Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Cancer Res ; 76(18): 5467-78, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27503926

ABSTRACT

SIRT2 is a protein deacetylase with tumor suppressor activity in breast and liver tumors where it is mutated; however, the critical substrates mediating its antitumor activity are not fully defined. Here we demonstrate that SIRT2 binds, deacetylates, and inhibits the peroxidase activity of the antioxidant protein peroxiredoxin (Prdx-1) in breast cancer cells. Ectopic overexpression of SIRT2, but not its catalytically dead mutant, increased intracellular levels of reactive oxygen species (ROS) induced by hydrogen peroxide, which led to increased levels of an overoxidized and multimeric form of Prdx-1 with activity as a molecular chaperone. Elevated levels of SIRT2 sensitized breast cancer cells to intracellular DNA damage and cell death induced by oxidative stress, as associated with increased levels of nuclear FOXO3A and the proapoptotic BIM protein. In addition, elevated levels of SIRT2 sensitized breast cancer cells to arsenic trioxide, an approved therapeutic agent, along with other intracellular ROS-inducing agents. Conversely, antisense RNA-mediated attenuation of SIRT2 reversed ROS-induced toxicity as demonstrated in a zebrafish embryo model system. Collectively, our findings suggest that the tumor suppressor activity of SIRT2 requires its ability to restrict the antioxidant activity of Prdx-1, thereby sensitizing breast cancer cells to ROS-induced DNA damage and cell cytotoxicity. Cancer Res; 76(18); 5467-78. ©2016 AACR.


Subject(s)
Breast Neoplasms/pathology , Peroxiredoxins/metabolism , Sirtuin 2/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Comet Assay , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Immunoblotting , Immunoprecipitation , Microscopy, Confocal , Oxidants/pharmacology , Oxidative Stress/physiology , Peroxidase/metabolism , Zebrafish
3.
Mar Drugs ; 13(3): 1552-68, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25803180

ABSTRACT

The biologically active lipopeptide kalkitoxin was previously isolated from the marine cyanobacterium Moorea producens (Lyngbya majuscula). Kalkitoxin exhibited N-methyl-D-aspartate (NMDA)-mediated neurotoxicity and acted as an inhibitory ligand for voltage-sensitive sodium channels in cultured rat cerebellar granule neurons. Subsequent studies revealed that kalkitoxin generated a delayed form of colon tumor cell cytotoxicity in 7-day clonogenic cell survival assays. Cell line- and exposure time-dependent cytostatic/cytotoxic effects were previously observed with mitochondria-targeted inhibitors of hypoxia-inducible factor-1 (HIF-1). The transcription factor HIF-1 functions as a key regulator of oxygen homeostasis. Therefore, we investigated the ability of kalkitoxin to inhibit hypoxic signaling in human tumor cell lines. Kalkitoxin potently and selectively inhibited hypoxia-induced activation of HIF-1 in T47D breast tumor cells (IC50 5.6 nM). Mechanistic studies revealed that kalkitoxin inhibits HIF-1 activation by suppressing mitochondrial oxygen consumption at electron transport chain (ETC) complex I (NADH-ubiquinone oxidoreductase). Further studies indicate that kalkitoxin targets tumor angiogenesis by blocking the induction of angiogenic factors (i.e., VEGF) in tumor cells.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Breast Neoplasms/drug therapy , Lipids/pharmacology , Neovascularization, Pathologic/drug therapy , Thiazoles/pharmacology , Angiogenesis Inhibitors/administration & dosage , Breast Neoplasms/pathology , Cell Hypoxia/drug effects , Cell Line, Tumor , Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Female , Humans , Hypoxia-Inducible Factor 1/metabolism , Inhibitory Concentration 50 , Lipids/administration & dosage , Mitochondria/drug effects , Mitochondria/metabolism , Signal Transduction/drug effects , Thiazoles/administration & dosage , Vascular Endothelial Growth Factor A/metabolism
4.
Biochem J ; 450(1): 169-78, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23167260

ABSTRACT

SLC5A8 (solute carrier gene family 5A, member 8) is a sodium-coupled transporter for monocarboxylates. Among its substrates are the HDAC (histone deacetylase) inhibitors butyrate, propionate and pyruvate. Expression of SLC5A8 is silenced in cancers via DNA methylation, and ectopic expression of SLC5A8 in cancer cells induces apoptosis in the presence of its substrates that are HDAC inhibitors. In the present study we show that ectopic expression of SLC5A8 in cancer cells translocates the anti-apoptotic protein survivin to the plasma membrane through protein-protein interaction resulting in depletion of nuclear survivin and also decreases cellular levels of survivin through inhibition of transcription. These SLC5A8-induced changes in the location and levels of survivin result in cell-cycle arrest, disruption of the chromosome passenger complex involved in mitosis, induction of apoptosis and enhancement in chemosensitivity. These effects are seen independently of the transport function of SLC5A8 and histone acetylation status of the cell; in the presence of pyruvate, a SLC5A8 substrate and also an HDAC inhibitor, these effects are amplified. Ectopic expression of SLC5A8 in the breast cancer cell line MB231 inhibits the ability of cells to form colonies in vitro and to form tumours in mouse xenografts in vivo. The suppression of survivin transcription occurs independently of HDAC inhibition, and the underlying mechanism is associated with decreased phosphorylation of STAT3 (signal transducer and activator of transcription 3). The observed effects are specific for survivin with no apparent changes in expression of other inhibitor-of-apoptosis proteins. The present study unravels a novel, hitherto unrecognized, mechanism for the tumour-suppressive role of a plasma membrane transporter independent of its transport function.


Subject(s)
Breast Neoplasms/pathology , Cation Transport Proteins/metabolism , Cell Membrane/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Pancreatic Neoplasms/pathology , Animals , Apoptosis , Biological Transport , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cation Transport Proteins/genetics , Cell Line, Tumor , Female , Humans , Inhibitor of Apoptosis Proteins/genetics , Mice , Mice, Nude , Monocarboxylic Acid Transporters , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Survivin , Transplantation, Heterologous
5.
Invest Ophthalmol Vis Sci ; 54(1): 63-71, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-23169885

ABSTRACT

PURPOSE: Hemochromatosis is a disorder of iron overload arising mostly from mutations in HFE. HFE is expressed in retinal pigment epithelium (RPE), and Hfe(-/-) mice develop age-related iron accumulation and retinal degeneration associated with RPE hyperproliferation. Here, the mechanism underlying the hyperproliferative phenotype in RPE was investigated. METHODS: Cellular senescence was monitored by ß-galactosidase activity. Gene expression was monitored by real-time PCR. Survivin was analyzed by Western blot and immunofluorescence. Migration and invasion were monitored using appropriate kits. Glucose transporters (GLUTs) were monitored by 3-O-methyl-D-glucose uptake. Histone deacetylases (HDACs) were studied by monitoring catalytic activity and acetylation status of histones H3/H4. RESULTS: Hfe(-/-) RPE cells exhibited slower senescence rate and higher survivin expression than wild type cells. Hfe(-/-) cells migrated faster and showed greater glucose uptake and increased expression of GLUTs. The expression of HDACs and DNA methyltransferase (DNMTs) also was increased. Similarly, RPE cells from hemojuvelin (Hjv)-knockout mice, another model of hemochromatosis, also had increased expression of GLUTs, HDACs, and DNMTs. The expression of Slc5a8 was decreased in Hfe(-/-) RPE cells, but treatment with a DNA methylation inhibitor restored the transporter expression, indicating involvement of DNA methylation in the silencing of Slc5a8 in Hfe(-/-) cells. CONCLUSIONS: RPE cells from iron-overloaded mice exhibit several features of tumor cells: decreased senescence, enhanced migration, increased glucose uptake, and elevated levels of HDACs and DNMTs. These features are seen in Hfe(-/-) RPE cells as well as in Hjv(-/-) RPE cells, providing a molecular basis for the hyperproliferative phenotype of Hfe(-/-) and Hjv(-/-) RPE cells.


Subject(s)
Eye Neoplasms , Hemochromatosis , Histocompatibility Antigens Class I/genetics , Membrane Proteins/genetics , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/physiopathology , Amino Acid Transport Systems/genetics , Animals , Cation Transport Proteins/genetics , Cell Movement/physiology , Cell Transformation, Neoplastic/pathology , Cellular Senescence/physiology , DNA Methylation/physiology , Disease Progression , Eye Neoplasms/genetics , Eye Neoplasms/pathology , Eye Neoplasms/physiopathology , Female , Glucose/pharmacokinetics , Hemochromatosis/genetics , Hemochromatosis/pathology , Hemochromatosis/physiopathology , Hemochromatosis Protein , Inhibitor of Apoptosis Proteins/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Monocarboxylic Acid Transporters , Phenotype , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Primary Cell Culture , Repressor Proteins/metabolism , Survivin , Transplantation, Heterologous
6.
PLoS One ; 7(1): e29916, 2012.
Article in English | MEDLINE | ID: mdl-22238673

ABSTRACT

Reporter-based assays underlie many high-throughput screening (HTS) platforms, but most are limited to in vitro applications. Here, we report a simple whole-organism HTS method for quantifying changes in reporter intensity in individual zebrafish over time termed, Automated Reporter Quantification in vivo (ARQiv). ARQiv differs from current "high-content" (e.g., confocal imaging-based) whole-organism screening technologies by providing a purely quantitative data acquisition approach that affords marked improvements in throughput. ARQiv uses a fluorescence microplate reader with specific detection functionalities necessary for robust quantification of reporter signals in vivo. This approach is: 1) Rapid; achieving true HTS capacities (i.e., >50,000 units per day), 2) Reproducible; attaining HTS-compatible assay quality (i.e., Z'-factors of ≥0.5), and 3) Flexible; amenable to nearly any reporter-based assay in zebrafish embryos, larvae, or juveniles. ARQiv is used here to quantify changes in: 1) Cell number; loss and regeneration of two different fluorescently tagged cell types (pancreatic beta cells and rod photoreceptors), 2) Cell signaling; relative activity of a transgenic Notch-signaling reporter, and 3) Cell metabolism; accumulation of reactive oxygen species. In summary, ARQiv is a versatile and readily accessible approach facilitating evaluation of genetic and/or chemical manipulations in living zebrafish that complements current "high-content" whole-organism screening methods by providing a first-tier in vivo HTS drug discovery platform.


Subject(s)
Gene Dosage , Genes, Reporter , High-Throughput Screening Assays/methods , Image Processing, Computer-Assisted/methods , Zebrafish , Animals , Animals, Genetically Modified , Automation/methods , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Embryo, Nonmammalian , Gene Dosage/physiology , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Osmolar Concentration , Reproducibility of Results , Validation Studies as Topic , Zebrafish/embryology , Zebrafish/genetics
7.
J Biol Chem ; 286(36): 31830-8, 2011 Sep 09.
Article in English | MEDLINE | ID: mdl-21771784

ABSTRACT

SLC6A14, also known as ATB(0,+), is an amino acid transporter with unique characteristics. It transports 18 of the 20 proteinogenic amino acids. However, this transporter is expressed only at low levels in normal tissues. Here, we show that the transporter is up-regulated specifically in estrogen receptor (ER)-positive breast cancer, demonstrable with primary human breast cancer tissues and human breast cancer cell lines. SLC6A14 is an estrogen/ER target. The transport features of SLC6A14 include concentrative transport of leucine (an activator of mTOR), glutamine (an essential amino acid for nucleotide biosynthesis and substrate for glutaminolysis), and arginine (an essential amino acid for tumor cells), suggesting that ER-positive breast cancer cells up-regulate SLC6A14 to meet their increased demand for these amino acids. Consequently, treatment of ER-positive breast cancer cells in vitro with α-methyl-DL-tryptophan (α-MT), a selective blocker of SLC6A14, induces amino acid deprivation, inhibits mTOR, and activates autophagy. Prolongation of the treatment with α-MT causes apoptosis. Addition of an autophagy inhibitor (3-methyladenine) during α-MT treatment also induces apoptosis. These effects of α-MT are specific to ER-positive breast cancer cells, which express the transporter. The ability of α-MT to cause amino acid deprivation is significantly attenuated in MCF-7 cells, an ER-positive breast cancer cell line, when SLC6A14 is silenced with shRNA. In mouse xenograft studies, α-MT by itself is able to reduce the growth of the ER-positive ZR-75-1 breast cancer cells. These studies identify SLC6A14 as a novel and effective drug target for the treatment of ER-positive breast cancer.


Subject(s)
Amino Acid Transport Systems, Neutral/antagonists & inhibitors , Breast Neoplasms/drug therapy , Amino Acid Transport Systems , Amino Acid Transport Systems, Neutral/genetics , Animals , Autophagy/drug effects , Breast Neoplasms/pathology , Female , Humans , Mice , Molecular Targeted Therapy/methods , Receptors, Estrogen , Transplantation, Heterologous , Tryptophan/analogs & derivatives , Tryptophan/pharmacology , Tumor Cells, Cultured
8.
Invest Ophthalmol Vis Sci ; 52(8): 5749-57, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21508099

ABSTRACT

PURPOSE: To evaluate the role of SLC5A8 in the transport of 2-oxothiazolidine-4-carboxylate (OTC) and to determine whether OTC augments glutathione production in RPE cells, thereby providing protection against oxidative stress. METHODS: SLC5A8-mediated transport of OTC was monitored in Xenopus laevis oocytes by electrophysiological means. Saturation kinetics, Na(+)-activation kinetics, and inhibition by ibuprofen were analyzed by monitoring OTC-induced currents as a measure of transport activity. Oxidative stress was induced in ARPE-19 cells and primary RPE cells isolated from wild type and Slc5a8(-/-) mouse retinas using H(2)O(2), and the effects of OTC on cell death and intracellular glutathione concentration were examined. RESULTS: Heterologous expression of human SLC5A8 in X. laevis oocytes induced Na(+)-dependent inward currents in the presence of OTC under voltage-clamp conditions. The transport of OTC via SLC5A8 was saturable, with a K(t) of 104 ± 3 µM. The Na(+)-activation kinetics was sigmoidal with a Hill coefficient of 1.9 ± 0.1, suggesting involvement of two Na(+) in the activation process. Ibuprofen, a blocker of SLC5A8, inhibited SLC5A8-mediated OTC transport; the concentration necessary for half-maximal inhibition was 17 ± 1 µM. OTC increased glutathione levels and protected ARPE-19 and primary RPE cells isolated from wild type mouse retinas from H(2)O(2)-induced cell death. These effects were abolished in primary RPE isolated from Slc5a8(-/-) mouse retinas. CONCLUSIONS: OTC is a transportable substrate for SLC5A8. OTC augments glutathione production in RPE cells, thereby protecting them from oxidative damage. Transport via SLC5A8 is obligatory for this process.


Subject(s)
Cation Transport Proteins/metabolism , Glutathione/metabolism , Pyrrolidonecarboxylic Acid/pharmacokinetics , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , Thiazolidines/pharmacokinetics , Animals , Apoptosis , Biological Transport/physiology , Cation Transport Proteins/genetics , Cell Survival/drug effects , Cyclooxygenase Inhibitors/pharmacology , Drug Interactions , Humans , Hydrogen Peroxide/toxicity , Ibuprofen/pharmacology , Kinetics , Mice , Mice, Mutant Strains , Monocarboxylic Acid Transporters , Oocytes/physiology , Oxidants/toxicity , Oxidative Stress/drug effects , Oxidative Stress/physiology , Pyrrolidonecarboxylic Acid/chemistry , Retinal Pigment Epithelium/cytology , Thiazolidines/chemistry , Xenopus laevis
9.
Bioorg Med Chem ; 18(16): 5988-94, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20637638

ABSTRACT

A natural product chemistry-based approach was applied to discover small-molecule inhibitors of hypoxia-inducible factor-1 (HIF-1). A Petrosaspongia mycofijiensis marine sponge extract yielded mycothiazole (1), a solid tumor selective compound with no known mechanism for its cell line-dependent cytotoxic activity. Compound 1 inhibited hypoxic HIF-1 signaling in tumor cells (IC(50) 1nM) that correlated with the suppression of hypoxia-stimulated tumor angiogenesis in vitro. However, 1 exhibited pronounced neurotoxicity in vitro. Mechanistic studies revealed that 1 selectively suppresses mitochondrial respiration at complex I (NADH-ubiquinone oxidoreductase). Unlike rotenone, MPP(+), annonaceous acetogenins, piericidin A, and other complex I inhibitors, mycothiazole is a mixed polyketide/peptide-derived compound with a central thiazole moiety. The exquisite potency and structural novelty of 1 suggest that it may serve as a valuable molecular probe for mitochondrial biology and HIF-mediated hypoxic signaling.


Subject(s)
Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Porifera/chemistry , Thiazoles/pharmacology , Animals , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Electron Transport Complex I/metabolism , Enzyme Inhibitors/isolation & purification , Female , Gene Expression Regulation/drug effects , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neovascularization, Pathologic/drug therapy , Neurons/drug effects , Rats , Thiazoles/isolation & purification
10.
Clin Cancer Res ; 16(19): 4742-54, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20647473

ABSTRACT

PURPOSE: Bortezomib induces unfolded protein response (UPR) and endoplasmic reticulum stress, as well as exhibits clinical activity in patients with relapsed and refractory mantle cell lymphoma (MCL). Here, we determined the molecular basis of the improved in vitro and in vivo activity of the combination of the pan-histone deacetylase inhibitor panobinostat and bortezomib against human, cultured, and primary MCL cells. EXPERIMENTAL DESIGN: Immunoblot analyses, reverse transcription-PCR, and immunofluorescent and electron microscopy were used to determine the effects of panobinostat on bortezomib-induced aggresome formation and endoplasmic reticulum stress in MCL cells. RESULTS: Treatment with panobinostat induced heat shock protein 90 acetylation; depleted the levels of heat shock protein 90 client proteins, cyclin-dependent kinase 4, c-RAF, and AKT; and abrogated bortezomib-induced aggresome formation in MCL cells. Panobinostat also induced lethal UPR, associated with induction of CAAT/enhancer binding protein homologous protein (CHOP). Conversely, knockdown of CHOP attenuated panobinostat-induced cell death of MCL cells. Compared with each agent alone, cotreatment with panobinostat increased bortezomib-induced expression of CHOP and NOXA, as well as increased bortezomib-induced UPR and apoptosis of cultured and primary MCL cells. Cotreatment with panobinostat also increased bortezomib-mediated in vivo tumor growth inhibition and improved survival of mice bearing human Z138C MCL cell xenograft. CONCLUSION: These findings suggest that increased UPR and induction of CHOP are involved in enhanced anti-MCL activity of the combination of panobinostat and bortezomib.


Subject(s)
Boronic Acids/pharmacology , Endoplasmic Reticulum/drug effects , Hydroxamic Acids/pharmacology , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/pathology , Pyrazines/pharmacology , Stress, Physiological/drug effects , Transcription Factor CHOP/metabolism , Acetylation/drug effects , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum/metabolism , Fluorescent Antibody Technique , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Indoles , Lymphoma, Mantle-Cell/metabolism , Mice , Microscopy, Confocal , Panobinostat , Protein Folding/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Transcription Factor CHOP/genetics , Xenograft Model Antitumor Assays
11.
J Nat Prod ; 73(5): 956-61, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20423107

ABSTRACT

Products that contain twig extracts of pawpaw (Asimina triloba) are widely consumed anticancer alternative medicines. Pawpaw crude extract (CE) and purified acetogenins inhibited hypoxia-inducible factor-1 (HIF-1)-mediated hypoxic signaling pathways in tumor cells. In T47D cells, pawpaw CE and the acetogenins 10-hydroxyglaucanetin (1), annonacin (2), and annonacin A (3) inhibited hypoxia-induced HIF-1 activation with IC(50) values of 0.02 microg/mL, 12 nM, 13 nM, and 31 nM, respectively. This inhibition correlates with the suppression of the hypoxic induction of HIF-1 target genes VEGF and GLUT-1. The induction of secreted VEGF protein represents a key event in hypoxia-induced tumor angiogenesis. Both the extract and the purified acetogenins blocked the angiogenesis-stimulating activity of hypoxic T47D cells in vitro. Pawpaw extract and acetogenins inhibited HIF-1 activation by blocking the hypoxic induction of nuclear HIF-1alpha protein. The inhibition of HIF-1 activation was associated with the suppression of mitochondrial respiration at complex I. Thus, the inhibition of HIF-1 activation and hypoxic tumor angiogenesis constitutes a novel mechanism of action for these anticancer alternative medicines.


Subject(s)
Acetogenins/isolation & purification , Acetogenins/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/pharmacology , Asimina/chemistry , Glucose Transporter Type 1/drug effects , Hypoxia-Inducible Factor 1/drug effects , Neovascularization, Physiologic/drug effects , Plants, Medicinal/chemistry , Vascular Endothelial Growth Factors/drug effects , Acetogenins/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Complementary Therapies , Drug Screening Assays, Antitumor , Glucose Transporter Type 1/analysis , Glucose Transporter Type 1/genetics , Humans , Inhibitory Concentration 50 , Molecular Structure , Vascular Endothelial Growth Factors/analysis , Vascular Endothelial Growth Factors/genetics
12.
Mol Cancer Ther ; 9(4): 942-52, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20371724

ABSTRACT

Increased levels of misfolded polypeptides in the endoplasmic reticulum (ER) triggers the dissociation of glucose-regulated protein 78 (GRP78) from the three transmembrane ER-stress mediators, i.e., protein kinase RNA-like ER kinase (PERK), activating transcription factor-6 (ATF6), and inositol-requiring enzyme 1alpha, which results in the adaptive unfolded protein response (UPR). In the present studies, we determined that histone deacetylase-6 (HDAC6) binds and deacetylates GRP78. Following treatment with the pan-histone deacetylase inhibitor panobinostat (Novartis Pharmaceuticals), or knockdown of HDAC6 by short hairpin RNA, GRP78 is acetylated in 11 lysine residues, which dissociates GRP78 from PERK. This is associated with the activation of a lethal UPR in human breast cancer cells. Coimmunoprecipitation studies showed that binding of HDAC6 to GRP78 requires the second catalytic and COOH-terminal BUZ domains of HDAC6. Treatment with panobinostat increased the levels of phosphorylated-eukaryotic translation initiation factor (p-eIF2alpha), ATF4, and CAAT/enhancer binding protein homologous protein (CHOP). Panobinostat treatment also increased the proapoptotic BIK, BIM, BAX, and BAK levels, as well as increased the activity of caspase-7. Knockdown of GRP78 sensitized MCF-7 cells to bortezomib and panobinostat-induced UPR and cell death. These findings indicate that enforced acetylation and decreased binding of GRP78 to PERK is mechanistically linked to panobinostat-induced UPR and cell death of breast cancer cells. Mol Cancer Ther; 9(4); 942-52. (c)2010 AACR.


Subject(s)
Breast Neoplasms/pathology , Endoplasmic Reticulum/pathology , Heat-Shock Proteins/metabolism , Hydroxamic Acids/pharmacology , Stress, Physiological/drug effects , Acetylation/drug effects , Amino Acid Sequence , Catalytic Domain , Cell Death/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Female , Gene Knockdown Techniques , Heat-Shock Proteins/chemistry , Histone Deacetylase 6 , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Humans , Indoles , Models, Biological , Molecular Sequence Data , Panobinostat , Protein Binding/drug effects , Unfolded Protein Response/drug effects , eIF-2 Kinase/metabolism
13.
J Nat Prod ; 72(12): 2104-9, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19921787

ABSTRACT

The transcription factor hypoxia-inducible factor-1 (HIF-1) represents an important molecular target for anticancer drug discovery. In a T47D cell-based reporter assay, the Caulerpa spp. algal pigment caulerpin (1) inhibited hypoxia-induced as well as 1,10-phenanthroline-induced HIF-1 activation. The angiogenic factor vascular endothelial growth factor (VEGF) is regulated by HIF-1. Caulerpin (10 microM) suppressed hypoxic induction of secreted VEGF protein and the ability of hypoxic T47D cell-conditioned media to promote tumor angiogenesis in vitro. Under hypoxic conditions, 1 (10 microM) blocked the induction of HIF-1alpha protein, the oxygen-regulated subunit that controls HIF-1 activity. Reactive oxygen species produced by mitochondrial complex III are believed to act as a signal of cellular hypoxia that leads to HIF-1alpha protein induction and activation. Further mechanistic studies revealed that 1 inhibits mitochondrial respiration at electron transport chain (ETC) complex I (NADH-ubiquinone oxidoreductase). Under hypoxic conditions, it is proposed that 1 may disrupt mitochondrial ROS-regulated HIF-1 activation and HIF-1 downstream target gene expression by inhibiting the transport or delivery of electrons to complex III.


Subject(s)
Caulerpa/chemistry , Hypoxia-Inducible Factor 1/drug effects , Indoles/pharmacology , Coloring Agents/chemistry , Coloring Agents/pharmacology , Electron Transport Complex I/antagonists & inhibitors , Female , Humans , Indoles/chemistry , Indoles/isolation & purification , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...