Abstract

Histone deacetylase inhibitors (HDACis) are a potent class of tumor-suppressive agents traditionally believed to exert their effects through loosening tightly-wound chromatin resulting in de-inhibition of various tumor suppressive genes. Recent literature however has shown altered intratumoral hypoxia signaling with HDACi administration not attributable to changes in chromatin structure. We sought to determine the precise mechanism of HDACi-mediated hypoxia signaling attenuation using vorinostat (SAHA), an FDA-approved class I/IIb/IV HDACi. Through an in-vitro and in-vivo approach utilizing cell lines for hepatocellular carcinoma (HCC), osteosarcoma (OS), and glioblastoma (GBM), we demonstrate that SAHA potently inhibits HIF-a nuclear translocation via direct acetylation of its associated chaperone, heat shock protein 90 (Hsp90). In the presence of SAHA we found elevated levels of acetyl-Hsp90, decreased interaction between acetyl-Hsp90 and HIF-a, decreased nuclear/cytoplasmic HIF-α expression, absent HIF-α association with its nuclear karyopharyin Importin, and markedly decreased HIF-a transcriptional activity. These changes were associated with downregulation of downstream hypoxia molecules such as endothelin 1, erythropoietin, glucose transporter 1, and vascular endothelial growth factor. Findings were replicated in an in-vivo Hep3B HRE-Luc expressing xenograft, and were associated with significant decreases in xenograft tumor size. Altogether, this study highlights a novel mechanism of action of an important class of chemotherapeutic.

Highlights

  • Over the last decade the field of targeted chemotherapy has yielded significant improvements in the prognosis of patients with incurable malignancies

  • We evaluated the effect of SAHA on HIF1/2α-associated transcriptional activation through a luciferase assay based on Hep3B cells carrying a stably www.impactjournals.com/oncotarget transfected Hypoxia Responsive Element (HRE) luciferase reporter (Hep3B HRE-Luc)

  • Effects of SAHA on hypoxia related gene expression in Hep3B cells exposed to SAHA for 16 hr under conditions of 21% or 1% O2 analyzed by qRT-PCR, showing significant suppression of endothelin 1 (EDN1), EPO, glucose transporter 1 (GLUT1), and vascular endothelial growth factor (VEGFA) in response to both 0.5 and 1 μM SAHA with no significant change in HIF1A or HIF2A expression

Read more

Summary

Introduction

Over the last decade the field of targeted chemotherapy has yielded significant improvements in the prognosis of patients with incurable malignancies. There still exists a need for an effective therapy that interferes with components of the hypoxia signaling pathway. Elevated intratumoral HIF levels carry a negative prognosis, and are an independent risk factor for decreased patient survival [8,9,10]. To this end, a variety of therapies inhibiting HIF and/or components of the hypoxia signaling cascade have been identified [11, 12]. Agents that only target specific components of the hypoxia signaling pathway generally fail to produce an enduring clinical response [13,14,15,16]. The vast majority of HIF inhibitors utilized for pre-clinical and clinical investigation are non-specific for the hypoxia pathway [11]

Methods
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.