Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
Apoptosis ; 26(5-6): 338-347, 2021 06.
Article En | MEDLINE | ID: mdl-33905036

Caspase-mediated cleavage of proteins ensures the irreversible commitment of cells to undergo apoptosis, and is thus a hallmark of apoptosis. Rapamycin-insensitive companion of mTOR (rictor) functions primarily as a core and essential component of mTOR complex 2 (mTORC2) to critically regulate cellular homeostasis. However, its role in the regulation of apoptosis is largely unknown. In the current study, we found that rictor was cleaved to generate two small fragments at ~ 50 kD and ~ 130 kD in cells undergoing apoptosis upon treatment with different stimuli such as the death ligand, TRAIL, and the small molecule, AZD9291. This cleavage was abolished when caspases were inhibited and could be reproduced when directly incubating rictor protein and caspase-3 in vitro. Furthermore, the cleavage site of caspase-3 on rictor was mapped at D1244 (VGVD). These findings together robustly demonstrate that rictor is a substrate of caspase-3 and undergoes cleavage during apoptosis. These results add new information for understanding the biology of rictor in the regulation of cell survival and growth.


Apoptosis/physiology , Caspases/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Acrylamides/pharmacology , Aniline Compounds/pharmacology , Apoptosis/drug effects , Caspase Inhibitors/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Humans , Mechanistic Target of Rapamycin Complex 2/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Receptors, Death Domain/metabolism , Signal Transduction/drug effects
2.
Biomolecules ; 11(4)2021 03 26.
Article En | MEDLINE | ID: mdl-33810241

Death ligands such as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL; TNFSF10) and their corresponding death receptors (e.g., DR5) not only initiate apoptosis through activation of the extrinsic apoptotic pathway but also exert non-apoptotic biological functions such as regulation of inflammation and cancer metastasis. The involvement of the TRAIL/death receptor signaling pathway in the regulation of cancer invasion and metastasis is complex as both positive and negative roles have been reported. The underlying molecular mechanisms are even more complicated. This review will focus on discussing current knowledge in our understanding of the involvement of TRAIL/death receptor-mediated signaling in the regulation of cancer cell invasion and metastasis.


Neoplasms/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Humans , Neoplasm Metastasis , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/genetics
3.
Cell Rep ; 34(2): 108625, 2021 01 12.
Article En | MEDLINE | ID: mdl-33440161

Radiation sensitive 52 (RAD52) is an important factor for double-strand break repair (DSBR). However, deficiency in vertebrate/mammalian Rad52 has no apparent phenotype. The underlying mechanism remains elusive. Here, we report that RAD52 deficiency increased cell survival after camptothecin (CPT) treatment. CPT generates single-strand breaks (SSBs) that further convert to double-strand breaks (DSBs) if they are not repaired. RAD52 inhibits SSB repair (SSBR) through strong single-strand DNA (ssDNA) and/or poly(ADP-ribose) (PAR) binding affinity to reduce DNA-damage-promoted X-Ray Repair Cross Complementing 1 (XRCC1)/ligase IIIα (LIG3α) co-localization. The inhibitory effects of RAD52 on SSBR neutralize the role of RAD52 in DSBR, suggesting that RAD52 may maintain a balance between cell survival and genomic integrity. Furthermore, we demonstrate that blocking RAD52 oligomerization that disrupts RAD52's DSBR, while retaining its ssDNA binding capacity that is required for RAD52's inhibitory effects on SSBR, sensitizes cells to different DNA-damaging agents. This discovery provides guidance for developing efficient RAD52 inhibitors in cancer therapy.


DNA Damage , DNA Ligase ATP/metabolism , DNA, Single-Stranded/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , Rad52 DNA Repair and Recombination Protein/metabolism , X-ray Repair Cross Complementing Protein 1/metabolism , Animals , DNA Ligase ATP/genetics , DNA, Single-Stranded/genetics , Humans , Mice , Rad52 DNA Repair and Recombination Protein/genetics , X-ray Repair Cross Complementing Protein 1/genetics
6.
DNA Repair (Amst) ; 78: 37-44, 2019 06.
Article En | MEDLINE | ID: mdl-30954901

DNA double strand breaks (DSBs) are a severe threat to genome integrity and a potential cause of tumorigenesis, which is a multi-stage process and involves many factors including the mutation of oncogenes and tumor suppressors, some of which are transcribed microRNAs (miRNAs). Among more than 2000 known miRNAs, miR-21 is a unique onco-miRNA that is highly expressed in almost all types of human tumors and is associated with tumorigenesis through its multiple targets. However, it remains unclear whether there is any functional link between DSBs and miR-21 expression and, if so, does the link contribute to DSB-induced genomic instability/tumorigenesis. To address this question, we used DNA-PKcs-/- (deficient in non-homologous end-joining (NHEJ)) and Rad54-/- (deficient in homologous recombination repair (HRR)) mouse embryonic fibroblasts (MEFs) since NHEJ and HRR are the major pathways for DSB repair in mammalian cells. Our results indicate that levels of miR-21 are elevated in these DSB repair (DSBR) deficient cells, and ionizing radiation (IR) further increases these levels in both wild-type (WT) and DSBR-deficient cells. Interestingly, IR stimulated growth in soft agar and this effect was greatly reduced by blocking miR-21 expression in both WT and DSBR-deficient cells. Taken together, our results suggest that either IR or DSBR-deficient can lead to an upregulation of miR-21 levels and that miR-21 is associated with IR-induced cell growth in soft agar. These results may help our understanding of DSB-induced tumorigenesis and provide information that could facilitate the development of new strategies to prevent DSB-induced carcinogenesis.


Agar , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , MicroRNAs/genetics , Up-Regulation/radiation effects , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Carcinogenesis/genetics , Carcinogenesis/radiation effects , Cell Line , Cell Proliferation/genetics , Cell Proliferation/radiation effects , DNA Repair/genetics , ErbB Receptors/metabolism , Mice
7.
Oncogene ; 37(25): 3415-3425, 2018 06.
Article En | MEDLINE | ID: mdl-29551769

Monocyte chemotactic protein-induced protein-1 (MCPIP1; also called Regnase-1) encoded by the ZC3H12A gene critically regulates inflammatory responses and immune homeostasis primarily by RNase-dependent and -independent mechanisms. However, the relationship of MCPIP1 with apoptosis and cancer and the underlying mechanisms are largely unclear. The current study has demonstrated a previously uncovered connection between MCPIP1 and the negative regulation of death receptor 5 (DR5; also known as TRAIL-R2 or killer/DR5), a cell surface receptor for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), which is produced endogenously by various immune cells such as T cells. Our findings have revealed that MCPIP1 decreases both total cellular and cell surface DR5, primarily through modulating DUB-mediated protein autophagic/lysosomal degradation. Suppression of MCPIP1 by gene knockdown induces the formation of death-induced signaling complex (DISC) and enhances TRAIL or DR5 activation-induced apoptosis in cancer cells. Moreover, we demonstrated an inverse correlation between MCPIP1 expression and DR5 expression/cell sensitivity to DR5 activation-induced apoptosis in cancer cells. Our findings warrant future investigation of the roles of negative regulation of DR5 by MCPIP1 in cancer and in T-cell immunity.


Apoptosis , Breast Neoplasms/pathology , Deubiquitinating Enzymes/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Ribonucleases/metabolism , Transcription Factors/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , Deubiquitinating Enzymes/genetics , Female , Humans , Proteolysis , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Ribonucleases/genetics , Signal Transduction , Transcription Factors/genetics , Tumor Cells, Cultured
8.
Clin Cancer Res ; 23(21): 6567-6579, 2017 Nov 01.
Article En | MEDLINE | ID: mdl-28765329

Purpose: The mechanisms accounting for anticancer activity of AZD9291 (osimertinib or TAGRISSO), an approved third-generation EGFR inhibitor, in EGFR-mutant non-small cell lung cancer (NSCLC) cells and particularly for the subsequent development of acquired resistance are unclear and thus are the focus of this study.Experimental Design: AZD9219-resistant cell lines were established by exposing sensitive cell lines to AZD9291. Protein alterations were detected with Western blotting. Apoptosis was measured with annexin V/flow cytometry. Growth-inhibitory effects of tested drugs were evaluated in vitro with cell number estimation and colony formation assay and in vivo with mouse xenograft models. Protein degradation was determined by comparing protein half-lives and inhibiting proteasome. Gene knockdown were achieved with siRNA or shRNA.Results: AZD9291 potently induced apoptosis in EGFR-mutant NSCLC cell lines, in which ERK phosphorylation was suppressed accompanied with Bim elevation and Mcl-1 reduction likely due to enhanced Mcl-1 degradation and increased Bim stability. Blocking Bim elevation by gene knockdown or enforcing Mcl-1 expression attenuated or abolished AZD9291-induced apoptosis. Moreover, AZD9291 lost its ability to modulate Bim and Mcl-1 levels in AZD9291-resistant cell lines. The combination of a MEK inhibitor with AZD9291 restores the sensitivity of AZD9291-resistant cells including those with C797S mutation to undergo apoptosis and growth regression in vitro and in vivoConclusions: Modulation of MEK/ERK-dependent Bim and Mcl-1 degradation critically mediates sensitivity and resistance of EGFR-mutant NSCLC cells to AZD9291 and hence is an effective strategy to overcome acquired resistance to AZD9291. Clin Cancer Res; 23(21); 6567-79. ©2017 AACR.


Acrylamides/administration & dosage , Aniline Compounds/administration & dosage , Bcl-2-Like Protein 11/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Acrylamides/adverse effects , Aniline Compounds/adverse effects , Animals , Apoptosis/drug effects , Bcl-2-Like Protein 11/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/antagonists & inhibitors , Humans , MAP Kinase Signaling System/drug effects , Mice , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Proteolysis/drug effects , Xenograft Model Antitumor Assays
9.
Sci Rep ; 7(1): 8027, 2017 08 14.
Article En | MEDLINE | ID: mdl-28808321

b-AP15 and its derivatives block proteasome deubiquitinase (DUB) activity and have been developed and tested in the clinic as potential cancer therapeutic agents. b-AP15 induces apoptosis in cancer cells, but the underlying mechanisms are largely undefined. The current study focuses on studying the modulatory effects of b-AP15 on death receptor 5 (DR5) levels and DR5 activation-induced apoptosis as well as on understanding the underlying mechanisms. Treatment with b-AP15 potently increased DR5 levels including cell surface DR5 in different cancer cell lines with limited or no effects on the levels of other related proteins including DR4, c-FLIP, FADD, and caspase-8. b-AP15 substantially slowed the degradation of DR5, suggesting that it stabilizes DR5. Moreover, b-AP15 effectively augmented apoptosis when combined with TRAIL or the DR5 agonistic antibody AMG655; these effects are DR5-dependent because DR5 deficiency abolished the ability of b-AP15 to enhance TRAIL- or AMG655-induced apoptosis. Therefore, it is clear that b-AP15, and possibly its derivatives, can stabilize DR5 and increase functional cell surface DR5 levels, resulting in enhancement of DR5 activation-induced apoptosis. Our findings suggest that b-AP15 and its derivatives may have potential in sensitizing cancer cells to DR5 activation-based cancer therapy.


Apoptosis/drug effects , Piperidones/pharmacology , Protease Inhibitors/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , HCT116 Cells , Humans , Protein Stability , Proteolysis
10.
Cell Commun Signal ; 15(1): 18, 2017 05 08.
Article En | MEDLINE | ID: mdl-28482915

BACKGROUND: Death receptor (DR5), a well-characterized death domain-containing cell surface pro-apoptotic protein, has been suggested to suppress cancer cell invasion and metastasis. However, the underlying mechanisms have not been fully elucidated. Our recent work demonstrates that DR5 suppression promotes cancer cell invasion and metastasis through caspase-8/TRAF2-mediated activation of ERK and JNK signaling and MMP1 elevation. The current study aimed at addressing the mechanism through which TRAF2 is activated in a caspase-8 dependent manner. RESULTS: DR5 knockdown increased TRAF2 polyubiquitination, a critical event for TRAF2-mediated JNK/AP-1 activation. Suppression of sphingosine-1-phosphate (S1P) generation or depletion of casapse-8 inhibited not only enhancement of cell invasion, but also elevation and polyubiquitination of TRAF2, activation of JNK/AP-1 activation and increased expression of MMP1 induced by DR5 knockdown. CONCLUSIONS: Both S1P and caspase-8 are critical for TRAF2 stabilization, polyubiquitination, subsequent activation of JNK/AP1 signaling and MMP1 expression and final promotion of cell invasion.


JNK Mitogen-Activated Protein Kinases/metabolism , Lysophospholipids/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Sphingosine/analogs & derivatives , TNF Receptor-Associated Factor 2/metabolism , Transcription Factor AP-1/metabolism , Ubiquitination , Caspase 8/metabolism , Cell Line, Tumor , Enzyme Activation , Gene Knockdown Techniques , Humans , Matrix Metalloproteinase 1/metabolism , Neoplasm Invasiveness , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/deficiency , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Signal Transduction , Sphingosine/metabolism
11.
J Biol Chem ; 291(41): 21694-21702, 2016 Oct 07.
Article En | MEDLINE | ID: mdl-27576686

Death receptor 4 (DR4) is a cell surface receptor for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and triggers apoptosis upon ligation with TRAIL or aggregation. MEK/ERK signaling is a well known and the best-studied effector pathway downstream of Ras and Raf. This study focuses on determining the impact of pharmacological MEK inhibition on DR4 expression and elucidating the underlying mechanism. We found that several MEK inhibitors including MEK162, AZD6244, and PD0325901 effectively decreased DR4 protein levels including cell surface DR4 in different cancer cell lines. Accordingly, pre-treatment of TRAIL-sensitive cancer cell lines with a MEK inhibitor desensitized them to TRAIL-induced apoptosis. These results indicate that MEK inhibition negatively regulates DR4 expression and cell response to TRAIL-induced apoptosis. MEK inhibitors did not alter DR4 protein stability, rather decreased its mRNA levels, suggesting a transcriptional regulation. In contrast, enforced activation of MEK/ERK signaling by expressing ectopic B-Raf (V600E) or constitutively activated MEK1 (MEK1-CA) or MEK2 (MEK2-CA) activated ERK and increased DR4 expression; these effects were inhibited when a MEK inhibitor was present. Promoter analysis through deletion and mutation identified the AP-1 binding site as an essential response element for enhancing DR4 transactivation by MEK1-CA. Furthermore, inhibition of AP-1 by c-Jun knockdown abrogated the ability of MEK1-CA to increase DR4 promoter activity and DR4 expression. These results suggest an essential role of AP-1 in mediating MEK/ERK activation-induced DR4 expression. Our findings together highlight a previously undiscovered mechanism that positively regulates DR4 expression through activation of the MEK/ERK/AP-1 signaling pathway.


Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Transcription Factor AP-1/metabolism , Transcriptional Activation , Amino Acid Substitution , Cell Line, Tumor , HEK293 Cells , Humans , Mutation, Missense , Neoplasms/genetics , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transcription Factor AP-1/genetics
12.
Cancer Lett ; 380(2): 494-504, 2016 10 01.
Article En | MEDLINE | ID: mdl-27450722

The 3rd generation epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs; e.g., AZD9291), which selectively and irreversibly inhibit EGFR activating and T790M mutants, represent very promising therapeutic options for patients with non-small cell lung cancer (NSCLC) that has become resistant to 1st generation EGFR-TKIs due to T790M mutation. However, eventual resistance to the 3rd generation EGFR-TKIs has already been described in the clinic, resulting in disease progression. Therefore, there is a great challenge and urgent need to understand how this resistance occurs and to develop effective strategies to delay or overcome the resistance. The current study has demonstrated that Met amplification and hyperactivation is a resistance mechanism to both 1st and 3rd generation EGFR-TKIs since both erlotinib- and AZD9291-resistant HCC827 cell lines possessed amplified Met gene and hyperactivated Met, and were cross-resistant to AZD9291 or erlotinib. Met inhibition overcame the resistance of these cell lines to AZD9291 both in vitro and in vivo, including enhancement of apoptosis or G1 cell cycle arrest. Hence, we suggest that Met inhibition is also an effective strategy to overcome resistance of certain EGFR-mutated NSCLCs with Met amplification to AZD9291, warranting the further clinical validation of our findings.


Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Gene Amplification , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Acrylamides/pharmacology , Aniline Compounds/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Crizotinib , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Nude , Molecular Targeted Therapy , Mutation , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA Interference , Signal Transduction/drug effects , Time Factors , Transfection , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
13.
Sci Rep ; 6: 26803, 2016 05 25.
Article En | MEDLINE | ID: mdl-27222248

B-Raf inhibitors have been used for the treatment of some B-Raf-mutated cancers. They effectively inhibit B-Raf/MEK/ERK signaling in cancers harboring mutant B-Raf, but paradoxically activates MEK/ERK in Ras-mutated cancers. Death receptor 5 (DR5), a cell surface pro-apoptotic protein, triggers apoptosis upon ligation with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) or aggregation. This study focused on determining the effects of B-Raf inhibition on DR5 expression and DR5 activation-induced apoptosis in Ras-mutant cancer cells. Using chemical and genetic approaches, we have demonstrated that the B-Raf inhibitor PLX4032 induces DR5 upregulation exclusively in Ras-mutant cancer cells; this effect is dependent on Ras/c-Raf/MEK/ERK signaling activation. PLX4032 induces DR5 expression at transcriptional levels, largely due to enhancing CHOP/Elk1-mediated DR5 transcription. Pre-exposure of Ras-mutated cancer cells to PLX4032 sensitizes them to TRAIL-induced apoptosis; this is also a c-Raf/MEK/ERK-dependent event. Collectively, our findings highlight a previously undiscovered effect of B-Raf inhibition on the induction of DR5 expression and the enhancement of DR5 activation-induced apoptosis in Ras-mutant cancer cells and hence may suggest a novel therapeutic strategy against Ras-mutated cancer cells by driving their death due to DR5-dependent apoptosis through B-Raf inhibition.


Gene Expression Regulation, Neoplastic/drug effects , Genes, ras , Indoles/pharmacology , MAP Kinase Signaling System/drug effects , Neoplasm Proteins/biosynthesis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , Sulfonamides/pharmacology , Apoptosis/drug effects , Benzimidazoles/pharmacology , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Imidazoles/pharmacology , Neoplasm Proteins/genetics , Oximes/pharmacology , Proto-Oncogene Proteins B-raf/physiology , Proto-Oncogene Proteins c-raf/physiology , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Recombinant Proteins/pharmacology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Transcription Factor CHOP/physiology , Transcription, Genetic/drug effects , Vemurafenib , ets-Domain Protein Elk-1/physiology
14.
Oncotarget ; 6(38): 41324-38, 2015 Dec 01.
Article En | MEDLINE | ID: mdl-26510914

The role of death receptor 5 (DR5), a well-known cell surface pro-apoptotic protein, in the negative regulation of invasion and metastasis of human cancer cells and the underlying mechanisms are largely unknown and were hence the focus of this study. In this report, we have demonstrated that DR5 functions to suppress invasion and metastasis of human cancer cells, as evidenced by enhanced cancer cell invasion and metastasis upon genetic suppression of DR5 either by gene knockdown or knockout. When DR5 is suppressed, FADD and caspase-8 may recruit and stabilize TRAF2 to form a metastasis and invasion signaling complex, resulting in activation of ERK and JNK/AP-1 signaling that mediate the elevation and activation of matrix metalloproteinase-1 (MMP1) and eventual promotion of cancer invasion and metastasis. Our findings thus highlight a novel non-apoptotic function of DR5 as a suppressor of human cancer cell invasion and metastasis and suggest a basic working model elucidating the underlying biology.


Caspase 8/metabolism , MAP Kinase Signaling System , Neoplasms/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF Receptor-Associated Factor 2/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Enzyme Activation , Fas-Associated Death Domain Protein/metabolism , Female , HCT116 Cells , HEK293 Cells , Humans , Matrix Metalloproteinase 1/metabolism , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/therapy , RNA Interference , RNAi Therapeutics/methods , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Xenograft Model Antitumor Assays/methods
15.
Oncotarget ; 6(19): 17532-42, 2015 Jul 10.
Article En | MEDLINE | ID: mdl-26009898

Carfilzomib (CFZ) is a second generation proteasome inhibitor approved for the treatment of patients with multiple myeloma. It induces apoptosis in human cancer cells; but the underlying mechanisms remain undefined. In the present study, we show that CFZ decreases the survival of several human cancer cell lines and induces apoptosis. Induction of apoptosis by CFZ occurs, at least in part, due to activation of the extrinsic apoptotic pathway, since FADD deficiency protected cancer cells from undergoing apoptosis. CFZ increased total and cell surface levels of DR5 in different cancer cell lines; accordingly it enhanced TRAIL-induced apoptosis. DR5 deficiency protected cancer cells from induction of apoptosis by CFZ either alone or in combination with TRAIL. These data together convincingly demonstrate that DR5 upregulation is a critical mechanism accounting for CFZ-induced apoptosis and enhancement of TRAIL-induced apoptosis. CFZ inhibited the degradation of DR5, suggesting that DR5 stabilization contributes to CFZ-induced DR5 upregulation. In summary, the present study highlights the important role of DR5 upregulation in CFZ-induced apoptosis and enhancement of TRAIL-induced apoptosis in human cancer cells.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Oligopeptides/pharmacology , Proteasome Inhibitors/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Blotting, Western , Cell Line, Tumor , Humans , Polymerase Chain Reaction , RNA, Small Interfering , Transfection , Up-Regulation
16.
J Biol Chem ; 287(1): 257-267, 2012 Jan 02.
Article En | MEDLINE | ID: mdl-22065586

Oncogenic mutations of ras and B-raf frequently occur in many cancer types and are critical for cell transformation and tumorigenesis. Death receptor 5 (DR5) is a cell surface pro-apoptotic death receptor for tumor necrosis factor-related apoptosis-inducing ligand and has been targeted in cancer therapy. The current study has demonstrated induction of DR5 expression by the oncogenic proteins Ras and B-Raf and revealed the underlying mechanisms. We demonstrated that both Ras and B-Raf induce DR5 expression by enforced expression of oncogenic Ras (e.g. H-Ras12V or K-Ras12V) or B-Raf (i.e. V600E) in cells and by analyzing gene expression array data generated from cancer cell lines and from human cancer tissues. This finding is further supported by our results that knockdown of endogenous K-Ras or B-Raf (V600E) reduced the expression of DR5. Importantly, we have elucidated that Ras induces DR5 expression through co-activation of ERK/RSK and JNK signaling pathways and subsequent cooperative effects among the transcriptional factors CHOP, Elk1, and c-Jun to enhance DR5 gene transcription. Moreover, we found that the majority of cancer cell lines highly sensitive to the DR5 agonistic antibody AMG655 have either Ras or B-Raf mutations. Our findings warrant further study on the biology of DR5 regulation by Ras and B-Raf, which may provide new insight into the biology of Ras and B-Raf, and on the potential impact of Ras or B-Raf mutations on the outcome of DR5-targeted cancer therapy.


Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation, Neoplastic , JNK Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Signal Transduction , ras Proteins/metabolism , Cell Line, Tumor , Humans , Mutation , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins B-raf/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Ribosomal Protein S6 Kinases/metabolism , Transcription Factor CHOP/metabolism , Transcription, Genetic , ets-Domain Protein Elk-1/metabolism
17.
Acta Virol ; 55(2): 155-9, 2011.
Article En | MEDLINE | ID: mdl-21692564

Many human immunodeficiency virus 1 (HIV-1) researchers focus on the developing new anti-reservoir therapy to eradicate HIV-1 provirus from the HIV-1-infected patients. HIV-1 provirus is the major obstacle for effective HIV-1 treatment because it integrates into the host genome and can produce a virus progeny after stopping highly active antiretroviral therapy (HAART). We established two novel cell lines latently infected with HIV-1 by limiting dilution cloning of A3.01 cells infected with HIV-1. Analysis of the flanking sequence of HIV-1 proviral DNA integrated into chromosomal cellular DNA revealed that proviral DNA was inserted into different sites of different chromosomes in the two examined cell lines. In these lines, virus reactivation could be induced by a phorbol 12-myristate 13-acetate (PMA) treatment that resulted in a marked increase of the production HIV-1 p24 antigen and appearance of the infectious virus. The novel cell lines latently infected with HIV-1 represent further tool for the study of molecular mechanisms of viral latency and development of anti-reservoir therapy of HIV-1 infection.


Cell Line/virology , HIV-1/physiology , Virus Latency , HIV Infections/virology , HIV-1/genetics , Humans , Virus Cultivation
18.
J Biol Chem ; 285(53): 41310-9, 2010 Dec 31.
Article En | MEDLINE | ID: mdl-21044953

Death receptor 5 (DR5) is a death domain-containing transmembrane receptor that triggers apoptosis upon binding to its ligand or when overexpressed. Its expression is induced by certain small molecule drugs, including celecoxib, through mechanisms that have not been fully elucidated. The current study has revealed a novel ERK/ribosomal S6 kinase (RSK)-dependent mechanism that regulates DR5 expression primarily using celecoxib as a DR5 inducer. Both C/EBP homologous protein (CHOP) and Elk1 are required for celecoxib-induced DR5 expression based on promoter deletion and mutation analysis and siRNA-mediated gene silencing results. Co-expression of both CHOP and Elk1 exhibited enhanced effects on increasing DR5 promoter activity and DR5 expression, indicating that CHOP and Elk1 co-operatively regulate DR5 expression. Because Elk1 is an ERK-regulated protein, we accordingly found that celecoxib increased the levels of phosphorylated ERK1/2, RSK2, and Elk1. Inhibition of either ERK signaling with a MEK inhibitor or ERK1/2 siRNA, or RSK2 signaling with an RSK2 inhibitor or RSK2 siRNA abrogated DR5 up-regulation by celecoxib as well as other agents. Moreover, these inhibitions suppressed celecoxib-induced CHOP up-regulation. Thus, ERK/RSK-dependent, CHOP and Elk1-mediated mechanisms are critical for DR5 induction. Additionally, celecoxib increased CHOP promoter activity in an ATF4-dependent manner, and siRNA-mediated blockade of ATF4 abrogated both CHOP induction and DR5 up-regulation, indicating that ATF4 is involved in celecoxib-induced CHOP and DR5 expression. Collectively, we conclude that small molecules such as celecoxib induce DR5 expression through activating ERK/RSK signaling and subsequent Elk1 activation and ATF4-dependent CHOP induction.


Activating Transcription Factor 4/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Pyrazoles/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/physiology , Sulfonamides/pharmacology , Transcription Factor CHOP/metabolism , ets-Domain Protein Elk-1/genetics , ets-Domain Protein Elk-1/metabolism , Apoptosis , Celecoxib , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Silencing , Humans , Signal Transduction
19.
AIDS ; 24(4): 609-11, 2010 Feb 20.
Article En | MEDLINE | ID: mdl-20154582

Histone deacetylase plays an important role in HIV latency. Novel histone deacetylase inhibitors, CG05 and CG06, were evaluated for their roles in HIV latency using ACH2 cells. Both inhibitors were highly efficient in reactivation of provirus and exerted lesser toxicity compared with other known histone deacetylase inhibitors. Histone acetylation increased when proviruses were reactivated by the compounds. These new inhibitors may contribute to the reduction of the HIV reservoir when used in conjunction with highly active antiretroviral therapy.


HIV Infections/drug therapy , HIV-1/drug effects , Histone Deacetylase Inhibitors/pharmacology , Virus Latency/drug effects , Antiretroviral Therapy, Highly Active , HIV Infections/genetics , HIV-1/genetics , HIV-1/physiology , Histone Deacetylases/metabolism , Humans , Virus Activation/drug effects , Virus Latency/genetics
20.
Apoptosis ; 12(7): 1339-47, 2007 Jul.
Article En | MEDLINE | ID: mdl-17431791

Cyclin-dependent kinase (CDK) inhibitor p21(WAF1/CIP1(-/-))-null mice have an increased incidence of tumor formation. Here, we demonstrate that p21(WAF1/CIP1) is unstable in HeLa cells treated with siRNA duplexes that target PKCdelta. PKCdelta phosphorylates p21(WAF1/CIP1 )at a serine residue ((146)Ser) located in its C-terminal domain. In cells treated with 12-O-tetradecanoylphorbol 13-acetate, the levels of both p21(WAF1/CIP1) and its (146)Ser-phosphorylated form increased significantly. We also show that a substitution, resulting from a single nucleotide polymorphism (SNP) at (149)Asp found in certain cancer patients, strongly compromises PKCdelta-mediated phosphorylation at (146)Ser and results in cells that are relatively resistant to TNFalpha-induced apoptosis. Thus, post-translational phosphorylation of p21(WAF1/CIP1) is important from an apoptotic cell death, and may also have patho-physiological relevance for the development of human cancer.


Apoptosis/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Protein Kinase C-delta/metabolism , Animals , Caspase 3/metabolism , Cell Line , HeLa Cells , Humans , Mice , Phosphorylation , Tumor Necrosis Factor-alpha/pharmacology
...