Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Proc Natl Acad Sci U S A ; 107(14): 6459-64, 2010 Apr 06.
Article in English | MEDLINE | ID: mdl-20308550

ABSTRACT

The phosphatase and tensin homolog (PTEN) is a tumor suppressor that is inactivated in many human cancers. PTEN loss has been associated with resistance to inhibitors of the epidermal growth factor receptor (EGFR), but the molecular basis of this resistance is unclear. It is believed that unopposed phosphatidylinositol-3-kinase (PI3K) activation through multiple receptor tyrosine kinases (RTKs) can relieve PTEN-deficient cancers from their "dependence" on EGFR or any other single RTK for survival. Here we report a distinct resistance mechanism whereby PTEN inactivation specifically raises EGFR activity by impairing the ligand-induced ubiquitylation and degradation of the activated receptor through destabilization of newly formed ubiquitin ligase Cbl complexes. PTEN-associated resistance to EGFR kinase inhibitors is phenocopied by expression of dominant negative Cbl and can be overcome by more complete EGFR kinase inhibition. PTEN inactivation does not confer resistance to inhibitors of the MET or PDGFRA kinase. Our study identifies a critical role for PTEN in EGFR signal termination and suggests that more potent EGFR inhibition should overcome resistance caused by PI3K pathway activation.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , PTEN Phosphohydrolase/metabolism , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis , Cell Line , Enzyme Activation , Humans , Mice , Mice, Knockout , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Protein Binding , Proto-Oncogene Proteins c-cbl/metabolism , RNA Interference , Signal Transduction/drug effects , Ubiquitination
2.
Exp Cell Res ; 317(6): 838-48, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21211535

ABSTRACT

High levels of the soluble form of E-cadherin can be found in the serum of cancer patients and are associated with poor prognosis. Despite the possible predictive value of soluble E-cadherin, little is understood concerning its patho-physiological consequences in tumor progression. In this study, we show that soluble E-cadherin facilitates cell survival via functional interaction with cellular E-cadherin. Exposure of cells to a recombinant form of soluble E-cadherin, at a concentration found in cancer patient's serum, prevents apoptosis due to serum/growth factor withdrawal, and inhibits epithelial lumen formation, a process that requires apoptosis. Further, soluble E-cadherin-mediated cell survival involves activation of the epidermal growth factor receptor (EGFR) and EGFR-mediated activation of both phosphoinositide-3 kinase (PI3K)/AKT and ERK1/2 signaling pathways. These results are evidence of a complex functional interplay between EGFR and E-cadherin and also suggest that the presence of soluble E-cadherin in cancer patients' sera might have relevance to cell survival and tumor progression.


Subject(s)
Apoptosis/drug effects , Cadherins/pharmacology , ErbB Receptors/metabolism , Gene Expression Regulation/drug effects , Cadherins/metabolism , Cell Survival/drug effects , HEK293 Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oncogene Protein v-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Solubility
3.
BMC Cancer ; 11: 144, 2011 Apr 18.
Article in English | MEDLINE | ID: mdl-21501498

ABSTRACT

BACKGROUND: Medulloblastoma is the most common brain tumor in children, and its prognosis is worse than for many other common pediatric cancers. Survivors undergoing treatment suffer from serious therapy-related side effects. Thus, it is imperative to identify safer, effective treatments for medulloblastoma. In this study we evaluated the anti-cancer potential of curcumin in medulloblastoma by testing its ability to induce apoptosis and inhibit tumor growth in vitro and in vivo using established medulloblastoma models. METHODS: Using cultured medulloblastoma cells, tumor xenografts, and the Smo/Smo transgenic medulloblastoma mouse model, the antitumor effects of curcumin were tested in vitro and in vivo. RESULTS: Curcumin induced apoptosis and cell cycle arrest at the G2/M phase in medulloblastoma cells. These effects were accompanied by reduced histone deacetylase (HDAC) 4 expression and activity and increased tubulin acetylation, ultimately leading to mitotic catastrophe. In in vivo medulloblastoma xenografts, curcumin reduced tumor growth and significantly increased survival in the Smo/Smo transgenic medulloblastoma mouse model. CONCLUSIONS: The in vitro and in vivo data suggest that curcumin has the potential to be developed as a therapeutic agent for medulloblastoma.


Subject(s)
Antineoplastic Agents/pharmacology , Cerebellar Neoplasms/drug therapy , Curcumin/pharmacology , Histone Deacetylases/metabolism , Medulloblastoma/drug therapy , Repressor Proteins/metabolism , Acetylation/drug effects , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Cell Growth Processes/drug effects , Cell Growth Processes/genetics , Cell Line, Tumor , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Curcumin/therapeutic use , Histone Deacetylases/genetics , Humans , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Mice, Transgenic , Receptors, G-Protein-Coupled/genetics , Repressor Proteins/genetics , Smoothened Receptor , Tubulin/metabolism , Xenograft Model Antitumor Assays
4.
J Mol Cell Cardiol ; 47(4): 552-60, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19683723

ABSTRACT

Na,K-ATPase is composed of two essential alpha- and beta-subunits, both of which have multiple isoforms. Evidence indicates that the Na,K-ATPase enzymatic activity as well as its alpha(1), alpha(3) and beta(1) isoforms are reduced in the failing human heart. The catalytic alpha-subunit is the receptor for cardiac glycosides such as digitalis, used for the treatment of congestive heart failure. The role of the Na,K-ATPase beta(1)-subunit (Na,K-beta(1)) in cardiac function is not known. We used Cre/loxP technology to inactivate the Na,K-beta(1) gene exclusively in the ventricular cardiomyocytes. Animals with homozygous Na,K-beta(1) gene excision were born at the expected Mendelian ratio, grew into adulthood, and appeared to be healthy until 10 months of age. At 13-14 months, these mice had 13% higher heart/body weight ratios, and reduced contractility as revealed by echocardiography compared to their wild-type (WT) littermates. Pressure overload by transverse aortic constriction (TAC) in younger mice, resulted in compensated hypertrophy in WT mice, but decompensation in the Na,K-beta(1) KO mice. The young KO survivors of TAC exhibited decreased contractile function and mimicked the effects of the Na,K-beta(1) KO in older mice. Further, we show that intact hearts of Na,K-beta(1) KO anesthetized mice as well as isolated cardiomyocytes were insensitive to ouabain-induced positive inotropy. This insensitivity was associated with a reduction in NCX1, one of the proteins involved in regulating cardiac contractility. In conclusion, our results demonstrate that Na,K-beta(1) plays an essential role in regulating cardiac contractility and that its loss is associated with significant pathophysiology of the heart.


Subject(s)
Gene Deletion , Myocardial Contraction/drug effects , Myocardium/enzymology , Ouabain/pharmacology , Protein Subunits/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Aging/drug effects , Animals , Calcium Signaling/drug effects , Cardiomegaly/enzymology , Cardiomegaly/physiopathology , Cell Separation , Heart Function Tests , Immunoblotting , Mice , Mice, Knockout , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Organ Specificity/drug effects , Pressure , Sodium-Calcium Exchanger/metabolism
5.
Biochim Biophys Acta ; 1778(3): 757-69, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18086552

ABSTRACT

Tight junctions are unique organelles in epithelial cells. They are localized to the apico-lateral region and essential for the epithelial cell transport functions. The paracellular transport process that occurs via tight junctions is extensively studied and is intricately regulated by various extracellular and intracellular signals. Fine regulation of this transport pathway is crucial for normal epithelial cell functions. Among factors that control tight junction permeability are ions and their transporters. However, this area of research is still in its infancy and much more needs to be learned about how these molecules regulate tight junction structure and functions. In this review we have attempted to compile literature on ion transporters and channels involved in the regulation of tight junctions.


Subject(s)
Tight Junctions/physiology , Animals , Epithelial Cells/physiology , Epithelial Cells/ultrastructure , Humans , Ion Channels/physiology , Membrane Proteins/chemistry , Membrane Proteins/physiology , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/physiology , Models, Biological , Models, Molecular , Paracrine Communication , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/physiology , Tight Junctions/ultrastructure
6.
Mol Cancer Ther ; 7(7): 2142-51, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18645024

ABSTRACT

Prostate-specific membrane antigen (PSMA) is a transmembrane protein highly expressed in advanced and metastatic prostate cancers. The pathologic consequence of elevated PSMA expression in not known. Here, we report that PSMA is localized to a membrane compartment in the vicinity of mitotic spindle poles and associates with the anaphase-promoting complex (APC). PSMA-expressing cells prematurely degrade cyclin B and exit mitosis due to increased APC activity and incomplete inactivation of APC by the spindle assembly checkpoint. Further, expression of PSMA in a karyotypically stable cell line induces aneuploidy. Thus, these findings provide the first evidence that PSMA has a causal role in the induction of aneuploidy and might play an etiologic role in the progression of prostate cancer.


Subject(s)
Chromosomal Instability , Prostate-Specific Antigen/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Centrosome/drug effects , Centrosome/ultrastructure , Chromosomal Instability/drug effects , Cyclin B/metabolism , Cyclin B1 , Dogs , Humans , Nocodazole/pharmacology , Prostate-Specific Antigen/ultrastructure , Protein Binding/drug effects , Protein Transport/drug effects , Spindle Apparatus/metabolism
7.
Mol Cancer Ther ; 7(6): 1386-97, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18566211

ABSTRACT

Loss of alpha-catenin is one of the characteristics of prostate cancer. The catenins (alpha and beta) associated with E-cadherin play a critical role in the regulation of cell-cell adhesion. Tyrosine phosphorylation of beta-catenin dissociates it from E-cadherin and facilitates its entry into the nucleus, where beta-catenin acts as a transcriptional activator inducing genes involved in cell proliferation. Thus, beta-catenin regulates cell-cell adhesion and cell proliferation. Mechanisms controlling the balance between these functions of beta-catenin invariably are altered in cancer. Although a wealth of information is available about beta-catenin deregulation during oncogenesis, much less is known about how or whether alpha-catenin regulates beta-catenin functions. In this study, we show that alpha-catenin acts as a switch regulating the cell-cell adhesion and proliferation functions of beta-catenin. In alpha-catenin-null prostate cancer cells, reexpression of alpha-catenin increased cell-cell adhesion and decreased beta-catenin transcriptional activity, cyclin D1 levels, and cell proliferation. Further, Src-mediated tyrosine phosphorylation of beta-catenin is a major mechanism for decreased beta-catenin interaction with E-cadherin in alpha-catenin-null cells. alpha-Catenin attenuated the effect of Src phosphorylation by increasing beta-catenin association with E-cadherin. We also show that alpha-catenin increases the sensitivity of prostate cancer cells to a Src inhibitor in suppressing cell proliferation. This study reveals for the first time that alpha-catenin is a key regulator of beta-catenin transcriptional activity and that the status of alpha-catenin expression in tumor tissues might have prognostic value for Src targeted therapy.


Subject(s)
Oncogene Proteins/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction , alpha Catenin/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cell Proliferation , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic , Humans , Intercellular Junctions/metabolism , Intercellular Junctions/ultrastructure , Male , Phosphorylation , Transcription, Genetic , beta Catenin/genetics
8.
J Mol Biol ; 365(3): 706-14, 2007 Jan 19.
Article in English | MEDLINE | ID: mdl-17078968

ABSTRACT

Na,K-ATPase is a hetero-oligomer of alpha and beta-subunits. The Na,K-ATPase beta-subunit (Na,K-beta) is involved in both the regulation of ion transport activity, and in cell-cell adhesion. By structure prediction and evolutionary analysis, we identified two distinct faces on the Na,K-beta transmembrane domain (TMD) that could mediate protein-protein interactions: a glycine zipper motif and a conserved heptad repeat. Here, we show that the heptad repeat face is involved in the hetero-oligomeric interaction of Na,K-beta with Na,K-alpha, and the glycine zipper face is involved in the homo-oligomerization of Na,K-beta. Point mutations in the heptad repeat motif reduced Na,K-beta binding to Na,K-alpha, and Na,K-ATPase activity. Na,K-beta TMD homo-oligomerized in biological membranes, and mutation of the glycine zipper motif affected oligomerization and cell-cell adhesion. These results provide a structural basis for understanding how Na,K-beta links ion transport and cell-cell adhesion.


Subject(s)
Models, Molecular , Protein Structure, Quaternary , Protein Subunits/chemistry , Protein Subunits/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/metabolism , Amino Acid Sequence , Animals , Cell Aggregation , Cell Membrane/enzymology , Dogs , Glycine/genetics , Leucine/genetics , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation/genetics , Protein Structure, Secondary , Protein Structure, Tertiary , Repetitive Sequences, Amino Acid
9.
J Urol ; 179(1): 338-45, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18006011

ABSTRACT

PURPOSE: Na,K-adenosine triphosphatase, which is composed of a catalytic alpha-subunit and a regulatory beta-subunit, generates an electrochemical gradient across the plasma membrane. Previous studies demonstrated altered Na,K-adenosine triphosphatase subunit expression in renal clear cell carcinoma and an association of subunit levels with the prediction of recurrent bladder cancer. We determined the clinical association of protein expression patterns of the Na,K-adenosine triphosphatase alpha1 and beta1-subunits in renal clear cell carcinoma using tissue microarrays with linked clinicopathological data. MATERIALS AND METHODS: The UCLA kidney cancer tissue microarray was used to investigate the protein expression of Na,K-adenosine triphosphatase alpha1 and beta1-subunits by immunohistochemistry in 342 patients with renal clear cell carcinoma who were treated with radical nephrectomy. Of these patients clinical outcomes studies were performed in 317. The resultant expression reactivity was correlated with clinicopathological variables. RESULTS: We found that the alpha1-subunit was a significant and independent predictor of disease specific death from renal clear cell carcinoma on multivariate Cox proportional hazards analysis that included established prognostic factors Eastern Cooperative Oncology Group performance status, pT status, metastasis status and tumor grade. Significance was found when examining all patients with clear cell renal cell carcinoma as well as patient substrata with low or high grade tumors and localized or metastatic disease, suggesting that the Na,K-adenosine triphosphatase alpha1-subunit could be used as a new prognosticator for disease specific death from renal clear cell carcinoma. CONCLUSIONS: These results suggest that Na,K-adenosine triphosphatase alpha1-subunit expression patterns may be a useful clinical prognosticator for renal clear cell carcinoma. The Na,K-adenosine triphosphatase beta1-subunit was not found to be a useful prognosticator in this setting.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/mortality , Kidney Neoplasms/enzymology , Kidney Neoplasms/mortality , Sodium-Potassium-Exchanging ATPase/biosynthesis , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Survival Rate
10.
Mol Biol Cell ; 16(3): 1082-94, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15616195

ABSTRACT

The Na,K-ATPase, consisting of alpha- and beta-subunits, regulates intracellular ion homeostasis. Recent studies have demonstrated that Na,K-ATPase also regulates epithelial cell tight junction structure and functions. Consistent with an important role in the regulation of epithelial cell structure, both Na,K-ATPase enzyme activity and subunit levels are altered in carcinoma. Previously, we have shown that repletion of Na,K-ATPase beta1-subunit (Na,K-beta) in highly motile Moloney sarcoma virus-transformed Madin-Darby canine kidney (MSV-MDCK) cells suppressed their motility. However, until now, the mechanism by which Na,K-beta reduces cell motility remained elusive. Here, we demonstrate that Na,K-beta localizes to lamellipodia and suppresses cell motility by a novel signaling mechanism involving a cross-talk between Na,K-ATPase alpha1-subunit (Na,K-alpha) and Na,K-beta with proteins involved in phosphatidylinositol 3-kinase (PI3-kinase) signaling pathway. We show that Na,K-alpha associates with the regulatory subunit of PI3-kinase and Na,K-beta binds to annexin II. These molecular interactions locally activate PI3-kinase at the lamellipodia and suppress cell motility in MSV-MDCK cells, independent of Na,K-ATPase ion transport activity. Thus, these results demonstrate a new role for Na,K-ATPase in regulating carcinoma cell motility.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Sodium-Potassium-Exchanging ATPase/physiology , Actins/chemistry , Actins/metabolism , Animals , Annexin A2/chemistry , Annexin A2/genetics , Cell Line , Cell Movement , Chromatography, Liquid , Chromones/pharmacology , Cloning, Molecular , Cytoplasm/metabolism , Cytoskeleton , Dogs , Epithelial Cells/cytology , Glutathione Transferase/metabolism , Immunoblotting , Immunoprecipitation , Ions , Mass Spectrometry , Microscopy, Confocal , Microscopy, Fluorescence , Models, Biological , Morpholines/pharmacology , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/metabolism , Phalloidine/pharmacology , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Signal Transduction , Sodium-Potassium-Exchanging ATPase/chemistry , Tight Junctions , rac1 GTP-Binding Protein/metabolism
11.
Mol Biol Cell ; 15(7): 3224-32, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15133131

ABSTRACT

The Na,K-ATPase consists of an alpha- and beta-subunit. Moloney sarcoma virus-transformed MDCK cells (MSV-MDCK) express low levels of Na,K-ATPase beta(1)-subunit. Ectopic expression of Na,K-ATPase beta(1)-subunit in these cells increased the protein levels of the alpha(1)-subunit of Na,K-ATPase. This increase was not due to altered transcription of the alpha(1)-subunit gene or half-life of the alpha(1)-subunit protein because both alpha(1)-subunit mRNA levels and half-life of the alpha(1)-subunit protein were comparable in MSV-MDCK and beta(1)-subunit expressing MSV-MDCK cells. However, short pulse labeling revealed that the initial translation rate of the alpha(1)-subunit in beta(1)-subunit expressing MSV-MDCK cells was six- to sevenfold higher compared with MSV-MDCK cells. The increased translation was specific to alpha(1)-subunit because translation rates of occludin and beta-catenin, membrane and cytosolic proteins, respectively, were not altered. In vitro cotranslation/translocation experiments using rabbit reticulocyte lysate and rough microsomes revealed that the alpha(1)-subunit mRNA is more efficiently translated in the presence of beta(1)-subunit. Furthermore, sucrose density gradient analysis revealed significantly more alpha(1)-subunit transcript associated with the polysomal fraction in beta(1)-subunit expressing MSV-MDCK cells compared with MSV-MDCK cells, indicating that in mammalian cells the Na,K-ATPase beta(1)-subunit is involved in facilitating the translation of the alpha(1)-subunit mRNA in the endoplasmic reticulum.


Subject(s)
Gene Expression Regulation, Enzymologic , Protein Biosynthesis , Protein Subunits/physiology , Sodium-Potassium-Exchanging ATPase/physiology , Animals , Cell Extracts/chemistry , Cell Line, Transformed , Cell Membrane/chemistry , Dogs , Moloney murine sarcoma virus/metabolism , Polyribosomes/chemistry , Polyribosomes/metabolism , Protein Subunits/analysis , Protein Subunits/biosynthesis , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/analysis , RNA, Messenger/metabolism , Sodium-Potassium-Exchanging ATPase/analysis , Sodium-Potassium-Exchanging ATPase/metabolism , Up-Regulation
12.
Mol Biol Cell ; 15(3): 1364-73, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14699059

ABSTRACT

The Na,K-ATPase consists of two essential alpha- and beta-subunits and regulates the intracellular Na+ and K+ homeostasis. Although the alpha-subunit contains the catalytic activity, it is not active without functional beta-subunit. Here, we report that poorly differentiated carcinoma cell lines derived from colon, breast, kidney, and pancreas show reduced expression of the Na,K-ATPase beta1-subunit. Decreased expression of beta1-subunit in poorly differentiated carcinoma cell lines correlated with increased expression of the transcription factor Snail known to down-regulate E-cadherin. Ectopic expression of Snail in well-differentiated epithelial cell lines reduced the protein levels of E-cadherin and beta1-subunit and induced a mesenchymal phenotype. Reduction of Snail expression in a poorly differentiated carcinoma cell line by RNA interference increased the levels of Na,K-ATPase beta1-subunit. Furthermore, Snail binds to a noncanonical E-box in the Na,K-ATPase beta1-subunit promoter and suppresses its promoter activity. These results suggest that down-regulation of Na,K-ATPase beta1-subunit and E-cadherin by Snail are associated with events leading to epithelial to mesenchymal transition.


Subject(s)
Carcinoma/enzymology , DNA-Binding Proteins/metabolism , Down-Regulation/genetics , Epithelial Cells/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Transcription Factors/metabolism , Animals , Cadherins/metabolism , Cell Differentiation/physiology , Dogs , E-Box Elements/genetics , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Enzymologic/physiology , Humans , Promoter Regions, Genetic/genetics , Protein Subunits/metabolism , Snail Family Transcription Factors , Tumor Cells, Cultured
13.
Mol Biol Cell ; 14(12): 4835-45, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14528023

ABSTRACT

Prostate-specific membrane antigen (PSMA) is a transmembrane protein expressed at high levels in prostate cancer and in tumor-associated neovasculature. In this study, we report that PSMA is internalized via a clathrin-dependent endocytic mechanism and that internalization of PSMA is mediated by the five N-terminal amino acids (MWNLL) present in its cytoplasmic tail. Deletion of the cytoplasmic tail abolished PSMA internalization. Mutagenesis of N-terminal amino acid residues at position 2, 3, or 4 to alanine did not affect internalization of PSMA, whereas mutation of amino acid residues 1 or 5 to alanine strongly inhibited internalization. Using a chimeric protein composed of Tac antigen, the alpha-chain of interleukin 2-receptor, fused to the first five amino acids of PSMA (Tac-MWNLL), we found that this sequence is sufficient for PSMA internalization. In addition, inclusion of additional alanines into the MWNLL sequence either in the Tac chimera or the full-length PSMA strongly inhibited internalization. From these results, we suggest that a novel MXXXL motif in the cytoplasmic tail mediates PSMA internalization. We also show that dominant negative micro2 of the adaptor protein (AP)-2 complex strongly inhibits the internalization of PSMA, indicating that AP-2 is involved in the internalization of PSMA mediated by the MXXXL motif.


Subject(s)
Adaptor Protein Complex 2/metabolism , Antigens, Surface/metabolism , Clathrin/metabolism , Endocytosis/physiology , Glutamate Carboxypeptidase II/metabolism , Amino Acid Motifs/physiology , Animals , Antigens, Surface/chemistry , Antigens, Surface/genetics , COS Cells , Chlorocebus aethiops , Glutamate Carboxypeptidase II/chemistry , Glutamate Carboxypeptidase II/genetics , HeLa Cells , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Models, Molecular , Mutation , Plasmids/genetics , Protein Binding , Receptors, Interleukin-2/metabolism
14.
Cancer Res ; 63(10): 2645-8, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12750292

ABSTRACT

Prostate-specific membrane antigen (PMSA) is an integral membrane protein highly expressed by prostate cancer cells. We reported previously that PSMA undergoes internalization via clathrin-coated pits (Liu et al., Cancer Res., 58: 4055-4060, 1998). In this study we demonstrate that filamin A, an actin cross-linking protein, associates with the cytoplasmic tail of PSMA and that this association of PSMA with filamin is involved in its localization to the recycling endosomal compartment. By ectopically expressing PSMA in filamin-negative and -positive cell lines, we additionally show that filamin binding to PSMA reduces the internalization rate of PSMA and its N-acelylated-alpha linked-acidic dipeptidase activity. These results suggest that filamin might be an important regulator of PSMA function.


Subject(s)
Antigens, Surface , Carboxypeptidases/metabolism , Contractile Proteins/metabolism , Microfilament Proteins/metabolism , Prostatic Neoplasms/metabolism , Amino Acid Sequence , Filamins , Glutamate Carboxypeptidase II , Humans , Male , Molecular Sequence Data , Prostatic Neoplasms/enzymology , Sequence Homology, Amino Acid , Tumor Cells, Cultured
15.
Mol Cancer Ther ; 4(5): 704-14, 2005 May.
Article in English | MEDLINE | ID: mdl-15897234

ABSTRACT

Prostate-specific membrane antigen (PSMA) is an important biomarker expressed in prostate cancer cells with levels proportional to tumor grade. The membrane association and correlation with disease stage portend a promising role for PSMA as an antigenic target for antibody-based therapies. Successful application of such modalities necessitates a detailed knowledge of the subcellular localization and trafficking of target antigen. In this study, we show that PSMA is expressed predominantly in the apical plasma membrane in epithelial cells of the prostate gland and in well-differentiated Madin-Darby canine kidney cells. We show that PSMA is targeted directly to the apical surface and that sorting into appropriate post-Golgi vesicles is dependent upon N-glycosylation of the protein. Integrity of the microtubule cytoskeleton is also essential for delivery and retention of PSMA at the apical plasma membrane domain, as destabilization of microtubules with nocodazole or commonly used chemotherapeutic Vinca alkaloids resulted in the basolateral expression of PSMA and increased the uptake of anti-PSMA antibody from the basolateral domain. These results may have important relevance to PSMA-based immunotherapy and imaging strategies, as prostate cancer cells can maintain a well-differentiated morphology even after metastasis to distal sites. In contrast to antigens on the basolateral surface, apical antigens are separated from the circulation by tight junctions that restrict transport of molecules across the epithelium. Thus, antigens expressed on the apical plasma membrane are not exposed to intravenously administered agents. The ability to reverse the polarity of PSMA from apical to basolateral could have significant implications for the use of PSMA as a therapeutic target.


Subject(s)
Antigens, Surface/metabolism , Cell Membrane/metabolism , Epithelial Cells/metabolism , Gene Targeting , Glutamate Carboxypeptidase II/metabolism , Kidney/metabolism , Microtubules/metabolism , Prostate/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Polarity/physiology , Dogs , Epithelial Cells/cytology , Glycosylation , Golgi Apparatus , Humans , Immunotherapy , Male , Nocodazole/pharmacology , Protein Transport
16.
Semin Nephrol ; 25(5): 328-34, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16139688

ABSTRACT

The Na,K-adenosine triphosphatase (ATPase), or sodium pump, has been well studied for its role in the regulation of ion homeostasis in mammalian cells. Recent studies suggest that Na,K-ATPase might have multiple functions such as a role in the regulation of tight junction structure and function, induction of polarity, regulation of actin dynamics, control of cell movement, and cell signaling. These functions appear to be modulated by Na,K-ATPase enzyme activity as well as protein-protein interactions of the alpha and beta subunits. In this review we attempt to differentiate functions associated with enzyme activity and subunit interactions. In addition, the consequence of impaired Na,K-ATPase function or reduced subunit expression levels in kidney diseases such as cancer, tubulointerstitial fibrosis, and ischemic nephropathy are discussed.


Subject(s)
Epithelial Cells/enzymology , Sodium-Potassium-Exchanging ATPase/physiology , Animals , Cell Membrane Permeability/physiology , Cell Movement/physiology , Cytoskeleton/physiology , Humans , Kidney/cytology , Kidney Diseases/metabolism , Kidney Diseases/physiopathology , Signal Transduction/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Stress Fibers/enzymology
17.
World J Gastrointest Pathophysiol ; 2(1): 1-14, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21607160

ABSTRACT

Curcumin, also known as diferuloylmethane, is derived from the plant Curcuma longa and is the active ingredient of the spice turmeric. The therapeutic activities of curcumin for a wide variety of diseases such as diabetes, allergies, arthritis and other chronic and inflammatory diseases have been known for a long time. More recently, curcumin's therapeutic potential for preventing and treating various cancers is being recognized. As curcumin's therapeutic promise is being explored more systematically in various diseases, it has become clear that, due to its increased bioavailability in the gastrointestinal tract, curcumin may be particularly suited to be developed to treat gastrointestinal diseases. This review summarizes some of the current literature of curcumin's anti-inflammatory, anti-oxidant and anti-cancer potential in inflammatory bowel diseases, hepatic fibrosis and gastrointestinal cancers.

18.
Biomaterials ; 32(25): 5906-14, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21601921

ABSTRACT

Curcumin, a hydrophobic polyphenol, is an extract of turmeric root with antioxidant, anti-inflammatory and anti-tumorigenic properties. Its lack of water solubility and relatively low bioavailability set major limitations for its therapeutic use. In this study, a self-assembling peptide hydrogel is demonstrated to be an effective vehicle for the localized delivery of curcumin over sustained periods of time. The curcumin-hydrogel is prepared in-situ where curcumin encapsulation within the hydrogel network is accomplished concurrently with peptide self-assembly. Physical and in vitro biological studies were used to demonstrate the effectiveness of curcumin-loaded ß-hairpin hydrogels as injectable agents for localized curcumin delivery. Notably, rheological characterization of the curcumin-loaded hydrogel before and after shear flow have indicated solid-like properties even at high curcumin payloads. In vitro experiments with a medulloblastoma cell line confirm that the encapsulation of the curcumin within the hydrogel does not have an adverse effect on its bioactivity. Most importantly, the rate of curcumin release and its consequent therapeutic efficacy can be conveniently modulated as a function of the concentration of the MAX8 peptide.


Subject(s)
Curcumin/administration & dosage , Drug Carriers , Drug Delivery Systems , Hydrogels , Peptides , Circular Dichroism , Microscopy, Electron, Transmission , Rheology
19.
Mol Cancer Ther ; 9(6): 1515-24, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20501797

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is an important developmental process, participates in tissue repair, and occurs during pathologic processes of tumor invasiveness, metastasis, and tissue fibrosis. The molecular mechanisms leading to EMT are poorly understood. Although it is well documented that transforming growth factor (TGF)-beta plays a central role in the induction of EMT, the targets of TGF-beta signaling are poorly defined. We have shown earlier that Na,K-ATPase beta(1)-subunit levels are highly reduced in poorly differentiated kidney carcinoma cells in culture and in patients' tumor samples. In this study, we provide evidence that Na,K-ATPase is a new target of TGF-beta(1)-mediated EMT in renal epithelial cells, a model system used in studies of both cancer progression and fibrosis. We show that following treatment with TGF-beta(1), the surface expression of the beta(1)-subunit of Na,K-ATPase is reduced, before well-characterized EMT markers, and is associated with the acquisition of a mesenchymal phenotype. RNAi-mediated knockdown confirmed the specific involvement of the Na,K-ATPase beta(1)-subunit in the loss of the epithelial phenotype and exogenous overexpression of the Na,K-ATPase beta(1)-subunit attenuated TGF-beta(1)-mediated EMT. We further show that both Na,K-ATPase alpha- and beta-subunit levels are highly reduced in renal fibrotic tissues. These findings reveal for the first time that Na,K-ATPase is a target of TGF-beta(1)-mediated EMT and is associated with the progression of EMT in cancer and fibrosis.


Subject(s)
Biomarkers, Tumor/metabolism , Epithelium/pathology , Mesoderm/pathology , Neoplasms/enzymology , Neoplasms/pathology , Protein Subunits/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Cell Differentiation/drug effects , Epithelial Cells/drug effects , Epithelial Cells/enzymology , Epithelium/enzymology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Gene Knockdown Techniques , Intracellular Space/drug effects , Intracellular Space/metabolism , Kidney Tubules, Proximal/cytology , LLC-PK1 Cells , MAP Kinase Signaling System/drug effects , Mesoderm/enzymology , Phenotype , Sodium/metabolism , Swine , Transforming Growth Factor beta/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL