Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Front Oncol ; 12: 841054, 2022.
Article in English | MEDLINE | ID: mdl-35223522

ABSTRACT

Kidney cancer is one of the top ten cancer diagnosed worldwide and its incidence has increased the last 20 years. Clear Cell Renal Cell Carcinoma (ccRCC) are characterized by mutations that inactivate the von Hippel-Lindau (VHL) tumor suppressor gene and evidence indicated alterations in metabolic pathways, particularly in glutamine metabolism. We previously identified a small molecule, STF-62247, which target VHL-deficient renal tumors by affecting late-stages of autophagy and lysosomal signaling. In this study, we investigated ccRCC metabolism in VHL-deficient and proficient cells exposed to the small molecule. Metabolomics profiling using 1H NMR demonstrated that STF-62247 increases levels of glucose, pyruvate, glycerol 3-phosphate while glutamate, asparagine, and glutathione significantly decreased. Diminution of glutamate and glutamine was further investigated using mass spectrometry, western blot analyses, enzymatic activities, and viability assays. We found that expression of SLC1A5 increases in VHL-deficient cells treated with STF-62247, possibly to stimulate glutamine uptake intracellularly to counteract the diminution of this amino acid. However, exogenous addition of glutamine was not able to rescue cell viability induced by the small molecule. Instead, our results showed that VHL-deficient cells utilize glutamine to produce fatty acid in response to STF-62247. Surprisingly, this occurs through oxidative phosphorylation in STF-treated cells while control cells use reductive carboxylation to sustain lipogenesis. We also demonstrated that STF-62247 stimulated expression of stearoyl-CoA desaturase (SCD1) and peripilin2 (PLIN2) to generate accumulation of lipid droplets in VHL-deficient cells. Moreover, the carnitine palmitoyltransferase 1A (CPT1A), which control the entry of fatty acid into mitochondria for ß-oxidation, also increased in response to STF-62247. CPT1A overexpression in ccRCC is known to limit tumor growth. Together, our results demonstrated that STF-62247 modulates cellular metabolism of glutamine, an amino acid involved in the autophagy-lysosome process, to support lipogenesis, which could be implicated in the signaling driving to cell death.

2.
Front Cell Dev Biol ; 9: 798262, 2021.
Article in English | MEDLINE | ID: mdl-34977038

ABSTRACT

For a long time, lysosomes were considered as mere waste bags for cellular constituents. Thankfully, studies carried out in the past 15 years were brimming with elegant and crucial breakthroughs in lysosome research, uncovering their complex roles as nutrient sensors and characterizing them as crucial multifaceted signaling organelles. This review presents the scientific knowledge on lysosome physiology and functions, starting with their discovery and reviewing up to date ground-breaking discoveries highlighting their heterogeneous functions as well as pending questions that remain to be answered. We also review the roles of lysosomes in anti-cancer drug resistance and how they undergo a series of molecular and functional changes during malignant transformation which lead to tumor aggression, angiogenesis, and metastases. Finally, we discuss the strategy of targeting lysosomes in cancer which could lead to the development of new and effective targeted therapies.

3.
Carcinogenesis ; 41(6): 828-840, 2020 07 10.
Article in English | MEDLINE | ID: mdl-31556451

ABSTRACT

The inactivation of the tumor suppressor gene, von Hippel-Lindau (VHL), has been identified as the earliest event in renal cell carcinoma (RCC) development. The loss of heterogeneity by chromosome 3p deletion followed by inactivating mutations on the second VHL copy are events present in close to 90% of patients. Our study illustrates a lysosomal vulnerability in VHL-inactivated RCC in vitro. By investigating the mechanism of action of the previously identified STF-62247, a small bioactive compound known for its selective cytotoxic properties towards VHL-defective models, we present the promising approach of targeting truncal-driven VHL inactivation through lysosome disruption. Furthermore, by analyzing the open platform for exploring cancer genomic data (cbioportal), we uncover the high alteration frequency of essential lysosomal and autophagic genes in sequenced biopsies from clear cell RCC patient primary tumors. By investigating lysosome physiology, we also identify VHL-inactivated cells' inability to maintain their lysosomes at the perinuclear localization in response to STF-62247-induced stress and accumulate cytoplasmic inclusion bodies in response to an inefficient lysosomal degradative capacity. Finally, by testing other known lysosomal-disrupting agents (LDAs), we show that these are selectively cytotoxic to cells lacking VHL functions. Our study builds a strong platform that could specifically link genetic clonal ccRCC evolution to lysosomal and trafficking vulnerabilities.


Subject(s)
Autophagy , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Lysosomes/pathology , Mutation , Pyridines/pharmacology , Thiazoles/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Cell Proliferation , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Tumor Cells, Cultured , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
4.
Eur J Med Chem ; 179: 347-357, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31260889

ABSTRACT

The inhibition of 5-lipoxygenase (5-LO), the key enzyme for the biosynthesis of leukotrienes (LTs), has generated increasing enthusiasm as anti-inflammatory and antitumor strategies in recent years. Based on our previous studies, we synthesized a series of dihydroxycinnamic acid-based analogs that might be 5-LO inhibitors. LTs biosynthesis inhibition in HEK293 cells and polymorphonuclear leukocytes (PMNL) was measured and antitumor activities were investigated in Renal Cell Carcinoma (RCC). Results showed that the 2,5-dihydroxycinnamic acid phenethyl ester (10b) was the best 5-LO inhibitor and was 7-fold more potent than Zileuton (1), the only clinically approved 5-LO inhibitor. 2,5-Dihydroxy substitution was more favorable to 5-LO inhibition since compound 10b is twice as active as CAPE (2) which is a 3,4-dihydroxylcinnamic acid ester. Meanwhile, 10b reduced the cell viability of renal cancer cells  and was more selective toward RCC4 and 786.0 cells which are deficient for the Von Hippel-Lindau (VHL) tumor suppressor gene. As to the underlying cell-death mechanisms, 10b induced apoptosis in VHL-deficient RCC4 cells. Also, increases in LC3B and p62 expression suggest a blockage of the autophagic flux in RCC in response to 10b.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arachidonate 5-Lipoxygenase/metabolism , Carcinoma, Renal Cell/drug therapy , Drug Discovery , Kidney Neoplasms/drug therapy , Lipoxygenase Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Arachidonate 5-Lipoxygenase/biosynthesis , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HEK293 Cells , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lipoxygenase Inhibitors/chemical synthesis , Lipoxygenase Inhibitors/chemistry , Molecular Structure , Neutrophils/drug effects , Neutrophils/metabolism , Structure-Activity Relationship
5.
Am J Physiol Cell Physiol ; 316(5): C605-C620, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30758995

ABSTRACT

Autophagy is a highly conserved, homeostatic process by which cytosolic components reach lysosomes for degradation. The roles played by different autophagic processes in cancer are complex and remain cancer type and stage dependent. Renal cell carcinoma (RCC) is the most common subtype of kidney cancer and is characterized by the inactivation of the von Hippel-Lindau (VHL) tumor suppressor. Our previous study identified a small compound, STF-62247, as an autophagy-modulating molecule causing selective cytotoxicity for VHL-inactivated cells. This present study investigates the effects of STF-62247 specifically on the macroautophagic flux to better characterize its mechanism of action in RCC. Our results clearly demonstrate that this compound is a potent blocker of late stages of autophagy. We show that inhibiting autophagy by CRISPR knockouts of autophagy-related genes rendered VHL-deficient cells insensitive to STF-62247, uncovering the importance of the autophagic pathway in STF-selective cell death. By exploiting the autofluorescence of STF-62247, we pinpointed its cellular localization to lysosomes. Finally, in response to prolonged STF treatments, we show that VHL-proficient cells are able to surmount the block in late stages of autophagy by restoring their lysosome numbers. Conversely, an increase in autophagic vesicles accompanied by a time-dependent decrease in lysosomes was observed in VHL-deficient cells. This is the first mechanistic study investigating STF-62447's effects on the autophagic flux in RCC. Importantly, our study reclassifies STF-62247 as a blocker of later stages of autophagy and highlights the possibility of blocking this process through lysosome disruption in VHL-mutated RCCs.


Subject(s)
Autophagy/physiology , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Lysosomes/metabolism , Pyridines/metabolism , Thiazoles/metabolism , von Hippel-Lindau Disease/metabolism , Autophagy/drug effects , Carcinoma, Renal Cell/drug therapy , Dose-Response Relationship, Drug , Drug Delivery Systems/methods , HeLa Cells , Humans , Kidney Neoplasms/drug therapy , Lysosomes/drug effects , Pyridines/administration & dosage , Thiazoles/administration & dosage , von Hippel-Lindau Disease/drug therapy
6.
Int J Cancer ; 141(4): 778-790, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28486780

ABSTRACT

Inactivation of the tumor suppressor gene, von Hippel-Lindau (VHL), is known to play an important role in the development of sporadic clear cell renal cell carcinomas (ccRCCs). Even if available targeted therapies for metastatic RCCs (mRCCs) have helped to improve progression-free survival rates, they have no durable clinical response. We have previously shown the feasibility of specifically targeting the loss of VHL with the identification of a small molecule, STF-62247. Understanding its functionality is crucial for developing durable personalized therapeutic agents differing from those available targeting hypoxia inducible factor (HIF-) pathways. By using SILAC proteomics, we identified 755 deregulated proteins in response to STF-62247 that were further analyzed by ingenuity pathway analysis (IPA). Bioinformatics analyses predicted alterations in 37 signaling pathways in VHL-null cells in response to treatment. Validation of some altered pathways shows that STF-62247's selectivity is linked to an important inhibition of mTORC1 activation in VHL-null cells leading to protein synthesis arrest, a mechanism differing from two allosteric inhibitors Rapamycin and Everolimus. Altogether, our study identified signaling cascades driving STF-62247 response and brings further knowledge for this molecule that shows selectivity for the loss of VHL. The use of a global SILAC approach was successful in identifying novel affected signaling pathways that could be exploited for the development of new personalized therapeutic strategies to target VHL-inactivated RCCs.


Subject(s)
Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Proteome/drug effects , Pyridines/metabolism , Thiazoles/metabolism , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , Humans , Isotope Labeling , Kidney Neoplasms/genetics , Proteomics/methods , Signal Transduction/drug effects , Von Hippel-Lindau Tumor Suppressor Protein/genetics
7.
Int J Cancer ; 138(10): 2439-49, 2016 May 15.
Article in English | MEDLINE | ID: mdl-26620126

ABSTRACT

Von Hippel-Lindau (VHL) is an onco-suppressor involved in oxygen and energy-dependent promotion of protein ubiquitination and proteosomal degradation. Loss of function mutations of VHL (VHL-cells) result in organ specific cancers with the best studied example in renal cell carcinomas. VHL has a well-established role in deactivation of hypoxia-inducible factor (HIF-1) and in regulation of PI3K/AKT/mTOR activity. Cell culture metabolomics analysis was utilized to determined effect of VHL and HIF-1α or HIF-2α on metabolism of renal cell carcinomas (RCC). RCC cells were stably transfected with VHL or shRNA designed to silence HIF-1α or HIF-2α genes. Obtained metabolic data was analysed qualitatively, searching for overall effects on metabolism as well as quantitatively, using methods developed in our group in order to determine specific metabolic changes. Analysis of the effect of VHL and HIF silencing on cellular metabolic footprints and fingerprints provided information about the metabolic pathways affected by VHL through HIF function as well as independently of HIF. Through correlation network analysis as well as statistical analysis of significant metabolic changes we have determined effects of VHL and HIF on energy production, amino acid metabolism, choline metabolism as well as cell regulation and signaling. VHL was shown to influence cellular metabolism through its effect on HIF proteins as well as by affecting activity of other factors.


Subject(s)
Carcinoma, Renal Cell/metabolism , Gene Silencing , Kidney Neoplasms/metabolism , Metabolome , Metabolomics , Proton Magnetic Resonance Spectroscopy , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Cluster Analysis , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Metabolomics/methods , Mutation , Proton Magnetic Resonance Spectroscopy/methods , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
8.
Adv Exp Med Biol ; 772: 167-88, 2014.
Article in English | MEDLINE | ID: mdl-24272359

ABSTRACT

Since 1940 chemotherapy has been one of the major therapies used to kill cancer cells. However, conventional standard cytotoxic agents have a low therapeutic index and often show toxicity in healthy cells. Over the past decade, progress in molecular biology and genomics has identified signaling pathways and mutations driving different types of cancer. Genetic and epigenetic alterations that characterize tumor cells have been used in the development of targeted therapy, a very active area of cancer research. Moreover, identification of synthetic lethal interactions between two altered genes in cancer cells shows much promise to target specifically tumor cells. For a long time, apoptosis was considered the principal mechanism by which cells die from chemotherapeutic agents. Autophagy, necroptosis (a programmed cell death mechanism of necrosis), and lysosomal-mediated cell death significantly improve our understanding of how malignancy can be targeted by anticancer treatments. Autophagy is a highly regulated process by which misfolded proteins and organelles reach lysosomes for their degradation. Alterations in this cellular process have been observed in several pathological conditions, including cancer. The role of autophagy in cancer raised a paradox wherein it can act as a tumor suppressor at early stage of tumor development but can also be used by cancer cells as cytoprotection to promote survival in established tumors. It is interesting that autophagy can be targeted by anticancer agents to provoke cancer cell death. This review focuses on the role of autophagy in cancer cells and its potential to therapeutically kill cancer cells.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Autophagy/physiology , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Humans , Lysosomes/physiology , Neoplasms/genetics , Neoplasms/pathology , Phagosomes/physiology , Signal Transduction/physiology
9.
Can Urol Assoc J ; 7(9-10): 319-23, 2013.
Article in English | MEDLINE | ID: mdl-24319509

ABSTRACT

BACKGROUND: Hereditary renal cell cancer (RCC) is an ideal model for germline genetic testing. We propose a guideline of hereditary RCC specific criteria to suggest referral for genetic assessment. METHODS: A review of the literature and stakeholder resources for existing guidelines or consensus statements was performed. Referral criteria were developed by expert consensus. RESULTS: The criteria included characteristics for patients with RCC (age ≤45 years, bilateral or multifocal tumours, associated medical conditions and non-clear cell histologies with unusual features) and for patients with or without RCC, but a family history of specific clinical or genetic diagnoses. CONCLUSIONS: This guideline represents a practical RCC-specific reference to allow healthcare providers to identify patients who may have a hereditary RCC syndrome, without extensive knowledge of each syndrome. RCC survivors and their families can also use the document to guide their discussions with healthcare providers about their need for referral. The criteria refer to the most common hereditary renal tumour syndromes and do not represent a comprehensive or exclusive list. Prospective validation of the criteria is warranted.

10.
Sci Transl Med ; 3(94): 94ra70, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21813754

ABSTRACT

Identifying new targeted therapies that kill tumor cells while sparing normal tissue is a major challenge of cancer research. Using a high-throughput chemical synthetic lethal screen, we sought to identify compounds that exploit the loss of the von Hippel-Lindau (VHL) tumor suppressor gene, which occurs in about 80% of renal cell carcinomas (RCCs). RCCs, like many other cancers, are dependent on aerobic glycolysis for ATP production, a phenomenon known as the Warburg effect. The dependence of RCCs on glycolysis is in part a result of induction of glucose transporter 1 (GLUT1). Here, we report the identification of a class of compounds, the 3-series, exemplified by STF-31, which selectively kills RCCs by specifically targeting glucose uptake through GLUT1 and exploiting the unique dependence of these cells on GLUT1 for survival. Treatment with these agents inhibits the growth of RCCs by binding GLUT1 directly and impeding glucose uptake in vivo without toxicity to normal tissue. Activity of STF-31 in these experimental renal tumors can be monitored by [(18)F]fluorodeoxyglucose uptake by micro-positron emission tomography imaging, and therefore, these agents may be readily tested clinically in human tumors. Our results show that the Warburg effect confers distinct characteristics on tumor cells that can be selectively targeted for therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Glucose Transporter Type 1/metabolism , Kidney Neoplasms/drug therapy , Adenosine Triphosphate/biosynthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Apoptosis , Carcinoma, Renal Cell/metabolism , Glucose/metabolism , Glycolysis , Humans , Kidney Neoplasms/metabolism
11.
Curr Opin Cell Biol ; 22(2): 246-51, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20056398

ABSTRACT

Autophagy is a cellular degradation process in which portions of the cell's cytoplasm and organelles are sequestered in a double-membrane bound vesicle called an autophagosome. Fusion of autophagosomes with lysosomes results in the formation of autolysosomes, where the proteins and organelles are degraded. This degradation pathway is induced under nutrient deprivation, metabolic stress or microenvironmental conditions to ensure energy balance, clearance of damaged proteins and adaptation to stress. Disruption of autophagy is involved in diverse human diseases including cancer. In particular, the regulation of autophagy in cancer cells is complex since it can enhance tumor cell survival in response to certain stresses, yet it can also act to suppress the initiation of tumor growth. Understanding the signaling pathways involved in the regulation of autophagy as well as the autophagy process itself represents new directions in the development of anticancer therapies. In this review, we discuss recent advances in our understanding the complexity of the autophagy process and the development of targeted therapies that modulate autophagy in cancer cells in the clinic.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Animals , Carcinoma, Renal Cell/drug therapy , Humans , Kidney Neoplasms/drug therapy
12.
J Med Chem ; 53(2): 787-97, 2010 Jan 28.
Article in English | MEDLINE | ID: mdl-19994864

ABSTRACT

Renal cell carcinomas (RCC) are refractory to standard therapy with advanced RCC having a poor prognosis; consequently treatment of advanced RCC represents an unmet clinical need. The von Hippel-Lindau (VHL) tumor suppressor gene is mutated or inactivated in a majority of RCCs. We recently identified a 4-pyridyl-2-anilinothiazole (PAT) with selective cytotoxicity against VHL-deficient renal cells mediated by induction of autophagy and increased acidification of autolysosomes. We report exploration of structure-activity relationships (SAR) around this PAT lead. Analogues with substituents on each of the three rings, and various linkers between rings, were synthesized and tested in vitro using paired RCC4 cell lines. A contour map describing the relative spatial contributions of different chemical features to potency illustrates a region, adjacent to the pyridyl ring, with potential for further development. Examples probing this domain validated this approach and may provide the opportunity to develop this novel chemotype as a targeted approach to the treatment of RCC.


Subject(s)
Aniline Compounds/pharmacology , Autophagy/drug effects , Carcinoma, Renal Cell/drug therapy , Thiazoles/pharmacology , von Hippel-Lindau Disease/drug therapy , Aniline Compounds/chemistry , Aniline Compounds/therapeutic use , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Drug Delivery Systems , Humans , Lysosomes/drug effects , Pyridines , Structure-Activity Relationship , Thiazoles/chemistry , Thiazoles/therapeutic use , von Hippel-Lindau Disease/pathology
13.
Autophagy ; 4(7): 944-6, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18769110

ABSTRACT

Radiation and conventional cytotoxic chemotherapies are ineffective in treating renal cancer. Approximately 75 percent of renal cell carcinoma (RCC) is associated with an inactivation of the tumor suppressor gene von Hippel-Lindau (VHL). We exploited the possibility of targeting VHL-deficient RCC through synthetic lethality using a high-throughput screening approach. In this screen, STF-62247 was identified to be selectively toxic and growth inhibitory to renal cells lacking VHL. We recently demonstrated that the cytotoxicity of STF-62247 is due to dysregulated autophagy. Furthermore, the reduction of protein levels of essential autophagy pathway components such as Atg5, Atg7 and Atg9 reduces sensitivity of VHL-deficient cells to killing by STF-62247. Loss of proteins involved in Golgi trafficking sensitized RCC with wild-type VHL to killing by STF-62247, indicating a potential role for these proteins as a target of the compound. Our study supports the concept of using synthetic lethality to selectively kill VHL-deficient cells that represents a new type of targeted therapy for the treatment of RCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Pyridines/therapeutic use , Thiazoles/therapeutic use , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Humans , Kidney Neoplasms/genetics
14.
Cancer Cell ; 14(1): 90-102, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18598947

ABSTRACT

Renal cell carcinomas (RCCs) are refractory to standard therapies. The von Hippel-Lindau (VHL) tumor suppressor gene is inactivated in 75% of RCCs. By screening for small molecules selectively targeting VHL-deficient RCC cells, we identified STF-62247. STF-62247 induces cytotoxicity and reduces tumor growth of VHL-deficient RCC cells compared to genetically matched cells with wild-type VHL. STF-62247-stimulated toxicity occurs in a HIF-independent manner through autophagy. Reduction of protein levels of essential autophagy pathway components reduces sensitivity of VHL-deficient cells to STF-62247. Using a yeast deletion pool, we show that loss of proteins involved in Golgi trafficking increases killing by STF-62247. Thus, we have found a small molecule that selectively induces cell death in VHL-deficient cells, representing a paradigm shift for targeted therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Pyridines/pharmacology , Thiazoles/pharmacology , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Antineoplastic Agents/chemistry , Autophagy-Related Protein 5 , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Gene Expression Regulation, Neoplastic , Gene Silencing , Golgi Apparatus/metabolism , Humans , Hydrogen-Ion Concentration , Hypoxia-Inducible Factor 1/metabolism , Inhibitory Concentration 50 , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Male , Mice , Mice, SCID , Microtubule-Associated Proteins/metabolism , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Protein Transport , Structure-Activity Relationship , Time Factors , Transfection , Vacuoles/drug effects , Vacuoles/metabolism , Vacuoles/pathology , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Yeasts/drug effects , Yeasts/growth & development , Yeasts/metabolism
15.
Biochem Biophys Res Commun ; 372(3): 440-6, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18485890

ABSTRACT

We have investigated the involvement of P-glycoprotein (P-gp)/caveolin-1 interaction in the regulation of brain endothelial cells (EC) migration and tubulogenesis. P-gp overexpression in MDCK-MDR cells was correlated with enhanced cell migration whereas treatment with P-gp inhibitors CsA or PSC833 reduced it. Transfection of RBE4 rat brain endothelial cells with mutated versions of MDR1, in the caveolin-1 interaction motif, decreased the interaction between P-gp and caveolin-1, enhanced P-gp transport activity and cell migration. Moreover, down-regulation of caveolin-1 in RBE4 cells by siRNA against caveolin-1 stimulated cell migration. Interestingly, the inhibition of P-gp/caveolin-1 interaction increased also EC tubulogenesis. Furthermore, decrease of P-gp expression by siRNA inhibited EC tubulogenesis. These data indicate that the level of P-gp/caveolin-1 interaction can modulate brain endothelial angiogenesis and P-gp dependent cell migration.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Brain/blood supply , Caveolin 1/metabolism , Cell Movement , Endothelial Cells/physiology , Neovascularization, Physiologic , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Caveolin 1/genetics , Cell Line , Cell Movement/drug effects , Cell Movement/genetics , Cyclosporine/pharmacology , Cyclosporins/pharmacology , Dogs , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/genetics , RNA, Small Interfering/genetics , Rats
16.
Methods Enzymol ; 435: 323-45, 2007.
Article in English | MEDLINE | ID: mdl-17998061

ABSTRACT

Tumor hypoxia is a feature common to almost all solid tumors due to malformed vasculature and inadequate perfusion. Tumor cells have evolved mechanisms that allow them to respond and adapt to a hypoxic microenvironment. The hypoxia-inducible transcription factor (HIF) family is comprised of oxygen-sensitive alpha (alpha) subunits that respond rapidly to decreased oxygen levels and oxygen-insensitive beta (beta) subunits. HIF binds to specific recognition sequences in the genome and increases the transcription of genes involved in a variety of metabolic and enzymatic pathways that are necessary for cells to respond to an oxygen-poor environment. The critical role of this family of transcriptional regulators in maintaining oxygen homeostasis is supported by multiple regulatory mechanisms that allow the cell to control the levels of HIF as well as its transcriptional activity. This review will focus on how the transcriptional activity of HIF is studied and how it can be exploited for cancer therapy.


Subject(s)
Genetic Techniques , Hypoxia-Inducible Factor 1/genetics , Neoplasms/therapy , Animals , Gene Expression , Humans , Hypoxia-Inducible Factor 1/analysis , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Mice , Mice, Knockout , Neoplasms/genetics
17.
Cancer Res ; 67(12): 5896-905, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575159

ABSTRACT

Late-stage clear cell renal carcinoma poses a formidable clinical challenge due to the high mortality rate associated with this disease. Molecular and genetic studies have identified functional loss of the von Hippel-Lindau (VHL) gene as a frequent and crucial event in the development of the malignant phenotype of clear cell renal carcinomas. Loss of VHL function thus represents a pathognomonic molecular defect for therapeutic exploitation. The objective of this study was to evaluate the possibility of targeting VHL loss through pharmacologic means. Chromomycin A3 (ChA3) was identified through in silico analysis of existing publicly available drug profiles from the National Cancer Institute as an agent that seemed to selectively target VHL-deficient clear cell renal carcinoma cells. Genotype-selective toxicity was first determined through short-term viability assays and then confirmed with clonogenic studies. Coculture of fluorescently labeled VHL-deficient and VHL-positive cells showed discriminate killing of the VHL-deficient cells with ChA3. Mechanistically, overexpression of hypoxia-inducible factor (HIF)-2alpha in VHL-positive clear cell renal carcinoma cells phenocopied loss of VHL with respect to ChA3 toxicity, establishing ChA3 as a HIF-dependent cytotoxin. This study shows the feasibility of selectively targeting the loss of the VHL tumor suppressor gene in clear cell renal carcinoma for potential clinical benefit and may have greater ramifications in the development of new targeted therapies for the treatment of cancer and other genetic diseases.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Carcinoma, Renal Cell/genetics , Chromomycin A3/administration & dosage , Drug Delivery Systems/methods , Kidney Neoplasms/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Algorithms , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Carcinoma, Renal Cell/drug therapy , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Kidney Neoplasms/drug therapy
18.
Int J Cancer ; 114(5): 702-12, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15609318

ABSTRACT

The ras oncogenes are among those most frequently found in human cancers. Blocking Ras farnesylation is a promising strategy for arresting cancer growth. Ras activates several signaling pathways with key roles in cellular proliferation, invasion, metastasis and angiogenesis. Furthermore, proteolytic activities of matrix proteinases such as urokinase-type plasminogen activator (uPA) and matrix metalloproteinases (MMPs) are regulated by Ras isoforms. Thus, we investigated the effects of SCH-66336, a farnesyltransferase inhibitor, on secretion of components of the plasminogen activation system as well as on the gelatinases MMP-2 and MMP-9, which play pivotal roles in matrix remodeling. SCH-66336 up to 5 microM did not significantly alter the viability of prostate (PC-3) and renal (Caki-1) cancer cells incubated in serum-depleted medium. SCH-66336 partly inhibited the processing of H-Ras, while levels of mature N-Ras and K-Ras remained unaffected. Under these noncytotoxic conditions, uPA and tPA levels were lowered in culture medium but raised in cell lysates, suggesting inhibition of trafficking pathways. In contrast, SCH-66336 had no effect on uPAR expression or on secreted PAI-1 levels. As expected, the reduction of uPA and tPA activities by SCH-66336 inhibited the conversion of plasminogen to plasmin by about 25% in PC-3 cells. SCH-66336 also inhibited the levels of secreted pro-MMP-2 and pro-MMP-9 as well as the release of their inhibitors TIMP-1 and TIMP-2. SCH-66336 decreased both the adhesion and even more so the migration of PC-3 cells on gelatin. Thus, SCH-66336 inhibited farnesylation in both cancer cell types, and H-Ras functions should be reduced by the drug. In addition, the lower levels of secreted proteinases in the presence of SCH-66336 suggest that reduced matrix remodeling and cell migration should occur in treated tumors.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Carcinoma/enzymology , Down-Regulation , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Matrix Metalloproteinases/biosynthesis , Piperidines/pharmacology , Pyridines/pharmacology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Survival , Culture Media/pharmacology , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Farnesyltranstransferase , Fibrinolysin/metabolism , Humans , Immunoblotting , Kinetics , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Plasminogen/metabolism , Protein Isoforms , Proto-Oncogene Proteins p21(ras)/metabolism , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator
19.
Biochem Biophys Res Commun ; 324(2): 937-45, 2004 Nov 12.
Article in English | MEDLINE | ID: mdl-15474518

ABSTRACT

The organosulfur compounds (OSCs), present in garlic, are studied for their protective effect against human cancers. P-glycoprotein (P-gp) and multidrug resistance protein 2 (Mrp2) are two transporters involved in the defense of cells and in the development of multidrug resistance. Whereas OSCs increase glutathione S-transferase activity (GST), Mrp2 plays a role in the transport of glutathione (GSH)-conjugates. In this study, we have investigated the effect of two OSCs, diallyl disulfide (DADS) and S-allyl cysteine (SAC), on P-gp and Mrp2 expression in renal brush-border membranes. By Western blot analysis, our results show that DADS induces Mrp2 expression (by 7-fold), which correlates with the rise of GST activity and GSH levels. Surprisingly, a co-administration of OSC with cisplatin, an anticancer drug, significantly increased Mrp2 gene and protein expression (by 30-fold), suggesting that DADS could potentiate the effects of cisplatin. Interestingly, SAC and cisplatin in co-treatment decreased P-gp protein expression and mdr1b isoform mRNA levels. In addition, modulation of the mdr1b isoform and Mrp2 by cisplatin was completely abolished by a glutathione precursor, N-acetyl cysteine. These results indicate that OSCs present in a garlic-rich diet might alter chemotherapeutic treatments using P-gp or Mrp2 substrates.


Subject(s)
Allyl Compounds/pharmacology , Anticarcinogenic Agents/pharmacology , Cysteine/analogs & derivatives , Disulfides/pharmacology , Garlic/metabolism , Mitochondrial Proteins/biosynthesis , Plant Extracts/pharmacology , Ribosomal Proteins/biosynthesis , Saccharomyces cerevisiae Proteins/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Acetylcysteine/metabolism , Allyl Compounds/metabolism , Animals , Blotting, Western , Cell Membrane/metabolism , Cisplatin/pharmacology , Cysteine/metabolism , Disulfides/metabolism , Glutathione/metabolism , Glutathione Transferase/metabolism , Kidney/metabolism , Kidney Cortex/metabolism , Liver/metabolism , Male , Microvilli/metabolism , Mitochondrial Proteins/genetics , Protein Isoforms , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Ribosomal Proteins/genetics , Saccharomyces cerevisiae Proteins/genetics
20.
Int J Cancer ; 112(5): 777-86, 2004 Dec 10.
Article in English | MEDLINE | ID: mdl-15386385

ABSTRACT

Inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene is associated with the development of vascular tumors including renal cell carcinoma. Aside from the role played by the VHL protein (pVHL) in negative regulation of hypoxia-inducible factor, 41F-1alpha, pVHL also takes part in cytoskeletal organization. Thrombin is a serine protease involved in angiogenesis and in cancer progression and its action is mediated by the protease-activated receptors (PARs). In several cell types, thrombin induces reorganization of the cytoskeleton along with RhoA activation. Thus, we conducted an investigation on the capacity of thrombin to regulate pVHL expression. Our results demonstrated that VHL mRNA and protein levels were increased by thrombin in cultured renal cancer cells. Cytoplasmic pVHL was redistributed to perinuclear regions and membrane fractions following thrombin treatments. Stimulation of Caki-1 cells with PAR1, PAR2 and PAR4 agonist peptides demonstrated that PAR1 was the receptor involved in thrombin-induced pVHL expression. Western blot analysis confirmed that these cells express PAR1 and that its expression was increased by thrombin. PAR1 activation by both thrombin and an agonist peptide stimulated renal cancer cell invasion through Matrigel. Interestingly, the upregulation of pVHL was dependent on RhoA because C3 exotoxin abolished pVHL induction. However, the pharmacological Rho kinase inhibitor, Y27632, did not influence pVHL expression in the presence of thrombin, suggesting that other RhoA effectors were involved in the process. Together, these results demonstrate that thrombin induces both pVHL expression via PAR1/RhoA activation as well as the stimulation of renal cancer cell invasion suggesting a role for thrombin in tumor invasion.


Subject(s)
Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/physiopathology , Receptor, PAR-1/physiology , Thrombin/pharmacology , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/pharmacology , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Tumor Cells, Cultured , Up-Regulation , Von Hippel-Lindau Tumor Suppressor Protein , rhoA GTP-Binding Protein/pharmacology , rhoA GTP-Binding Protein/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...