Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cells ; 13(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38727288

ABSTRACT

Glioblastoma (GBM) is a devastating brain cancer for which new effective therapies are urgently needed. GBM, after an initial response to current treatment regimens, develops therapeutic resistance, leading to rapid patient demise. Cancer cells exhibit an inherent elevation of endoplasmic reticulum (ER) stress due to uncontrolled growth and an unfavorable microenvironment, including hypoxia and nutrient deprivation. Cancer cells utilize the unfolded protein response (UPR) to maintain ER homeostasis, and failure of this response promotes cell death. In this study, as integrins are upregulated in cancer, we have evaluated the therapeutic potential of individually targeting all αß1 integrin subunits using RNA interference. We found that GBM cells are uniquely susceptible to silencing of integrin α3. Knockdown of α3-induced proapoptotic markers such as PARP cleavage and caspase 3 and 8 activation. Remarkably, we discovered a non-canonical function for α3 in mediating the maturation of integrin ß1. In its absence, generation of full length ß1 was reduced, immature ß1 accumulated, and the cells underwent elevated ER stress with upregulation of death receptor 5 (DR5) expression. Targeting α3 sensitized TRAIL-resistant GBM cancer cells to TRAIL-mediated apoptosis and led to growth inhibition. Our findings offer key new insights into integrin α3's role in GBM survival via the regulation of ER homeostasis and its value as a therapeutic target.


Subject(s)
Endoplasmic Reticulum Stress , Glioblastoma , Integrin alpha3 , Integrin beta1 , TNF-Related Apoptosis-Inducing Ligand , Humans , Apoptosis/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Cell Line, Tumor , Glioblastoma/pathology , Glioblastoma/metabolism , Glioblastoma/genetics , Integrin alpha3/metabolism , Integrin alpha3/genetics , Integrin beta1/metabolism , Integrin beta1/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology
3.
Mol Biol Cell ; 33(2): ar13, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34818063

ABSTRACT

ELMODs are a family of three mammalian paralogues that display GTPase-activating protein (GAP) activity toward a uniquely broad array of ADP-ribosylation factor (ARF) family GTPases that includes ARF-like (ARL) proteins. ELMODs are ubiquitously expressed in mammalian tissues, highly conserved across eukaryotes, and ancient in origin, being present in the last eukaryotic common ancestor. We described functions of ELMOD2 in immortalized mouse embryonic fibroblasts (MEFs) in the regulation of cell division, microtubules, ciliogenesis, and mitochondrial fusion. Here, using similar strategies with the paralogues ELMOD1 and ELMOD3, we identify novel functions and locations of these cell regulators and compare them to those of ELMOD2, allowing the determination of functional redundancy among the family members. We found strong similarities in phenotypes resulting from deletion of either Elmod1 or Elmod3 and marked differences from those arising in Elmod2 deletion lines. Deletion of either Elmod1 or Elmod3 results in the decreased ability of cells to form primary cilia, loss of a subset of proteins from cilia, and accumulation of some ciliary proteins at the Golgi, predicted to result from compromised traffic from the Golgi to cilia. These phenotypes are reversed upon activating mutant expression of either ARL3 or ARL16, linking their roles to ELMOD1/3 actions.


Subject(s)
GTPase-Activating Proteins/metabolism , ADP-Ribosylation Factors/metabolism , Animals , Cilia/metabolism , Cytoskeletal Proteins/metabolism , Fibroblasts/metabolism , GTPase-Activating Proteins/physiology , Golgi Apparatus/metabolism , Mice , Microtubules/metabolism , Mitochondrial Dynamics , Signal Transduction/genetics
4.
Oncogene ; 40(33): 5182-5191, 2021 08.
Article in English | MEDLINE | ID: mdl-34218269

ABSTRACT

Uveal melanoma (UM) is the most prevalent primary intraocular malignancy in adults, and patients that develop metastases (~50%) survive <1 year, highlighting the urgent need for new therapies. TCGA has recently revealed that a hypoxia gene signature is associated with poor UM patient prognosis. Here we show that expression of hypoxia-regulated collagen prolyl-4-hydroxylase genes P4HA1 and P4HA2 is significantly upregulated in UM patients with metastatic disease and correlates with poor prognosis, suggesting these enzymes might be key tumor drivers. We targeted hypoxia-induced expression of P4HA1/2 in UM with KCN1, a hypoxia inducible factor-1 (HIF-1) pathway inhibitor and found potent inhibition of primary and metastatic disease and extension of animal survival, without overt side effects. At the molecular level, KCN1 antagonized hypoxia-induced expression of P4HA1 and P4HA2, which regulate collagen maturation and deposition in the extracellular matrix. The treatment decreased prolyl hydroxylation, induced proteolytic cleavage and rendered a disordered structure to collagen VI, the main collagen produced by UM, and reduced UM cell invasion. Together, these data demonstrate that extracellular collagen matrix formation can be targeted in UM by inhibiting hypoxia-induced P4HA1 and P4HA2 expression, warranting further development of this strategy in patients with uveal melanoma.


Subject(s)
Hypoxia-Inducible Factor-Proline Dioxygenases , Extracellular Matrix , Humans , Hydroxylation , Melanoma , Transcriptional Activation , Up-Regulation , Uveal Neoplasms
5.
Clin Cancer Res ; 26(11): 2711-2724, 2020 06 01.
Article in English | MEDLINE | ID: mdl-31969339

ABSTRACT

PURPOSE: Exploitation of altered glycosylation in cancer is a major goal for the design of new cancer therapy. Here, we designed a novel secreted chimeric signal peptide-Galectin-3 conjugate (sGal-3) and investigated its ability to induce cancer-specific cell death by targeting aberrantly N-glycosylated cell surface receptors on cancer cells. EXPERIMENTAL DESIGN: sGal-3 was genetically engineered from Gal-3 by extending its N-terminus with a noncleavable signal peptide from tissue plasminogen activator. sGal-3 killing ability was tested on normal and tumor cells in vitro and its antitumor activity was evaluated in subcutaneous lung cancer and orthotopic malignant glioma models. The mechanism of killing was investigated through assays detecting sGal-3 interaction with specific glycans on the surface of tumor cells and the elicited downstream proapoptotic signaling. RESULTS: We found sGal-3 preferentially binds to ß1 integrin on the surface of tumor cells due to aberrant N-glycosylation resulting from cancer-associated upregulation of several glycosyltransferases. This interaction induces potent cancer-specific death by triggering an oncoglycan-ß1/calpain/caspase-9 proapoptotic signaling cascade. sGal-3 could reduce the growth of subcutaneous lung cancers and malignant gliomas in brain, leading to increased animal survival. CONCLUSIONS: We demonstrate that sGal-3 kills aberrantly glycosylated tumor cells and antagonizes tumor growth through a novel integrin ß1-dependent cell-extrinsic apoptotic pathway. These findings provide proof-of-principle that aberrant N-oncoglycans represent valid cancer targets and support further translation of the chimeric sGal-3 peptide conjugate for cancer therapy.


Subject(s)
Apoptosis , Blood Proteins/metabolism , Galectins/metabolism , Glioma/drug therapy , Integrin beta1/metabolism , Peptide Fragments/pharmacology , Protein Sorting Signals , Animals , Blood Proteins/genetics , Cell Proliferation , Female , Galectins/genetics , Glioma/metabolism , Glioma/pathology , Glycosylation , Humans , Integrin beta1/genetics , Mice , Mice, Nude , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Oncogene ; 39(5): 1041-1048, 2020 01.
Article in English | MEDLINE | ID: mdl-31582835

ABSTRACT

Medulloblastoma (MB) is a malignant pediatric brain tumor for which new therapies are urgently needed. We demonstrate that treatment with EPZ-6438 (Tazemetostat), an enhancer of zeste homolog 2 (EZH2) inhibitor approved for clinical trials, blocks MB cell growth in vitro and in vivo, and prolongs survival in orthotopic xenograft models. We show that the therapeutic effect is dependent on epigenetic reactivation of adhesion G-protein-coupled receptor B1 (BAI1/ADGRB1), a tumor suppressor that controls p53 stability by blocking Mdm2. Histone 3 trimethylated on lysine 27 (H3K27me3), a marker of silent chromatin conformation is present at the ADGRB1 promoter, and inhibition of EZH2, the catalytic component of the Polycomb Repressive complex 2 (PRC2) that methylates H3K27, switches the gene into an active chromatin status and reactivates BAI1 expression. Mechanistically, targeting EZH2 promotes transition from H3K27me3 to H3K27ac at the promoter, recruits the C/EBPß (CREB-binding protein) and CBP transcription factors and activates ADGRB1 gene transcription. Taken together, our results identify key molecular players that regulate ADGRB1 gene expression in MB, demonstrate that reactivation of BAI1 expression underlies EPZ-6438 antitumorigenic action, and provide preclinical proof-of-principle evidence for targeting EZH2 in patients with MB.


Subject(s)
Angiogenic Proteins/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Medulloblastoma/pathology , Receptors, G-Protein-Coupled/genetics , Tumor Suppressor Protein p53/genetics , Angiogenic Proteins/deficiency , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic , Gene Silencing , Histones/metabolism , Humans , Methylation , Peptide Fragments/metabolism , Receptors, G-Protein-Coupled/deficiency , Sialoglycoproteins/metabolism
7.
Oncogene ; 39(5): 1165, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31676871

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Clin Cancer Res ; 25(7): 2206-2218, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30563937

ABSTRACT

PURPOSE: Uveal melanoma (UM) is the most prevalent and lethal intraocular malignancy in adults. Here, we examined the importance of hypoxia in UM growth and tested the antitumor effects of arylsulfonamide 64B, an inhibitor of the hypoxia-induced factor (HIF) pathway in animal models of UM and investigated the related mechanisms. EXPERIMENTAL DESIGN: UM cells were implanted in the uvea of mice eyes and mice systemically treated with 64B. Drug effect on primary eye tumor growth, circulating tumor cells, metastasis formation in liver, and survival were examined. 64B effects on UM cell growth, invasion and hypoxia-induced expression of C-X-C chemokine receptor type 4 (CXCR4) and mesenchymal-epithelial transition factor (c-Met) were measured. Luciferase reporter assays, chromatin immunoprecipitation, co-immunoprecipitation, and cellular thermal shift assays were used to determine how 64B interferes with the HIF transcriptional complex. RESULTS: Systemic administration of 64B had potent antitumor effects against UM in several orthotopic mouse models, suppressing UM growth in the eye (∼70% reduction) and spontaneous liver metastasis (∼50% reduction), and extending mice survival (P < 0.001) while being well tolerated. 64B inhibited hypoxia-induced expression of CXCR4 and c-Met, 2 key drivers of tumor invasion and metastasis. 64B disrupted the HIF-1 complex by interfering with HIF-1α binding to p300/CBP co-factors, thus reducing p300 recruitment to the MET and CXCR4 gene promoters. 64B could thermostabilize p300, supporting direct 64B binding to p300. CONCLUSIONS: Our preclinical efficacy studies support the further optimization of the 64B chemical scaffold toward a clinical candidate for the treatment of UM.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Melanoma/etiology , Melanoma/metabolism , Proto-Oncogene Proteins c-met/genetics , Receptors, CXCR4/genetics , Sulfonamides/pharmacology , Uveal Neoplasms/etiology , Uveal Neoplasms/metabolism , Animals , Biomarkers, Tumor , Biopsy , Cell Line, Tumor , Disease Models, Animal , E1A-Associated p300 Protein/metabolism , Humans , Liver Neoplasms/secondary , Melanoma/drug therapy , Melanoma/pathology , Mice , Prognosis , Protein Binding , Proto-Oncogene Proteins c-met/metabolism , Receptors, CXCR4/metabolism , Sulfonamides/chemistry , Uveal Neoplasms/drug therapy , Uveal Neoplasms/pathology , Xenograft Model Antitumor Assays
9.
Cancer Cell ; 33(6): 1004-1016.e5, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29894688

ABSTRACT

Adhesion G protein-coupled receptors (ADGRs) encompass 33 human transmembrane proteins with long N termini involved in cell-cell and cell-matrix interactions. We show the ADGRB1 gene, which encodes Brain-specific angiogenesis inhibitor 1 (BAI1), is epigenetically silenced in medulloblastomas (MBs) through a methyl-CpG binding protein MBD2-dependent mechanism. Knockout of Adgrb1 in mice augments proliferation of cerebellar granule neuron precursors, and leads to accelerated tumor growth in the Ptch1+/- transgenic MB mouse model. BAI1 prevents Mdm2-mediated p53 polyubiquitination, and its loss substantially reduces p53 levels. Reactivation of BAI1/p53 signaling axis by a brain-permeable MBD2 pathway inhibitor suppresses MB growth in vivo. Altogether, our data define BAI1's physiological role in tumorigenesis and directly couple an ADGR to cancer formation.


Subject(s)
Angiogenic Proteins/metabolism , Cerebellar Neoplasms/metabolism , Medulloblastoma/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Angiogenic Proteins/genetics , Animals , Cell Line, Tumor , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , HCT116 Cells , Humans , Kaplan-Meier Estimate , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Proto-Oncogene Proteins c-mdm2/genetics , RNA Interference , Receptors, G-Protein-Coupled , Small Molecule Libraries/pharmacology , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
10.
Bioorg Med Chem Lett ; 27(8): 1731-1736, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28285917

ABSTRACT

Many forms of solid tumor have a characteristic feature known as hypoxia, which describes a low or non-existent presence of oxygen in the cellular microenvironment. This decrease in oxygen causes activation of the hypoxia inducible factor (HIF) pathway, which activates the transcription of many genes that cause cell proliferation, metastasis, increased glycolysis and angiogenesis. Increased HIF expression has been linked with poor patient prognosis, increased malignancy, and therapeutic resistance. Previous work in our lab has identified 1 and 2 as inhibitors of the HIF pathway, specifically as disrupters of the p300-HIF-1α complex formation. A library of sulfonamide analogs has been designed and synthesized with the intent of examining the SAR of this series of compounds and improving potency and physicochemical properties as compared with lead compounds 1 and 2. At the end, we have achieved a thorough understanding of the structural features critical for future optimization work.


Subject(s)
Benzopyrans/chemistry , Benzopyrans/pharmacology , E1A-Associated p300 Protein/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , E1A-Associated p300 Protein/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Molecular Docking Simulation , Signal Transduction/drug effects , Structure-Activity Relationship
11.
Clin Cancer Res ; 22(24): 6256-6265, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27340277

ABSTRACT

PURPOSE: Elevation in D-2-Hydroxyglutarate (D-2HG) has recently emerged as a mandatory byproduct of mutated Isocitrate Dehydrogenase (IDH) genes 1 and 2 in glioma patients. The goal of the present study was to demonstrate the feasibility of detection of elevated levels of D-2HG in the cerebrospinal fluid (CSF) of glioma patients that carry point substitutions in the IDH gene. EXPERIMENTAL DESIGN: We developed a mass spectrometry (MS)-based platform to detect and quantify the D- and L-forms of 2HG in the CSF of glioma patients. Three independent cohorts of patients were analyzed, comprising a total of 176 samples derived from 84 patients. The levels of D- and L-2HG were used to stratify patients into IDH wild-type or IDH-mutated groups using an empirically obtained threshold of 0.69 µmol/L. RESULTS: Using this platform, a greater than 17-fold mean increase in D-2HG was observed in the CSF of patients with IDH mutant versus wild-type gliomas. The means for the D-2HG levels in CSF were 0.427 µmol/L in wild-type and 7.439 µmol/L in mutant groups. The C statistic for the receiver operator curve was 0.938, with 84% sensitivity, 90% specificity, and 89% accuracy to detect D-2HG. The levels of D- and L-2HG in CSF from wild-type patients varied by location of CSF draw (cisternal > ventricular > lumbar). CONCLUSIONS: Our findings demonstrate that the CSF of patients harboring IDH mutant gliomas contain increased levels of D-2HG, which can be reliably detected with a MS-based platform. Clin Cancer Res; 22(24); 6256-65. ©2016 AACR.


Subject(s)
Brain Neoplasms/genetics , Cerebrospinal Fluid/metabolism , Glioma/genetics , Glutarates/cerebrospinal fluid , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Adult , Aged , Aged, 80 and over , Brain/metabolism , Brain Neoplasms/cerebrospinal fluid , Female , Glioma/cerebrospinal fluid , Humans , Male , Mass Spectrometry/methods , Middle Aged , Young Adult
12.
Mol Cell ; 53(4): 534-48, 2014 Feb 20.
Article in English | MEDLINE | ID: mdl-24486017

ABSTRACT

Mitochondrial pyruvate dehydrogenase complex (PDC) is crucial for glucose homeostasis in mammalian cells. The current understanding of PDC regulation involves inhibitory serine phosphorylation of pyruvate dehydrogenase (PDH) by PDH kinase (PDK), whereas dephosphorylation of PDH by PDH phosphatase (PDP) activates PDC. Here, we report that lysine acetylation of PDHA1 and PDP1 is common in epidermal growth factor (EGF)-stimulated cells and diverse human cancer cells. K321 acetylation inhibits PDHA1 by recruiting PDK1, and K202 acetylation inhibits PDP1 by dissociating its substrate PDHA1, both of which are important in promoting glycolysis in cancer cells and consequent tumor growth. Moreover, we identified mitochondrial ACAT1 and SIRT3 as the upstream acetyltransferase and deacetylase, respectively, of PDHA1 and PDP1, while knockdown of ACAT1 attenuates tumor growth. Furthermore, Y381 phosphorylation of PDP1 dissociates SIRT3 and recruits ACAT1 to PDC. Together, hierarchical, distinct posttranslational modifications act in concert to control molecular composition of PDC and contribute to the Warburg effect.


Subject(s)
Acetyl-CoA C-Acetyltransferase/metabolism , Pyruvate Dehydrogenase (Lipoamide)-Phosphatase/metabolism , Pyruvate Dehydrogenase (Lipoamide)/metabolism , Sirtuin 3/metabolism , Tyrosine/chemistry , Animals , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glycolysis , Humans , Lysine/chemistry , Male , Mice , Mice, Nude , Mitochondria/metabolism , Neoplasm Transplantation , Neoplasms/metabolism , Phosphorylation
13.
Clin Cancer Res ; 18(24): 6623-33, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22923450

ABSTRACT

PURPOSE: The hypoxia-inducible factor-1 (HIF-1) plays a critical role in tumor adaptation to hypoxia, and its elevated expression correlates with poor prognosis and treatment failure in patients with cancer. In this study, we determined whether 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide, KCN1, the lead inhibitor in a novel class of arylsulfonamide inhibitors of the HIF-1 pathway, had antitumorigenic properties in vivo and further defined its mechanism of action. EXPERIMENTAL DESIGN: We studied the inhibitory effect of systemic KCN1 delivery on the growth of human brain tumors in mice. To define mechanisms of KCN1 anti-HIF activities, we examined its influence on the assembly of a functional HIF-1α/HIF-1ß/p300 transcription complex. RESULTS: KCN1 specifically inhibited HIF reporter gene activity in several glioma cell lines at the nanomolar level. KCN1 also downregulated transcription of endogenous HIF-1 target genes, such as VEGF, Glut-1, and carbonic anhydrase 9, in a hypoxia-responsive element (HRE)-dependent manner. KCN1 potently inhibited the growth of subcutaneous malignant glioma tumor xenografts with minimal adverse effects on the host. It also induced a temporary survival benefit in an intracranial model of glioma but had no effect in a model of melanoma metastasis to the brain. Mechanistically, KCN1 did not downregulate the levels of HIF-1α or other components of the HIF transcriptional complex; rather, it antagonized hypoxia-inducible transcription by disrupting the interaction of HIF-1α with transcriptional coactivators p300/CBP. CONCLUSIONS: Our results suggest that the new HIF pathway inhibitor KCN1 has antitumor activity in mouse models, supporting its further translation for the treatment of human tumors displaying hypoxia or HIF overexpression.


Subject(s)
Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Brain Neoplasms/drug therapy , CREB-Binding Protein/antagonists & inhibitors , E1A-Associated p300 Protein/antagonists & inhibitors , Glioma/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Sulfonamides/pharmacology , Animals , Brain Neoplasms/pathology , CREB-Binding Protein/metabolism , Cell Line, Tumor , E1A-Associated p300 Protein/metabolism , Gene Expression/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Glioma/pathology , Humans , Inhibitory Concentration 50 , Luciferases, Renilla/biosynthesis , Luciferases, Renilla/genetics , Mice , Mice, Inbred C57BL , Mice, Nude , Protein Binding/drug effects , Response Elements , Signal Transduction/drug effects , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
J Med Chem ; 55(15): 6738-50, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22746274

ABSTRACT

The hypoxia inducible factor (HIF) pathway is an attractive target for cancer, as it controls tumor adaptation to growth under hypoxia and mediates chemotherapy and radiation resistance. We previously discovered 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide as a novel, small-molecule HIF-1 pathway inhibitor in a high-throughput cell-based assay, but its in vivo delivery is hampered by poor aqueous solubility (0.009 µM in water; log P(7.4) = 3.7). Here we describe the synthesis of 12 N-alkyl-N-[(8-R-2,2-dimethyl-2H-chromen-6-yl)methyl]heteroarylsulfonamides, which were designed to possess optimal lipophilicities and aqueous solubilities by in silico calculations. Experimental log P(7.4) values of 8 of the 12 new analogs ranged from 1.2-3.1. Aqueous solubilities of three analogs were measured, among which the most soluble N-[(8-methoxy-2,2-dimethyl-2H-chromen-6-yl)methyl]-N-(propan-2-yl)pyridine-2-sulfonamide had an aqueous solubility of 80 µM, e.g., a solubility improvement of ∼9000-fold. The pharmacological optimization had limited impact on drug efficacy as the compounds retained IC(50) values at or below 5 µM in our HIF-dependent reporter assay.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzopyrans/chemical synthesis , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Sulfonamides/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzopyrans/chemistry , Benzopyrans/pharmacology , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Signal Transduction , Solubility , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology , Transcription, Genetic/drug effects
15.
Bioorg Med Chem ; 20(14): 4590-7, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22682301

ABSTRACT

We have discovered that 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide, a novel small molecule HIF-1 pathway inhibitor, can antagonize tumor growth in animal models of cancer, but the treatment necessitates its delivery in a formulation, due to poor water solubility (<15 µg/mL; pH 7.4), evidencing that the chemotype needs further exploration of its amenability to additional chemical modifications for ultimate optimization of function and pharmacology. As a first step towards this goal we investigated the structure-activity relationships of 15 lipophilic 2,2-dimethyl-2H-chromene based arylsulfonamide analogs of 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide to find out strategies of modification. A 3,4-dimethoxybenzenesulfonyl group in region 1 showed the strongest inhibition among five arylsulfonyl groups tested. The presence of propan-2-amine in region 2 conferred the strongest inhibitory effect of the compound on HIF-1 activated transcription in a reporter assay. These findings are important as they help define the structural motifs where the 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide can be chemically modified to improve its pharmacological properties towards development as a cancer therapeutic.


Subject(s)
Antineoplastic Agents/chemistry , Benzopyrans/chemistry , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Sulfonamides/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Genes, Reporter , Humans , Hypoxia-Inducible Factor 1/metabolism , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/pharmacology , Transcription, Genetic/drug effects
16.
ACS Med Chem Lett ; 3(8): 620-5, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-24936238

ABSTRACT

Hypoxia inducible factors (HIFs) are transcription factors that activate expression of multiple gene products and promote tumor adaptation to a hypoxic environment. To become transcriptionally active, HIFs associate with cofactors p300 or CBP. Previously, we found that arylsulfonamides can antagonize HIF transcription in a bioassay, block the p300/HIF-1α interaction, and exert potent anticancer activity in several animal models. In the present work, KCN1-bead affinity pull down, (14)C-labeled KCN1 binding, and KCN1-surface plasmon resonance measurements provide initial support for a mechanism in which KCN1 can bind to the CH1 domain of p300 and likely prevent the p300/HIF-1α assembly. Using a previously reported NMR structure of the p300/HIF-1α complex, we have identified potential binding sites in the p300-CH1 domain. A two-site binding model coupled with IC50 values has allowed establishment of a modest ROC-based enrichment and creation of a guide for future analogue synthesis.

17.
J Med Chem ; 54(24): 8471-89, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22032632

ABSTRACT

Hypoxia, a reduction in partial oxygen pressure, is a salient property of solid tumors. Hypoxia drives malignant progression and metastasis in tumors and participates in tumor resistance to radio- and chemotherapies. Hypoxia activates the hypoxia-inducible factor (HIF) family of transcription factors, which induce target genes that regulate adaptive biological processes such as anaerobic metabolism, cell motility, and angiogenesis. Clinical evidence has demonstrated that expression of HIF-1 is strongly associated with poor patient prognosis and activation of HIF-1 contributes to malignant behavior and therapeutic resistance. Consequently, HIF-1 has become an important therapeutic target for inhibition by small molecules. Herein, we describe the design and synthesis of small molecules that inhibit the HIF-1 signaling pathway. Many of these compounds exhibit inhibitory activity in the nanomolar range. Separate mechanistic studies indicate that these inhibitors do not alter HIF-1 levels but interfere with the ability of HIF-1α/HIF-1ß to interact with cofactors p300/CBP to form an active transcriptional complex.


Subject(s)
Antineoplastic Agents/chemical synthesis , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Aryl Hydrocarbon Receptor Nuclear Translocator/antagonists & inhibitors , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Benzofurans/chemical synthesis , Benzofurans/chemistry , Benzofurans/pharmacology , Benzopyrans/chemical synthesis , Benzopyrans/chemistry , Benzopyrans/pharmacology , CREB-Binding Protein/metabolism , Cell Hypoxia , Cell Line, Tumor , Drug Design , Drug Screening Assays, Antitumor , Humans , Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Pyridines/chemical synthesis , Pyridines/chemistry , Pyridines/pharmacology , Quinolines/chemical synthesis , Quinolines/chemistry , Quinolines/pharmacology , Signal Transduction/drug effects , Structure-Activity Relationship , p300-CBP Transcription Factors/metabolism
18.
Bioorg Med Chem Lett ; 21(18): 5528-32, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21831638

ABSTRACT

Solid tumors generally grow under hypoxic conditions, a pathophysiological change, which activates the expression of genes responsible for malignant, aggressive, and treatment-refractory properties. Hypoxia inducible factor (HIF) is the chief transcription factor regulating hypoxia-driven gene expression. Therefore, the HIF pathway has become a critical target for cancer therapeutics development. We screened a privileged library of about 10,000 natural-product-like compounds using a cell-based assay for HIF-dependent transcriptional activity and identified several arylsulfonamide HIF pathway inhibitors. Among these compounds, the most potent ones showed an IC(50) of ∼0.5 µM in the hypoxia-responsive element (HRE)-luciferase reporter system. Further studies are needed to fully elucidate the mechanism of action of this class of compounds and their structure-activity relationship.


Subject(s)
Sulfonamides/pharmacology , Transcription Factors/antagonists & inhibitors , Alkaline Phosphatase/antagonists & inhibitors , Alkaline Phosphatase/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Hypoxia-Inducible Factor 1 , Molecular Structure , Small Molecule Libraries , Stereoisomerism , Structure-Activity Relationship , Sulfonamides/chemistry , Transcription Factors/metabolism
19.
Cancer Res ; 69(3): 1212-20, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19176395

ABSTRACT

Angiogenesis is a critical physiologic process that is appropriated during tumorigenesis. Little is known about how this process is specifically regulated in the brain. Brain angiogenesis inhibitor-1 (BAI1) is a brain-predominant seven-transmembrane protein that contains five antiangiogenic thrombospondin type-1 repeats (TSR). We recently showed that BAI1 is cleaved at a conserved proteolytic cleavage site releasing a soluble, 120 kDa antiangiogenic factor called vasculostatin (Vstat120). Vstat120 has been shown to inhibit in vitro angiogenesis and suppress subcutaneous tumor growth. Here, we examine its effect on the intracranial growth of malignant gliomas and further study its antitumor mechanism. First, we show that expression of Vstat120 strongly suppresses the intracranial growth of malignant gliomas, even in the presence of the strong proangiogenic stimulus mediated by the oncoprotein epidermal growth factor receptor variant III (EGFRvIII). This tumor-suppressive effect is accompanied by a decrease in tumor vascular density, suggesting a potent antiangiogenic effect in the brain. Second, and consistent with this interpretation, we find that treatment with Vstat120 reduces the migration of cultured microvascular endothelial cells in vitro and inhibits corneal angiogenesis in vivo. Third, we show that these antivascular effects critically depend on the presence of the cell surface receptor CD36 on endothelial cells in vitro and in vivo, supporting the role of Vstat120 TSRs in mediating these effects. These results advance the understanding of brain-specific angiogenic regulation, and suggest that Vstat120 has therapeutic potential in the treatment of brain tumors and other intracerebral vasculopathies.


Subject(s)
Angiogenic Proteins/biosynthesis , Brain Neoplasms/blood supply , Brain Neoplasms/therapy , CD36 Antigens/metabolism , Glioblastoma/blood supply , Glioblastoma/therapy , Peptide Fragments/biosynthesis , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Movement/physiology , Corneal Neovascularization/drug therapy , DNA, Complementary/administration & dosage , DNA, Complementary/genetics , Endothelial Cells/pathology , Glioblastoma/genetics , Humans , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/therapy , Peptide Fragments/genetics , Peptide Fragments/metabolism , Rats , Receptors, G-Protein-Coupled , Transfection , Xenograft Model Antitumor Assays
20.
Cancer Res ; 68(17): 6953-62, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18757409

ABSTRACT

BRCA1-associated protein-1 (BAP1), a deubiquitinating enzyme of unknown cellular function, is mutated in breast and lung cancers. In this study, we have shown for the first time that BAP1 has tumor suppressor activity in vivo by showing that BAP1 can suppress tumorigenicity of lung cancer cells in athymic nude mice. We show that BAP1 fulfills another criterion of a genuine tumor suppressor because cancer-associated BAP1 mutants are deficient in deubiquitinating activity. We show for the first time that one of the two predicted nuclear targeting motifs is required for nuclear localization of BAP1 and that a truncation mutant found in a lung cancer cell line results in BAP1 that fails to localize to the nucleus. Furthermore, we show that deubiquitinating activity and nuclear localization are both required for BAP1-mediated tumor suppression in nude mice. We show that BAP1 exerts its tumor suppressor functions by affecting the cell cycle, speeding the progression through the G(1)-S checkpoint, and inducing cell death via a process that has characteristics of both apoptosis and necrosis. Surprisingly, BAP1-mediated growth suppression is independent of wild-type BRCA1. Because deubiquitinating enzymes are components of the ubiquitin proteasome system, this pathway has emerged as an important target for anticancer drugs. The identification of the deubiquitinating enzyme BAP1 as a tumor suppressor may lead to further understanding of how the ubiquitin proteasome system contributes to cancer and aid in the identification of new targets for cancer therapy.


Subject(s)
Cell Nucleus/metabolism , Tumor Suppressor Proteins/physiology , Ubiquitin Thiolesterase/physiology , Ubiquitin/metabolism , Amino Acid Sequence , Animals , Cell Cycle , Cell Division , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Molecular Sequence Data , Sequence Homology, Amino Acid , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/chemistry , Ubiquitin Thiolesterase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...