Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Eye Res ; 93(6): 889-97, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22019372

ABSTRACT

Dying cells release pro-inflammatory molecules, functioning as cytokines to trigger cell/tissue inflammation that is relevant to disease pathology. Heat-shock protein 90 (HSP90) is believed to act as a danger signal for tissue damage once released extracellularly. Potential roles of HSP90 were explored in retinal pigment epithelial (RPE) inflammatory responses to necrosis. Cellular extracts can trigger ARPE-19 cell inflammatory responses, producing cytokines that lead to an increase in ARPE-19 cell monolayer permeability. Addition of recombinant HSP90ß mimics the induction of chemokines IL-8 and MCP-1 in cultured RPE cells, suggesting that released HSP90 can incite RPE cell sterile inflammatory responses. Consistent with this, classical HSP90 inhibitors were shown to substantially reduce necrosis-induced cytokine production and permeability increases in ARPE-19 cells. Moreover, a cell-impermeable inhibitor, 17-N,N-dimethylaminoethylamino-17-demethoxy-geldanamycin-N-oxide, also efficiently inhibited necrosis-induced cytokine production and TNF-α/IL-1ß-induced increase in ARPE-19 cell permeability in vitro and endotoxin-induced development of uveitis in vivo, suggesting that HSP90 can contribute to necrosis-induced RPE inflammatory responses. Collectively, our data identify HSP90 as a pro-inflammatory molecule in RPE cell sterile inflammatory responses.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzoquinones/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Heterocyclic Compounds, 2-Ring/pharmacology , Inflammation Mediators/antagonists & inhibitors , Lactams, Macrocyclic/pharmacology , Pyrazoles/pharmacology , Retinal Pigment Epithelium/drug effects , Uveitis/prevention & control , Animals , Anti-Inflammatory Agents/metabolism , Benzoquinones/metabolism , Cell Line , Chemokine CCL2/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , HSP90 Heat-Shock Proteins/metabolism , Heterocyclic Compounds, 2-Ring/metabolism , Humans , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Interleukin-8/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lactams, Macrocyclic/metabolism , Lipopolysaccharides , Male , Necrosis , Permeability , Protein Kinase Inhibitors/pharmacology , Pyrazoles/metabolism , Rats , Rats, Inbred Lew , Retinal Pigment Epithelium/immunology , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Signal Transduction/drug effects , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Uveitis/chemically induced , Uveitis/immunology , Uveitis/metabolism , Uveitis/pathology
2.
Invest Ophthalmol Vis Sci ; 52(2): 890-903, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-20847119

ABSTRACT

PURPOSE: To investigate the role of the peroxisome proliferator-activated receptor (PPAR)-γ in modulating retinal pigmented epithelium (RPE) responses to oxidative stress. METHODS: ARPE-19 cells were treated with the oxidant, t-butylhydroperoxide (tBH) to induce apoptosis. Cells pretreated with synthetic PPARγ agonists of the antidiabetic thiazolidinediones class before tBH challenge were assessed for viability and, by microarray analysis, for effects on gene expression. RESULTS: Treatment of ARPE-19 cells with tBH resulted in a loss of viability and global changes in the pattern of gene expression. PPARγ ligands were found to have differential modulatory effects on tBH-induced apoptosis of RPE cells. Whereas rosiglitazone and pioglitazone potentiated cell death, troglitazone acted as a potent cytoprotective agent. Downregulation of PPARγ expression by an siRNA resulted in enhanced cell death in response to tBH treatment and blocked the cytoprotective effect of troglitazone consistent with a role of PPARγ in mediating this response. Microarray analysis revealed that while rosiglitazone and pioglitazone had little effect on gene changes induced by tBH treatment, troglitazone dramatically reduced the number of changes caused by oxidative stress. A unique subset of genes that were deregulated by tBH and selectively normalized by troglitazone were identified. CONCLUSIONS: These findings demonstrate that PPARγ agonists can have differential effects on RPE survival in response to oxidative stress. Oxidative stress leads to deregulation of a large set of genes in ARPE-19 cells. A specific subset of these genes can be selectively modulated by troglitazone and represent potential novel targets for cytoprotective therapies.


Subject(s)
Apoptosis/drug effects , Oxidative Stress/drug effects , PPAR gamma/metabolism , Retinal Pigment Epithelium/pathology , Thiazolidinediones/pharmacology , Blotting, Western , Caspase 3/metabolism , Cell Line , Cell Survival , Drug Synergism , Gene Silencing/physiology , Humans , Ligands , Microarray Analysis , PPAR gamma/agonists , Pioglitazone , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Retinal Pigment Epithelium/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rosiglitazone , Transfection , tert-Butylhydroperoxide/toxicity
3.
Int J Cancer ; 115(6): 917-23, 2005 Jul 20.
Article in English | MEDLINE | ID: mdl-15729717

ABSTRACT

The novel synthetic retinoid-related molecule 4-[3-(1-heptyl-4,4-dimethyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-3-oxo-propenyl]benzoic acid (AGN193198) neither binds effectively to retinoic acid receptors (RARs) and retinoid X receptors (RXRs) nor transactivates in RAR- and RXR-mediated reporter assays. Even so, AGN193198 is potent in inducing apoptosis in human prostate and breast carcinoma cells (Keedwell et al., Cancer Res 2004;64:3302-12). Here, we extend these findings to show that AGN193198 potently and rapidly induces apoptosis in bladder carcinoma cell lines. One micromolar of AGN193198 completely abolished the growth of the transitional cell carcinoma lines UM-UC-3 and J82, and the squamous cell carcinoma line SCaBER; the transitional cell papilloma line RT-4 was slightly less sensitive to the growth inhibitory effect of AGN193198. Treated cells accumulated in the G2M phase of the cell cycle. This was accompanied by apoptosis, as revealed by staining cells for exposure of phosphatidylserine at their surface (binding of Annexin V) and FACS analysis of propidium iodide labeled cells. As reported for prostate cancer cells, AGN193198 provoked rapid activation of caspases-3 (by 6 hr), -8 (by 16 hr) and -9 (by 6 hr) in bladder cancer cells. These findings suggest that AGN193198 and related compounds, whose mechanism of action does not appear to involve RARs and RXRs, may be useful in the treatment of bladder cancer.


Subject(s)
Apoptosis/drug effects , Carcinoma, Transitional Cell/pathology , Papilloma/pathology , Quinolines/pharmacology , Urinary Bladder Neoplasms/pathology , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Humans , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/metabolism , Tumor Cells, Cultured
4.
Cancer Res ; 64(9): 3302-12, 2004 May 01.
Article in English | MEDLINE | ID: mdl-15126374

ABSTRACT

Synthetic retinoid-related molecules, such as N-(4-hydroxyphenyl)retinamide (fenretinide) and 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphthalene carboxylic acid (CD437) induce apoptosis in a variety of malignant cells. The mechanism(s) of action of these compounds does not appear to involve retinoic acid receptors (RARs) and retinoid X receptors (RXRs), although some investigators disagree with this view. To clarify whether some retinoid-related molecules can induce apoptosis without involving RARs and/or RXRs, we used 4-[3-(1-heptyl-4,4-dimethyl-2-oxo-1,2,3,4-tetrahydroquinolin-6-yl)-3-oxo-E-propenyl] benzoic acid (AGN193198) that neither binds effectively to RARs and RXRs nor transactivates in RAR- and RXR-mediated reporter assays. AGN193198 potently induced apoptosis in prostate, breast, and gastrointestinal carcinoma cells and in leukemia cells. AGN193198 also abolished growth (by 50% at 130-332 nM) and induced apoptosis in primary cultures established from prostatic carcinoma (13 patients) and gastrointestinal carcinoma (1 patient). Apoptosis was induced rapidly, as indicated by mitochondrial depolarization and DNA fragmentation. Molecular events provoked by AGN193198 included activation of caspase-3, -8, -9, and -10 (by 4-6 h) and the production of BID/p15 (by 6 h). These findings show that caspase-mediated induction of apoptosis by AGN193198 is RAR/RXR-independent and suggest that this compound may be useful in the treatment of prostate cancer.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Prostatic Neoplasms/drug therapy , Quinolines/pharmacology , Receptors, Retinoic Acid/metabolism , Retinoids/pharmacology , Transcription Factors/metabolism , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/physiology , Cell Division/drug effects , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Isoenzymes/metabolism , Jurkat Cells , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Quinolines/metabolism , Retinoid X Receptors , Retinoids/metabolism , Transcriptional Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...