and in animals. The apoptotic effect of NuBCPs and their activation of Bax are not inhibited but rather potentiated by Bcl-2. NuBCP-9 and its enantiomer bind to the Bcl-2 loop, which shares the characteristics of structurally adaptable regions with many cancer-associated and signaling proteins. NuBCP-9s act as molecular switches to dislodge the Bcl-2 BH4 domain, exposing its BH3 domain, which in turn blocks the activity of antiapoptotic Bcl-X(L).


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , DNA-Binding Proteins/metabolism , Neoplasms, Experimental/drug therapy , Oligopeptides/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Steroid/metabolism , Animals , Antineoplastic Agents/chemistry , BH3 Interacting Domain Death Agonist Protein/metabolism , Binding Sites , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Female , HeLa Cells , Humans , Jurkat Cells , Mice , Mice, Knockout , Mice, SCID , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1 , Oligopeptides/chemistry , Peptide Fragments/pharmacology , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Proteins/pharmacology , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Steroid/genetics , Stereoisomerism , Time Factors , Transfection , Xenograft Model Antitumor Assays , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism
9.
Mol Cell Biol ; 24(22): 9705-25, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15509776

ABSTRACT

Retinoid X receptor (RXR) plays a central role in the regulation of intracellular receptor signaling pathways by acting as a ubiquitous heterodimerization partner of many nuclear receptors, including the orphan receptor Nur77 (also known as TR3 [corrected] or NGFI-B), which translocates from the nucleus to mitochondria, where it interacts with Bcl-2 to induce apoptosis. Here, we report that RXRalpha is required for nuclear export and mitochondrial targeting of Nur77 through their unique heterodimerization that is mediated by dimerization interfaces located in their DNA-binding domain. The effects of RXRalpha are attributed to a putative nuclear export sequence (NES) present in its carboxyl-terminal region. RXRalpha ligands suppress NES activity by inducing RXRalpha homodimerization or altering RXRalpha/Nur77 heterodimerization. The RXRalpha NES is also silenced by RXRalpha heterodimerization with retinoic acid receptor or vitamin D receptor. Consistently, we were able to show that the mitochondrial targeting of the RXRalpha/Nur77 heterodimer and its induction of apoptosis are potently inhibited by RXR ligands. Together, our results reveal a novel nongenotropic function of RXRalpha and its involvement in the regulation of the Nur77-dependent apoptotic pathway [corrected]


Subject(s)
Apoptosis/physiology , DNA-Binding Proteins/metabolism , Retinoid X Receptor alpha/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Apoptosis/drug effects , Base Sequence , Cell Line , DNA/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dimerization , Humans , Mitochondria/metabolism , Molecular Sequence Data , Mutation , Nuclear Receptor Subfamily 4, Group A, Member 1 , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , Receptors, Cytoplasmic and Nuclear , Receptors, Steroid , Receptors, Thyroid Hormone/chemistry , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoid X Receptor alpha/chemistry , Retinoid X Receptor alpha/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transcription Factors/chemistry , Transcription Factors/genetics
10.
Cell ; 116(4): 527-40, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14980220

ABSTRACT

The Bcl-2 family proteins are key regulators of apoptosis in human diseases and cancers. Though known to block apoptosis, Bcl-2 promotes cell death through an undefined mechanism. Here, we show that Bcl-2 interacts with orphan nuclear receptor Nur77 (also known as TR3), which is required for cancer cell apoptosis induced by many antineoplastic agents. The interaction is mediated by the N-terminal loop region of Bcl-2 and is required for Nur77 mitochondrial localization and apoptosis. Nur77 binding induces a Bcl-2 conformational change that exposes its BH3 domain, resulting in conversion of Bcl-2 from a protector to a killer. These findings establish the coupling of Nur77 nuclear receptor with the Bcl-2 apoptotic machinery and demonstrate that Bcl-2 can manifest opposing phenotypes, induced by interactions with proteins such as Nur77, suggesting novel strategies for regulating apoptosis in cancer and other diseases.


Subject(s)
DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription Factors/metabolism , Apoptosis , Cell Death , Cell Line , Cytochromes c/metabolism , Glutathione Transferase/metabolism , Green Fluorescent Proteins , Humans , Ligands , Luminescent Proteins/metabolism , Lymphocytes/metabolism , Microscopy, Fluorescence , Mitochondria/metabolism , Mutation , Nuclear Receptor Subfamily 4, Group A, Member 1 , Oligonucleotides, Antisense/pharmacology , Phenotype , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear , Receptors, Steroid , Transfection , Two-Hybrid System Techniques
11.
Mol Cell Biol ; 23(23): 8651-67, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14612408

ABSTRACT

TR3, also known as NGFI-B or nur77, is an immediate-early response gene and an orphan member of the steroid/thyroid/retinoid receptor superfamily. We previously reported that TR3 expression was induced by apoptotic stimuli and was required for their apoptotic effect in lung cancer cells. Here, we present evidence that TR3 was also induced by epidermal growth factor (EGF) and serum and was required for their mitogenic effect in lung cancer cells. Ectopic expression of TR3 in both H460 and Calu-6 lung cancer cell lines promoted their cell cycle progression and BrdU incorporation, while inhibition of TR3 expression by the small interfering RNA approach suppressed the mitogenic effect of EGF and serum. Analysis of TR3 mutants showed that both TR3 DNA binding and transactivation were required for its mitogenic effect. In contrast, they were dispensable for its apoptotic activity. Furthermore, confocal microscopy analysis demonstrated that TR3 functioned in the nucleus to induce cell proliferation, whereas it acted on mitochondria to induce apoptosis. In examining the signaling that regulates the mitogenic function of TR3, we observed that coexpression of constitutive-active MEKK1 inhibited TR3 transcriptional activity and TR3-induced proliferation. The inhibitory effect of MEKK1 was mediated through activation of Jun N-terminal kinase, which efficiently phosphorylated TR3, resulting in loss of its DNA binding. Together, our results demonstrate that TR3 is capable of inducing both proliferation and apoptosis in the same cells depending on the stimuli and its cellular localization.


Subject(s)
Lung Neoplasms/metabolism , MAP Kinase Kinase Kinase 1 , Mitosis/physiology , Protein Serine-Threonine Kinases/metabolism , Receptors, Steroid/metabolism , Receptors, Thyroid Hormone/metabolism , Apoptosis , Base Sequence , Cell Line, Tumor , Culture Media , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Epidermal Growth Factor/pharmacology , Gene Expression/drug effects , Humans , JNK Mitogen-Activated Protein Kinases , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mitogen-Activated Protein Kinases/metabolism , Mitosis/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1 , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Receptors, Steroid/genetics , Receptors, Thyroid Hormone/genetics , Subcellular Fractions/metabolism , Transcriptional Activation
12.
Int J Cancer ; 99(2): 171-8, 2002 May 10.
Article in English | MEDLINE | ID: mdl-11979430

ABSTRACT

Epidemiological and animal studies have demonstrated that vitamin A and its natural and synthetic derivatives, retinoids, are effective agents in preventing the development of tobacco-associated cancers. Unfortunately, clinical trials of retinoids on cigarette smokers have shown lack of efficacy in preventing lung cancer. In our study, we investigated the effect of nicotine on the anti-cancer activity of all trans-retinoic acid (trans-RA) in human lung cancer cells. Our results demonstrated that nicotine could abrogate the growth inhibitory effect of trans-RA by suppressing its ability to induce the expression of RA receptor beta (RAR beta), a tumor suppressor. The inhibitory effect of nicotine was accompanied with induction of orphan receptor TR3. Inhibition of TR3 expression by overexpression of TR3 anti-sense RNA in H460 lung cancer cells strongly prevented the suppressive effect of nicotine on trans-RA activity. Treatment with nicotine or the cotransfection of TR3 expression vector inhibited the induction of RAR beta promoter activity by trans-RA in transient transfection assays. The inhibition of RAR beta promoter activity was due to the interaction of TR3 with orphan receptor COUP-TF, resulting in inhibition of COUP-TF DNA binding and transactivation on the RAR beta promoter. Furthermore, we found that nicotine failed to suppress the effect of a retinoid X receptor (RXR)-selective retinoid SR11237 on inducing both growth inhibition and RAR beta promoter activity, due to the ability of SR11237 to activate the RAR beta promoter through the RXR/TR3 heterodimer. Together, our results demonstrate that nicotine suppresses the growth inhibitory effects of trans-RA by inhibiting RAR beta expression through its induction of TR3 expression and suggest that RXR-selective retinoids may be more effective than classical retinoids for preventing and treating tobacco-associated cancers.


Subject(s)
Cell Division/drug effects , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Nicotine/pharmacology , Receptors, Retinoic Acid/genetics , Receptors, Steroid , Retinoids/pharmacology , Benzoates/pharmacology , Blotting, Northern , COUP Transcription Factors , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Drug Interactions , Gene Expression/drug effects , Humans , Nuclear Receptor Subfamily 4, Group A, Member 1 , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear , Receptors, Retinoic Acid/drug effects , Retinoid X Receptors , Retinoids/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Transcription Factors/drug effects , Transcription Factors/genetics , Transfection , Tretinoin/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL