Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters











Publication year range
1.
J Med Chem ; 67(13): 10795-10830, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38913996

ABSTRACT

Clinical and biological studies have shown that overexpression of BFL-1 is one contributing factor to venetoclax resistance. The resistance might be overcome by a potent BFL-1 inhibitor, but such an inhibitor is rare. In this study, we show that 56, featuring an acrylamide moiety, inhibited the BFL-1/BID interaction with a Ki value of 105 nM. More interestingly, 56 formed an irreversible conjugation adduct at the C55 residue of BFL-1. 56 was a selective BFL-1 inhibitor, and its MCL-1 binding affinity was 10-fold weaker, while it did not bind BCL-2 and BCL-xL. Mechanistic studies showed that 56 overcame venetoclax resistance in isogenic AML cell lines MOLM-13-OE and MV4-11-OE, which both overexpressed BFL-1. More importantly, 56 and venetoclax combination promoted stronger apoptosis induction than either single agent. Collectively, our data show that 56 overcame resistance to venetoclax in AML cells overexpressing BFL-1. These attributes make 56 a promising candidate for future optimization.


Subject(s)
Antineoplastic Agents , Bridged Bicyclo Compounds, Heterocyclic , Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute , Proto-Oncogene Proteins c-bcl-2 , Sulfonamides , Humans , Sulfonamides/pharmacology , Sulfonamides/chemistry , Sulfonamides/chemical synthesis , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Drug Resistance, Neoplasm/drug effects , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cell Line, Tumor , Minor Histocompatibility Antigens/metabolism , Apoptosis/drug effects , Drug Discovery , Structure-Activity Relationship
2.
Eur J Med Chem ; 270: 116366, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38581730

ABSTRACT

Inhibition of MDM2/p53 interaction with small-molecule inhibitors stabilizes p53 from MDM2 mediated degradation, which is a promising strategy for the treatment of cancer. In this report, a novel series of 4-imidazolidinone-containing compounds have been synthesized and tested in MDM2/p53 and MDM4/p53 FP binding assays. Upon SAR studies, compounds 2 (TB114) and 22 were identified as the most potent inhibitors of MDM2/p53 but not MDM4/p53 interactions. Both 2 and 22 exhibited strong antiproliferative activities in HCT-116 and MOLM-13 cell lines harboring wild type p53. Mechanistic studies show that 2 and 22 dose-dependently activated p53 and its target genes and induced apoptosis in cells based on the Western blot, qPCR, and flow cytometry assays. In addition, the antiproliferative activities of 2 and 22 were dependent on wild type p53, while they were not toxic to HEK-293 kidney cells. Furthermore, the on-target activities of 2 were general and applicable to other cancer cell lines with wild type p53. These attributes make 2 a good candidate for future optimization to discover a potential treatment of wild-type p53 cancer.


Subject(s)
Antineoplastic Agents , Tumor Suppressor Protein p53 , Humans , Tumor Suppressor Protein p53/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , HEK293 Cells , Cell Line, Tumor , Apoptosis , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Proto-Oncogene Proteins/metabolism , Cell Cycle Proteins/metabolism
3.
J Med Chem ; 67(2): 1079-1092, 2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38166388

ABSTRACT

The DNA-encoded library (DEL) is a powerful hit generation tool for chemical biology and drug discovery; however, the optimization of DEL hits remained a daunting challenge for the medicinal chemistry community. In this study, hit compounds targeting the WIN binding domain of WDR5 were discovered by the initial three-cycle linear DEL selection, and their potency was further enhanced by a cascade DEL selection from the focused DEL designed based on the original first run DEL hits. As expected, these new compounds from the second run of focused DEL were more potent WDR5 inhibitors in the protein binding assay confirmed by the off-DNA synthesis. Interestingly, selected inhibitors exhibited good antiproliferative activity in two human acute leukemia cell lines. Taken together, this new cascade DEL selection strategy may have tremendous potential for finding high-affinity leads against WDR5 and provide opportunities to explore and optimize inhibitors for other targets.


Subject(s)
DNA , Drug Discovery , Humans , Gene Library , Protein Binding , DNA/metabolism , Intracellular Signaling Peptides and Proteins/metabolism
4.
J Med Chem ; 66(24): 16991-17025, 2023 12 28.
Article in English | MEDLINE | ID: mdl-38062557

ABSTRACT

MDM2 and MDM4 cooperatively and negatively regulate p53, while this pathway is often hijacked by cancer cells in favor of their survival. Blocking MDM2/p53 interaction with small-molecule inhibitors liberates p53 from MDM2 mediated degradation, which is an attractive strategy for drug discovery. We reported herein structure-based discovery of highly potent spiroindoline-containing MDM2 inhibitor (-)60 (JN122), which also exhibited moderate activities against MDM4/p53 interactions. In a panel of cancer cell lines harboring wild type p53, (-)60 efficiently promoted activation of p53 and its target genes, inhibited cell cycle progression, and induced cell apoptosis. Interestingly, (-)60 also promoted degradation of MDM4. More importantly, (-)60 exhibited good PK properties and exerted robust antitumor efficacies in a systemic mouse xenograft model of MOLM-13. Taken together, our study showcases a class of potent MDM2 inhibitors featuring a novel spiro-indoline scaffold, which is promising for future development targeting cancer cells with wild-type p53.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Mice , Animals , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Apoptosis , Cell Line, Tumor , Proto-Oncogene Proteins/metabolism , Cell Cycle Proteins/metabolism
5.
Eur J Med Chem ; 254: 115381, 2023 Jun 05.
Article in English | MEDLINE | ID: mdl-37084596

ABSTRACT

Targeted degradation of BET family proteins BRD2/3/4 or only BRD4 with PROTAC molecules has been a promising strategy for the treatment of human cancer. Meanwhile, selective degradation of cellular BRD3 and BRD4-L remains a challenging task. We report herein a novel PROTAC molecule 24 that promoted selective degradation of cellular BRD3 and BRD4-L, but not BRD2 or BRD4-S, in a panel of six cancer cell lines. The observed target selectivity was partially attributed to differences in protein degradation kinetics and in types of cell lines. In a MM.1S mouse xenograft model, an optimized lead compound 28 promoted selective degradation of BRD3 and BRD4-L in vivo and exhibited robust antitumor activity. In summary, we have demonstrated that selective degradation of BRD3 and BRD4-L over BRD2 and BRD4-S is a feasible and robust approach in multiple cancer cell lines and an animal model, which could be helpful for further investigations on BRD3 and BRD4-L that ultimately benefitting cancer research and therapeutics.


Subject(s)
Neoplasms , Nuclear Proteins , Humans , Mice , Animals , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Cell Line , Neoplasms/drug therapy , Neoplasms/metabolism , Cell Cycle Proteins
6.
Molecules ; 28(4)2023 Feb 11.
Article in English | MEDLINE | ID: mdl-36838727

ABSTRACT

Sclareolide is a sesquiterpene lactone isolated from various plant sources in tons every year and is commercially used as a flavor ingredient in the cosmetic and food industries. Antitumor and antiviral activities of sclareolide have been previously reported. However, biological studies of sclareolide synthetic analogous are few. In view of these, we developed a robust synthetic method that allows the assembly of 36 novel sclareolide-indole conjugates and their derivatives. The synthetic method was based on TiCl4-promoted nucleophilic substitution of sclareolide-derived hemiacetal 4, while electron-rich aryles including indoles, polyphenol ethers, and pyrazolo [1,5-a]pyridine were good substrates. The stereochemistry of the final products was confirmed by single-crystal X-ray diffraction analysis, while the antiproliferative activities of selected final products were tested in K562 and MV4-11 cancer cell lines. Cytometric flow analysis shows that lead compounds 8k- and 10-induced robust apoptosis in MV4-11 cancer cells, while they exhibited weak impact on cell cycle progression. Taken together, our study suggests that sclareolide could be a good template and substrate for the synthesis of novel antiproliferative compounds.


Subject(s)
Antineoplastic Agents , Diterpenes , Antineoplastic Agents/pharmacology , Indoles/chemistry , Diterpenes/pharmacology , Cell Proliferation , Drug Screening Assays, Antitumor , Molecular Structure , Structure-Activity Relationship , Cell Line, Tumor
7.
Eur J Med Chem ; 236: 114327, 2022 Jun 05.
Article in English | MEDLINE | ID: mdl-35385805

ABSTRACT

Induction of apoptosis by the FDA-approved drug Venetoclax in cancer cells mainly derives from blocking the interactions between BCL-2 and BH3-only proteins. Anti-apoptotic BFL-1, a homolog of BCL-2, also competitively binds to the BH3-only proteins and is responsible for Venetoclax-induced drug resistance. Compared to BCL-2, small-molecule inhibitors of BFL-1 are relatively underexplored. In order to tackle this issue, in-house compound library was screened and a hit compound was identified and optimized to obtain 12 (ZH97) functioning as a covalent and selective inhibitor of BFL-1. 12 modifies BFL-1 at the C55 residue, blocks BFL-1/BID interaction in vitro, promotes cellular cytochrome c release from mitochondria, and induced apoptosis in BFL-1 overexpressing cancer cells. Mechanistic studies show that 12 inhibited BFL-1/PUMA interaction in cell lysate and is effective in cancer cells that harboring high expression level of BFL-1. In summary, blockade of BFL-1/BH3-only proteins interactions with a covalent small-molecule inhibitor induced apoptosis and elicited antitumor activity. Thus, our study demonstrates an appealing strategy for selective modulation of cellular BFL-1 for cancer therapy.


Subject(s)
Neoplasms , Proto-Oncogene Proteins c-bcl-2 , Apoptosis , Methylcellulose/metabolism , Minor Histocompatibility Antigens/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
8.
J Med Chem ; 65(8): 6207-6230, 2022 04 28.
Article in English | MEDLINE | ID: mdl-35420431

ABSTRACT

Despite recent clinical progress in peptide-based dual inhibitors of MDM2/4, small-molecule ones with robust antitumor activities remain challenging. To tackle this issue, 31 (YL93) was structure-based designed and synthesized, which had MDM2/4 binding Ki values of 1.1 and 642 nM, respectively. In three MDM4-overexpressing cancer cell lines harboring wild-type p53, 31 shows improved cell growth inhibition activities compared to RG7388, an MDM2-selective inhibitor in late-stage clinical trials. Mechanistic studies show that 31 increased cellular protein levels of p53 and p21 and upregulated the expression of p53-targeted genes in RKO cells with MDM4 amplification. In addition, 31 induced cell-cycle arrest and apoptosis in western blot and flow cytometry assays. Taken together, dual inhibition of MDM2/4 by 31 elicited stronger antitumor activities in vitro compared to selective MDM2 inhibitors in wild-type p53 and MDM4-overexpressing cancer cells.


Subject(s)
Antineoplastic Agents , Neoplasms , Antineoplastic Agents/pharmacology , Apoptosis , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , Neoplasms/drug therapy , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism
9.
J Med Chem ; 64(15): 10621-10640, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34286973

ABSTRACT

MDM4 is a homologue of MDM2, serving cooperatively as the negative regulator of tumor suppressor p53. Under the shadow of MDM2 inhibitors, limited efforts had been put into the discovery of MDM4 modulators. Recent studies of the experimental drug ALRN-6924, a dual MDM4 and MDM2 inhibitor, suggest that concurrent inhibition of MDM4 and MDM2 might be beneficial over only MDM2 inhibition. In view of the present research progress, we summarized published inhibitors of MDM4/p53 interactions including both peptide-based compounds and small molecules. Cocrystal structures of ligand/MDM4 complexes have been examined, and their structural features were compiled and compared in order to show the molecular basis required for high MDM4 binding affinities. Representative examples of small-molecule MDM4 inhibitors were discussed, followed by clinical results of ALRN-6924, together, providing a consolidated reference for further development of MDM4 inhibitors, either dual or selective.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Drug Development , Proto-Oncogene Proteins/antagonists & inhibitors , Tumor Suppressor Protein p53/antagonists & inhibitors , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Humans , Molecular Structure , Protein Binding/drug effects , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism
10.
Technol Cancer Res Treat ; 19: 1533033820967473, 2020.
Article in English | MEDLINE | ID: mdl-33073697

ABSTRACT

MiR-124-3p has been identified as a novel tumor suppressor and a potential therapeutic target in hepatocellular carcinoma (HCC) through regulating its target genes. However, the upstream regulatory mechanisms of mir-124-3p in HCC has not been fully understood. The transcription factor liver X receptor (LXR) plays a critical role in suppressing the proliferation of HCC cells, but it is unclear whether LXR is involved in the regulation of mir-124-3p. In the present study, we demonstrated that the expression of mir-124-3p was positively correlated with that of LXR in HCC, and the cell growth of HCC was significantly inhibited by LXR agonists. Moreover, activation of LXR with the agonists up-regulated the expression of mir-124-3p, and in turn down-regulated cyclin D1 and cyclin-dependent kinase 6 (CDK6) expression, which are the target genes of mir-124-3p. Mechanistically, miR-124-3p mediates LXR induced inhibition of HCC cell growth and down-regulation of cyclin D1 and CDK6 expression. In vivo experiments also confirmed that LXR induced miR-124-3p expression inhibited the growth of HCC xenograft tumors, as well as cyclin D1 and CDK6 expression. Our findings revealed that miR-124-3p is a novel target gene of LXR, and regulation of the miR-124-3p-cyclin D1/CDK6 pathway by LXR plays a crucial role in the proliferation of HCC cells. LXR-miR-124-3p-cyclin D1/CDK6 pathway may be a novel potential therapeutic target for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cyclin D1/genetics , Cyclin-Dependent Kinase 6/genetics , Liver Neoplasms/genetics , Liver X Receptors/genetics , MicroRNAs/genetics , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Heterografts , Humans , Liver Neoplasms/pathology , Mice
11.
Eur J Med Chem ; 195: 112277, 2020 Jun 01.
Article in English | MEDLINE | ID: mdl-32289582

ABSTRACT

Inhibition of MAP3K kinase ASK1 has been an attractive strategy for the treatment of nonalcoholic steatohepatitis and multiple sclerosis, among others. Herein, we reported the discovery of 2-pyridinyl urea-containing compound 14l (YD57) as a potent, small-molecule inhibitor of ASK1. 14l was selective against MAP3K kinases ASK2 and TAK1 (>140-fold), while it also inhibited several cell cycle regulating kinases with IC50 values in a range of 90-400 nM (<20-fold selectivity). As a consequence, 14l had stronger apoptosis induction, more potent G1 cell cycle arrest activities, and lower IC50 value of cell growth inhibition than that of GS4997 in HepG2 cancer cell line. On the other hand, 14l did not inhibit ASK1 and p38 phosphorylation in intact cells. We reason that the multi-target effects of 14l likely neutralized the activities caused by inhibition of cellular ASK1. Future studies of these ASK1 inhibitors should pay close attention to their kinome selectivity profile.


Subject(s)
Drug Design , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyridines/chemistry , Urea/chemistry , Urea/pharmacology , Cell Cycle/drug effects , Hep G2 Cells , Humans , Inhibitory Concentration 50
12.
J Med Chem ; 63(8): 4388-4407, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32216342

ABSTRACT

The overexpression of NIK plays a critical role in liver inflammatory diseases. Treatment of such diseases with small-molecule NIK inhibitors is a reasonable but underexplored approach. In this paper, we reported the discovery of a potent and selective NIK inhibitor 46 (XT2). 46 inhibited the NIK kinase with an IC50 value of 9.1 nM in vitro, and it also potently suppressed NIK activities in intact cells. In isogenic primary hepatocytes, treatment of 46 efficiently suppressed the expressions of NIK-induced genes. 46 was orally bioavailable in mice with moderate systemic exposure. In a NIK-associated mouse liver inflammation model, 46 suppressed CCl4-induced upregulation of ALT, a key biomarker of acute liver injury. 46 also decreased immune cell infiltration into the injured liver tissue. Overall, these studies provide examples that an NIK inhibitor is able to suppress toxin-induced liver inflammations, which indicates its therapeutic potentials for the treatment of liver inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Drug Discovery/methods , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Dose-Response Relationship, Drug , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Secondary , NF-kappaB-Inducing Kinase
13.
Eur J Med Chem ; 182: 111633, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31461688

ABSTRACT

Recently, selective inhibition of BET BD2 is emerging as a promising strategy for drug discovery. Despite significant progress in this area, systematic studies of selective BET BD2 inhibitors are still few. In this study, we report the discovery of a potent and selective BET BD2 inhibitor BY27 (47). Our high resolution co-crystal structures of 47/BRD2 BD1 and BD2 showed that the triazole group of 47, water molecules, H433 and N429 in BRD2 BD2 established a water-bridged H-bonding network, which is responsible for the observed selectivities. DNA microarray analysis of HepG2 cells treated with 47 or OTX015 demonstrated the transcriptome impact differences between a BET BD2 selective inhibitor and a pan BET inhibitor. In a MV4-11 mouse xenograft model, 47 caused 67% of tumor growth inhibition and was less toxic than a pan BET inhibitor 1 at high doses. We conclude that the improved safety profile of selective BET BD2 inhibitors warrant future studies in BET associated diseases.


Subject(s)
Azepines/chemistry , Drug Discovery , Proteins/antagonists & inhibitors , Pyrazoles/chemistry , Animals , Azepines/chemical synthesis , Azepines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Crystallography, X-Ray , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Molecular , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Domains , Proteins/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Structure-Activity Relationship
14.
J Immunol Res ; 2019: 8042097, 2019.
Article in English | MEDLINE | ID: mdl-31240234

ABSTRACT

B cell activating factor (BAFF), a member of the tumor necrosis factor (TNF) family, plays a critical role in the pathogenesis and progression of rheumatoid arthritis (RA). Chlorogenic acid (CGA) is a phenolic compound and exerts antiarthritic activities in arthritis. However, it is not clear whether the anti-inflammatory property of CGA is associated with the regulation of BAFF expression. In this study, we found that treatment of the collagen-induced arthritis (CIA) mice with CGA significantly attenuated arthritis progression and markedly inhibited BAFF production in serum as well as the production of serum TNF-α. Furthermore, CGA inhibits TNF-α-induced BAFF expression in a dose-dependent manner and apoptosis in MH7A cells. Mechanistically, we found the DNA-binding site for the transcription factor NF-κB in the BAFF promoter region is required for this regulation. Moreover, CGA reduces the DNA-binding activity of NF-κB to the BAFF promoter region and suppresses BAFF expression through the NF-κB pathway in TNF-α-stimulated MH7A cells. These results suggest that CGA may serve as a novel therapeutic agent for the treatment of RA by targeting BAFF.


Subject(s)
B-Cell Activating Factor/genetics , Chlorogenic Acid/pharmacology , Gene Expression Regulation/drug effects , NF-kappa B/metabolism , Signal Transduction/drug effects , Synoviocytes/drug effects , Synoviocytes/metabolism , Animals , Apoptosis/drug effects , Arthritis, Experimental , B-Cell Activating Factor/metabolism , Biomarkers , Cell Survival/drug effects , Cytokines/metabolism , Humans , Immunohistochemistry , Inflammation Mediators/metabolism , Male , Mice , Promoter Regions, Genetic , Transcriptional Activation , Tumor Necrosis Factor-alpha/metabolism
15.
Cancer Lett ; 409: 81-90, 2017 11 28.
Article in English | MEDLINE | ID: mdl-28889000

ABSTRACT

Considerable evidences have shown that both heat shock transcription factor 1 (HSF1) and autophagy can attenuate the sensitivity of hepatocellular carcinoma (HCC) cells to chemotherapeutic reagents. However, it is still little known whether HSF1 is associated with autophagy in regulating the chemosensitivity of HCC cells. In this study, we for the first time demonstrated that HSF1 markedly attenuated the killing effect of epirubicin (EPI) to HCC cells via enhancing the EPI-induced protective autophagy. Mechanistically, HSF1 upregulated autophagy related 4B (ATG4B) in HCC cells, which enhanced the EPI-triggered protective autophagy. Reporter assay showed that HSF1 increased the transcriptional activity of ATG4B gene promoter, and chromatin immunoprecipitation assay verified that HSF1 bound to the site (-1429 to -1417) in ATG4B gene promoter region. The experiments in nude mice showed that knockdown of HSF1 or ATG4B strengthened the anti-HCC effect of EPI in vivo. Collectively, these results revealed that HSF1 elevates ATG4B via promoting its transcription, which alleviates the sensitivity of EPI in HCC cells through enhancing protective autophagy, suggesting that the "HSF1/ATG4B/protective autophagy" pathway may be a novel target for developing sensitizing strategy to HCC chemotherapy.


Subject(s)
Autophagy-Related Proteins/biosynthesis , Autophagy/drug effects , Carcinoma, Hepatocellular/drug therapy , Cysteine Endopeptidases/biosynthesis , DNA-Binding Proteins/metabolism , Epirubicin/pharmacology , Liver Neoplasms/drug therapy , Transcription Factors/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Autophagy-Related Proteins/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Chloroquine/pharmacology , Cysteine Endopeptidases/metabolism , DNA-Binding Proteins/genetics , Drug Synergism , Heat Shock Transcription Factors , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/genetics , Transfection , Up-Regulation , Xenograft Model Antitumor Assays
16.
Mol Cancer Ther ; 16(9): 1806-1818, 2017 09.
Article in English | MEDLINE | ID: mdl-28533436

ABSTRACT

p53 deficiency, a frequent event in multiple kinds of malignancies, decreases the sensitivity of diverse targeted chemotherapeutics including the BCL-XL/BCL-2 inhibitor ABT-263. Loss of p53 function can activate mTOR complex 1 (mTORC1), which may make it a vulnerable target. Metformin has shown anti-neoplastic efficiency partially through suppressing mTORC1. However, it remains unknown whether mTORC1 activation confers ABT-263 resistance and whether metformin can overcome it in the p53-defective contexts. In this study, we for the first time demonstrated that metformin and ABT-263 synergistically elicited remarkable apoptosis through orchestrating the proapoptotic machineries in various p53-defective cancer cells. Mechanistic studies revealed that metformin sensitized ABT-263 via attenuating mTORC1-mediated cap-dependent translation of MCL-1 and survivin and weakening internal ribosome entry site (IRES)-dependent translation of XIAP Meanwhile, ABT-263 sensitized metformin through disrupting the BCL-XL/BIM complex. However, metformin and ABT-263 had no synergistic killing effect in p53 wild-type (p53-WT) cancer cells because the cotreatment dramatically induced the senescence-associated secretory phenotype (SASP) in the presence of wild type p53, and SASP could aberrantly activate the AKT/ERK-mTORC1-4EBP1-MCL-1/survivin signaling axis. Blocking the axis using corresponding kinase inhibitors or neutralizing antibodies against different SASP components sensitized the cotreatment effect of metformin and ABT-263 in p53-WT cancer cells. The in vivo experiments showed that metformin and ABT-263 synergistically inhibited the growth of p53-defective (but not p53-WT) cancer cells in tumor xenograft nude mice. These results suggest that the combination of metformin and ABT-263 may be a novel targeted therapeutic strategy for p53-defective cancers. Mol Cancer Ther; 16(9); 1806-18. ©2017 AACR.


Subject(s)
Aniline Compounds/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Metformin/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , Tumor Suppressor Protein p53/deficiency , bcl-X Protein/antagonists & inhibitors , Animals , Biomarkers , Cell Line, Tumor , Cellular Senescence/drug effects , Cellular Senescence/genetics , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Drug Synergism , Humans , Internal Ribosome Entry Sites , Male , Mice , Models, Biological , Protein Biosynthesis/drug effects , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics
17.
Oncotarget ; 7(37): 59458-59470, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27449090

ABSTRACT

Both dichloroacetate (DCA) and metformin (Met) have shown promising antitumor efficacy by regulating cancer cell metabolism. However, the DCA-mediated protective autophagy and Met-induced lactate accumulation limit their tumor-killing potential respectively. So overcoming the corresponding shortages will improve their therapeutic effects. In the present study, we found that DCA and Met synergistically inhibited the growth and enhanced the apoptosis of ovarian cancer cells. Interestingly, we for the first time revealed that Met sensitized DCA via dramatically attenuating DCA-induced Mcl-1 protein and protective autophagy, while DCA sensitized Met through markedly alleviating Met-induced excessive lactate accumulation and glucose consumption. The in vivo experiments in nude mice also showed that DCA and Met synergistically suppressed the growth of xenograft ovarian tumors. These results may pave a way for developing novel strategies for the treatment of ovarian cancer based on the combined use of DCA and Met.


Subject(s)
Antineoplastic Agents/therapeutic use , Dichloroacetic Acid/therapeutic use , Growth Inhibitors/therapeutic use , Metformin/therapeutic use , Ovarian Neoplasms/drug therapy , Animals , Apoptosis , Autophagy , Cell Line, Tumor , Drug Synergism , Drug Therapy, Combination , Female , Humans , Lactic Acid/metabolism , Mice , Mice, Nude , Xenograft Model Antitumor Assays
18.
Clin Immunol ; 161(2): 339-47, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26431776

ABSTRACT

B-lymphocyte stimulator (BLyS) plays a critical role in the pathogenesis and progression of rheumatoid arthritis (RA). Liver X receptor (LXR), a nuclear receptor, has an important anti-inflammatory effect. However, it is unclear whether the BLyS expression is regulated by LXR. In this study, we found that treatment with LXR agonist in collagen-induced arthritis (CIA) mice significantly attenuated arthritis progression, and markedly decreased BLyS production in serum and splenocytes as well as the production of serum IFNγ and TGFß. Activation of LXR in B lymphocytes dramatically suppressed the basal and IFNγ/TGFß-induced BLyS expression. Moreover, LXR agonist prominently suppressed the binding of NF-κB to BLyS promoter region, and decreased the promoter's transcriptional activity. Additionally, activation of LXR obviously repressed IFNγ-induced STAT1 activation and TGFß-induced SMAD3 activation. These results indicated that downregulation of BLyS may be a novel mechanism by which LXR ameliorates RA, and LXR/BLyS pathway may serve as a novel target for the treatment of RA.


Subject(s)
Arthritis, Experimental/metabolism , B-Cell Activating Factor/metabolism , B-Lymphocytes/metabolism , Orphan Nuclear Receptors/metabolism , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/prevention & control , B-Cell Activating Factor/genetics , B-Lymphocytes/drug effects , Benzoates/pharmacology , Benzylamines/pharmacology , Blotting, Western , Cells, Cultured , Gene Expression/drug effects , Interferon-gamma/blood , Interferon-gamma/metabolism , Liver X Receptors , Male , Mice, Inbred DBA , NF-kappa B/metabolism , Orphan Nuclear Receptors/agonists , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Transforming Growth Factor beta/blood , Transforming Growth Factor beta/metabolism
19.
Oncotarget ; 6(33): 34606-16, 2015 Oct 27.
Article in English | MEDLINE | ID: mdl-26416445

ABSTRACT

Suppressor of cytokine signaling 3 (SOCS3) is regarded as a vital repressor in the liver carcinogenesis mainly by inhibiting signal transducer and activator of transcription 3 (STAT3) activity. Farnesoid X Receptor (FXR), highly expressed in liver, has an important role in protecting against hepatocellular carcinoma (HCC). However, it is unclear whether the tumor suppressive activity of FXR involves the regulation of SOCS3. In the present study, we found that activation of FXR by its specific agonist GW4064 in HCC cells inhibited cell growth, induced cell cycle arrest at G1 phase, elevated p21 expression and repressed STAT3 activity. The above anti-tumor effects of FXR were dramatically alleviated by knockdown of SOCS3 with siRNA. Reporter assay revealed that FXR activation enhanced the transcriptional activity of SOCS3 promoter. Electrophoretic mobility shift assay (EMSA) and chromatin immunoprecipitation (ChIP) assay displayed that FXR directly bound to IR9 DNA motif within SOCS3 promoter region. The in vivo study in nude mice showed that treatment with FXR ligand GW4064 could decelerate the growth of HCC xenografts, up-regulate SOCS3 and p21 expression and inhibit STAT3 phosphorylation in the xenografts. These results suggest that induction of SOCS3 may be a novel mechanism by which FXR exerts its anti-HCC effects, and the FXR-SOCS3 signaling may serve as a new potential target for the prevention/treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Receptors, Cytoplasmic and Nuclear/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Humans , Immunohistochemistry , Isoxazoles/pharmacology , Liver Neoplasms/metabolism , Mice , Mice, Nude , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Suppressor of Cytokine Signaling 3 Protein , Transfection , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL