Abstract

The objective of this study was to examine the combined effect of Interferon-gamma (IFN-γ) and Tumor Necrosis factor-alpha (TNF-α) on cytotoxicity and expression of prostate apoptosis response-4 (Par-4) and Par-4 interacting proteins B-cell lymphoma (Bcl-2), nuclear factor kappa-light-chain-enhancer of activated B cells/p65 subunit (NF-κB/p65), Ak mouse strain thymoma (Akt) in human neuroblastoma (NB) cells. Materials and methods included human neuroblastoma cell lines-SK-N-MC, SK-N-SH, and SH-SY5Y, which were treated with IFN-γ and TNF-α individually, or in combination, and were assessed for viability by tetrazolium (MTT) assay. Apoptosis was monitored by hypodiploid population (by flow cytometry), DNA fragmentation, Poly (ADP-ribose) polymerase (PARP) cleavage, and caspase-8 activity. Transcript level of Par-4 was measured by RT-PCR. Protein levels of Par-4 and suppressor of cytokine signaling 3 (SOCS-3) were assessed by immunoblotting. Cellular localization of Par-4 and p65 was examined by immunofluorescence. Unbiased transcript analysis for IFN-γ, TNF-α, and Par-4 were analyzed from three independent clinical datasets from neuroblastoma patients. In terms of results, SK-N-MC cells treated with a combination of, but not individually with, IFN-γ and TNF-α induced apoptosis characterized by hypodiploidy, DNA fragmentation, PARP cleavage, and increased caspase-8 activity. Apoptosis was associated with up-regulation of Par-4 mRNA and protein expression. Immunofluorescence studies revealed that Par-4 was localized exclusively in cytoplasm in SK-N-MC cells cultured for 24 h. but showed nuclear localization at 48 h. Treatment with IFN-γ and TNF-α together enhanced the intensity of nuclear Par-4. In gene expression, data from human neuroblastoma patients, levels of IFN-γ, and TNF-α have strong synergy with Par-4 expression and provide good survival advantage. The findings also demonstrated that apoptosis was associated with reduced level of pro-survival proteins–Bcl-2 and Akt and NF-κB/p65. Furthermore, the apoptotic effect induced by IFN-γ-induced Signal Transducer and Activator of Transcription-1(STAT-1), and could be due to down-regulation of suppressor of cytokine signaling-3 (SOCS3). The study concludes that a combinatorial approach using IFN-γ and TNF-α can be explored to maximize the effect in chemotherapy in neuroblastoma, and implies a role for Par-4 in the process.

Highlights

  • Neuroblastoma (NB) is the most common extracranial solid tumor in children and accounts for 8 to 10% of pediatric tumors [1]

  • Our results demonstrate that IFN-γ and TNF-α up-regulate pro-apoptotic apoptotic protein-prostate apoptosis response-4 (Par-4) with a concomitant decrease in the pro-survivaland protein-Bcl-2, protein-Par-4 with a concomitant decrease in the expression of expression pro-survivalofprotein-Bcl-2, activation and activation of

  • We show that IFN-γ and TNF-α together down-regulated suppressor of cytokine signaling-3 (SOCS3)

Read more

Summary

Introduction

Neuroblastoma (NB) is the most common extracranial solid tumor in children and accounts for 8 to 10% of pediatric tumors [1]. Neuroblastoma is characterized by high rate of spontaneous regression suggesting activation of an apoptotic/differentiation program; advanced stages are associated with poor prognosis and resistance to chemotherapy [2]. Despite advancements in conventional therapies and the development of a recent line of treatments that include blood stem cell transplantation, differentiation therapy with retinoic acid, and passive immunotherapy with anti-disialoganglioside (GD2) antibodies, there is no significant improvement in the survival rate [3]. Tumor necrosis factor-alpha (TNF-α) is a pleiotropic cytokine with multifaceted functions. In vitro and in vivo studies have demonstrated strong anti-tumor activity of TNF-α in different types of cancers [4,5,6]. Interferon-gamma (IFN-γ) is a pro-inflammatory cytokine belonging to the type II class of interferons. The biological activity of IFN-γ as an anticancer agent is attributed to inhibition of proliferation, induction, and modulation of gene expression mediated by activation of Janus

Objectives
Methods
Conclusion
Full Text
Published version (Free)

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