Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
PLoS One ; 8(4): e61836, 2013.
Article in English | MEDLINE | ID: mdl-23637916

ABSTRACT

Epidemiological studies have shown that the regular use of non-steroidal anti-inflammatory (NSAIDs) drugs is associated with a reduced risk of various cancers. In addition, in vitro and experiments in mouse models have demonstrated that NSAIDs decrease tumor initiation and/or progression of several cancers. However, there are limited preclinical studies investigating the effects of NSAIDs in ovarian cancer. Here, we have studied the effects of two NSAIDs, diclofenac and indomethacin, in ovarian cancer cell lines and in a xenograft mouse model. Diclofenac and indomethacin treatment decreased cell growth by inducing cell cycle arrest and apoptosis. In addition, diclofenac and indomethacin reduced tumor volume in a xenograft model of ovarian cancer. To identify possible molecular pathways mediating the effects of NSAID treatment in ovarian cancer, we performed microarray analysis of ovarian cancer cells treated with indomethacin or diclofenac. Interestingly, several of the genes found downregulated following diclofenac or indomethacin treatment are transcriptional target genes of E2F1. E2F1 was downregulated at the mRNA and protein level upon treatment with diclofenac and indomethacin, and overexpression of E2F1 rescued cells from the growth inhibitory effects of diclofenac and indomethacin. In conclusion, NSAIDs diclofenac and indomethacin exert an anti-proliferative effect in ovarian cancer in vitro and in vivo and the effects of NSAIDs may be mediated, in part, by downregulation of E2F1.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , E2F1 Transcription Factor/metabolism , Ovarian Neoplasms/metabolism , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Diclofenac/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , E2F1 Transcription Factor/genetics , Female , Gene Expression , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indomethacin/pharmacology , Mice , Mice, Nude , Ovarian Neoplasms/genetics , Xenograft Model Antitumor Assays
2.
Crit Care Nurse ; 33(2): 46-56, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23547125

ABSTRACT

According to estimates, 1.2 million Americans are infected with human immuno deficiency virus (HIV). Because of antiretroviral therapy, persons who have HIV infection or have progressed to AIDS are living longer. As a result, the likelihood that they will need critical care nursing is increasing. Unlike in years past, when these patients were often admitted because of the consequences of immunosuppression, today they are also being cared for in critical care units for other conditions associated with aging, other chronic health conditions, and trauma. When persons who have HIV disease or AIDS are admitted to the intensive care unit, nurses must be prepared to provide care, especially management of complexities associated with antiretroviral therapy. Therefore, this article examines critical care nurses' role in initiating and administering antiretroviral therapy in the intensive care unit and reducing the risk of drug interactions associated with the therapy.


Subject(s)
Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active/nursing , Critical Care/methods , HIV Infections/nursing , Intensive Care Units , Acquired Immunodeficiency Syndrome/nursing , Adult , Anti-HIV Agents/adverse effects , Antiretroviral Therapy, Highly Active/adverse effects , Female , HIV Infections/drug therapy , Humans , Life Expectancy , Male , Middle Aged , Quality of Life , Risk Factors , Treatment Outcome
3.
J Gerontol A Biol Sci Med Sci ; 64(11): 1146-53, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19692671

ABSTRACT

Tight junctions (TJs) play crucial roles in tissue homeostasis and inflammation through their roles in the control of paracellular transport and barrier function. There is evidence that these functions are compromised in older organisms, but the exact mechanisms leading to TJ deterioration are not well understood. Claudin proteins are a family of membrane proteins that constitute the structural barrier elements of TJs and therefore play a major role in their formation and function. Using immunohistochemistry and immunoblotting, we have studied the expression of six different claudin proteins (claudin-1, -2, -3, -4, -5, and -7) in three tissues (liver, kidney, and pancreas) of aging male and female mice. In general, we find an age-dependent decrease in the expression of several claudin proteins in all three tissues observed, although the exact changes are tissue specific. Our findings provide a possible basis for the decrease in tissue barrier function in older organisms.


Subject(s)
Aging/metabolism , Kidney/chemistry , Liver/chemistry , Membrane Proteins/analysis , Pancreas/chemistry , Tight Junctions/chemistry , Animals , Claudin-1 , Claudin-3 , Claudin-4 , Claudin-5 , Claudins , Female , Immunoblotting , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Tight Junctions/physiology
4.
Cancer Biol Ther ; 6(11): 1733-42, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17986852

ABSTRACT

The claudin (CLDN) genes encode a family of proteins involved in the formation and function of tight junctions. CLDN gene expression is frequently altered in several human cancers, and in particular, CLDN3 and CLDN4 are commonly overexpressed in ovarian cancer. However, the mechanisms leading to the deregulation of these genes in cancer remain unclear. In the present study, we have examined the CLDN3 promoter and have identified a minimal region containing an Sp1 site crucial for its activity. In addition, we find that the CLDN3 promoter is regulated through epigenetic processes. Cells that express high levels of CLDN3 exhibit low DNA methylation and high histone H3 acetylation of the critical CLDN3 promoter region, and the reverse is observed in cells that do not express this gene. CLDN3-negative cells can be induced to express CLDN3 through treatment with DNA methyltransferase or histone deacetylase inhibitors. Interestingly, in vitro binding experiments, as well as chip assays show that Sp1 binds the unmethylated promoter much more efficiently, providing a mechanism for CLDN3 silencing in non-expressing cells. Finally, siRNA-mediated knockdown of Sp1 led to a significant decrease of CLDN3 expression at both the mRNA and protein levels, demonstrating a crucial role for this transcription factor in the regulation of CLDN3. Our data provide a basis for CLDN3 expression in ovarian cancer cells, as well as a mechanism for the silencing of this promoter in tumors lacking expression of claudin-3.


Subject(s)
Membrane Proteins/genetics , Ovarian Neoplasms/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Binding Sites , Cell Line, Tumor , Claudin-3 , Claudin-4 , CpG Islands , DNA Methylation , Decitabine , Female , Humans , Hydroxamic Acids/pharmacology , Mutagenesis, Site-Directed , Ovarian Neoplasms/pathology , Promoter Regions, Genetic , RNA, Messenger/analysis , Sp1 Transcription Factor/metabolism , Sp3 Transcription Factor/metabolism
5.
Exp Cell Res ; 313(15): 3364-75, 2007 Sep 10.
Article in English | MEDLINE | ID: mdl-17678893

ABSTRACT

Claudin proteins belong to a large family of transmembrane proteins essential to the formation and maintenance of tight junctions (TJs). In ovarian cancer, TJ protein claudin-4 is frequently overexpressed and may have roles in survival and invasion, but the molecular mechanisms underlying its regulation are poorly understood. In this report, we show that claudin-4 can be phosphorylated by protein kinase C (PKC) at Thr189 and Ser194 in ovarian cancer cells and overexpression of a claudin-4 mutant protein mimicking the phosphorylated state results in the disruption of the barrier function. Furthermore, upon phorbol ester-mediated PKC activation of OVCA433 cells, TJ strength is decreased and claudin-4 localization is altered. Analyses using PKC inhibitors and siRNA suggest that PKCepsilon, an isoform typically expressed in ovarian cancer cells, may be important in the TPA-mediated claudin-4 phosphorylation and weakening of the TJs. Furthermore, immunofluorescence studies showed that claudin-4 and PKCepsilon are co-localized at the TJs in these cells. The modulation of claudin-4 activity by PKCepsilon may not only provide a mechanism for disrupting TJ function in ovarian cancer, but may also be important in the regulation of TJ function in normal epithelial cells.


Subject(s)
Membrane Proteins/metabolism , Ovarian Neoplasms/metabolism , Protein Kinase C-epsilon/metabolism , Tight Junctions/physiology , Cell Line, Tumor , Claudin-4 , Female , Humans , Indoles/pharmacology , Maleimides/pharmacology , Membrane Proteins/genetics , Mutation , Phosphorylation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/metabolism , Protein Kinase C-epsilon/antagonists & inhibitors , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
6.
J Biol Chem ; 282(20): 14816-26, 2007 May 18.
Article in English | MEDLINE | ID: mdl-17389601

ABSTRACT

The actin-binding protein filamin A (FLNa) is associated with diverse cellular processes such as cell motility and signaling through its scaffolding properties. Here we examine the effect of FLNa on the regulation of signaling pathways that control the expression of matrix metalloproteinases (MMPs). The lack of FLNa in human M2 melanoma cells was associated with constitutive and phorbol ester-induced expression and secretion of active MMP-9 in the absence of MMP-2 up-regulation. M2 cells displayed stronger MMP-9 production and activity than their M2A7 counterparts where FLNa had been stably reintroduced. Using an MMP-9 promoter construct (pMMP-9-Luc), in vitro kinase assays, and genetic and pharmacological approaches, we demonstrate that FLNa mediated transcriptional down-regulation of pMMP-9-Luc by suppressing the constitutive hyperactivity of the Ras/MAPK extracellular signal-regulated kinase (ERK) cascade. Experimental evidence indicated that this phenomenon was associated with destabilization and ubiquitylation of Ras-GRF1, a guanine nucleotide exchange factor that activates H-Ras by facilitating the release of GDP. Ectopic expression of Ras-GRF1 was accompanied by ERK activation and elevated levels of MMP-9 in M2A7 cells, whereas a catalytically inactive dominant negative Ras-GRF1, which prevented ERK activation, reduced MMP-9 expression in M2 cells. Our results indicate that expression of FLNa regulates constitutive activation of the Ras/ERK pathway partly through a Ras-GRF1 mechanism to modulate the production of MMP-9.


Subject(s)
Contractile Proteins/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System , Matrix Metalloproteinase 9/biosynthesis , Microfilament Proteins/biosynthesis , ras-GRF1/metabolism , Carcinogens/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation/drug effects , Filamins , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 2/biosynthesis , Melanoma , Proto-Oncogene Proteins p21(ras)/metabolism , Tetradecanoylphorbol Acetate/pharmacology
7.
Cancer Res ; 65(16): 7378-85, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-16103090

ABSTRACT

Claudin proteins form a large family of integral membrane proteins crucial for tight junction formation and function. Our previous studies have revealed that claudin-3 and claudin-4 proteins are highly overexpressed in ovarian cancer. To clarify the roles of claudins in ovarian tumorigenesis, we have generated human ovarian surface epithelial (HOSE) cells constitutively expressing wild-type claudin-3 and claudin-4. Expression of these claudins in HOSE cells increased cell invasion and motility as measured by Boyden chamber assays and wound-healing experiments. Conversely, small interfering RNA (siRNA)-mediated knockdown of claudin-3 and claudin-4 expression in ovarian cancer cell lines reduced invasion. Claudin expression also increased cell survival in HOSE cells but did not significantly affect cell proliferation. Moreover, the claudin-expressing ovarian epithelial cells were found to have increased matrix metalloproteinase-2 (MMP-2) activity indicating that claudin-mediated increased invasion might be mediated through the activation of MMP proteins. However, siRNA inactivation of claudins in ovarian cancer cell lines did not have a significant effect on the high endogenous MMP-2 activity present in these cells, showing that malignant cells have alternative or additional pathways to fully activate MMP-2. Taken together, our results suggest that claudin overexpression may promote ovarian tumorigenesis and metastasis through increased invasion and survival of tumor cells.


Subject(s)
Matrix Metalloproteinase 2/metabolism , Membrane Proteins/biosynthesis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Animals , Cell Growth Processes/physiology , Cell Survival/physiology , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Claudin-3 , Claudin-4 , Dogs , Female , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Neoplasm Invasiveness , Ovarian Neoplasms/enzymology , Ovary/enzymology , Ovary/metabolism , Ovary/pathology , RNA, Small Interfering/genetics , Transfection
8.
J Biol Chem ; 280(28): 26233-40, 2005 Jul 15.
Article in English | MEDLINE | ID: mdl-15905176

ABSTRACT

Claudins are integral membrane proteins essential in the formation and function of tight junctions (TJs). Disruption of TJs, which have essential roles in cell permeability and polarity, is thought to contribute to epithelial tumorigenesis. Claudin-3 and -4 are frequently overexpressed in ovarian cancer, but the molecular pathways involved in the regulation of these proteins are unclear. Interestingly, several studies have demonstrated a role for phosphorylation in the regulation of TJ complexes, although evidence for claudin phosphorylation is scarce. Here, we showed that claudin-3 and -4 can be phosphorylated in ovarian cancer cells. In vitro phosphorylation assays using glutathione S-transferase fusion constructs demonstrated that the C terminus of claudin-3 is an excellent substrate for cAMP-dependent protein kinase (PKA). Using site-directed mutagenesis, we identified a PKA phosphorylation site at amino acid 192 in the C terminus of claudin-3. Overexpression of the protein containing a T192D mutation, mimicking the phosphorylated state, resulted in a decrease in TJ strength in ovarian cancer cell line OVCA433. Our results suggest that claudin-3 phosphorylation by PKA, a kinase frequently activated in ovarian cancer, may provide a mechanism for the disruption of TJs in this cancer. In addition, our findings may have general implications for the regulation of TJs in normal epithelial cells.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/physiology , Glutathione Transferase/metabolism , Membrane Proteins/chemistry , Membrane Proteins/physiology , Ovarian Neoplasms/metabolism , Threonine/chemistry , Tight Junctions/chemistry , Binding Sites , Calcium/metabolism , Cell Line , Cell Line, Tumor , Claudin-3 , Claudin-4 , Cyclic AMP-Dependent Protein Kinases/chemistry , Cyclic AMP-Dependent Protein Kinases/metabolism , Electric Impedance , Electrophoresis, Polyacrylamide Gel , Electrophysiology , Enzyme Inhibitors/pharmacology , Female , Humans , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Membrane Proteins/metabolism , Microscopy, Fluorescence , Models, Genetic , Mutation , Permeability , Phosphorylation , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Time Factors , Transfection
9.
Pflugers Arch ; 448(6): 613-20, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15243742

ABSTRACT

Intermediate-conductance (IK) Ca(2+)-activated K(+) channels are expressed in many different cell types where they perform a variety of functions including cell volume regulation, transepithelial secretion, lymphocyte activation and cell cycle progression. IK channels are thought to be regulated by phosphorylation; however, whether kinases act directly on the channel is unclear. Using IK channels heterologously expressed in Xenopus oocytes, we demonstrate that IK channels are potently inhibited (60%) by the catalytic subunit of protein kinase A (PKA). Inhibition of IK channel current by PKA is abolished by mutation of four phosphorylation residues (S312, T327, S332, and T348) in the putative calmodulin-binding region of the channel. Evidence for direct modulation of the IK channel by PKA was further demonstrated using GST fusion proteins. The major site of phosphorylation was found to be serine 332; however, other residues were also phosphorylated. We conclude that IK channels can be directly regulated by the cAMP second-messenger system. The mechanism appears to involve direct phosphorylation by PKA of a modulatory locus in the cytoplasmic region of the channel, the site at which calmodulin is thought to interact. Modulation of IK channels by protein kinases may be an important mechanism regulating cell function.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/pharmacology , Oocytes/drug effects , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Amino Acid Sequence , Animals , Consensus Sequence , Electric Conductivity , Gene Expression , Glutathione/genetics , Glutathione/metabolism , In Vitro Techniques , Molecular Sequence Data , Mutagenesis , Oocytes/metabolism , Phosphorylation , Potassium Channels, Calcium-Activated/genetics , Potassium Channels, Calcium-Activated/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Xenopus
10.
Clin Cancer Res ; 9(7): 2567-75, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12855632

ABSTRACT

PURPOSE: Claudin proteins represent a large family of integral membrane proteins crucial for tight junction (TJ) formation and function. Claudins have been shown to be up-regulated in various cancers and have been suggested as possible biomarkers and targets for cancer therapy. Because claudin-3 and claudin-4 have been proposed to be expressed in epithelial ovarian cancer, we have performed a detailed analysis of CLDN3 and CLDN4 expression in a panel of ovarian tumors of various subtypes and cell lines. We also investigated whether high expression of claudin-3 and claudin-4 was associated with TJ function in ovarian cancer cells. EXPERIMENTAL DESIGN: RNA was obtained from a panel of 39 microdissected epithelial ovarian tumors of various histological subtypes for real-time reverse transcription-PCR analysis. In addition, a total of 70 cases of ovarian carcinomas, ovarian cysts, and normal ovarian epithelium from a tissue array were analyzed by immunohistochemistry. Finally, a panel of cell lines was used for Western analysis of claudin expression and TJ permeability studies. RESULTS: Although expressed at low levels in some normal human tissues, including the ovary, CLDN3 and CLDN4 are highly up-regulated in epithelial ovarian cancers of all subtypes. Immunohistochemical analyses using our ovarian tissue array confirmed the high level of expression of claudin-3 and claudin-4 in the majority of ovarian carcinomas, including many tumors exhibiting cytoplasmic staining. Ovarian cystadenoma did not frequently overexpress these proteins, suggesting that the expression of these proteins is associated with malignancy. In ovarian cancer cell lines, claudin-3 and claudin-4 expression was not associated with functional TJs as measured by transepithelial electrical resistance. CONCLUSIONS: These results show that CLDN3 and CLDN4 are frequently up-regulated in ovarian tumors and cell lines and may represent novel markers for this disease. Overexpression of these genes in ovarian cancer also suggests interesting scenarios for the involvement of TJ in tumorigenesis. A better knowledge of the mechanisms underlying ovarian tumorigenesis will likely result in the development of novel approaches for the diagnosis and therapy of this deadly disease.


Subject(s)
Cystadenoma, Mucinous/metabolism , Cystadenoma, Serous/metabolism , Membrane Proteins/biosynthesis , Ovarian Neoplasms/metabolism , Blotting, Northern , Cell Line, Tumor , Claudin-3 , Claudin-4 , Cystadenoma/metabolism , Cytoplasm/metabolism , Epithelium/metabolism , Female , Humans , Immunoblotting , Immunohistochemistry , Reverse Transcriptase Polymerase Chain Reaction , Tight Junctions/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...