Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
J Pineal Res ; 72(1): e12781, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34826170

ABSTRACT

Melatonin, secreted by the pineal gland, regulates the circadian rhythms and also plays an oncostatic role in cancer cells. Previously, we showed that melatonin induces the expression of Bim, a pro-apoptotic Bcl-2 protein, at both the transcriptional and post-translational levels. In the present study, we investigated the molecular mechanisms underlying the melatonin-mediated Bim upregulation through post-translational regulation. We found that ovarian tumor domain-containing protein 1 (OTUD1), a deubiquitinase belonging to the OTU protein family, is upregulated by melatonin at the mRNA and protein levels. OTUD1 knockdown inhibited melatonin-induced Bim upregulation and apoptosis in cancer cells. OTUD1 directly interacted with Bim and inhibited its ubiquitination. Melatonin-induced OTUD1 upregulation caused deubiquitination at the lysine 3 residue of Bim, resulting in its stabilization. In addition, melatonin-induced activation of Sp1 was found to be involved in OTUD1 upregulation at the transcriptional level, and pharmacological inhibition and genetic ablation of Sp1 (siRNA) interrupted melatonin-induced OTUD1-mediated Bim upregulation. Furthermore, melatonin reduced tumor growth and induced upregulation of OTUD1 and Bim in a mouse xenograft model. Notably, Bim expression levels correlated with OTUD1 levels in patients with renal clear cell carcinoma. Thus, our results demonstrated that melatonin induces apoptosis by stabilizing Bim via Sp1-mediated OTUD1 upregulation.


Subject(s)
Melatonin , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11/genetics , Cell Line, Tumor , Humans , Melatonin/pharmacology , Mice , Sp1 Transcription Factor/genetics , Transcriptional Activation , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Up-Regulation
2.
Molecules ; 25(19)2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33050112

ABSTRACT

Magnolol is a biologically active compound, isolated from the Chinese herb Magnolia, that regulates antiproliferative, anticancer, antiangiogenic and antimetastatic activities. We found that magnolol sensitizes TRAIL-induced apoptotic cell death via upregulation of DR5 and downregulation of cellular FLICE-inhibitory protein (c-FLIP) and Mcl-1 in cancer cells, but not in normal cells. Mechanistically, magnolol increased ATF4-dependent DR5 expression at the transcription level, and knockdown of ATF4 markedly inhibited magnolol-induced DR5 upregulation. Silencing DR5 with siRNA prevented combined treatment with magnolol and TRAIL-induced apoptosis and PARP cleavage. Magnolol induced proteasome-mediated Mcl-1 downregulation, while magnolol-induced c-FLIP downregulation was regulated, at least in part, by lysosomal degradation. Our results revealed that magnolol enhanced TRAIL-induced apoptosis via ATF4-dependent DR5 upregulation and downregulation of c-FLIP and Mcl-1 proteins.


Subject(s)
Biphenyl Compounds/pharmacology , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Down-Regulation/drug effects , Lignans/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , Up-Regulation/drug effects , A549 Cells , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Down-Regulation/genetics , HeLa Cells , Humans , Neoplasms/genetics , Proteasome Endopeptidase Complex/genetics , Up-Regulation/genetics
3.
Molecules ; 25(24)2020 Dec 08.
Article in English | MEDLINE | ID: mdl-33302576

ABSTRACT

Mitochondrial fragmentation occurs during the apoptosis. Dynamin-related protein 1 (Drp1) acts as an important component in mitochondrial fission machinery and can regulate various biological processes including apoptosis, cell cycle, and proliferation. The present study demonstrates that dysfunction of mitochondrial dynamics plays a pivotal role in cisplatin-induced apoptosis. Inhibiting the mitochondrial fission with the specific inhibitor (Mdivi-1) did not affect apoptotic cell death in low concentrations (<10 µM). Interestingly, mdivi-1 enhanced cisplatin-induced apoptosis in cancer cells, but not in normal cells. Particularly in the presence of mdivi-1, several human cancer cell lines, including renal carcinoma cell line Caki-1, became vulnerable to cisplatin by demonstrating the traits of caspase 3-dependent apoptosis. Combined treatment induced downregulation of c-FLIP expression transcriptionally, and ectopic expression of c-FLIP attenuated combined treatment-induced apoptotic cell death with mdivi-1 plus cisplatin. Collectively, our data provide evidence that mdivi-1 might be a cisplatin sensitizer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cisplatin/pharmacology , Drug Resistance, Neoplasm/genetics , Transcription, Genetic/drug effects , Caspases/metabolism , Cell Line, Tumor , Humans , Kidney Neoplasms/genetics , Quinazolinones/pharmacology
4.
Int J Mol Sci ; 20(13)2019 Jul 02.
Article in English | MEDLINE | ID: mdl-31269715

ABSTRACT

R428, a selective small molecule Axl inhibitor, is known to have anti-cancer effects, such as inhibition of invasion and proliferation and induction of cell death in cancer cells. The Axl receptor tyrosine kinase is highly expressed in cancer cells and the level of Axl expression is associated with survival, metastasis, and drug resistance of many cancer cells. However, the effect of Axl inhibition on overcoming anti-cancer drugs resistance is unclear. Therefore, we investigated the capability of Axl inhibition as a therapeutic agent for the induction of TRAIL (tumor necrosis factor-related apoptosis-inducing ligand) sensitivity. In this study, R428 markedly sensitized cancer cells to TRAIL-induced apoptotic cell death, but not in normal human skin fibroblast (HSF) and human umbilical vein cells (EA.hy926). Moreover, knockdown of Axl by siRNA also increased TRAIL-induced apoptosis. R428 decreased c-FLIP proteins levels via induction of miR-708 expression and survivin protein levels at the post-translational level, and we found that knockdown of Axl also decreased both c-FLIP and survivin protein expression. Overexpression of c-FLIP and survivin markedly inhibited R428 plus TRAIL-induced apoptosis. Furthermore, R428 sensitized cancer cells to multiple anti-cancer drugs-mediated cell death. Our results provide that inhibition of Axl could improve sensitivity to TRAIL through downregulation of c-FLIP and survivin expression in renal carcinoma cells. Taken together, Axl may be a tempting target to overcome TRAIL resistance.


Subject(s)
Apoptosis/drug effects , Benzocycloheptenes/pharmacology , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Triazoles/pharmacology , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Down-Regulation/drug effects , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Survivin/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Axl Receptor Tyrosine Kinase
5.
Molecules ; 24(11)2019 Jun 05.
Article in English | MEDLINE | ID: mdl-31195760

ABSTRACT

Hispidulin (4',5,7-trihydroxy-6-methoxyflavone) is a natural compound derived from traditional Chinese medicinal herbs, and it is known to have an anti-inflammatory effect. Here, we investigated the effect of hispidulin on the immunoglobulin E (IgE)-mediated allergic responses in rat basophilic leukemia (RBL)-2H3 mast cells. When RBL-2H3 cells were sensitized with anti-dinitrophenyl (anti-DNP) IgE and subsequently stimulated with DNP-human serum albumin (HSA), histamine and ß-hexosaminidase were released from the cells by degranulation of activated mast cells. However, pretreatment with hispidulin before the stimulation of DNP-HSA markedly attenuated release of both in anti-DNP IgE-sensitized cells. Furthermore, we investigated whether hispidulin inhibits anti-DNP IgE and DNP-HSA-induced passive cutaneous anaphylaxis (PCA), as an animal model for Type I allergies. Hispidulin markedly decreased the PCA reaction and allergic edema of ears in mice. In addition, activated RBL-2H3 cells induced the expression of inflammatory cytokines (tumor necrosis factor-α and interleukin-4), which are critical for the pathogenesis of allergic disease, through the activation of c-Jun N-terminal kinase (JNK). Inhibition of JNK activation by hispidulin treatment reduced the induction of cytokine expression in the activated mast cells. Our results indicate that hispidulin might be a possible therapeutic candidate for allergic inflammatory diseases through the suppression of degranulation and inflammatory cytokines expression.


Subject(s)
Cytokines/metabolism , Down-Regulation , Flavones/therapeutic use , Histamine Release , Hypersensitivity/drug therapy , Inflammation Mediators/metabolism , Inflammation/drug therapy , Mast Cells/pathology , Animals , Cell Degranulation/drug effects , Down-Regulation/drug effects , Flavones/chemistry , Flavones/pharmacology , Histamine Release/drug effects , Hypersensitivity/complications , Immunoglobulin E/metabolism , Inflammation/complications , Inflammation/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Mast Cells/drug effects , Mice, Inbred ICR , Passive Cutaneous Anaphylaxis/drug effects , Phosphorylation/drug effects
6.
Biochem Biophys Res Commun ; 498(4): 849-854, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29534961

ABSTRACT

Inhibition of cathespsin S not only inhibits invasion and angiogenesis, but also induces apoptosis and autophagy in cancer cells. In present study, we revealed that pharmacological inhibitor [Z-FL-COCHO (ZFL)] of cathepsin S up-regulates pro-apoptotic protein Bim expression at the posttranslational levels. These effects were not associated with MAPKs and AMPK signal pathways. Interestingly, pretreatment with the chemical chaperones (TUDCA and PBA) and knockdown of protein phosphatase 2A (PP2A) markedly inhibited ZFL-induced Bim upregulation. ZFL enhances oxaliplatin-mediated apoptosis through ER stress-induced Bim upregulation in cancer cells. Collectively, our results suggest that inhibition of cathepsin S-induced Bim upregulation contribute to anti-cancer drug-induced apoptotic cell death in renal carcinoma Caki cells.


Subject(s)
Apoptosis/drug effects , Bcl-2-Like Protein 11/metabolism , Cathepsins/antagonists & inhibitors , Organoplatinum Compounds/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Endoplasmic Reticulum Stress , Humans , Oxaliplatin , Protein Phosphatase 2/metabolism , Protein Processing, Post-Translational , Up-Regulation/drug effects
7.
Drug Dev Res ; 79(1): 3-10, 2018 02.
Article in English | MEDLINE | ID: mdl-29044596

ABSTRACT

Preclinical Research & Development Angelicin is a furocoumarin derived from Psoralea corylifolia L. fruit that has anti-inflammatory and anti-tumor activity. In the present study, the effect of angelicin in enhancing tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptotic cell death was studied in Caki (renal carcinoma) cells. Angelicin alone and TRAIL alone had no effect on apoptosis, but in combination these compounds markedly induced apoptosis in the cancer cell lines while not inducing apoptosis in normal cells. The combination treatment induced accumulation of the sub-G1 population, DNA fragmentation, and activated caspase 3 activity in Caki cells, induced down-regulation of c-FLIP expression post-translationally, and over-expression of c-FLIP markedly blocked apoptosis induced by combined treatment with angelicin plus TRAIL. This study provides evidence that angelicin might be a TRAIL sensitizer.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 3/metabolism , Furocoumarins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Apoptosis/drug effects , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cells, Cultured , DNA Fragmentation , Down-Regulation , Drug Synergism , Fibroblasts , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism
8.
Int J Mol Sci ; 19(10)2018 Oct 22.
Article in English | MEDLINE | ID: mdl-30360403

ABSTRACT

Cepharanthine (CEP) is a natural plant alkaloid, and has anti-inflammatory, antineoplastic, antioxidative and anticancer properties. In this study, we investigated whether CEP could sensitize renal carcinoma Caki cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis. CEP alone and TRAIL alone had no effect on apoptosis. However, combined CEP and TRAIL treatment markedly enhanced apoptotic cell death in cancer cells, but not in normal cells. CEP induced downregulation of survivin and cellular-FLICE inhibitory protein (c-FLIP) expression at post-translational levels. Ectopic expression of survivin blocked apoptosis by combined treatment with CEP plus TRAIL, but not in c-FLIP overexpression. Interestingly, CEP induced survivin downregulation through downregulation of deubiquitin protein of STAM-binding protein-like 1 (STAMBPL1). Overexpression of STAMBPL1 markedly recovered CEP-mediated survivin downregulation. Taken together, our study suggests that CEP sensitizes TRAIL-mediated apoptosis through downregulation of survivin expression at the post-translational levels in renal carcinoma cells.


Subject(s)
Apoptosis/drug effects , Benzylisoquinolines/pharmacology , Carcinoma, Renal Cell/metabolism , Survivin/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Ubiquitin Thiolesterase/metabolism , Apoptosis/genetics , Cell Line, Tumor , Humans , Peptide Hydrolases , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , Ubiquitin Thiolesterase/genetics
9.
Int J Mol Sci ; 19(5)2018 Apr 27.
Article in English | MEDLINE | ID: mdl-29702597

ABSTRACT

Corosolic acid is one of the pentacyclic triterpenoids isolated from Lagerstroemia speciose and has been reported to exhibit anti-cancer and anti-proliferative activities in various cancer cells. In the present study, we investigated the molecular mechanisms of corosolic acid in cancer cell death. Corosolic acid induces a decrease of cell viability and an increase of cell cytotoxicity in human renal carcinoma Caki cells. Corosolic acid-induced cell death is not inhibited by apoptosis inhibitor (z-VAD-fmk, a pan-caspase inhibitor), necroptosis inhibitor (necrostatin-1), or ferroptosis inhibitors (ferrostatin-1 and deferoxamine (DFO)). Furthermore, corosolic acid significantly induces reactive oxygen species (ROS) levels, but antioxidants (N-acetyl-l-cysteine (NAC) and trolox) do not inhibit corosolic acid-induced cell death. Interestingly, corosolic acid induces lipid oxidation, and α-tocopherol markedly prevents corosolic acid-induced lipid peroxidation and cell death. Anti-chemotherapeutic effects of α-tocopherol are dependent on inhibition of lipid oxidation rather than inhibition of ROS production. In addition, corosolic acid induces non-apoptotic cell death in other renal cancer (ACHN and A498), breast cancer (MDA-MB231), and hepatocellular carcinoma (SK-Hep1 and Huh7) cells, and α-tocopherol markedly inhibits corosolic acid-induced cell death. Therefore, our results suggest that corosolic acid induces non-apoptotic cell death in cancer cells through the increase of lipid peroxidation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Renal Cell/pathology , Cell Death/drug effects , Kidney Neoplasms/pathology , Lipid Peroxidation/drug effects , Reactive Oxygen Species/metabolism , Triterpenes/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Lagerstroemia/chemistry , alpha-Tocopherol/pharmacology
10.
Molecules ; 23(9)2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30189637

ABSTRACT

Niclosamide is used to treat intestinal parasite infections, as being an anthelmintic drug. Recently, several papers suggest the niclosamide inhibits multiple signaling pathways, which are highly activated and mutated in cancer. Here, niclosamide was evaluated for identifying strategies to overcome tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) resistance. Although niclosamide (100⁻200 nM) alone did not bring about cell death, combinations of niclosamide and TRAIL led to apoptotic cell death in carcinoma cells, but not in normal cells. Niclosamide markedly increased DR5 protein levels, including cell-surface DR5, and decreased c-FLIP protein levels. Down-regulation of DR5 by specific small interfering RNA (siRNA) and ectopic expression of c-FLIP markedly blocked niclosamide plus TRAIL-induced apoptosis. Our findings provide that niclosamide could overcome resistance to TRAIL through up-regulating DR5 on the cell surface and down-regulating c-FLIP in cancer cells. Taken together, niclosamide may be an attractive candidate to overcome TRAIL resistance.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Niclosamide/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Humans , Reactive Oxygen Species/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics
11.
Molecules ; 23(11)2018 Oct 24.
Article in English | MEDLINE | ID: mdl-30356017

ABSTRACT

We previously reported that PP242 (dual inhibitor of mTORC1/2) plus curcumin induced apoptotic cell death through lysosomal membrane permeabilization (LMP)-mediated autophagy. However, the relationship between ER stress and apoptotic cell death by combined PP242 and curcumin treatment remains unknown. In the present study, we found that combined PP242 and curcumin treatment induced cytosolic Ca2+ release and ER stress. Interestingly, pretreatment with the chemical chaperones (TUDCA and 4-PBA) and knockdown of CHOP and ATF4 by siRNA did not abolish combined treatment-induced apoptosis in renal carcinoma cells. These results suggest that combined treatment with mTORC1/2 inhibitor and curcumin induces ER stress which is not essential for apoptotic cell death. Furthermore, overexpression of HSP70 significantly inhibited PP242 plus curcumin-induced LMP and apoptosis, but the protective effect was abolished by K77R mutation of acetylation site of HSP70. Taken together, our results reveal that regulation of HSP70 through K77 acetylation plays role in combined PP242 and curcumin treatment-induced apoptosis.


Subject(s)
Curcumin/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Acetylation , Apoptosis/drug effects , Calcium/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Curcumin/chemistry , Drug Synergism , Endoplasmic Reticulum Stress/drug effects , Humans , Protein Kinase Inhibitors/chemistry , Signal Transduction/drug effects
12.
Molecules ; 23(7)2018 Jul 02.
Article in English | MEDLINE | ID: mdl-30004456

ABSTRACT

Garcinol is a polyisoprenylated benzophenone derived from the Garcinia indica fruit that possess potential therapeutic effects such as inhibition of inflammation and tumor expansion. Here, we investigated whether garcinol induces TRAIL sensitization in renal carcinoma cells. Single treatment with garcinol or TRAIL did not effect on apoptosis. However, combined treatment with garcinol plus TRAIL significantly induced apoptosis in renal carcinoma (Caki, ACHN and A498), lung carcinoma (A549), and hepatoma (SK-Hep1) cells. In contrast, garcinol plus TRAIL did not alter cell viability in normal cells. Garcinol plus TRAIL induced up-regulation of DR5 and down-regulation of c-FLIP expression at post-translational levels. Furthermore, knock-down of DR5 by siRNA and ectopic expression of c-FLIP blocked apoptotic cell death induced by garcinol plus TRAIL. Overall, our study provides evidence that garcinol can be exploited as a potential TRAIL sensitizer.


Subject(s)
Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Down-Regulation/drug effects , TNF-Related Apoptosis-Inducing Ligand/metabolism , Terpenes/pharmacology , Up-Regulation/drug effects , A549 Cells , Animals , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cell Line , Cell Line, Tumor , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , RNA, Small Interfering/metabolism
13.
Molecules ; 23(11)2018 Nov 20.
Article in English | MEDLINE | ID: mdl-30463333

ABSTRACT

Maritoclax, an active constituent isolated from marine bacteria, has been known to induce Mcl-1 downregulation through proteasomal degradation. In this study, we investigated the sensitizing effect of maritoclax on tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in human renal carcinoma cells. We found that combined treatment with maritoclax and TRAIL markedly induced apoptosis in renal carcinoma (Caki, ACHN and A498), lung cancer (A549) and hepatocellular carcinoma (SK-Hep1) cells. The upregulation of death receptor 5 (DR5) and downregulation of cellular FLICE-inhibitory protein (cFLIP) were involved in maritoclax plus TRAIL-induced apoptosis. Maritoclax-induced DR5 upregulation was regulated by induction of C/EBP homologous protein (CHOP) expression. Interestingly, maritoclax induced cFLIP downregulation through the increased expression of miR-708. Ectopic expression of cFLIP prevented combined maritoclax and TRAIL-induced apoptosis. Taken together, maritoclax sensitized TRAIL-induced apoptosis through CHOP-mediated DR5 upregulation and miR-708-mediated cFLIP downregulation.


Subject(s)
MicroRNAs/genetics , Neoplasms/metabolism , Pyrroles/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Transcription Factor CHOP/metabolism , A549 Cells , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Transcription Factor CHOP/genetics , Up-Regulation
14.
Mol Cell Biochem ; 435(1-2): 15-24, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28466458

ABSTRACT

Artesunate is a well-known anti-malarial drug originated from artemisinin as a Chinese herb and has been reported to have anti-cancer potential in many cancer cells. In the present study, we examined the efficacy of artesunate against the renal carcinoma Caki cells and explored its mechanism of cytotoxicity. A steep decline in cell viability within 18 h was recorded upon artesunate exposure, but pretreatment of z-VAD-FMK had no effect on the loss of the cell viability by artesunate. On the other hand, necrostatin-1 pretreatment and knockdown of RIP-1 significantly reduced the cytotoxicity of artesunate against Caki cell. Moreover, the generation of mitochondrial ROS prompted by artesunate was found to be the principle mechanism of cell death. Pretreatment with necrostatin-1 or knockdown of RIP-1 inhibited the generation of ROS by artesunate, resulting in the protection of the cells from artesunate toxicity. Moreover, the similar results were observed in the case of other renal carcinoma cell lines (ACHN and A498). The results suggest that artesunate induces the generation of ROS and cell death in RIP1-dependent manner. Therefore, our data suggest that artesunate could induce RIP1-dependent cell death in human renal carcinoma.


Subject(s)
Artemisinins/pharmacology , Kidney Neoplasms/metabolism , Neoplasm Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Reactive Oxygen Species/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Artesunate , Cell Death/drug effects , Cell Line, Tumor , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Neoplasm Proteins/genetics , Nuclear Pore Complex Proteins/genetics , RNA-Binding Proteins/genetics
15.
Mol Cell Biochem ; 429(1-2): 91-102, 2017 May.
Article in English | MEDLINE | ID: mdl-28120212

ABSTRACT

ABT-737 is a BH3 mimetic inhibitor of Bcl-xL, Bcl-2, and Bcl-w, and it has been reported for anti-cancer effects in various types of cancer cells. However, ABT-737 fails to induce apoptosis in cancer cell with high levels of Mcl-1 expression. The pharmacological survivin inhibitor YM155 has been reported to induce downregulation of Mcl-1 expression. Therefore, we investigated the effect of YM155 to sensitize resistance against ABT-737 in Mcl-1-overexpressed human renal carcinoma Caki cells. We found that ABT-737 alone and YM155 alone did not induce apoptosis, but YM155 markedly sensitized ABT-737-mediated apoptosis in Mcl-1-overexpressed Caki cells, human glioma cells (U251MG), and human lung carcinoma cells (A549). In contrast, combined treatment with ABT-737 and YM155 did not increase apoptosis in normal mouse kidney cells (TCMK-1) and human mesangial cells (MC). YM155 induced lysosome-dependent downregulation of Mcl-1 expression in Mcl-1-overexpressed Caki cells. In addition, combined treatment with ABT-737 and YM155 induced loss of mitochondrial membrane potential and inhibited interaction of Bcl-xL and Bax. Taken together, our results suggested that YM155 effectively improves sensitivity to ABT-737 through downregulation of Mcl-1 expression.


Subject(s)
Biphenyl Compounds/pharmacology , Down-Regulation , Drug Resistance, Neoplasm/drug effects , Imidazoles/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Naphthoquinones/pharmacology , Neoplasms/metabolism , Nitrophenols/pharmacology , Sulfonamides/pharmacology , A549 Cells , Animals , Apoptosis , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Neoplasms/drug therapy , Piperazines/pharmacology
16.
Int J Mol Sci ; 18(12)2017 Nov 29.
Article in English | MEDLINE | ID: mdl-29186071

ABSTRACT

Polo-like kinase 1 (PLK1) plays major roles in cell cycle control and DNA damage response. Therefore, PLK1 has been investigated as a target for cancer therapy. Volasertib is the second-in class dihydropteridinone derivate that is a specific PLK1 inhibitor. In this study, we examined that combining PLK1 inhibitor with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) would have an additive and synergistic effect on induction of apoptosis in cancer cells. We found that volasertib alone and TRAIL alone had no effect on apoptosis, but the combined treatment of volasertib and TRAIL markedly induced apoptosis in Caki (renal carcinoma), A498 (renal carcinoma) and A549 (lung carcinoma) cells, but not in normal cells (human skin fibroblast cells and mesangial cells). Combined treatment induced accumulation of sub-G1 phase, DNA fragmentation, cleavage of poly (ADP-ribose) polymerase (PARP) and activation of caspase 3 activity in Caki cells. Interestingly, combined treatment induced downregulation of cellular-FLICE-inhibitory protein (c-FLIP) expression and ectopic expression of c-FLIP markedly blocked combined treatment-induced apoptosis. Therefore, this study demonstrates that volasertib may sensitize TRAIL-induced apoptosis in Caki cells via downregulation of c-FLIP.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Pteridines/pharmacology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Humans
17.
Molecules ; 22(8)2017 Aug 02.
Article in English | MEDLINE | ID: mdl-28767099

ABSTRACT

Fisetin is a natural compound found in fruits and vegetables such as strawberries, apples, cucumbers, and onions. Since fisetin can elicit anti-cancer effects, including anti-proliferation and anti-migration, we investigated whether fisetin induced apoptosis in human renal carcinoma (Caki) cells. Fisetin markedly induced sub-G1 population and cleavage of poly (ADP-ribose) polymerase (PARP), which is a marker of apoptosis, and increased caspase activation. We found that pan-caspase inhibitor (z-VAD-fmk) inhibited fisetin-induced apoptosis. In addition, fisetin induced death receptor 5 (DR5) expression at the transcriptional level, and down-regulation of DR5 by siRNA blocked fisetin-induced apoptosis. Furthermore, fisetin induced p53 protein expression through up-regulation of protein stability, whereas down-regulation of p53 by siRNA markedly inhibited fisetin-induced DR5 expression. In contrast, fisetin induced up-regulation of CHOP expression and reactive oxygen species production, which had no effect on fisetin-induced apoptosis. Taken together, our study demonstrates that fisetin induced apoptosis through p53 mediated up-regulation of DR5 expression at the transcriptional level.


Subject(s)
Antineoplastic Agents/pharmacology , Flavonoids/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Apoptosis , Carcinoma , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Down-Regulation , Flavonols , Humans , Kidney Neoplasms , Mice , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Up-Regulation
18.
J Cell Biochem ; 117(2): 361-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26174226

ABSTRACT

RU486 (Mifepristone) is known as an antagonist of the progesterone receptor and glucocorticoid receptor. Here, we investigated the mechanism underlying anti-tumor activity of RU486 in renal carcinoma Caki cells. Treatment of Caki cells with RU486 was found to induce several signature ER stress markers; including ER stress-specific XBP1 splicing, and the up-regulation of glucose-regulated protein (GRP)-78 and CCAAT/enhancer-binding protein homologous protein (CHOP) expression. RU486-induced expression of CHOP involves the putative C/EBPδ site within the CHOP promoter region. Using a combination of C/EBPδ cDNA transfection, the luciferase assay with a mutated C/EBPδ binding site and siRNA-mediated C/EBPδ knockdown, we found that the C/EBPδ site is required for RU486-mediated activation of the CHOP promoter. In addition, RU486-induced CHOP expression is down-regulated by inhibition of the p38 MAPK and JNK signaling pathways at the post-translational levels. RU486 dose-dependently induced apoptotic cell death in renal carcinoma cells. Suppression of CHOP expression by CHOP siRNA attenuated RU486-induced apoptosis. Taken together, RU486 induces pro-apoptotic ER stress through the induction of CHOP expression.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , CCAAT-Enhancer-Binding Protein-delta/metabolism , Endoplasmic Reticulum Stress/drug effects , Mifepristone/pharmacology , Transcription Factor CHOP/metabolism , Base Sequence , Binding Sites , CCAAT-Enhancer-Binding Protein-delta/genetics , Carcinoma, Renal Cell , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kidney Neoplasms , MAP Kinase Signaling System , Promoter Regions, Genetic , RNA Splicing , Regulatory Factor X Transcription Factors , Transcription Factor CHOP/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation , X-Box Binding Protein 1
19.
Mol Carcinog ; 54(3): 216-28, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24127286

ABSTRACT

Silymarin has been known as a chemopreventive agent, and possesses multiple anti-cancer activities including induction of apoptosis, inhibition of proliferation and growth, and blockade of migration and invasion. However, whether silymarin could inhibit prostaglandin (PG) E2 -induced renal cell carcinoma (RCC) migration and what are the underlying mechanisms are not well elucidated. Here, we found that silymarin markedly inhibited PGE2 -stimulated migration. PGE2 induced G protein-dependent CREB phosphorylation via protein kinase A (PKA) signaling, and PKA inhibitor (H89) inhibited PGE2 -mediated migration. Silymarin reduced PGE2 -induced CREB phosphorylation and CRE-promoter activity. PGE2 also activated G protien-independent signaling pathways (Src and STAT3) and silymarin reduced PGE2 -induced phosphorylation of Src and STAT3. Inhibitor of Src (Saracatinib) markedly reduced PGE2 -mediated migration. We found that EP2, a PGE2 receptor, is involved in PGE2 -mediated cell migration. Down regulation of EP2 by EP2 siRNA and EP2 antagonist (AH6809) reduced PGE2 -inudced migration. In contrast, EP2 agonist (Butaprost) increased cell migration and silymarin effectively reduced butaprost-mediated cell migration. Moreover, PGE2 increased EP2 expression through activation of positive feedback mechanism, and PGE2 -induced EP2 expression, as well as basal EP2 levels, were reduced in silymarin-treated cells. Taken together, our study demonstrates that silymarin inhibited PGE2 -induced cell migration through inhibition of EP2 signaling pathways (G protein dependent PKA-CREB and G protein-independent Src-STAT3).


Subject(s)
Antioxidants/pharmacology , Carcinoma, Renal Cell/pathology , Cell Movement/drug effects , Dinoprostone/antagonists & inhibitors , Kidney Neoplasms/pathology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Silymarin/pharmacology , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Benzodioxoles/pharmacology , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dinoprostone/metabolism , Down-Regulation , Enzyme Activation , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/metabolism , HCT116 Cells , HT29 Cells , Humans , Integrases/genetics , Kidney Neoplasms/metabolism , Phosphorylation , Prostaglandin Antagonists/pharmacology , Quinazolines/pharmacology , RNA Interference , RNA, Small Interfering , Receptors, Prostaglandin E, EP2 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP2 Subtype/biosynthesis , Receptors, Prostaglandin E, EP2 Subtype/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Wound Healing , Xanthones/pharmacology , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
20.
J Pineal Res ; 58(3): 310-20, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25711465

ABSTRACT

Melatonin is involved in many physiological functions, and it has differential effects on apoptosis in normal and cancer cells. However, the mechanism of its antitumor roles is not well understood. In this study, we show that melatonin enhances tunicamycin-induced apoptosis in human breast carcinoma MDA-MB-231 cells. Melatonin up-regulates pro-apoptotic protein Bim expression at the transcriptional levels in the presence of tunicamycin. Melatonin inhibits tunicamycin-induced COX-2 expression in MDA-MB-231 cells. Furthermore, inhibition of COX-2 activity using the COX-2 inhibitor, NS398, increases tunicamycin-induced apoptosis. Interestingly, these effects were not associated with melatonin receptor signal pathways. Pertussis toxin (a general Gi protein inhibitor) or luzindole (a nonspecific melatonin receptor antagonist) did not reverse the effect of melatonin. In addition, melatonin blocked tunicamycin-induced NF-κB transcriptional activity, p65 nuclear translocation, and p38 MAPK activation. Melatonin-mediated p38 MAPK inhibition contributed to decreased COX-2 mRNA stability. Taken together, our results suggest that melatonin enhances antitumor function through up-regulation of Bim expression and down-regulation of COX-2 expression in tunicamycin-treated MDA-MB-231 cells.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Cyclooxygenase 2/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Melatonin/pharmacology , Membrane Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Tunicamycin/pharmacology , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Line, Tumor , Cyclooxygenase 2/genetics , Down-Regulation/drug effects , Humans , Membrane Proteins/genetics , NF-kappa B/metabolism , Proto-Oncogene Proteins/genetics , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL