Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
EMBO J ; 40(12): e106412, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33988249

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) integrates nutrients, growth factors, stress, and energy status to regulate cell growth and metabolism. Amino acids promote mTORC1 lysosomal localization and subsequent activation. However, the subcellular location or interacting proteins of mTORC1 under amino acid-deficient conditions is not completely understood. Here, we identify ADP-ribosylation factor GTPase-activating protein 1 (ArfGAP1) as a crucial regulator of mTORC1. ArfGAP1 interacts with mTORC1 in the absence of amino acids and inhibits mTORC1 lysosomal localization and activation. Mechanistically, the membrane curvature-sensing amphipathic lipid packing sensor (ALPS) motifs that bind to vesicle membranes are crucial for ArfGAP1 to interact with and regulate mTORC1 activity. Importantly, ArfGAP1 represses cell growth through mTORC1 and is an independent prognostic factor for the overall survival of pancreatic cancer patients. Our study identifies ArfGAP1 as a critical regulator of mTORC1 that functions by preventing the lysosomal transport and activation of mTORC1, with potential for cancer therapeutics.


Subject(s)
GTPase-Activating Proteins/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Pancreatic Neoplasms/metabolism , Animals , Cell Line , Female , GTPase-Activating Proteins/genetics , Humans , Kaplan-Meier Estimate , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Prognosis
2.
Proc Natl Acad Sci U S A ; 119(20): e2123261119, 2022 05 17.
Article in English | MEDLINE | ID: mdl-35561222

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) senses amino acids to control cell growth, metabolism, and autophagy. Some amino acids signal to mTORC1 through the Rag GTPase, whereas glutamine and asparagine activate mTORC1 through a Rag GTPase-independent pathway. Here, we show that the lysosomal glutamine and asparagine transporter SNAT7 activates mTORC1 after extracellular protein, such as albumin, is macropinocytosed. The N terminus of SNAT7 forms nutrient-sensitive interaction with mTORC1 and regulates mTORC1 activation independently of the Rag GTPases. Depletion of SNAT7 inhibits albumin-induced mTORC1 lysosomal localization and subsequent activation. Moreover, SNAT7 is essential to sustain KRAS-driven pancreatic cancer cell growth through mTORC1. Thus, SNAT7 links glutamine and asparagine signaling from extracellular protein to mTORC1 independently of the Rag GTPases and is required for macropinocytosis-mediated mTORC1 activation and pancreatic cancer cell growth.


Subject(s)
Amino Acid Transport Systems, Neutral , Lysosomes , Mechanistic Target of Rapamycin Complex 1 , Pinocytosis , Amino Acid Transport Systems, Neutral/chemistry , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Asparagine/metabolism , Glutamine/metabolism , Humans , Lysosomes/enzymology , Mechanistic Target of Rapamycin Complex 1/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction
3.
PLoS Genet ; 17(10): e1009832, 2021 10.
Article in English | MEDLINE | ID: mdl-34673774

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) senses multiple stimuli to regulate anabolic and catabolic processes. mTORC1 is typically hyperactivated in multiple human diseases such as cancer and type 2 diabetes. Extensive research has focused on signaling pathways that can activate mTORC1 such as growth factors and amino acids. However, less is known about signaling cues that can directly inhibit mTORC1 activity. Here, we identify A-kinase anchoring protein 13 (AKAP13) as an mTORC1 binding protein, and a crucial regulator of mTORC1 inhibition by G-protein coupled receptor (GPCR) signaling. GPCRs paired to Gαs proteins increase cyclic adenosine 3'5' monophosphate (cAMP) to activate protein kinase A (PKA). Mechanistically, AKAP13 acts as a scaffold for PKA and mTORC1, where PKA inhibits mTORC1 through the phosphorylation of Raptor on Ser 791. Importantly, AKAP13 mediates mTORC1-induced cell proliferation, cell size, and colony formation. AKAP13 expression correlates with mTORC1 activation and overall lung adenocarcinoma patient survival, as well as lung cancer tumor growth in vivo. Our study identifies AKAP13 as an important player in mTORC1 inhibition by GPCRs, and targeting this pathway may be beneficial for human diseases with hyperactivated mTORC1.


Subject(s)
A Kinase Anchor Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Minor Histocompatibility Antigens/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction/physiology , A549 Cells , Animals , Cell Line , Cell Line, Tumor , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , HCT116 Cells , HEK293 Cells , Humans , Mice , PC-3 Cells , Phosphorylation/physiology
4.
Biochem Soc Trans ; 51(2): 655-664, 2023 04 26.
Article in English | MEDLINE | ID: mdl-36929165

ABSTRACT

The Rag GTPases are an evolutionarily conserved family that play a crucial role in amino acid sensing by the mammalian target of rapamycin complex 1 (mTORC1). mTORC1 is often referred to as the master regulator of cell growth. mTORC1 hyperactivation is observed in multiple diseases such as cancer, obesity, metabolic disorders, and neurodegeneration. The Rag GTPases sense amino acid levels and form heterodimers, where RagA or RagB binds to RagC or RagD, to recruit mTORC1 to the lysosome where it becomes activated. Here, we review amino acid signaling to mTORC1 through the Rag GTPases.


Subject(s)
Monomeric GTP-Binding Proteins , Multiprotein Complexes , Multiprotein Complexes/metabolism , Monomeric GTP-Binding Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction/physiology , Amino Acids/metabolism , Lysosomes/metabolism
5.
Int J Mol Sci ; 24(2)2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36674523

ABSTRACT

Three-dimensional (3D) culture platforms have been adopted in a high-throughput screening (HTS) system to mimic in vivo physiological microenvironments. The automated dispenser has been established commercially to enable spotting or distributing non-viscous or viscous biomaterials onto microplates. However, there are still challenges to the precise and accurate dispensation of cells embedded in hydrogels such as Alginate- and Matrigel-extracellular matrices. We developed and improved an automated contact-free dispensing machine, the ASFA SPOTTER (V5 and V6), which is compatible with 96- and 384-pillar/well plates and 330- and 532-micropillar/well chips for the support of 3D spheroid/organoid models using bioprinting techniques. This enables the distribution of non-viscous and viscous biosamples, including chemical drugs and cancer cells, for large-scale drug screening at high speed and small volumes (20 to 4000 nanoliters) with no damage to cells. The ASFA SPOTTER (V5 and V6) utilizes a contact-free method that minimizes cross-contamination for the dispensation of encapsulated tissue cells with highly viscous scaffolds (over 70%). In particular, the SPOTTER V6 does not require a washing process and offers the advantage of almost no dead volume (defined as additional required sample volume, including a pre-shot and flushing shot for dispensing). It can be successfully applied for the achievement of an organoid culture in automation, with rapid and easy operation, as well as miniaturization for high-throughput screening. In this study, we report the advantages of the ASFA SPOTTER, which distributes standard-sized cell spots with hydrogels onto a 384-pillar/well plate with a fast dispensing speed, small-scale volume, accuracy, and precision.


Subject(s)
High-Throughput Screening Assays , Neoplasms , Humans , High-Throughput Screening Assays/methods , Cell Culture Techniques/methods , Drug Evaluation, Preclinical/methods , Hydrogels , Spheroids, Cellular , Tumor Microenvironment
6.
Proc Natl Acad Sci U S A ; 115(18): 4672-4677, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29588416

ABSTRACT

Programmed cell death 5 (PDCD5) has been associated with human cancers as a regulator of cell death; however, the role of PDCD5 in the endothelium has not been revealed. Thus, we investigated whether PDCD5 regulates protein kinase B (PKB/AKT)-endothelial nitric oxide synthase (eNOS)-dependent signal transduction in the endothelium and affects atherosclerosis. Endothelial-specific PDCD5 knockout mice showed significantly reduced vascular remodeling compared with wild-type (WT) mice after partial carotid ligation. WT PDCD5 competitively inhibited interaction between histone deacetylase 3 (HDAC3) and AKT, but PDCD5L6R, an HDAC3-binding-deficient mutant, did not. Knockdown of PDCD5 accelerated HDAC3-AKT interaction, AKT and eNOS phosphorylation, and nitric oxide (NO) production in human umbilical vein endothelial cells. Moreover, we found that serum PDCD5 levels reflect endothelial NO production and are correlated with diabetes mellitus, high-density lipoprotein cholesterol, and coronary calcium in human samples obtained from the cardiovascular high-risk cohort. Therefore, we conclude that PDCD5 is associated with endothelial dysfunction and may be a novel therapeutic target in atherosclerosis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vascular Remodeling , Animals , Apoptosis Regulatory Proteins/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cholesterol, HDL/genetics , Cholesterol, HDL/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Endothelium, Vascular/pathology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics
7.
J Cell Physiol ; 234(5): 6428-6436, 2019 05.
Article in English | MEDLINE | ID: mdl-30317579

ABSTRACT

The role of histone deacetylase 3 (HDAC3) is to repress the expression of various genes by eliminating acetyl group from histone. Thus, the regulation of HDAC3 activity is essential to maintain cellular homeostasis. In this study, we found that HDAC3 interacts with c-Src kinase. However, the interaction between HDAC3 and c-Src was previously reported, it has still been ambiguous whether c-Src phosphorylates HDAC3 and affects the function of HDAC3. First, we confirmed that HDAC3 directly binds to c-Src, and c-Src identified to interact with C-terminal domain (277-428 a.a.) of HDAC3. c-Src also phosphorylated three tyrosine sites of HDAC3 at tyrosine 325, 328, and 331. Importantly, wild-type c-Src increases HDAC3 activity, but not mutant c-SrcK298M (kinase inactive form). When these tyrosine residues are all substituted for alanine residues, the deacetylase activity of mutant HDAC3 was abolished. In addition, a proliferation of HER2-positive breast cancer cells expressing phosphorylation deficient mutant HDAC3 is decreased in comparison with control cells. Thus, our findings suggested that phosphorylation of HDAC3 by c-Src kinase regulates the HDAC3 activity and the proliferation of breast cancer cells.


Subject(s)
Breast Neoplasms/pathology , CSK Tyrosine-Protein Kinase/metabolism , Cell Proliferation/physiology , Histone Deacetylases/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Phosphorylation , Receptor, ErbB-2/genetics , Tyrosine/metabolism
8.
J Cell Physiol ; 234(3): 2649-2658, 2019 03.
Article in English | MEDLINE | ID: mdl-30144069

ABSTRACT

Although programed cell death 5 (PDCD5) is an important protein in p53-mediated proapoptotic signaling, very little is known about PDCD5-related cell death. In this study, we report that serine/threonine kinase 31 (STK31) interacts with PDCD5, which maintains the stability of PDCD5. STK31 overexpression significantly activated PDCD5 stabilization and p53-mediated apoptosis in response to etoposide (ET). However, STK31 knockdown did not enhance apoptosis by ET treatment. Moreover, when STK31 was depleted, PDCD5 inhibited the activation of the p53 signaling pathway with ET, indicating that the PDCD5-STK31 network has an essential role in p53 activation. Importantly, STK31 activated the p53 signaling pathway by genotoxic stress through positive regulation of PDCD5-mediated apoptosis. We thus demonstrated that overexpression of STK31 greatly inhibited tumorigenic growth and increased the chemosensitivity of HCT116 human colorectal carcinoma cells. Taken together, these findings demonstrate that the STK31-PDCD5 complex network regulates apoptosis of cancer cells, and STK31 is a positive apoptosis regulator that inhibits tumorigenesis of colon cancer cells by inducing PDCD5-mediated apoptosis in response to genotoxic stress.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Etoposide/pharmacology , Protein Serine-Threonine Kinases/drug effects , Tumor Suppressor Protein p53/drug effects , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , DNA Damage/drug effects , Humans , Neoplasm Proteins/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism
9.
J Cell Physiol ; 228(6): 1159-65, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23129261

ABSTRACT

Protein kinase A (PKA) phosphorylates diverse protein substrates to modulate their function. In this study, we found that PKA specifically phosphorylates the RD1 (repression domain 1) domain of nuclear receptor corepressor (NCoR). We demonstrated that the Serine-70 of NCoR is identified the critical amino acid for PKA-dependent NCoR phosphorylation. Importantly, we found that PKA-dependent phosphorylation enhances the nuclear translocation of NCoR. More importantly, the activation of PKA enhanced the repressive activity of NCoR in a reporter assay and potentiated the antagonist activity in the androgen receptor (AR)-mediated transcription. Taken together, these results uncover a regulatory mechanism by which PKA positively modulates NCoR function in transcriptional regulation in prostate cancer.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Prostatic Neoplasms/enzymology , Active Transport, Cell Nucleus , Enzyme Activation , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Kallikreins/genetics , Kallikreins/metabolism , Male , Mutation , Nuclear Receptor Co-Repressor 1/genetics , Phosphorylation , Promoter Regions, Genetic , Prostate-Specific Antigen/genetics , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/genetics , Protein Binding , Protein Structure, Tertiary , RNA Interference , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Serine , Signal Transduction , Transcription, Genetic , Transfection
10.
JCI Insight ; 8(13)2023 07 10.
Article in English | MEDLINE | ID: mdl-37427586

ABSTRACT

The mammalian target of rapamycin complex 1 (mTORC1) senses multiple upstream stimuli to orchestrate anabolic and catabolic events that regulate cell growth and metabolism. Hyperactivation of mTORC1 signaling is observed in multiple human diseases; thus, pathways that suppress mTORC1 signaling may help to identify new therapeutic targets. Here, we report that phosphodiesterase 4D (PDE4D) promotes pancreatic cancer tumor growth by increasing mTORC1 signaling. GPCRs paired to Gαs proteins activate adenylyl cyclase, which in turn elevates levels of 3',5'-cyclic adenosine monophosphate (cAMP), whereas PDEs catalyze the hydrolysis of cAMP to 5'-AMP. PDE4D forms a complex with mTORC1 and is required for mTORC1 lysosomal localization and activation. Inhibition of PDE4D and the elevation of cAMP levels block mTORC1 signaling via Raptor phosphorylation. Moreover, pancreatic cancer exhibits an upregulation of PDE4D expression, and high PDE4D levels predict the poor overall survival of patients with pancreatic cancer. Importantly, FDA-approved PDE4 inhibitors repress pancreatic cancer cell tumor growth in vivo by suppressing mTORC1 signaling. Our results identify PDE4D as an important activator of mTORC1 and suggest that targeting PDE4 with FDA-approved inhibitors may be beneficial for the treatment of human diseases with hyperactivated mTORC1 signaling.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4 , Pancreatic Neoplasms , Humans , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Pancreatic Neoplasms/drug therapy , Proteins , Signal Transduction
11.
Sci Rep ; 11(1): 20812, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675258

ABSTRACT

Translationally controlled tumor protein (TCTP) is expressed in many tissues, particularly in human tumors. It plays a role in malignant transformation, apoptosis prevention, and DNA damage repair. The signaling mechanisms underlying TCTP regulation in cancer are only partially understood. Here, we investigated the role of mTORC1 in regulating TCTP protein levels, thereby modulating chemosensitivity, in human lung cancer cells and an A549 lung cancer xenograft model. The inhibition of mTORC1, but not mTORC2, induced ubiquitin/proteasome-dependent TCTP degradation without a decrease in the mRNA level. PLK1 activity was required for TCTP ubiquitination and degradation and for its phosphorylation at Ser46 upon mTORC1 inhibition. Akt phosphorylation and activation was indispensable for rapamycin-induced TCTP degradation and PLK1 activation, and depended on S6K inhibition, but not mTORC2 activation. Furthermore, the minimal dose of rapamycin required to induce TCTP proteolysis enhanced the efficacy of DNA-damaging drugs, such as cisplatin and doxorubicin, through the induction of apoptotic cell death in vitro and in vivo. This synergistic cytotoxicity of these drugs was induced irrespective of the functional status of p53. These results demonstrate a new mechanism of TCTP regulation in which the mTORC1/S6K pathway inhibits a novel Akt/PLK1 signaling axis and thereby induces TCTP protein stabilization and confers resistance to DNA-damaging agents. The results of this study suggest a new therapeutic strategy for enhancing chemosensitivity in lung cancers regardless of the functional status of p53.


Subject(s)
Antineoplastic Agents/pharmacology , Lung Neoplasms/drug therapy , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction/drug effects , Tumor Protein, Translationally-Controlled 1/metabolism , A549 Cells , Animals , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/metabolism , DNA Damage/drug effects , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Protein Serine-Threonine Kinases/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ribosomal Protein S6 Kinases/metabolism , Polo-Like Kinase 1
12.
Clin Cancer Res ; 25(12): 3617-3629, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30814110

ABSTRACT

PURPOSE: Because of disease heterogeneity, limited studies on effective chemotherapies and therapeutic agents for advanced gastric cancer are available. Erythrocyte membrane protein band 4.1-like 5 (EPB41L5) has critical roles in renal and breast cancer metastasis. However, its role in metastatic gastric cancer remains unknown. EXPERIMENTAL DESIGN: The specimens of 78 gastric cancer patients were analyzed by oligonucleotide microarray and survival analysis. In vitro experiments and metastatic mice models were used to assess the effects of EPB41L5 on gastric cancer metastasis. RESULTS: Gastric cancer patients with high EPB41L5 levels had poor prognosis and low survival rate. Further, TGFß1-induced EPB41L5 expression promoted gastric cancer cell migration and invasion by Smad-dependent TGFß signaling. Phospho-Smad3 recruitment to the EPB41L5 promoter was significantly inhibited by a TGFß inhibitor. EPB41L5 overexpression increased lung metastasis of gastric cancer cells in nude mice, which was completely reversed by anti-EPB41L5 monoclonal antibody treatment. Importantly, p120-catenin knockdown abolished EPB41L5-enhanced gastric cancer cell metastasis. Anti-EPB41L5 monoclonal antibody treatment blocked the association of EPB41L5 with p120-catenin. CONCLUSIONS: TGFß/EPB41L5/p120-catenin axis regulates gastric cancer cell metastasis, and EPB41L5 is a promising therapeutic target for advanced gastric cancer.


Subject(s)
Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Membrane Proteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transforming Growth Factor beta/metabolism , Animals , Cell Line, Tumor , Cell Movement/physiology , Chick Embryo , Female , Heterografts , Humans , Lung Neoplasms/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis , Promoter Regions, Genetic , Signal Transduction , Stomach Neoplasms/genetics , Survival Rate
13.
BMB Rep ; 51(5): 255-260, 2018 May.
Article in English | MEDLINE | ID: mdl-29555015

ABSTRACT

Wntless/GPR177 functions as WNT ligand carrier protein and activator of WNT/ß-catenin signaling, however, its molecular role in gastric cancer (GC) has remained elusive. We investigated the role of GPR177 in gastric tumorigenesis and provided the therapeutic potential of a clinical development of anti-GPR177 monoclonal antibodies. GPR177 mRNA expression was assessed in GC transcriptome data sets (GSE15459, n = 184; GSE66229, n = 300); protein expression was assessed in independent patient tumor tissues (Yonsei TMA, n = 909). GPR177 expression were associated with unfavorable prognosis [log-rank test, GSE15459 (P = 0.00736), GSE66229 (P = 0.0142), and Yonsei TMA (P = 0.0334)] and identified as an independent risk predictor of clinical outcomes: GSE15459 [hazard ratio (HR) 1.731 (95% confidence interval; CI; 1.103- 2.715), P = 0.017], GSE66229 [HR 1.54 (95% CI, 1.10-2.151), P = 0.011], and Yonsei TMA [HR 1.254 (95% CI, 1.049- 1.500), P = 0.013]. Either antibody treatment or GPR177 knockdown suppressed proliferation of GC cells and sensitized cells to apoptosis. And also inhibition of GPR177 suppresses in vitro and in vivo tumorogenesis in GC cells and inhibits WNT/ß-catenin signaling. Finally, targeting and inhibition of GPR177 with antibody suppressed tumorigenesis in PDX model. Together, these results suggest GPR177 as a novel candidate for prognostic marker as well as a promising target for treatment of GC patients. [BMB Reports 2018; 51(5): 255-260].


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Animals , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Humans , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
14.
Oncotarget ; 7(35): 56767-56780, 2016 Aug 30.
Article in English | MEDLINE | ID: mdl-27462923

ABSTRACT

Delphinidin is a major anthocyanidin compound found in various fruits. It has anti-inflammatory, anti-oxidant, and various other biological activities. In this study, we identified the epigenetic modulators that mediate the apoptotic effect of delphinidin in human prostate cancer cells. We found that treatment of LNCaP cells (a p53 wild-type, human prostate cancer cell line) with delphinidin increased caspase-3, -7, and -8 activity, whereas it decreased histone deacetylase activity. Among class I HDACs, the activity of HDAC3 was specifically inhibited by delphinidin. Moreover, the induction of apoptosis by delphinidin was dependent on caspase-mediated cleavage of HDAC3, which results in the acetylation and stabilization of p53. We also observed that delphinidin potently upregulated pro-apoptotic genes that are positively regulated by p53, and downregulated various anti-apoptotic genes. Taken together, these results show that delphinidin induces p53-mediated apoptosis by suppressing HDAC activity and activating p53 acetylation in human prostate cancer LNCaP cells. Therefore, delphinidin may be useful in the prevention of prostate cancer.


Subject(s)
Anthocyanins/pharmacology , Apoptosis , Histone Deacetylases/metabolism , Prostatic Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Antineoplastic Agents, Phytogenic/pharmacology , Caspase 3/metabolism , Caspase 7/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival , Epigenesis, Genetic , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Humans , Male , Plant Extracts/pharmacology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , RNA Interference , Tumor Suppressor Protein p53/genetics
15.
Oncotarget ; 6(12): 9970-84, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25991668

ABSTRACT

TRAIL can induce apoptosis in some cancer cells and is an immune effector in the surveillance and elimination of developing tumors. Yes, some cancers are resistant to TRAIL. Delphinidin, a polyphenolic compound contained in brightly colored fruits and vegetables, has anti-inflammatory, anti-oxidant, and anti-tumorigenic activities. Here we showed that delphinidin sensitized TRAIL-resistant human prostate cancer cells to undergo apoptosis. Cells treated with delphinidin and TRAIL activated the extrinsic and intrinsic pathways of caspase activation. TRAIL-induced apoptosis in prostate cancer cells pretreated with delphinidin was dependent on death receptor 5 (DR5) and downstream cleavage of histone deacetylase 3 (HDAC3). In conclusion, delphinidin sensitizes prostate cancer cells to TRAIL-induced apoptosis by inducing DR5, thus causing caspase-mediated HDAC3 cleavage. Our data reveal a potential way of chemoprevention of prostate cancer by enabling TRAIL-mediated apoptosis.


Subject(s)
Anthocyanins/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Histone Deacetylases/metabolism , Prostatic Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Anthocyanins/administration & dosage , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Drug Synergism , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Transfection
16.
Nat Commun ; 6: 7390, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26077467

ABSTRACT

The inhibition of p53 activity by histone deacetylase 3 (HDAC3) has been reported, but the precise molecular mechanism is unknown. Here we show that programmed cell death 5 (PDCD5) selectively mediates HDAC3 dissociation from p53, which induces HDAC3 cleavage and ubiquitin-dependent proteasomal degradation. Casein kinase 2 alpha phosphorylates PDCD5 at Ser-119 to enhance its stability and importin 13-mediated nuclear translocation of PDCD5. Genetic deletion of PDCD5 abrogates etoposide (ET)-induced p53 stabilization and HDAC3 cleavage, indicating an essential role of PDCD5 in p53 activation. Restoration of PDCD5(WT) in PDCD5(-/-) MEFs restores ET-induced HDAC3 cleavage. Reduction of both PDCD5 and p53, but not reduction of either protein alone, significantly enhances in vivo tumorigenicity of AGS gastric cancer cells and correlates with poor prognosis in gastric cancer patients. Our results define a mechanism for p53 activation via PDCD5-dependent HDAC3 decay under genotoxic stress conditions.


Subject(s)
Apoptosis Regulatory Proteins/genetics , DNA Damage/genetics , Fibroblasts/metabolism , Histone Deacetylases/metabolism , Neoplasm Proteins/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Casein Kinase II/metabolism , Cell Line, Tumor , Chromatin Immunoprecipitation , Gene Expression Profiling , HCT116 Cells , Humans , Immunoprecipitation , In Situ Nick-End Labeling , Karyopherins/metabolism , Mass Spectrometry , Mice , Mice, Knockout , Neoplasm Transplantation , Phosphorylation , Prognosis , Proteasome Endopeptidase Complex , Real-Time Polymerase Chain Reaction , Stomach Neoplasms/metabolism
17.
Cancer Lett ; 335(1): 205-13, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23419524

ABSTRACT

Transforming growth factor-ß1, the key ligand of Smad-dependent signaling pathway, is critical for epithelial-mesenchymal transition during embryo-morphogenesis, fibrotic diseases, and tumor metastasis. In this study, we found that activation of p300/CBP, a histone acetyltransferase, by TGF-ß1 mediates Epithelial-mesenchymal transition (EMT) via acetylating Smad2 and Smad3 in TGF-ß1 signaling pathway. We demonstrated that treatment with EGCG inhibited p300/CBP activity in human lung cancer cells. Also, we observed that EGCG potently inhibited TGF-ß1-induced EMT and reversed the up-regulation of various genes during EMT. Our findings suggest that EGCG inhibits the induction of p300/CBP activity by TGF-ß1. Therefore, EGCG inhibits TGF-ß1-mediated EMT by suppressing the acetylation of Smad2 and Smad3 in human lung cancer cells.


Subject(s)
Anticarcinogenic Agents/pharmacology , Catechin/analogs & derivatives , Epithelial-Mesenchymal Transition/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/physiology , Acetylation , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Catechin/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms , Protein Binding , Protein Processing, Post-Translational/drug effects , Signal Transduction , Vimentin/metabolism , p300-CBP Transcription Factors/antagonists & inhibitors , p300-CBP Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL