Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Carcinogenesis ; 41(10): 1421-1431, 2020 10 15.
Article in English | MEDLINE | ID: mdl-31917403

ABSTRACT

The repurposing of existing drugs has emerged as an attractive additional strategy to the development of novel compounds in the fight against cancerous diseases. Inhibition of phosphodiesterase 5 (PDE5) has been claimed as a potential approach to target various cancer subtypes in recent years. However, data on the treatment of tumors with PDE5 inhibitors as well as the underlying mechanisms are as yet very scarce. Here, we report that treatment of tumor cells with low concentrations of Sildenafil was associated with decreased cancer cell proliferation and augmented apoptosis in vitro and resulted in impaired tumor growth in vivo. Notably, incubation of cancer cells with Sildenafil was associated with altered expression of HSP90 chaperone followed by degradation of protein kinase D2, a client protein previously reported to be involved in tumor growth. Furthermore, the involvement of low doses of PU-H71, an HSP90 inhibitor currently under clinical evaluation, in combination with low concentrations of Sildenafil, synergistically and negatively impacted on the viability of cancer cells in vivo. Taken together, our study suggests that repurposing of already approved drugs, alone or in combination with oncology-dedicated compounds, may represent a novel cancer therapeutic strategy.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Neoplasms/pathology , Phosphodiesterase 5 Inhibitors/pharmacology , Proteolysis , Sildenafil Citrate/pharmacology , TRPP Cation Channels/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Humans , Neoplasms/metabolism
2.
Mol Cancer ; 15: 3, 2016 Jan 06.
Article in English | MEDLINE | ID: mdl-26739387

ABSTRACT

BACKGROUND: Initially identified as a molecule that regulates the final step of glycolysis, the M2 isoform of pyruvate kinase (PKM2) was recently reported to have a central role in the metabolic reprogramming of cancer cells as well as participating in cell cycle progression and gene transcription. Despite intensive efforts, the intricate molecular mechanisms through which PKM2 regulates tumor progression remain elusive. METHODS: The proliferation and apoptosis of various pancreatic cancer cells using lentiviral-mediated PKM2 abrogation were assessed in vitro via Western blot and flow cytometric assay while the in vivo experiments involved tumor xenograft on chicken chorionallantoic membranes and immunohistochemistry on human tissue specimens. In order to decipher the molecular mechanism of HIF-1α and p65/RelA regulation by PKM2 in cancer cells cultivated in hypoxic atmosphere or normoxia we involved various biochemical assays such as Western blotting, immunoprecipitation, reporter gene assay and ELISA. RESULTS: Strong expression of PKM2 was observed in 68 % of human pancreatic adenocarcinoma specimens and almost all analyzed pancreatic cancer cell lines. Abrogation of PKM2 resulted in impaired proliferation and augmented apoptosis in vitro as well as impaired tumor growth and decreased blood vessel formation in vivo. Furthermore, deletion of PKM2 negatively impacted hypoxia-induced HIF-1α accumulation and promoter activity ultimately resulting in impaired secretion of VEGF. CONCLUSIONS: Our study suggests that in hypoxic pancreatic tumors PKM2 interferes both with NF-κB/p65 and HIF-1α activation that ultimately triggers VEGF-A secretion and subsequent blood vessel formation.


Subject(s)
Carrier Proteins/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Membrane Proteins/metabolism , NF-kappa B/metabolism , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/metabolism , Thyroid Hormones/metabolism , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Cell Hypoxia , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Proliferation , Cell Survival , Chickens , Female , Gene Knockdown Techniques , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Middle Aged , Models, Biological , Neovascularization, Pathologic/genetics , Pancreatic Neoplasms/genetics , Protein Binding , Protein Transport , Signal Transduction/genetics , Transcription Factor RelA/metabolism , Transcription, Genetic , Vascular Endothelial Growth Factor A/metabolism , Thyroid Hormone-Binding Proteins
3.
Sci Rep ; 9(1): 4112, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30858416

ABSTRACT

HSP90 is a ubiquitously expressed molecular chaperone implicated in the correct folding and maturation of a plethora of proteins including protein kinases and transcription factors. While disruption of chaperone activity was associated with augmented cancer cell death and decreased tumor growth both in vitro and in vivo, the regulation of HSP90 is not clearly understood. Here we report that treatment of cancer cells with cold physical plasma, an emerging and less aggressive tumor therapy, resulted in ROS generation which subsequently triggered the cleavage of HSP90. Notably, cleavage of HSP90 was followed by the degradation of PKD2, a crucial regulator of tumor growth and angiogenesis. Pre-sensitization of cancer cells with subliminal doses of PU-H71, an HSP90 inhibitor currently under clinical evaluation, followed by treatment with cold-plasma, synergistically and negatively impacted on the viability of cancer cells. Taken together, cold-plasma can be used in conjunction with pharmacologic treatment in order to target the expression and activity of HSP90 and the downstream client proteins implicated in various cancer cell capabilities.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Neoplasms/pathology , Plasma Gases/pharmacology , Reactive Oxygen Species/metabolism , Argon/pharmacology , Atmospheric Pressure , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Models, Biological , Protein Kinase D2 , Protein Kinases/metabolism , Proteolysis/drug effects
4.
Oncotarget ; 8(44): 77474-77488, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-29100402

ABSTRACT

Lately, the HSP90 client serine/threonine kinase STK33 emerged to be required by cancer cells for their viability and proliferation. However, its mechanistic contribution to carcinogenesis is not clearly understood. Here we report that elevated STK33 expression correlates with advanced stages of human pancreatic and colorectal carcinomas. Impaired proliferation and augmented apoptosis associated with genetic abrogation of STK33 were paralleled by decreased vascularization in tumor xenografts. In line with this, ectopic STK33 not only promoted tumor growth after pharmacologic inhibition of HSP90 using structurally divergent small molecules currently in clinical development, but also restored blood vessel formation in vivo. Mechanistic studies demonstrated that HSP90-stabilized STK33 interacts with and regulates hypoxia-driven accumulation and activation of HIF-1α as well as secretion of VEGF-A in hypoxic cancer cells. In addition, our study reveals a putative cooperation between STK33 and other HSP90 client protein kinases involved in molecular and cellular events through which cancer cells ensure their survival by securing the oxygen and nutrient supply. Altogether, our findings indicate that STK33 interferes with signals from hypoxia and HSP90 to promote tumor angiogenesis and tumor growth.

5.
Cancer Res ; 74(23): 7125-36, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25297628

ABSTRACT

The kinase PRKD2 (protein kinase D) is a crucial regulator of tumor cell-endothelial cell communication in gastrointestinal tumors and glioblastomas, but its mechanistic contributions to malignant development are not understood. Here, we report that the oncogenic chaperone HSP90 binds to and stabilizes PRKD2 in human cancer cells. Pharmacologic inhibition of HSP90 with structurally divergent small molecules currently in clinical development triggered proteasome-dependent degradation of PRKD2, augmenting apoptosis in human cancer cells of various tissue origins. Conversely, ectopic expression of PRKD2 protected cancer cells from the apoptotic effects of HSP90 abrogation, restoring blood vessel formation in two preclinical models of solid tumors. Mechanistic studies revealed that PRKD2 is essential for hypoxia-induced accumulation of hypoxia-inducible factor-1α (HIF1α) and activation of NF-κB in tumor cells. Notably, ectopic expression of PRKD2 was able to partially restore HIF1α and secreted VEGF-A levels in hypoxic cancer cells treated with HSP90 inhibitors. Taken together, our findings indicate that signals from hypoxia and HSP90 pathways are interconnected and funneled by PRKD2 into the NF-κB/VEGF-A signaling axis to promote tumor angiogenesis and tumor growth.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Neovascularization, Pathologic/metabolism , Protein Kinase C/metabolism , Animals , Apoptosis/physiology , Cell Hypoxia/physiology , Cell Line, Tumor , Female , HCT116 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Nude , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL