Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters











Publication year range
1.
Nat Commun ; 15(1): 2485, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38509117

ABSTRACT

Proteasome subunit hRpn13 is partially proteolyzed in certain cancer cell types to generate hRpn13Pru by degradation of its UCHL5/Uch37-binding DEUBAD domain and retention of an intact proteasome- and ubiquitin-binding Pru domain. By using structure-guided virtual screening, we identify an hRpn13 binder (XL44) and solve its structure ligated to hRpn13 Pru by integrated X-ray crystallography and NMR to reveal its targeting mechanism. Surprisingly, hRpn13Pru is depleted in myeloma cells following treatment with XL44. TMT-MS experiments reveal a select group of off-targets, including PCNA clamp-associated factor PCLAF and ribonucleoside-diphosphate reductase subunit M2 (RRM2), that are similarly depleted by XL44 treatment. XL44 induces hRpn13-dependent apoptosis and also restricts cell viability by a PCLAF-dependent mechanism. A KEN box, but not ubiquitination, is required for XL44-induced depletion of PCLAF. Here, we show that XL44 induces ubiquitin-dependent loss of hRpn13Pru and ubiquitin-independent loss of select KEN box containing proteins.


Subject(s)
Membrane Glycoproteins , Proteasome Endopeptidase Complex , Proteasome Endopeptidase Complex/metabolism , Membrane Glycoproteins/metabolism , Intracellular Signaling Peptides and Proteins , Ubiquitin/metabolism , Cytoplasm/metabolism , Transcription Factors
2.
Aging (Albany NY) ; 14(3): 1068-1086, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35158337

ABSTRACT

Radiation therapy is a commonly used treatment modality for cancer. Although effective in providing local tumor control, radiation causes oxidative stress, inflammation, immunomodulatory and mitogenic cytokine production, extracellular matrix production, and premature senescence in lung parenchyma. The senescence associated secretory phenotype (SASP) can promote inflammation and stimulate alterations in the surrounding tissue. Therefore, we hypothesized that radiation-induced senescent parenchymal cells in irradiated lung would enhance tumor growth. Using a murine syngeneic tumor model of melanoma and non-small cell lung cancer lung metastasis, we demonstrate that radiation causes a significant increase in markers of premature senescence in lung parenchyma within 4 to 8 weeks. Further, injection of B16F0 (melanoma) or Lewis Lung carcinoma (epidermoid lung cancer) cells at these time points after radiation results in an increase in the number and size of pulmonary tumor nodules relative to unirradiated mice. Treatment of irradiated mice with a senolytic agent (ABT-737) or agents that prevent senescence (rapamycin, INK-128) was sufficient to reduce radiation-induced lung parenchymal senescence and to mitigate radiation-enhanced tumor growth. These agents abrogated radiation-induced expression of 12-Lipoxygenase (12-LOX), a molecule implicated in several deleterious effects of senescence. Deficiency of 12-LOX prevented radiation-enhanced tumor growth. Together, these data demonstrate the pro-tumorigenic role of radiation-induced senescence, introduces the dual TORC inhibitor INK-128 as an effective agent for prevention of radiation-induced normal tissue senescence, and identifies senescence-associated 12-LOX activity as an important component of the pro-tumorigenic irradiated tissue microenvironment. These studies suggest that combining senotherapeutic agents with radiotherapy may decrease post-therapy tumor growth.


Subject(s)
Carcinoma, Lewis Lung , Lung Neoplasms , Melanoma, Experimental , Animals , Arachidonate 12-Lipoxygenase/pharmacology , Carcinoma, Lewis Lung/enzymology , Carcinoma, Lewis Lung/pathology , Cell Growth Processes , Cellular Senescence , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Melanoma, Experimental/enzymology , Melanoma, Experimental/pathology , Mice , Tumor Microenvironment
3.
Clin Cancer Res ; 24(13): 3137-3148, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29599412

ABSTRACT

Purpose: To evaluate the potential of hyperpolarized [1-13C]-pyruvate magnetic resonance spectroscopic imaging (MRSI) of prostate cancer as a predictive biomarker for targeting the Warburg effect.Experimental Design: Two human prostate cancer cell lines (DU145 and PC3) were grown as xenografts. The conversion of pyruvate to lactate in xenografts was measured with hyperpolarized [1-13C]-pyruvate MRSI after systemic delivery of [1-13C] pyruvic acid. Steady-state metabolomic analysis of xenograft tumors was performed with mass spectrometry and steady-state lactate concentrations were measured with proton (1H) MRS. Perfusion and oxygenation of xenografts were measured with electron paramagnetic resonance (EPR) imaging with OX063. Tumor growth was assessed after lactate dehydrogenase (LDH) inhibition with FX-11 (42 µg/mouse/day for 5 days × 2 weekly cycles). Lactate production, pyruvate uptake, extracellular acidification rates, and oxygen consumption of the prostate cancer cell lines were analyzed in vitro LDH activity was assessed in tumor homogenates.Results: DU145 tumors demonstrated an enhanced conversion of pyruvate to lactate with hyperpolarized [1-13C]-pyruvate MRSI compared with PC3 and a corresponding greater sensitivity to LDH inhibition. No difference was observed between PC3 and DU145 xenografts in steady-state measures of pyruvate fermentation, oxygenation, or perfusion. The two cell lines exhibited similar sensitivity to FX-11 in vitro LDH activity correlated to FX-11 sensitivity.Conclusions: Hyperpolarized [1-13C]-pyruvate MRSI of prostate cancer predicts efficacy of targeting the Warburg effect. Clin Cancer Res; 24(13); 3137-48. ©2018 AACR.


Subject(s)
Carbon-13 Magnetic Resonance Spectroscopy , Magnetic Resonance Imaging , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Pyruvic Acid , Animals , Biomarkers , Carbon-13 Magnetic Resonance Spectroscopy/methods , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Glycolysis , Heterografts , Humans , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Lactic Acid/metabolism , Magnetic Resonance Imaging/methods , Male , Metabolome , Metabolomics/methods , Mice , Oxygen Consumption , Pyruvic Acid/metabolism
4.
Oncotarget ; 9(12): 10723-10733, 2018 Feb 13.
Article in English | MEDLINE | ID: mdl-29535838

ABSTRACT

Papillary renal cell carcinomas (PRCC) are a histologically and genetically heterogeneous group of tumors that represent 15-20% of all kidney neoplasms and may require diverse therapeutic approaches. Alteration of the NF2 tumor suppressor gene, encoding a key regulator of the Hippo signaling pathway, is observed in 22.5% of PRCC. The Hippo signaling pathway controls cell proliferation by regulating the transcriptional activity of Yes-Associated Protein, YAP1. Loss of NF2 results in aberrant YAP1 activation. The Src family kinase member Yes also regulates YAP1 transcriptional activity. This study investigated the importance of YAP and Yes activity in three NF2-deficient PRCC cell lines. NF2-deficency correlated with increased expression of YAP1 transcriptional targets and siRNA-based knockdown of YAP1 and Yes1 downregulated this pathway and dramatically reduced cell viability. Dasatinib and saracatinib have potent inhibitory effects on Yes and treatment with either resulted in downregulation of YAP1 transcription targets, reduced cell viability, and G0-G1 cell cycle arrest. Xenograft models for NF2-deficient PRCC also demonstrated reduced tumor growth in response to dasatinib. Thus, inhibiting Yes and the subsequent transcriptional activity of YAP1 had a substantial anti-tumor cell effect both in vitro and in vivo and may provide a viable therapeutic approach for patients with NF2-deficient PRCC.

5.
Int J Radiat Oncol Biol Phys ; 100(2): 344-352, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29157749

ABSTRACT

PURPOSE: Specificity protein 1 (SP1) is involved in the transcription of several genes implicated in tumor maintenance. We investigated the effects of mithramycin A (MTA), an inhibitor of SP1 DNA binding, on radiation response. METHODS AND MATERIALS: Clonogenic survival after irradiation was assessed in 2 tumor cell lines (A549, UM-UC-3) and 1 human fibroblast line (BJ) after SP1 knockdown or MTA treatment. DNA damage repair was evaluated using γH2AX foci formation, and mitotic catastrophe was assessed using nuclear morphology. Gene expression was evaluated using polymerase chain reaction arrays. In vivo tumor growth delay was used to evaluate the effects of MTA on radiosensitivity. RESULTS: Targeting of SP1 with small interfering RNA or MTA sensitized A549 and UM-UC-3 to irradiation, with no effect on the BJ radiation response. MTA did not alter γH2AX foci formation after irradiation in tumor cells but did enhance mitotic catastrophe. Treatment with MTA suppressed transcription of genes involved in cell death. MTA administration to mice bearing A549 and UM-UC-3 xenografts enhanced radiation-induced tumor growth delay. CONCLUSIONS: These results support SP1 as a target for radiation sensitization and confirm MTA as a radiation sensitizer in human tumor models.


Subject(s)
DNA Damage , Neoplasms, Experimental/radiotherapy , Plicamycin/analogs & derivatives , Radiation-Sensitizing Agents/pharmacology , Sp1 Transcription Factor/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , DNA Repair/drug effects , Humans , Mice , Mitosis/drug effects , Neoplasms, Experimental/pathology , Plicamycin/pharmacology
6.
Sci Rep ; 6: 39714, 2016 12 22.
Article in English | MEDLINE | ID: mdl-28004808

ABSTRACT

Pulmonary fibrosis is a potentially lethal late adverse event of thoracic irradiation. Prior research indicates that unrestrained TGF-ß1 and/or type 2 cytokine-driven immune responses promote fibrosis following radiation injury, but the full spectrum of factors governing this pathology remains unclear. Interleukin 13 (IL-13) is a key factor in fibrotic disease associated with helminth infection, but it is unclear whether it plays a similar role in radiation-induced lung fibrosis. Using a mouse model, we tested the hypothesis that IL-13 drives the progression of radiation-induced pulmonary fibrosis. Irradiated lungs from wild-type c57BL/6NcR mice accumulated alternatively-activated macrophages, displayed elevated levels of IL-13, and extensive fibrosis, whereas IL-13 deficient mice were resistant to these changes. Furthermore, plasma from irradiated wild-type mice showed a transient increase in the IL-13 saturated fraction of the circulating decoy receptor IL-13Rα2. Finally, we determined that therapeutic neutralization of IL-13, during the period of IL-13Rα2 saturation was sufficient to protect mice from lung fibrosis. Taken together, our results demonstrate that IL-13 is a major regulator of radiation-induced lung injury and demonstrates that strategies focusing on IL-13 may be useful in screening for timely delivery of anti-IL-13 therapeutics.


Subject(s)
Interleukin-13/pharmacology , Lung Injury , Pulmonary Fibrosis , Radiation Injuries, Experimental , Animals , Lung Injury/immunology , Lung Injury/pathology , Lung Injury/prevention & control , Mice , Mice, Knockout , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/prevention & control , Radiation Injuries, Experimental/immunology , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/prevention & control
7.
Int J Radiat Oncol Biol Phys ; 94(5): 1163-72, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26883561

ABSTRACT

PURPOSE: To determine whether the delivery of recombinant truncated plasminogen activator inhibitor-1 (PAI-1) protein (rPAI-1(23)) would protect from the development of radiation-induced lung injury. METHODS AND MATERIALS: C57Bl/6 mice received intraperitoneal injections of rPAI-1(23) (5.4 µg/kg/d) or vehicle for 18 weeks, beginning 2 days before irradiation (IR) (5 daily fractions of 6 Gy). Cohorts of mice were followed for survival (n=8 per treatment) and tissue collection (n=3 per treatment and time point). Fibrosis in lung was assessed with Masson-Trichrome staining and measurement of hydroxyproline content. Senescence was assessed with staining for ß-galactosidase activity in lung and primary pneumocytes. RESULTS: Hydroxyproline content in irradiated lung was significantly reduced in mice that received rPAI-1(23) compared with mice that received vehicle (IR+vehicle: 84.97 µg/lung; IR+rPAI-1(23): 56.2 µg/lung, P=.001). C57Bl/6 mice exposed to IR+vehicle had dense foci of subpleural fibrosis at 19 weeks, whereas the lungs of mice exposed to IR+rPAI-1(23) were largely devoid of fibrotic foci. Cellular senescence was significantly decreased by rPAI-1(23) treatment in primary pneumocyte cultures and in lung at multiple time points after IR. CONCLUSIONS: These studies identify that rPAI-1(23) is capable of preventing radiation-induced fibrosis in murine lungs. These antifibrotic effects are associated with increased fibrin metabolism, enhanced matrix metalloproteinase-3 expression, and reduced senescence in type 2 pneumocytes. Thus, rPAI-1(23) is a novel therapeutic option for radiation-induced fibrosis.


Subject(s)
Alveolar Epithelial Cells/drug effects , Cellular Senescence/drug effects , Plasminogen Activator Inhibitor 1/therapeutic use , Pulmonary Fibrosis/prevention & control , Radiation Pneumonitis/complications , Recombinant Proteins/therapeutic use , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/radiation effects , Animals , Cell Proliferation , Cellular Senescence/radiation effects , Collagen/metabolism , Cytokines/metabolism , Female , Fibrin/metabolism , Hydroxyproline/analysis , Hydroxyproline/metabolism , Lung/metabolism , Lung/radiation effects , Matrix Metalloproteinase 3/metabolism , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Plasminogen Activator Inhibitor 1/metabolism , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , Radiation Pneumonitis/metabolism , Real-Time Polymerase Chain Reaction
8.
PLoS One ; 10(10): e0141786, 2015.
Article in English | MEDLINE | ID: mdl-26517842

ABSTRACT

The two cytosolic/nuclear isoforms of the molecular chaperone HSP90, stress-inducible HSP90α and constitutively expressed HSP90ß, fold, assemble and maintain the three-dimensional structure of numerous client proteins. Because many HSP90 clients are important in cancer, several HSP90 inhibitors have been evaluated in the clinic. However, little is known concerning possible unique isoform or conformational preferences of either individual HSP90 clients or inhibitors. In this report, we compare the relative interaction strength of both HSP90α and HSP90ß with the transcription factors HSF1 and HIF1α, the kinases ERBB2 and MET, the E3-ubiquitin ligases KEAP1 and RHOBTB2, and the HSP90 inhibitors geldanamycin and ganetespib. We observed unexpected differences in relative client and drug preferences for the two HSP90 isoforms, with HSP90α binding each client protein with greater apparent affinity compared to HSP90ß, while HSP90ß bound each inhibitor with greater relative interaction strength compared to HSP90α. Stable HSP90 interaction was associated with reduced client activity. Using a defined set of HSP90 conformational mutants, we found that some clients interact strongly with a single, ATP-stabilized HSP90 conformation, only transiently populated during the dynamic HSP90 chaperone cycle, while other clients interact equally with multiple HSP90 conformations. These data suggest different functional requirements among HSP90 clientele that, for some clients, are likely to be ATP-independent. Lastly, the two inhibitors examined, although sharing the same binding site, were differentially able to access distinct HSP90 conformational states.


Subject(s)
Benzoquinones/pharmacology , HSP90 Heat-Shock Proteins/chemistry , Lactams, Macrocyclic/pharmacology , Mutation , Protein Kinase Inhibitors/pharmacology , Triazoles/pharmacology , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Binding Sites , DNA-Binding Proteins/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Kelch-Like ECH-Associated Protein 1 , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptor, ErbB-2/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
9.
Oncotarget ; 6(30): 29963-74, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26298773

ABSTRACT

Elucidating the targets and mechanism of action of natural products is strategically important prior to drug development and assessment of potential clinical applications. In this report, we elucidated the main targets and mechanism of action of the natural product tonantzitlolone (TZL) in clear cell renal cell carcinoma (CCRCC). We identified TZL as a dual PKCα and PKCθ activator in vitro, although in CCRCC cells its activity was mostly PKCθ-dependent. Through activation of PKCθ, TZL induced an insulin resistant phenotype by inhibiting IRS1 and the PI3K/Akt pathway. Simultaneously, TZL activated the heat shock factor 1 (HSF1) transcription factor driving glucose dependency. Thus, similar to the selective PKCθ activator englerin A, TZL induces a metabolic catastrophe in CCRCC, starving cells of glucose while simultaneously increasing their glycolytic dependency.


Subject(s)
DNA-Binding Proteins/metabolism , Diterpenes/pharmacology , Isoenzymes/metabolism , Macrocyclic Compounds/pharmacology , Protein Kinase C/metabolism , Transcription Factors/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Survival/drug effects , DNA-Binding Proteins/genetics , Diterpenes/chemistry , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Glucose/pharmacology , HEK293 Cells , Heat Shock Transcription Factors , Humans , Immunoblotting , Isoenzymes/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Macrocyclic Compounds/chemistry , Molecular Structure , Phosphorylation/drug effects , Protein Kinase C/genetics , Protein Kinase C-theta , RNA Interference , Sesquiterpenes, Guaiane/chemistry , Sesquiterpenes, Guaiane/pharmacology , Transcription Factors/genetics
10.
Radiat Res ; 182(3): 350-62, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25117621

ABSTRACT

Radiation fibrosis of the lung is a late toxicity of thoracic irradiation. Epidermal growth factor (EGF) signaling has previously been implicated in radiation lung injury. We hypothesized that TGF-α, an EGF receptor ligand, plays a key role in radiation-induced fibrosis in lung. Mice deficient in transforming growth factor (TGF-α(-/-)) and control C57Bl/6J (C57-WT) mice were exposed to thoracic irradiation in 5 daily fractions of 6 Gy. Cohorts of mice were followed for survival (n ≥ 5 per group) and tissue collection (n = 3 per strain and time point). Collagen accumulation in irradiated lungs was assessed by Masson's trichrome staining and analysis of hydroxyproline content. Cytokine levels in lung tissue were assessed with ELISA. The effects of TGF-α on pneumocyte and fibroblast proliferation and collagen production were analyzed in vitro. Lysyl oxidase (LOX) expression and activity were measured in vitro and in vivo. Irradiated C57-WT mice had a median survival of 24.4 weeks compared to 48.2 weeks for irradiated TGF-α(-/-) mice (P = 0.001). At 20 weeks after irradiation, hydroxyproline content was markedly increased in C57-WT mice exposed to radiation compared to TGF-α(-/-) mice exposed to radiation or unirradiated C57-WT mice (63.0, 30.5 and 37.6 µg/lung, respectively, P = 0.01). C57-WT mice exposed to radiation had dense foci of subpleural fibrosis at 20 weeks after exposure, whereas the lungs of irradiated TGF-α (-/-) mice were largely devoid of fibrotic foci. Lung tissue concentrations of IL-1ß, IL-4, TNF-α, TGF-ß and EGF at multiple time points after irradiation were similar in C57-WT and TGF-α(-/-) mice. TGF-α in lung tissue of C57-WT mice rose rapidly after irradiation and remained elevated through 20 weeks. TGF-α(-/-) mice had lower basal LOX expression than C57-WT mice. Both LOX expression and LOX activity were increased after irradiation in all mice but to a lesser degree in TGF-α(-/-) mice. Treatment of NIH-3T3 fibroblasts with TGF-α resulted in increases in proliferation, collagen production and LOX activity. These studies identify TGF-α as a critical mediator of radiation-induced lung injury and a novel therapeutic target in this setting. Further, these data implicate TGF-α as a mediator of collagen maturation through a TGF-ß independent activation of lysyl oxidase.


Subject(s)
Lung/radiation effects , Radiation Injuries/etiology , Transforming Growth Factor alpha/physiology , Animals , Collagen/metabolism , Cytokines/biosynthesis , Female , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Protein-Lysine 6-Oxidase/metabolism , Pulmonary Fibrosis/etiology , Radiation Injuries/metabolism , Radiation Injuries/pathology
11.
Stem Cells ; 31(10): 2231-41, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23897677

ABSTRACT

Exposure to ionizing radiation (IR) can result in the development of cutaneous fibrosis, for which few therapeutic options exist. We tested the hypothesis that bone marrow-derived mesenchymal stem cells (BMSC) would favorably alter the progression of IR-induced fibrosis. We found that a systemic infusion of BMSC from syngeneic or allogeneic donors reduced skin contracture, thickening, and collagen deposition in a murine model. Transcriptional profiling with a fibrosis-targeted assay demonstrated increased expression of interleukin-10 (IL-10) and decreased expression of IL-1ß in the irradiated skin of mice 14 days after receiving BMSC. Similarly, immunoassay studies demonstrated durable alteration of these and several additional inflammatory mediators. Immunohistochemical studies revealed a reduction in infiltration of proinflammatory classically activated CD80(+) macrophages and increased numbers of anti-inflammatory regulatory CD163(+) macrophages in irradiated skin of BMSC-treated mice. In vitro coculture experiments confirmed that BMSC induce expression of IL-10 by activated macrophages, suggesting polarization toward a regulatory phenotype. Furthermore, we demonstrated that tumor necrosis factor-receptor 2 (TNF-R2) mediates IL-10 production and transition toward a regulatory phenotype during coculture with BMSC. Taken together, these data demonstrate that systemic infusion of BMSC can durably alter the progression of radiation-induced fibrosis by altering macrophage phenotype and suppressing local inflammation in a TNF-R2-dependent fashion.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Radiation Injuries, Experimental/therapy , Skin Diseases/therapy , Animals , Cells, Cultured , Coculture Techniques , Female , Gene Expression , Inflammation Mediators/physiology , Interleukin-10/genetics , Interleukin-10/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Radiation Injuries, Experimental/immunology , Radiation Injuries, Experimental/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Skin/immunology , Skin/pathology , Skin/radiation effects , Skin Diseases/immunology , Skin Diseases/metabolism
12.
Oncotarget ; 4(7): 1065-74, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23867252

ABSTRACT

Heat shock protein 90 (Hsp90) is an essential molecular chaperone in eukaryotes that facilitates the conformational maturation and function of a diverse protein clientele, including aberrant and/or over-expressed proteins that are involved in cancer growth and survival. A role for Hsp90 in supporting the protein homeostasis of cancer cells has buoyed interest in the utility of Hsp90 inhibitors as anti-cancer drugs. Despite the fact that all clinically evaluated Hsp90 inhibitors target an identical nucleotide-binding pocket in the N domain of the chaperone, the precise determinants that affect drug binding in the cellular environment remain unclear, and it is possible that chemically distinct inhibitors may not share similar binding preferences. Here we demonstrate that two chemically unrelated Hsp90 inhibitors, the benzoquinone ansamycin geldanamycin and the purine analog PU-H71, select for overlapping but not identical subpopulations of total cellular Hsp90, even though both inhibitors bind to an amino terminal nucleotide pocket and prevent N domain dimerization. Our data also suggest that PU-H71 is able to access a broader range of N domain undimerized Hsp90 conformations than is geldanamycin and is less affected by Hsp90 phosphorylation, consistent with its broader and more potent anti-tumor activity. A more complete understanding of the impact of the cellular milieu on small molecule inhibitor binding to Hsp90 should facilitate their more effective use in the clinic.


Subject(s)
Benzodioxoles/metabolism , Benzodioxoles/pharmacology , Benzoquinones/metabolism , Benzoquinones/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Lactams, Macrocyclic/metabolism , Lactams, Macrocyclic/pharmacology , Protein Processing, Post-Translational , Purines/metabolism , Purines/pharmacology , Benzodioxoles/chemistry , Benzoquinones/chemistry , Binding Sites , Cell Line, Tumor , HEK293 Cells , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/genetics , Humans , Lactams, Macrocyclic/chemistry , Phosphorylation , Protein Binding , Protein Conformation , Purines/chemistry , Transfection , Tumor Cells, Cultured
13.
Int J Oncol ; 42(6): 2028-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23588995

ABSTRACT

The inhibition of the Ras/mitogen-activated protein kinase (Ras/MAPK) pathway through the suppression of mutated Ras or MAPK/extracellular signal-regulated kinase 1/2 (MEK1/2) has been shown to sensitize tumor cells to ionizing radiation (IR). The molecular mechanisms of this sensitization however, are not yet fully understood. In this study, we investigated the role of transforming growth factor-α (TGF-α) in the radiosensitizing effects of selumetinib, a selective inhibitor of MEK1/2. The expression of epidermal growth factor receptor (EGFR) ligands was assessed by ELISA in both Ras wild-type and Ras mutant cells that were exposed to radiation with or without selumetinib. The effects of selumetinib on the TGF-α/EGFR signaling cascade in response to radiation were examined by western blot analysis, clonogenic assay and by determing the yield of mitotic catastrophe. The treatment of cells with selumetinib reduced the basal and IR-induced secretion of TGF-α in both Ras wild-type and Ras mutant cell lines in vitro and in vivo. The reduction of TGF-α secretion was accompanied with a reduction in phosphorylated tumor necrosis factor-α converting enzyme (TACE) in the cells treated with selumetinib with or without IR. The treatment of cells with selumetinib with or without IR inhibited the phosphorylation of EGFR and checkpoint kinase 2 (Chk2), and reduced the expression of survivin. Supplementation with exogenous TGF-α partially rescued the selumetinib-treated cells from IR-induced cell death, restored EGFR and Chk2 phosphorylation and increased survivin expression. These data suggest that the inhibition of MEK1/2 with selumetinib may provide a mechanism to sensitize tumor cells to IR in a fashion that prevents the activation of the TGF-α autocrine loop following IR.


Subject(s)
Benzimidazoles/pharmacology , ErbB Receptors/metabolism , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , ADAM Proteins/metabolism , ADAM17 Protein , Animals , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Cell Survival/drug effects , Cell Survival/radiation effects , Humans , Ligands , Mice , Mice, Nude , Mutation , Neoplasms/metabolism , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Radiation Tolerance/drug effects , Radiation, Ionizing , Signal Transduction/drug effects , Transforming Growth Factor alpha/metabolism , Transforming Growth Factor alpha/pharmacology , Xenograft Model Antitumor Assays , ras Proteins/genetics
14.
Cancer Cell ; 23(2): 228-37, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23352416

ABSTRACT

The natural product englerin A (EA) binds to and activates protein kinase C-θ (PKCθ). EA-dependent activation of PKCθ induces an insulin-resistant phenotype, limiting the access of tumor cells to glucose. At the same time, EA causes PKCθ-mediated phosphorylation and activation of the transcription factor heat shock factor 1, an inducer of glucose dependence. By promoting glucose addiction, while simultaneously starving cells of glucose, EA proves to be synthetically lethal to highly glycolytic tumors.


Subject(s)
DNA-Binding Proteins/metabolism , Glucose/metabolism , Insulin Resistance , Insulin/metabolism , Isoenzymes/metabolism , Kidney Neoplasms/drug therapy , Protein Kinase C/metabolism , Sesquiterpenes, Guaiane/pharmacology , Signal Transduction/drug effects , Transcription Factors/metabolism , Animals , Apoptosis , Blotting, Western , Cell Proliferation , Heat Shock Transcription Factors , Humans , Immunoprecipitation , Insulin/chemistry , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Mice , Phosphorylation/drug effects , Protein Kinase C-theta , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Proc Natl Acad Sci U S A ; 109(32): 12866-72, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22753480

ABSTRACT

The "apoptotic ring" is characterized by the phosphorylation of histone H2AX at serine 139 (γ-H2AX) by DNA-dependent protein kinase (DNA-PK). The γ-H2AX apoptotic ring differs from the nuclear foci patterns observed in response to DNA-damaging agents. It contains phosphorylated DNA damage response proteins including activated Chk2, activated ATM, and activated DNA-PK itself but lacks MDC1 and 53BP1, which are required to initiate DNA repair. Because DNA-PK can phosphorylate heat shock protein 90α (HSP90α) in biochemical assays, we investigated whether HSP90α is involved in the apoptotic ring. Here we show that HSP90α is phosphorylated by DNA-PK on threonines 5 and 7 early during apoptosis and that both phosphorylated HSP90α and DNA-PK colocalize in the apoptotic ring. We also show that DNA-PK is a client of HSP90α and that HSP90α is required for full DNA-PK activation, γ-H2AX formation, DNA fragmentation, and apoptotic body formation. In contrast, HSP90 inhibition by geldanamycin markedly enhances TRAIL-induced DNA-PK and H2AX activation. Together, our results reveal that HSP90α is a substrate and chaperone of DNA-PK in the apoptotic response. The response of phosphorylated HSP90α to TRAIL and its localization to the γ-H2AX ring represent epigenetic features of apoptosis that offer insights for studying and monitoring nuclear apoptosis.


Subject(s)
Apoptosis/physiology , DNA-Activated Protein Kinase/metabolism , HSP90 Heat-Shock Proteins/metabolism , Histones/metabolism , Blotting, Western , Cell Line, Tumor , DNA Fragmentation , DNA-Activated Protein Kinase/genetics , Enzyme Activation/physiology , Flow Cytometry , Fluorometry , Humans , In Situ Nick-End Labeling , Microscopy, Fluorescence , Phosphorylation , RNA, Small Interfering/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism
16.
Mol Cell ; 47(3): 434-43, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22727666

ABSTRACT

Many critical protein kinases rely on the Hsp90 chaperone machinery for stability and function. After initially forming a ternary complex with kinase client and the cochaperone p50(Cdc37), Hsp90 proceeds through a cycle of conformational changes facilitated by ATP binding and hydrolysis. Progression through the chaperone cycle requires release of p50(Cdc37) and recruitment of the ATPase activating cochaperone AHA1, but the molecular regulation of this complex process at the cellular level is poorly understood. We demonstrate that a series of tyrosine phosphorylation events, involving both p50(Cdc37) and Hsp90, are minimally sufficient to provide directionality to the chaperone cycle. p50(Cdc37) phosphorylation on Y4 and Y298 disrupts client-p50(Cdc37) association, while Hsp90 phosphorylation on Y197 dissociates p50(Cdc37) from Hsp90. Hsp90 phosphorylation on Y313 promotes recruitment of AHA1, which stimulates Hsp90 ATPase activity, furthering the chaperoning process. Finally, at completion of the chaperone cycle, Hsp90 Y627 phosphorylation induces dissociation of the client and remaining cochaperones.


Subject(s)
Cell Cycle Proteins/metabolism , Chaperonins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Tyrosine/metabolism , Animals , COS Cells , Cell Cycle Proteins/genetics , Chaperonins/genetics , Chlorocebus aethiops , Humans , Mice , Molecular Chaperones/genetics , NIH 3T3 Cells , Phosphorylation/physiology
17.
Biochim Biophys Acta ; 1823(6): 1092-101, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22504172

ABSTRACT

The activator of Hsp90 ATPase, Aha1, is an Hsp90 co-chaperone that has been suggested to act as a general stimulator of Hsp90 function. In this report, we have characterized the interaction of Aha1 with Hsp90 and its co-chaperones in rabbit reticulocyte lysate (RRL) and in HeLa cell extracts. Complexes formed by Aha1 with Hsp90 in RRL were stabilized by molybdate and contained the co-chaperones FKBP52 and p23/Sba1, but lacked HOP/Sti1 and Cdc37. Aha1 complexes isolated from HeLa cell extracts also contained Hsp70 and DNAJA1. Over-expression of Aha1 has been reported to stimulate the activity of v-Src and steroid hormone receptors ectopically expressed in yeast, however, no interaction between Aha1 and nascent v-Src or the progesterone receptor could be detected in RRL. Contrary to expectations, over-expression of Aha1 also inhibited the rate of Hsp90-dependent refolding of denatured luciferase. A number of potential client proteins that specifically associated with Aha1 were identified by liquid chromatography/ tandem mass spectrometry (LC-MS/MS) and verified by Western blotting. The proteins identified suggest that Aha1 may play roles in modulating RNA splicing and DNA repair, in addition to other cellular processes.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Multiprotein Complexes/metabolism , Animals , Cell Extracts , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HeLa Cells , Humans , Luciferases/metabolism , Oncogene Protein pp60(v-src)/metabolism , Protein Binding , Protein Renaturation , Rabbits , Receptors, Progesterone/metabolism
18.
Mol Pharmacol ; 78(6): 1072-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20813864

ABSTRACT

Inactivating mutations of the von Hippel-Lindau (VHL) tumor suppressor gene are associated with inherited VHL syndrome, which is characterized by susceptibility to a variety of neoplasms, including central nervous system hemangioblastoma and clear cell renal cell carcinoma (CCRCC). Mutations in the VHL gene are also found in the majority of sporadic clear cell renal carcinoma, the most common malignant neoplasm of the human kidney. Inactivation of VHL ubiquitin ligase is associated with normoxic stabilization of hypoxia-inducible factor-1α and 2-α (HIF-1α and HIF-2α), transcriptional regulators of tumor angiogenesis, invasion, survival, and glucose utilization. HIF-2α has been particularly implicated in the development of CCRCC. Although several inhibitors of HIF-1α have been described, these drugs typically have a minimal affect on HIF-2α. 786-O is a VHL-deficient CCRCC cell line that constitutively expresses only HIF-2α and is therefore suitable for the screening of novel HIF-2α inhibitors. Using this cell line, we have identified emetine as a specific inhibitor of HIF-2α protein stability and transcriptional activity. Without altering HIF-2α mRNA level, emetine rapidly and dramatically down-regulated HIF-2α protein expression in 786-O cells. HIF-2α down-regulation was accompanied by HIF-2α ubiquitination and was reversed by proteasome inhibition. Emetine-induced HIF-2α down-regulation was confirmed in three additional VHL-renal cancer cell lines, was insensitive to the prolyl hydroxylase inhibitor dimethyloxaloyl glycine, and did not require neural precursor cell expressed developmentally down-regulated-8, suggesting that emetine accesses a previously undescribed cullin-independent proteasome degradation pathway for HIF-2α. These data support the use of emetine or structurally related compounds as useful leads for the identification of novel HIF-2α inhibitors.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/metabolism , Emetine/pharmacology , Kidney Neoplasms/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Cell Line, Tumor , Down-Regulation/drug effects , Humans
19.
Mol Cell ; 37(3): 333-43, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-20159553

ABSTRACT

Saccharomyces WEE1 (Swe1), the only "true" tyrosine kinase in budding yeast, is an Hsp90 client protein. Here we show that Swe1(Wee1) phosphorylates a conserved tyrosine residue (Y24 in yeast Hsp90 and Y38 in human Hsp90alpha) in the N domain of Hsp90. Phosphorylation is cell-cycle associated and modulates the ability of Hsp90 to chaperone a selected clientele, including v-Src and several other kinases. Nonphosphorylatable mutants have normal ATPase activity, support yeast viability, and productively chaperone the Hsp90 client glucocorticoid receptor. Deletion of SWE1 in yeast increases Hsp90 binding to its inhibitor geldanamycin, and pharmacologic inhibition/silencing of Wee1 sensitizes cancer cells to Hsp90 inhibitor-induced apoptosis. These findings demonstrate that Hsp90 chaperoning of distinct client proteins is differentially regulated by specific posttranslational modification of a unique subcellular pool of the chaperone, and they provide a strategy to increase the cellular potency of Hsp90 inhibitors.


Subject(s)
Cell Cycle Proteins/physiology , HSP90 Heat-Shock Proteins/metabolism , Protein-Tyrosine Kinases/physiology , Saccharomyces cerevisiae Proteins/physiology , Tyrosine/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Dimerization , HSP90 Heat-Shock Proteins/physiology , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , RNA Interference , Saccharomyces cerevisiae Proteins/antagonists & inhibitors , Saccharomyces cerevisiae Proteins/genetics , Ubiquitination
20.
Nat Struct Mol Biol ; 16(11): 1141-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19838189

ABSTRACT

Heat shock protein 90 (Hsp90) is an essential molecular chaperone in eukaryotes, as it regulates diverse signal transduction nodes that integrate numerous environmental cues to maintain cellular homeostasis. Hsp90 also is secreted from normal and transformed cells and regulates cell motility. Here, we have identified a conserved hydrophobic motif in a beta-strand at the boundary between the N domain and charged linker of Hsp90, whose mutation not only abrogated Hsp90 secretion but also inhibited its function. These Hsp90 mutants lacked chaperone activity in vitro and failed to support yeast viability. Notably, truncation of the charged linker reduced solvent accessibility of this beta-strand and restored chaperone activity to these mutants. These data underscore the importance of beta-strand 8 for Hsp90 function and demonstrate that the functional consequences of weakened hydrophobic contacts in this region are reversed by charged-linker truncation.


Subject(s)
HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/metabolism , Hydrophobic and Hydrophilic Interactions , Amino Acid Motifs/genetics , Amino Acid Motifs/physiology , Animals , COS Cells , Chlorocebus aethiops , Enzyme-Linked Immunosorbent Assay , Fluorescence Resonance Energy Transfer , HSP90 Heat-Shock Proteins/genetics , Humans , Immunoblotting , Immunoprecipitation , Mutation , Protein Binding/genetics , Protein Structure, Tertiary , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL