Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Oncotarget ; 8(63): 106807-106819, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29290990

ABSTRACT

The Eph receptor tyrosine kinase family member EphA2 plays a pivotal role in modulating cytoskeletal dynamics to control cancer cell motility and invasion. EphA2 is frequently upregulated in diverse solid tumors and has emerged as a viable druggable target. We previously reported that extracellular Hsp90 (eHsp90), a known pro-motility and invasive factor, collaborates with EphA2 to regulate tumor invasion in the absence of its cognate ephrin ligand. Here, we aimed to further define the molecular and functional relationship between EphA2 and eHsp90. Ligand dependent ephrin A1 signaling promotes RhoA activation and altered cell morphology to favor transient cell rounding, retraction, and diminished adhesion. Exposure of EphA2-expressing cancer cells to ligand herein revealed a unique role for eHsp90 as an effector of cytoskeletal remodeling. Notably, blockade of eHsp90 via either neutralizing antibodies or administration of cell-impermeable Hsp90-targeted small molecules significantly attenuated ligand dependent cell rounding in diverse tumor types. Although eHsp90 blockade did not appear to influence receptor internalization, downstream signaling events were augmented. In particular, eHsp90 activated a Src-RhoA axis to enhance ligand dependent cell rounding, retraction, and ECM detachment. Moreover, eHsp90 signaling via this axis stimulated activation of the myosin pathway, culminating in formation of an EphA2-myosin complex. Inhibition of either eHsp90 or Src was sufficient to impair ephrin A1 mediated Rho activation, activation of myosin intermediates, and EphA2-myosin complex formation. Collectively, our data support a paradigm whereby eHsp90 and EphA2 exhibit molecular crosstalk and functional cooperation within a ligand dependent context to orchestrate cytoskeletal events controlling cell morphology and attachment.

2.
Oncotarget ; 8(12): 19323-19341, 2017 Mar 21.
Article in English | MEDLINE | ID: mdl-28038472

ABSTRACT

Heat-shock protein 90 (Hsp90), a highly conserved molecular chaperone, is frequently upregulated in tumors, and remains an attractive anti-cancer target. Hsp90 is also found extracellularly, particularly in tumor models. Although extracellular Hsp90 (eHsp90) action is not well defined, eHsp90 targeting attenuates tumor invasion and metastasis, supporting its unique role in tumor progression. We herein investigated the potential role of eHsp90 as a modulator of cancer stem-like cells (CSCs) in prostate cancer (PCa). We report a novel function for eHsp90 as a facilitator of PCa stemness, determined by its ability to upregulate stem-like markers, promote self-renewal, and enhance prostasphere growth. Moreover, eHsp90 increased the side population typically correlated with the drug-resistant phenotype. Intriguingly, tumor cells with elevated surface eHsp90 exhibited a marked increase in stem-like markers coincident with increased expression of the epithelial to mesenchymal (EMT) effector Snail, indicating that surface eHsp90 may enrich for a unique CSC population. Our analysis of distinct effectors modulating the eHsp90-dependent CSC phenotyperevealed that eHsp90 is a likely facilitator of stem cell heterogeneity. Taken together, our findings provide unique functional insights into eHsp90 as a modulator of PCa plasticity, and provide a framework towards understanding its role as a driver of tumor progression.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Self Renewal , Epithelial-Mesenchymal Transition , HSP90 Heat-Shock Proteins/metabolism , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/pathology , Humans , Male , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms/metabolism , Signal Transduction , Tumor Cells, Cultured
3.
J Proteome Res ; 15(9): 3196-203, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27431976

ABSTRACT

Histone variants are known to play a central role in genome regulation and maintenance. However, many variants are inaccessible by antibody-based methods or bottom-up tandem mass spectrometry due to their highly similar sequences. For many, the only tractable approach is with intact protein top-down tandem mass spectrometry. Here, ultra-high-resolution FT-ICR MS and MS/MS yield quantitative relative abundances of all detected HeLa H2A and H2B isobaric and isomeric variants with a label-free approach. We extend the analysis to identify and relatively quantitate 16 proteoforms from 12 sequence variants of histone H2A and 10 proteoforms of histone H2B from three other cell lines: human embryonic stem cells (WA09), U937, and a prostate cancer cell line LaZ. The top-down MS/MS approach provides a path forward for more extensive elucidation of the biological role of many previously unstudied histone variants and post-translational modifications.


Subject(s)
Histones/analysis , Proteomics/methods , Cell Line , Cyclotrons , Genetic Variation , Genomic Structural Variation , Histones/genetics , Humans , Protein Processing, Post-Translational , Tandem Mass Spectrometry/methods
4.
Adv Cancer Res ; 129: 107-40, 2016.
Article in English | MEDLINE | ID: mdl-26916003

ABSTRACT

The cellular functions of Hsp90 have historically been attributed to its ability to chaperone client proteins involved in signal transduction. Although numerous stimuli and the signaling cascades they activate contribute to cancer progression, many of these pathways ultimately require transcriptional effectors to elicit tumor-promoting effects. Despite this obvious connection, the majority of studies evaluating Hsp90 function in malignancy have focused upon its regulation of cytosolic client proteins, and particularly members of receptor and/or kinase families. However, in recent years, Hsp90 has emerged as a pivotal orchestrator of nuclear events. Discovery of an expanding repertoire of Hsp90 clients has illuminated a vital role for Hsp90 in overseeing nuclear events and influencing gene transcription. Hence, this chapter will cast a spotlight upon several regulatory themes involving Hsp90-dependent nuclear functions. Highlighted topics include a summary of chaperone-dependent regulation of key transcription factors (TFs) and epigenetic effectors in malignancy, as well as a discussion of how the complex interplay among a subset of these TFs and epigenetic regulators may generate feed-forward loops that further support cancer progression. This chapter will also highlight less recognized indirect mechanisms whereby Hsp90-supported signaling may impinge upon epigenetic regulation. Finally, the relevance of these nuclear events is discussed within the framework of Hsp90's capacity to enable phenotypic variation and drug resistance. These newly acquired insights expanding our understanding of Hsp90 function support the collective notion that nuclear clients are major beneficiaries of Hsp90 action, and their impairment is likely responsible for many of the anticancer effects elicited by Hsp90-targeted approaches.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , HSP90 Heat-Shock Proteins/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/genetics , Transcription Factors/metabolism , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Chromatin/metabolism , DNA Methylation , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , Humans , Signal Transduction
5.
J Biol Chem ; 290(13): 8271-82, 2015 Mar 27.
Article in English | MEDLINE | ID: mdl-25670862

ABSTRACT

Prostate cancer remains the second highest contributor to male cancer-related lethality. The transition of a subset of tumors from indolent to invasive disease is associated with a poor clinical outcome. Activation of the epithelial to mesenchymal transition (EMT) genetic program is a major risk factor for cancer progression. We recently reported that secreted extracellular Hsp90 (eHsp90) initiates EMT in prostate cancer cells, coincident with its enhanced expression in mesenchymal models. Our current work substantially extended these findings in defining a pathway linking eHsp90 signaling to EZH2 function, a methyltransferase of the Polycomb repressor complex. EZH2 is also implicated in EMT activation, and its up-regulation represents one of the most frequent epigenetic alterations during prostate cancer progression. We have now highlighted a novel epigenetic function for eHsp90 via its modulation of EZH2 expression and activity. Mechanistically, eHsp90 initiated sustained activation of MEK/ERK, a signal critical for facilitating EZH2 transcriptional up-regulation and recruitment to the E-cadherin promoter. We further demonstrated that an eHsp90-EZH2 pathway orchestrates an expanded repertoire of EMT-related events including Snail and Twist expression, tumor cell motility, and anoikis resistance. To evaluate the role of eHsp90 in vivo, eHsp90 secretion was stably enforced in a prostate cancer cell line resembling indolent disease. Remarkably, eHsp90 was sufficient to induce tumor growth, suppress E-cadherin, and initiate localized invasion, events that are exquisitely dependent upon EZH2 function. In summary, our findings illuminate a hitherto unknown epigenetic function for eHsp90 and support a model wherein tumor eHsp90 functions as a rheostat for EZH2 expression and activity to orchestrate mesenchymal properties and coincident aggressive behavior.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Polycomb-Group Proteins/physiology , Prostatic Neoplasms/pathology , Animals , Antigens, CD , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Epithelial-Mesenchymal Transition , Gene Expression , Gene Expression Regulation, Neoplastic , HEK293 Cells , HSP90 Heat-Shock Proteins/physiology , Humans , MAP Kinase Signaling System , Male , Mice, SCID , Neoplasm Invasiveness , Neoplasm Transplantation , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Tumor Burden
6.
Cancers (Basel) ; 6(2): 1065-97, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24805867

ABSTRACT

Heat shock proteins (Hsps) represent a diverse group of chaperones that play a vital role in the protection of cells against numerous environmental stresses. Although our understanding of chaperone biology has deepened over the last decade, the "atypical" extracellular functions of Hsps have remained somewhat enigmatic and comparatively understudied. The heat shock protein 90 (Hsp90) chaperone is a prototypic model for an Hsp family member exhibiting a duality of intracellular and extracellular functions. Intracellular Hsp90 is best known as a master regulator of protein folding. Cancers are particularly adept at exploiting this function of Hsp90, providing the impetus for the robust clinical development of small molecule Hsp90 inhibitors. However, in addition to its maintenance of protein homeostasis, Hsp90 has also been identified as an extracellular protein. Although early reports ascribed immunoregulatory functions to extracellular Hsp90 (eHsp90), recent studies have illuminated expanded functions for eHsp90 in wound healing and cancer. While the intended physiological role of eHsp90 remains enigmatic, its evolutionarily conserved functions in wound healing are easily co-opted during malignancy, a pathology sharing many properties of wounded tissue. This review will highlight the emerging functions of eHsp90 and shed light on its seemingly dichotomous roles as a benevolent facilitator of wound healing and as a sinister effector of tumor progression.

7.
J Biol Chem ; 287(45): 37732-44, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-22989880

ABSTRACT

Prostate cancer (PCa) is the most frequently diagnosed malignancy in men, and the second highest contributor of male cancer related lethality. Disease mortality is due primarily to metastatic spread, highlighting the urgent need to identify factors involved in this progression. Activation of the genetic epithelial to mesenchymal transition (EMT) program is implicated as a major contributor of PCa progression. Initiation of EMT confers invasive and metastatic behavior in preclinical models and is correlated with poor clinical prognosis. Extracellular Hsp90 (eHsp90) promotes cell motility and invasion in cancer cells and metastasis in preclinical models, however, the mechanistic basis for its widespread tumorigenic function remains unclear. We have identified a novel and pivotal role for eHsp90 in driving EMT events in PCa. In support of this notion, more metastatic PCa lines exhibited increased eHsp90 expression relative to their lineage-related nonmetastatic counterparts. We demonstrate that eHsp90 promoted cell motility in an ERK and matrix metalloproteinase-2/9-dependent manner, and shifted cellular morphology toward a mesenchymal phenotype. Conversely, inhibition of eHsp90 attenuated pro-motility signaling, blocked PCa migration, and shifted cell morphology toward an epithelial phenotype. Last, we report that surface eHsp90 was found in primary PCa tumor specimens, and elevated eHsp90 expression was associated with increased levels of matrix metalloproteinase-2/9 transcripts. We conclude that eHsp90 serves as a driver of EMT events, providing a mechanistic basis for its ability to promote cancer progression and metastasis in preclinical models. Furthermore, its newly identified expression in PCa specimens, and potential regulation of pro-metastatic genes, supports a putative clinical role for eHsp90 in PCa progression.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , HSP90 Heat-Shock Proteins/genetics , Prostatic Neoplasms/genetics , Signal Transduction/genetics , Antibodies, Blocking/immunology , Antibodies, Blocking/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Dipeptides/pharmacology , Disease Progression , Gene Expression Regulation, Neoplastic , HEK293 Cells , HSP90 Heat-Shock Proteins/immunology , HSP90 Heat-Shock Proteins/metabolism , Humans , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protease Inhibitors/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
8.
BMC Cancer ; 11: 520, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22172030

ABSTRACT

BACKGROUND: Perturbing Hsp90 chaperone function targets hypoxia inducible factor (HIF) function in a von Hippel-Lindau (VHL) independent manner, and represents an approach to combat the contribution of HIF to cell renal carcinoma (CCRCC) progression. However, clinical trials with the prototypic Hsp90 inhibitor 17-AAG have been unsuccessful in halting the progression of advanced CCRCC. METHODS: Here we evaluated a novel next generation small molecule Hsp90 inhibitor, EC154, against HIF isoforms and HIF-driven molecular and functional endpoints. The effects of EC154 were compared to those of the prototypic Hsp90 inhibitor 17-AAG and the histone deacetylase (HDAC) inhibitor LBH589. RESULTS: The findings indicate that EC154 is a potent inhibitor of HIF, effective at doses 10-fold lower than 17-AAG. While EC154, 17-AAG and the histone deacetylase (HDAC) inhibitor LBH589 impaired HIF transcriptional activity, CCRCC cell motility, and angiogenesis; these effects did not correlate with their ability to diminish HIF protein expression. Further, our results illustrate the complexity of HIF targeting, in that although these agents suppressed HIF transcripts with differential dynamics, these effects were not predictive of drug efficacy in other relevant assays. CONCLUSIONS: We provide evidence for EC154 targeting of HIF in CCRCC and for LBH589 acting as a suppressor of both HIF-1 and HIF-2 activity. We also demonstrate that 17-AAG and EC154, but not LBH589, can restore endothelial barrier function, highlighting a potentially new clinical application for Hsp90 inhibitors. Finally, given the discordance between HIF activity and protein expression, we conclude that HIF expression is not a reliable surrogate for HIF activity. Taken together, our findings emphasize the need to incorporate an integrated approach in evaluating Hsp90 inhibitors within the context of HIF suppression.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/drug therapy , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Blotting, Western , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Disease Progression , Electric Impedance , HSP90 Heat-Shock Proteins/metabolism , Humans , Hydroxamic Acids/pharmacology , Hypoxia-Inducible Factor 1/metabolism , Indoles , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Luciferases/metabolism , Neoplasm Proteins/metabolism , Panobinostat , Real-Time Polymerase Chain Reaction , Transcription, Genetic/drug effects , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/metabolism
9.
Am J Cancer Res ; 1(6): 763-72, 2011.
Article in English | MEDLINE | ID: mdl-22016826

ABSTRACT

Oncogenic viruses are the etiologic agents for a significant proportion of human cancers, but effective therapies and preventative strategies are lacking for the majority of virus-associated cancers. Targeting of virus-induced signal transduction or virus-host protein interactions may offer novel therapeutic strategies for viral cancers. Heat shock protein 90 (Hsp90) is a well-characterized, multifunctional molecular chaperone involved in regulation of signal transduction, transcriptional activation, oncogenic protein stabilization, and neovascularization-pathogenic elements relevant to viral cancer pathogenesis. This review will summarize mechanistic concepts involving regulation of viral oncogenesis by both intracellular and extracellular Hsp90, as well as current therapeutic implications of these data.

10.
Am J Cancer Res ; 1(5): 687-700, 2011.
Article in English | MEDLINE | ID: mdl-21994906

ABSTRACT

The Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiologic agent of Kaposi's sarcoma (KS)-the most common tumor associated with HIV infection and an important cause of morbidity and mortality in this patient population. The majority of patients with KS exhibit little or no clinical response to existing therapies. The nuclear factor-kappaB (NF-κB) family of transcription factors plays a critical role in facilitating cancer pathogenesis associated with oncogenic viruses, and a better understanding of how cellular factors regulate NF-κB activation in the context of KSHV infection may facilitate development of new therapies for KS. Existing data implicate heat shock protein-90 associated with the cell surface (csHsp90) as a co-factor in cancer cell migration and invasion, and we recently reported that csHsp90 serves as a co-factor for mitogen-activated protein kinase (MAPK) activation during de novo KSHV infection. However, whether csHsp90 regulates NF-κB activation, or cellular pathogenesis associated with KS, has not been established. We have found that csHsp90 serves as an important co-factor for canonical NF-κB activation by KSHV during de novo infection of primary human cells relevant to KS. Furthermore, our correlative functional studies reveal that csHsp90 inhibition suppresses KSHV-induced, NF-κB-dependent secretion of the pro-migratory factors interleukin-8 and vascular endothelial growth factor as well as invasiveness for primary cells following de novo infection. These data implicate csHsp90 in KSHV-mediated activation of NF-κB and associated pathogenesis, and support the potential utility of targeting csHsp90 as a therapeutic approach for KS.

11.
PLoS One ; 6(3): e17649, 2011 Mar 08.
Article in English | MEDLINE | ID: mdl-21408136

ABSTRACT

BACKGROUND: Extracellular Hsp90 protein (eHsp90) potentiates cancer cell motility and invasion through a poorly understood mechanism involving ligand mediated function with its cognate receptor LRP1. Glioblastoma multiforme (GBM) represents one of the most aggressive and lethal brain cancers. The receptor tyrosine kinase EphA2 is overexpressed in the majority of GBM specimens and is a critical mediator of GBM invasiveness through its AKT dependent activation of EphA2 at S897 (P-EphA2(S897)). We explored whether eHsp90 may confer invasive properties to GBM via regulation of EphA2 mediated signaling. PRINCIPAL FINDINGS: We find that eHsp90 signaling is essential for sustaining AKT activation, P-EphA2(S897), lamellipodia formation, and concomitant GBM cell motility and invasion. Furthermore, eHsp90 promotes the recruitment of LRP1 to EphA2 in an AKT dependent manner. A finding supported by biochemical methodology and the dual expression of LRP1 and P-EphA2(S897) in primary and recurrent GBM tumor specimens. Moreover, hypoxia mediated facilitation of GBM motility and invasion is dependent upon eHsp90-LRP1 signaling. Hypoxia dramatically elevated surface expression of both eHsp90 and LRP1, concomitant with eHsp90 dependent activation of src, AKT, and EphA2. SIGNIFICANCE: We herein demonstrate a novel crosstalk mechanism involving eHsp90-LRP1 dependent regulation of EphA2 function. We highlight a dual role for eHsp90 in transducing signaling via LRP1, and in facilitating LRP1 co-receptor function for EphA2. Taken together, our results demonstrate activation of the eHsp90-LRP1 signaling axis as an obligate step in the initiation and maintenance of AKT signaling and EphA2 activation, thereby implicating this pathway as an integral component contributing to the aggressive nature of GBM.


Subject(s)
Antigens, CD/metabolism , Glioblastoma/enzymology , Glioblastoma/pathology , HSP90 Heat-Shock Proteins/metabolism , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Receptor, EphA2/metabolism , Cell Hypoxia/drug effects , Cell Movement/drug effects , Enzyme Activation/drug effects , Glyoxylates/pharmacology , Humans , Neoplasm Invasiveness , Phosphorylation/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Pseudopodia/drug effects , Pseudopodia/metabolism , Signal Transduction/drug effects
12.
Virology ; 403(1): 92-102, 2010 Jul 20.
Article in English | MEDLINE | ID: mdl-20451233

ABSTRACT

The Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma (KS), an important cause of morbidity and mortality in immunocompromised patients. KSHV interaction with the cell membrane triggers activation of specific intracellular signal transduction pathways to facilitate virus entry, nuclear trafficking, and ultimately viral oncogene expression. Extracellular heat shock protein 90 localizes to the cell surface (csHsp90) and facilitates signal transduction in cancer cell lines, but whether csHsp90 assists in the coordination of KSHV gene expression through these or other mechanisms is unknown. Using a recently characterized non-permeable inhibitor specifically targeting csHsp90 and Hsp90-specific antibodies, we show that csHsp90 inhibition suppresses KSHV gene expression during de novo infection, and that this effect is mediated largely through the inhibition of mitogen-activated protein kinase (MAPK) activation by KSHV. Moreover, we show that targeting csHsp90 reduces constitutive MAPK expression and the release of infectious viral particles by patient-derived, KSHV-infected primary effusion lymphoma cells. These data suggest that csHsp90 serves as an important co-factor for KSHV-initiated MAPK activation and provide proof-of-concept for the potential benefit of targeting csHsp90 for the treatment or prevention of KSHV-associated illnesses.


Subject(s)
Gene Expression Regulation, Viral , HSP90 Heat-Shock Proteins/metabolism , Herpesvirus 8, Human/pathogenicity , Host-Pathogen Interactions , Mitogen-Activated Protein Kinases/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Lymphocytes/virology , Lymphoma/virology , Sarcoma, Kaposi/virology
13.
Mol Cancer Res ; 7(7): 1021-32, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19567782

ABSTRACT

A subset of Eph receptors and their corresponding ligands are commonly expressed in tumor cells where they mediate biological processes such as cell migration and adhesion, whereas their expression in endothelial cells promotes angiogenesis. In particular, the tumor-specific up-regulation of EphA2 confers properties of increased cellular motility, invasiveness, tumor angiogenesis, and tumor progression, and its overexpression correlates with poor prognosis in several cancer types. The cellular chaperone Hsp90 also plays a significant role in regulating cell migration and angiogenesis, although the full repertoire of motility driving proteins dependent on Hsp90 function remain poorly defined. We explored the hypothesis that Hsp90 may regulate the activity of EphA2 and examined the potential relationship between EphA2 receptor signaling and chaperone function. We show that geldanamycin, an Hsp90 antagonist, dramatically destabilizes newly synthesized EphA2 protein and diminishes receptor levels in a proteasome-dependent pathway. In addition, geldanamycin treatment impairs EphA2 signaling, as evidenced by a decrease in ligand-dependent receptor phosphorylation and subsequent cell rounding. Therefore, Hsp90 exerts a dual role in regulating the stability of nascent EphA2 protein and maintaining the signaling capacity of the mature receptor. Our findings also suggest that the geldanamycin-dependent mitigation of EphA2 signaling in receptor-overexpressing cancer cells may be sufficient to recapitulate the antimotility effects of this drug. Finally, the identification of a pharmacologic approach to suppress EphA2 expression and signaling highlights the attractive possibility that Hsp90 inhibitors may have clinical utility in antagonizing EphA2-dependent tumorigenic progression.


Subject(s)
Cell Movement/physiology , HSP90 Heat-Shock Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Receptor, EphA2/metabolism , Cell Line , Cell Line, Tumor , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Microscopy, Fluorescence , Neoplasm Metastasis , Phosphorylation , Protein Multimerization , Protein Stability , Protein Structure, Tertiary , Signal Transduction , Ubiquitination
14.
J Biol Chem ; 281(19): 13047-13056, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16543242

ABSTRACT

A recent study of breast cancer patients with and without BRCA1 gene mutations found significantly lower levels of VEGF in serum from patients with BRCA1 mutations (Tarnowski, B., Chudecka-Glaz, A., Gorski, B., and Rzepka-Gorska, I. (2004) Breast Cancer Res. Treat. 88, 287-288). Here, we describe a possible mechanistic explanation for this correlation. Because hypoxia in tumors stimulates VEGF expression and secretion we hypothesized that altered BRCA1 protein levels in breast tumors could affect hypoxia-stimulated VEGF promoter activity. This possibility was tested in cells transfected with various combinations of expression plasmids for BRCA1, BRCA1 specific inhibitory RNAs (BRCA1-siRNAs), HIF-1alpha, and a VEGF promoter-reporter and then incubated in normoxia (21%, O2) or hypoxia (0.1%, O2). As predicted, increased BRCA1 levels enhanced both hypoxia-stimulated VEGF promoter activity and the amounts of VEGF mRNA, as determined by semiquantitative RT-PCR and quantitative real time PCR. Using the ChIP assay, we discovered that BRCA1 could be recruited to the endogenous human VEGF promoter along with HIF-1alpha following hypoxia. An interaction between BRCA1 and HIF-1alpha was found in human breast cancer cells. We also found that hypoxia-stimulated VEGF promoter activity and secretion was reduced in cells containing reduced amounts of endogenous BRCA1 protein (obtained by transfecting with BRCA1 siRNAs). A mechanistic explanation for these effects is provided by our finding a reduced half-life and reduced accumulation of HIF-1alpha in hypoxic cells transfected with BRCA1-siRNAs and that proteasome inhibitors blocked these effects of BRCA1-siRNAs. Thus, our results suggest that normal amounts of BRCA1 function in hypoxia to regulate HIF-1alpha stability, probably by interacting with HIF-1alpha.


Subject(s)
BRCA1 Protein/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Oxygen/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Promoter Regions, Genetic , Transcription, Genetic , Vascular Endothelial Growth Factor A/genetics
15.
Cancer Cell ; 8(2): 143-53, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16098467

ABSTRACT

Individuals with hemizygous germline fumarate hydratase (FH) mutations are predisposed to renal cancer. These tumors predominantly exhibit functional inactivation of the remaining wild-type allele, implicating FH inactivation as a tumor-promoting event. Hypoxia-inducible factors are expressed in many cancers and are increased in clear cell renal carcinomas. Under normoxia, the HIFs are labile due to VHL-dependent proteasomal degradation, but stabilization occurs under hypoxia due to inactivation of HIF prolyl hydroxylase (HPH), which prevents HIF hydroxylation and VHL recognition. We demonstrate that FH inhibition, together with elevated intracellular fumarate, coincides with HIF upregulation. Further, we show that fumarate acts as a competitive inhibitor of HPH. These data delineate a novel fumarate-dependent pathway for regulating HPH activity and HIF protein levels.


Subject(s)
Carcinoma, Renal Cell/metabolism , DNA-Binding Proteins/metabolism , Fumarate Hydratase/genetics , Fumarates/metabolism , Kidney Neoplasms/metabolism , Leiomyomatosis/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Adult , Alleles , Basic Helix-Loop-Helix Transcription Factors , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/genetics , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Female , Fumarate Hydratase/antagonists & inhibitors , Fumarate Hydratase/metabolism , Fumarates/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Ketoglutaric Acids/pharmacology , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Male , Middle Aged , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Syndrome , Transcription Factors/analysis , Transcription Factors/genetics , Up-Regulation
16.
Expert Opin Investig Drugs ; 14(6): 569-89, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16004589

ABSTRACT

Heat-shock protein (Hsp)-90 belongs to the class of molecular chaperone proteins that are capable of sensing cellular stress. Although Hsp90 is essential for viability, the pharmacological inhibition of this chaperone has emerged as an attractive means to inhibit tumorigenesis. This phenomenon is due to a unique property of Hsp90; its 'client proteins' are universally involved in signal transduction pathways commonly dysregulated in, and contributing to, cancer. The natural product geldanamycin, a potent ansamycin Hsp90 inhibitor, has served as a lead compound for the development of several derivatives that are currently undergoing clinical trials. Inhibition of Hsp90 with geldanamycin simultaneously depletes Hsp90-associated clients and impairs numerous signalling cascades that depend on chaperone function. Importantly, tumour cells are exquisitely sensitive to Hsp90 inhibition, lending credence to the feasibility of selectively targeting cancer tissue via the pharmacological modulation of Hsp90 function. Even more remarkably, Hsp90 inhibitors sensitise tumour cells to the cytotoxic effects of a variety of standard therapeutics, and thus, they are likely to have broad utility in combination therapy. Although these are promising developments, much remains to be discovered about client-chaperone biology and the tumour-specific effects of Hsp90 blockade. This information is required to fully grasp the multi-faceted roles of Hsp90 in cancer biology towards the goal of optimising the use of these agents in the clinic. Elucidation of these nuances will undoubtedly lead to better targeting of relevant oncogenic pathways and translate into the development of more effective anticancer regimens.


Subject(s)
Antineoplastic Agents/therapeutic use , Drugs, Investigational/therapeutic use , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Animals , Clinical Trials as Topic/trends , HSP90 Heat-Shock Proteins/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/metabolism
17.
Clin Cancer Res ; 10(18 Pt 2): 6282S-9S, 2004 Sep 15.
Article in English | MEDLINE | ID: mdl-15448018

ABSTRACT

Studies during the past two decades have shown that kidney cancer is not a single disease; it is made up of a number of different types of cancer that occur in this organ. Clear cell renal carcinoma is characterized by mutation of the VHL gene. The VHL gene product forms a heterotrimeric complex with elongin C, elongin B, and Cul-2 to target hypoxia-inducible factors 1 and 2alpha for ubiquitin-mediated degradation. VHL-/- clear cell renal carcinoma overexpresses epidermal growth factor receptor and transforming growth factor alpha. Both hypoxia-inducible factor 1alpha and the epidermal growth factor receptor are potential therapeutic targets in clear cell renal carcinoma. Studies of the hereditary form of renal cell carcinoma (RCC) associated with hereditary papillary renal carcinoma (HPRC) determined that the c-Met proto-oncogene on chromosome 7 is the gene for HPRC and for a number of sporadic papillary RCCs. The HPRC c-Met mutations are activating mutations in the tyrosine kinase domain of the gene. The gene for a new form of hereditary RCC (Birt Hogg Dubé syndrome) associated with cutaneous tumors, lung cysts, and colon polyps or cancer has recently been identified. Studies are currently under way to determine what type of gene BHD is and how damage to this gene leads to kidney cancer. Individuals affected with hereditary leiomyomatosis renal cell carcinoma are at risk for the development of cutaneous leiomyomas, uterine leiomyomas (fibroids), and type 2 papillary RCC. The HLRC gene has been found to be the Krebs cycle enzyme, fumarate hydratase. Studies are under way to understand the downstream pathway of this cancer gene.


Subject(s)
Genetic Therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/therapy , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/pathology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Female , Humans , Kidney Neoplasms/pathology , Male , Proteins/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins , Von Hippel-Lindau Tumor Suppressor Protein , von Hippel-Lindau Disease/genetics
18.
Mol Cancer Ther ; 3(5): 551-66, 2004 May.
Article in English | MEDLINE | ID: mdl-15141013

ABSTRACT

The ansamycin antibiotic, geldanamycin, targets the hsp 90 protein chaperone and promotes ubiquitin-dependent proteasomal degradation of its numerous client proteins. Bortezomib is a specific and potent proteasome inhibitor. Both bortezomib and the geldanamycin analogue, 17-N-allylamino-17-demethoxy geldanamycin, are in separate clinical trials as new anticancer drugs. We hypothesized that destabilization of hsp 90 client proteins with geldanamycin, while blocking their degradation with bortezomib, would promote the accumulation of aggregated, ubiquitinated, and potentially cytotoxic proteins. Indeed, geldanamycin plus bortezomib inhibited MCF-7 tumor cell proliferation significantly more than either drug alone. Importantly, while control cells were unaffected, human papillomavirus E6 and E7 transformed fibroblasts were selectively sensitive to geldanamycin plus bortezomib. Geldanamycin alone slightly increased protein ubiquitination, but when geldanamycin was combined with bortezomib, protein ubiquitination was massively increased, beyond the amount stabilized by bortezomib alone. In geldanamycin plus bortezomib-treated cells, ubiquitinated proteins were mostly detergent insoluble, indicating that they were aggregated. Individually, both geldanamycin and bortezomib induced hsp 90, hsp 70, and GRP78 stress proteins, but the drug combination superinduced these chaperones and caused them to become detergent insoluble. Geldanamycin plus bortezomib also induced the formation of abundant, perinuclear vacuoles, which were neither lysosomes nor autophagosomes and did not contain engulfed cytosolic ubiquitin or hsp 70. Fluorescence marker experiments indicated that these vacuoles were endoplasmic reticulum derived and that their formation was prevented by cycloheximide, suggesting a role for protein synthesis in their genesis. These observations support a mechanism whereby the geldanamycin plus bortezomib combination simultaneously disrupts hsp 90 and proteasome function, promotes the accumulation of aggregated, ubiquitinated proteins, and results in enhanced antitumor activity.


Subject(s)
Antineoplastic Agents/pharmacology , Endoplasmic Reticulum/drug effects , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Proteasome Inhibitors , Proteins/metabolism , Rifabutin/analogs & derivatives , Ubiquitins/metabolism , Vacuoles/drug effects , Benzoquinones , Boronic Acids/pharmacology , Bortezomib , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Transformation, Viral , Cycloheximide/pharmacology , Detergents/pharmacology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , HSP70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Humans , Lactams, Macrocyclic , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins , Proteasome Endopeptidase Complex/metabolism , Pyrazines/pharmacology , Quinones/pharmacology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Rifabutin/pharmacology , Solubility , Vacuoles/metabolism
19.
J Biol Chem ; 279(16): 16128-35, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-14764593

ABSTRACT

Hypoxia-inducible factor-1 (HIF-1) is a potent cellular survival factor contributing to tumorigenesis in a broad range of cancers. The functional transcription factor exists as a heterodimeric complex consisting of HIF-1alpha and the aryl hydrocarbon receptor nuclear translocator (ARNT). Association of HIF-1 with ARNT is required for its activity; however, no other role has been ascribed to this interaction. We demonstrated previously that pharmacologic inhibition of Hsp90 by geldanamycin (GA) impairs HIF transcription and promotes VHL (Von Hippel-Lindau)-independent degradation of the protein, thus implicating Hsp90 as an essential interacting partner for HIF. In this study, we further explore the physiological role for Hsp90 in HIF function. We establish that the PAS (Per-ARNT-Sim) domain of HIF is required both to promote association with Hsp90 and confer sensitivity to GA. Coincidentally, this domain also associates with ARNT. Overexpression of ARNT in a VHL-deficient background resulted in substantially increased HIF-1 protein concomitant with increased protein stability. Conversely, down-regulation of endogenous ARNT protein by RNA interference decreased the steady-state HIF protein. ARNT-mediated stabilization of HIF is specific for the Hsp90-dependent pathway, as ARNT was unable to protect HIF from VHL-mediated degradation. We establish that the ability of ARNT to up-regulate HIF and diminish HIF sensitivity to GA is due to its ability to compete for the Hsp90 binding site on HIF. These data elucidate novel functions for ARNT and Hsp90 in regulating HIF function and further illustrate that cofactor association may significantly impact upon the sensitivity of Hsp90 clients to chaperone inhibitors.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Transcription Factors/metabolism , Animals , Aryl Hydrocarbon Receptor Nuclear Translocator , Benzoquinones , Cell Line , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1 , Hypoxia-Inducible Factor 1, alpha Subunit , Lactams, Macrocyclic , Oxygen/metabolism , Protein Binding , Protein Structure, Tertiary , Quinones/pharmacology , Signal Transduction/drug effects , Transcriptional Activation , Up-Regulation
20.
Cancer Res ; 63(24): 8984-95, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14695217

ABSTRACT

Ansamycin antibiotics inhibit function of the heat shock protein (HSP) 90, causing selective degradation of several intracellular proteins regulating such processes as proliferation, cell cycle regulation, and prosurvival signaling cascades. HSP90 has been identified previously as a molecular target for anticancer agents, including ionizing radiation (IR). Therefore, we hypothesized that the ansamycin geldanamycin and its 17-allylamino-17-demethoxy analog (17-AAG), which inhibit HSP90, would enhance tumor cell susceptibility to the cytotoxicity of IR. Treatment of two human cervical carcinoma cell lines (HeLa and SiHa) with geldanamycin and 17-AAG resulted in cytotoxicity and, when combined with IR, enhanced the radiation response, each effect with a temporal range from 6 to 48 h after drug exposure. In addition, mouse in vivo models using 17-AAG at clinically achievable concentrations yielded results that paralleled the in vitro radiosensitization studies of both single and fractioned courses of irradiation. The increase in IR-induced cell death appears to be attributable to a combination of both programmed and nonprogrammed cell death. We also measured total levels of several prosurvival and apoptotic signaling proteins. Akt1, extracellular signal-regulated kinase-1, Glut-1, HER-2/neu, Lyn, cAMP-dependent protein kinase, Raf-1, and vascular endothelial growth factor expression were down-regulated in 17-AAG-treated cells, identifying these factors as molecular markers and potential therapeutic targets. Finally, a series of immortalized and human papillomavirus-transformed cell lines were used to demonstrate that the radiosensitizing effects of 17-AAG were limited to transformed cells, suggesting a possible differential cytotoxic effect. This work shows that altered HSP90 function induces significant tumor cytotoxicity and radiosensitization, suggesting a potential therapeutic utility.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , HSP90 Heat-Shock Proteins/physiology , Quinones/pharmacology , Radiation-Sensitizing Agents/pharmacology , Rifabutin/analogs & derivatives , Rifabutin/pharmacology , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/radiotherapy , Animals , Benzoquinones , Combined Modality Therapy , Dose-Response Relationship, Drug , Drug Synergism , Female , HeLa Cells , Humans , Lactams, Macrocyclic , Mice , Mice, Inbred C3H , Mice, Nude , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...