Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Pharmacol Exp Ther ; 341(3): 646-55, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22393247

ABSTRACT

Diltiazem is a calcium channel blocker used to treat cardiovascular ailments. In addition, reports suggest that diltiazem induces cell death, which could make it a drug of choice for the treatment of cancer associated with hypertension. The goal of this research was to determine whether diltiazem is capable of inducing apoptosis in prostate cancer cells, either alone or in combination with the proteasome inhibitors, lactacystin and bortezomib (Velcade). Bortezomib is approved for the treatment of multiple myeloma; unfortunately, it has side effects that limit its utility. Presumably these side effects could be decreased by reducing its dose in combination with another drug. We have previously shown that lactacystin induces apoptosis in LNCaP cells; here, we show that this effect was enhanced by diltiazem. Furthermore, in proteasome inhibitor-resistant DU145 cells, diltiazem alone did not induce apoptosis but decreased cytosolic calcium levels and induced mitochondrial fission; likewise, lactacystin did not induce apoptosis but up-regulated the proapoptotic protein Bik. However, increasing concentrations of diltiazem in combination with lactacystin or bortezomib induced apoptosis in a dose-dependent and synergistic manner. The combination of diltiazem and lactacystin also up-regulated the levels of Bik and released Bak from Bcl-xL, indicating the involvement of the Bcl2 family pathway in this apoptosis. In addition, the drug combination up-regulated GRP78, suggesting also the involvement of endoplasmic reticulum stress in the apoptotic response. Thus, our results demonstrate a potential therapeutic advantage of combining a frequently used calcium channel blocker with proteasome inhibitors in the treatment of prostate cancer.


Subject(s)
Apoptosis/drug effects , Calcium Channel Blockers/pharmacology , Diltiazem/pharmacology , Prostatic Neoplasms/pathology , Protease Inhibitors/therapeutic use , Acetylcysteine/analogs & derivatives , Acetylcysteine/therapeutic use , Blotting, Western , Boronic Acids/therapeutic use , Bortezomib , Calcium/metabolism , Clinical Trials as Topic , Dose-Response Relationship, Drug , Drug Interactions , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Humans , Male , Prostatic Neoplasms/metabolism , Pyrazines/therapeutic use , Transfection , Tumor Cells, Cultured/drug effects
2.
Mol Cancer Res ; 9(8): 1067-77, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21724752

ABSTRACT

Androgen and androgen receptors (AR) play critical roles in the proliferation of prostate cancer through transcriptional regulation of target genes. Here, we found that androgens upregulated the expression of dynamin-related protein 1 (Drp1), which is involved in the induction of mitochondrial fission, a common event in mitosis and apoptosis. Clinical tissue samples and various prostate cancer cell lines revealed a positive correlation between Drp1 and AR levels. Treatment of androgen-sensitive cells with an AR agonist, R1881, and antagonist, bicalutamide, showed that Drp1 is transcriptionally regulated by androgens, as confirmed by an AR ChIP-seq assay. Live imaging experiments using pAcGFP1-Mito stably transfected LNCaP (mito-green) cells revealed that androgen did not induce significant mitochondrial fission by itself, although Drp1 was upregulated. However, when treated with CGP37157 (CGP), an inhibitor of mitochondrial Ca²âº efflux, these cells exhibited mitochondrial fission, which was further enhanced by pretreatment with R1881, suggesting that androgen-induced Drp1 expression facilitated CGP-induced mitochondrial fission. This enhanced mitochondrial fission was correlated with increased apoptosis. Transfection with dominant-negative (DN-Drp1, K38A) rescued cells from increased apoptosis, confirming the role of androgen-induced Drp1 in the observed apoptosis with combination treatment. Furthermore, we found that CGP reduced the expression of Mfn1, a protein that promotes mitochondrial fusion, a process which opposes fission. We suggest that androgen-increased Drp1 enhanced mitochondrial fission leading to apoptosis. The present study shows a novel role for androgens in the regulation of mitochondrial morphology that could potentially be utilized in prostate cancer therapy.


Subject(s)
Androgens/metabolism , GTP Phosphohydrolases/metabolism , Microtubule-Associated Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Androgens/physiology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Dynamins , GTP Phosphohydrolases/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Metribolone/metabolism , Microtubule-Associated Proteins/genetics , Mitochondria/physiology , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics
3.
Int J Oncol ; 36(6): 1437-44, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20428767

ABSTRACT

Mitochondria are structurally complex organelles that undergo fragmentation or fission in apoptotic cells. Mitochondrial fission requires the cytoplasmic dynamin-related protein, Drp1, which translocates to the mitochondria during apoptosis and interacts with the mitochondrial protein, Fis1. Finely tuned changes in cellular calcium modulate a variety of intracellular functions; in resting cells, the level of mitochondrial calcium is low, while it is higher during apoptosis. Mitochondria take up Ca(2+) via the Uniporter and extrude it to the cytoplasm through the mitochondrial Na+/Ca(2+) exchanger. Overload of Ca(2+) in the mitochondria leads to their damage, affecting cellular function and survival. The mitochondrial Na+/Ca2+ exchanger was blocked by benzodiazepine, CGP37157 (CGP) leading to increased mitochondrial calcium and enhancing the apoptotic effects of TRAIL, TNFalpha related apoptosis inducing ligand. In the present study, we observed that increasing mitochondrial calcium induced mitochondrial fragmentation, which correlated with the presence of Drp1 at the mitochondria in CGP treated cells. Under these conditions, we observed interactions between Drp1 and Fis1. The importance of Drp1 in fragmentation was confirmed by transfection of dominant negative Drp1 construct. However, fragmentation of the mitochondria was not sufficient to induce apoptosis, although it enhanced TRAIL-induced apoptosis. Furthermore, oligomerization of Bak was partially responsible for the increased apoptosis in cells treated with both CGP and TRAIL. Thus, our results show that combination of an apoptogenic agent and an appropriate calcium channel blocker provide therapeutic advantages.


Subject(s)
Calcium/metabolism , GTP Phosphohydrolases/metabolism , Microtubule-Associated Proteins/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/metabolism , Prostatic Neoplasms/pathology , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Clonazepam/analogs & derivatives , Clonazepam/pharmacology , Dynamins , GTP Phosphohydrolases/genetics , Humans , Immunohistochemistry , Immunoprecipitation , Male , Membrane Proteins/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/drug effects , Mitochondrial Proteins/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Thiazepines/pharmacology , Transfection , bcl-2 Homologous Antagonist-Killer Protein/drug effects , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism
4.
Mol Cancer Res ; 8(4): 569-77, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20353998

ABSTRACT

Many human cancers express elevated levels of cyclooxygenase-2 (COX-2), an enzyme responsible for the biosynthesis of prostaglandins. Available clinical data establish the protective effect of COX-2 inhibition on human cancer progression. However, despite these encouraging outcomes, the appearance of unwanted side effects remains a major hurdle for the general application of COX-2 inhibitors as effective cancer drugs. Hence, a better understanding of the molecular signals downstream of COX-2 is needed for the elucidation of drug targets that may improve cancer therapy. Here, we show that the COX-2 product prostaglandin E(2) (PGE(2)) acts on cognate receptor EP4 to promote the migration of A549 lung cancer cells. Treatment with PGE(2) enhances tyrosine kinase c-Src activation, and blockade of c-Src activity represses the PGE(2)-mediated lung cancer cell migration. PGE(2) affects target cells by activating four receptors named EP1 to EP4. Use of EP subtype-selective ligand agonists suggested that EP4 mediates prostaglandin-induced A549 lung cancer cell migration, and this conclusion was confirmed using a short hairpin RNA approach to specifically knock down EP4 expression. Proximal EP4 effectors include heterotrimeric Gs and betaArrestin proteins. Knockdown of betaArrestin1 expression with shRNA significantly impaired the PGE(2)-induced c-Src activation and cell migration. Together, these results support the idea that increased expression of the COX-2 product PGE(2) in the lung tumor microenvironment may initiate a mitogenic signaling cascade composed of EP4, betaArrestin1, and c-Src which mediates cancer cell migration. Selective targeting of EP4 with a ligand antagonist may provide an efficient approach to better manage patients with advanced lung cancer.


Subject(s)
Arrestins/metabolism , Carcinoma/metabolism , Cell Movement/genetics , Dinoprostone/metabolism , Lung Neoplasms/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptors, Prostaglandin E/metabolism , Arrestins/drug effects , Arrestins/genetics , CSK Tyrosine-Protein Kinase , Carcinoma/genetics , Carcinoma/physiopathology , Cell Movement/drug effects , Cyclooxygenase 2/metabolism , Down-Regulation/genetics , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Lung Neoplasms/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/physiopathology , Protein-Tyrosine Kinases/drug effects , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/pharmacology , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E, EP4 Subtype , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Cells, Cultured , beta-Arrestins , src-Family Kinases
5.
Proc Natl Acad Sci U S A ; 106(23): 9379-84, 2009 Jun 09.
Article in English | MEDLINE | ID: mdl-19458261

ABSTRACT

Androgen receptor (AR) signaling regulates the development and homeostasis of male reproductive organs, including the prostate. Deregulation of AR and AR coregulators, expression, or activity is involved in the initiation of prostate cancer and contributes to the transition of the disease to hormone-refractory stage. The ubiquitous betaArrestin proteins are now recognized as bona fide adapters and signal transducers with target effectors found in both the cytosol and nucleus. Here, we provide evidence that betaArrestin2 forms a complex with AR and acts as an AR corepressor in androgen-dependent prostate cancer cells. Accordingly, the forced overexpression of betaArrestin2 diminishes, and knockdown of betaArrestin2 expression with RNAi increases the androgen-induced prostate-specific antigen (PSA) gene expression. betaArrestin2 serves as an adapter, bringing into close proximity the Mdm2 E3 ligase and AR, thereby promoting AR ubiquitylation and degradation. Human prostate tissues evidence an inverse relationship between the expression of betaArrestin2 and AR activity: glands that express high levels of betaArrestin2 exhibit low expression of PSA, and those glands that express low levels of betaArrestin2 evidence elevated PSA levels. We conclude that betaArrestin2 acts as a corepressor of AR by serving as a scaffold for Mdm2 leading to the AR ubiquitylation and degradation.


Subject(s)
Arrestins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Signal Transduction , Humans , Male , Prostate-Specific Antigen , Prostatic Neoplasms/pathology , Protein Stability , Proto-Oncogene Proteins c-mdm2/metabolism , Receptors, Androgen/analysis , Ubiquitination , beta-Arrestins
6.
Mol Cancer Ther ; 5(8): 1958-66, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16928816

ABSTRACT

Disruption of intracellular calcium initiates multiple cell-damaging processes, such as apoptosis. In normal cells, the levels of Ca(2+) are low in the mitochondria, whereas in apoptotic cells, Ca(2+) increases. Mitochondria uptake Ca(2+) via an inner membrane channel called the uniporter and extrude it into the cytoplasm through a Na(+)/Ca(2+) exchanger. Overload of Ca(2+) in the mitochondria in CGP-treated cells leads to its damage, thus affecting cellular function and survival. The goal of these experiments was to determine the importance of mitochondrial calcium ([Ca(2+)](m)) in apoptosis of prostate cancer cells. Furthermore, we have examined the advantages of increasing the [Ca(2+)](m) and treating the cells with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a potent apoptotic agent. Our results show that, under these treatment conditions, inhibiting the Na(+)/Ca(2+) exchanger using benzothiazepin CGP-37157 (CGP) did not induce apoptosis. However, combination of CGP and TRAIL increased the apoptotic response approximately 25-fold compared with control. Increase in apoptosis followed enhanced levels of [Ca(2+)](m) and was accompanied by pronounced mitochondrial changes characteristic of mitochondria-mediated apoptosis. Experiments with calcium ionophores showed that mere increase in cytosolic and/or mitochondrial Ca(2+) was not sufficient to induce apoptosis. These results have therapeutic implications as inhibitors of Na(+)/Ca(2+) exchanger are being used for treating some neurologic and cardiologic ailments, and TRAIL induces apoptosis preferentially in cancer cells. Furthermore, this system provides an excellent model to investigate the role of [Ca(2+)](m) in apoptosis.


Subject(s)
Apoptosis/drug effects , Calcium Channel Blockers/pharmacology , Prostatic Neoplasms/drug therapy , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Androgens/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Calcium/metabolism , Calcium Channel Blockers/administration & dosage , Clonazepam/analogs & derivatives , Clonazepam/pharmacology , Drug Synergism , Humans , Male , Mitochondria/drug effects , Mitochondria/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/metabolism , Thiazepines/pharmacology , Tumor Cells, Cultured
7.
Int J Cancer ; 119(1): 221-8, 2006 Jul 01.
Article in English | MEDLINE | ID: mdl-16450389

ABSTRACT

Treatment of cancer cells with histone deacetylase inhibitors (HDACi) such as suberolylanilide hydroxamic acid (SAHA) activates genes that promote apoptosis. To enhance proapoptotic efficiency, SAHA has been used in combination with radiation, kinase inhibitors and cytotoxic drugs. Although several prostate cells respond to TNFalpha-Related Apoptosis-Inducing Ligand (TRAIL), LNCaP are resistant. This model system was utilized to examine the advantages of combined treatment with SAHA and TRAIL. In LNCaP cells, TRAIL induced synergistic apoptosis when combined even with the lowest dose of SAHA. Treatment with caspase inhibitor confirmed that SAHA-induced apoptosis was mediated through caspases. In addition to induction of apoptosis, SAHA and TRAIL decreased the levels of proapoptotic proteins IKKalpha, IKKbeta and IKKgamma, suggesting that SAHA treatment may reduce the activity of NFkappaB. However, assay for NFkappaB luciferase reporter activity showed highly significant increase in SAHA-treated cells, supporting earlier suggestions that HDACi promotes NFkappaB transcriptional activity. Further analyses to determine the mechanisms by which the combination of SAHA and TRAIL led to synergistic apoptosis indicated that the apoptotic response of LNCaP is due to a complex regulation of death receptor pathway and alterations of NFkappaB activity at several regulatory steps.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/pharmacology , Apoptosis , Hydroxamic Acids/pharmacology , Membrane Glycoproteins/pharmacology , Prostatic Neoplasms/drug therapy , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis/drug effects , Caspases/drug effects , Caspases/metabolism , Cell Line, Tumor , Drug Synergism , Enzyme Inhibitors/pharmacology , Humans , Luciferases/metabolism , Male , NF-kappa B/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand , Vorinostat
8.
Mol Cancer Ther ; 5(1): 170-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16432176

ABSTRACT

Due to its specificity and effectiveness, tumor necrosis factor-alpha-related apoptosis-inducing ligand (TRAIL) is being tested for cancer therapy. Inhibition of the function of heat shock protein 90 (HSP90) is under clinical trials for cancer therapy. However, some cancer cells are resistant to TRAIL, and at the dose required for inducing apoptosis, geldanamycin, a drug that inhibits HSP90 function, has shown adverse effects. Therefore, our working plan was to identify a sublethal dose of geldanamycin and combine it with TRAIL to induce apoptosis in TRAIL-resistant prostate cancer cells. Treatment of LNCaP with 250 nmol/L geldanamycin inhibited HSP90 function but did not induce significant apoptosis. However, combination of geldanamycin and TRAIL induced highly significant apoptosis in TRAIL-resistant LNCaP cells. In addition to inducing caspase activity and apoptosis, treatment with geldanamycin and TRAIL decreased inhibitor of kappaB (IkappaB) kinase (IKK) complex proteins, IKKalpha, IKKbeta, and IKKgamma. The loss of IKK affected IkappaBalpha/nuclear factor-kappaB (NF-kappaB) interaction and reduced nuclear transport of NF-kappaB, resulting in reduced NF-kappaB activity. Our data show increase in apoptosis using low, suboptimal dose of geldanamycin when used with TRAIL. These results provide a means to alleviate two problems: resistance to TRAIL and adverse effects of high-dose geldanamycin.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Apoptosis Regulatory Proteins/administration & dosage , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Membrane Glycoproteins/administration & dosage , Quinones/pharmacology , Tumor Necrosis Factor-alpha/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/pharmacology , Benzoquinones , Dose-Response Relationship, Drug , HSP90 Heat-Shock Proteins/metabolism , Humans , I-kappa B Kinase/drug effects , I-kappa B Kinase/metabolism , I-kappa B Proteins/drug effects , I-kappa B Proteins/metabolism , Lactams, Macrocyclic , Male , Membrane Glycoproteins/pharmacology , NF-KappaB Inhibitor alpha , NF-kappa B/drug effects , NF-kappa B/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Quinones/administration & dosage , TNF-Related Apoptosis-Inducing Ligand , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/pharmacology
9.
Neoplasia ; 7(12): 1104-11, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16354593

ABSTRACT

The proteasome inhibitor Velcade (bortezomib/PS-341) has been shown to block the targeted proteolytic degradation of short-lived proteins that are involved in cell maintenance, growth, division, and death, advocating the use of proteasomal inhibitors as therapeutic agents. Although many studies focused on the use of one proteasomal inhibitor for therapy, we hypothesized that the combination of proteasome inhibitors Lactacystin (AG Scientific, Inc., San Diego CA) and MG132 (Biomol International, Plymouth Meeting, PA) may be more effective in inducing apoptosis. Additionally, this regimen would enable the use of sublethal doses of individual drugs, thus reducing adverse effects. Results indicate a significant increase in apoptosis when LNCaP prostate cancer cells were treated with increasing levels of Lactacystin, MG132, or a combination of sublethal doses of these two inhibitors. Furthermore, induction in apoptosis coincided with a significant loss of IKKalpha, IKKbeta, and IKKgamma proteins and NFkappaB activity. In addition to describing effective therapeutic agents, we provide a model system to facilitate the investigation of the mechanism of action of these drugs and their effects on the IKK-NFkappaB axis.


Subject(s)
Acetylcysteine/analogs & derivatives , Apoptosis/drug effects , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Prostatic Neoplasms/pathology , Acetylcysteine/pharmacology , Antineoplastic Combined Chemotherapy Protocols , Drug Synergism , Humans , I-kappa B Kinase/antagonists & inhibitors , I-kappa B Kinase/metabolism , Male , NF-kappa B/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Proteasome Inhibitors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...