Introduction:

Multiple myeloma (MM) is a uniformly fatal disorder of B cells characterized by the clonal expansion of plasma cells in the bone marrow. The treatment of MM patients has been dramatically changed by new agents such as proteasome inhibitors and immunomodulatory drugs, however, many patients will relapse even if new agents provide therapeutic advantages. Therefore, a new strategy is still needed to increase MM patient survival. Metabolic reprogramming is recognized as one of the hallmarks of cancer cells. Glutamine is the most abundant circulating amino acid in blood, glutamine metabolism through glutaminolysis may be associated with myeloma cell maintenance and survival.

Materials and Methods:

In this study, we investigated whether glutaminolysis was involved the proliferation in myeloma cells. We also investigated whether glutaminase (GLS) inhibitor, CB-839 could suppress myeloma cells and enhance the sensitivity of myeloma cells to histone deacetylase (HDAC) inhibition.

Results:

We first investigated the relationship between glutamine transporter or GLS gene expression and MM patients by microarray gene expression data from the online Gene Expression Omnibus (GEO). Glutamine transporter genes such as SLC38A1 and SLC1A5 were increased in myeloma and plasma cell leukemia cells (GSE13591). In contrast, GLS1 expression was not changed. We next investigated the glutaminolysis in myeloma cells. Deprivation of glutamine in culture medium revealed that cellular growth inhibition and cell cycle arrest at G0/G1 phase. Gene expression of AURKA (aurora kinase A), AURKB (aurora kinase B), HSP90AA1 (Heat Shock Protein 90 Alpha Family Class A Member 1) and CCNB1 (cyclin B1) were reduced from the public microarray datasets (GSE59931) and protein expressions were also reduced by immunoblot analysis. We next evaluated the effect of GLS inhibitor, CB-839. 72 h treatment of MM cells were inhibited by CB-839 in a dose dependent manner. Cellular cytotoxicity was also increased. Glutamine is converted by GLS into glutamate and alpha-ketoglutarate (α-KG), and related nicotinamide adenine dinucleotide phosphate (NADP) production. Intracellular α-KG and NADPH were reduced by CB-839. As metabolites are the substrates used to generate chromatin modification including acetylation of histone, we investigated HDAC inhibitor, panobinostat in myeloma cells. 72 h treatment of MM cells were inhibited by panobinostat and histone acetylation was increased. Combined treatment with panobinostat and CB-839 caused more cytotoxicity than each drug alone. Panobinostat and CB-839 also inhibited bortezomib resistant cells. Caspase 3/7 activity and cellular cytotoxicity were also increased. Proteasomal activity was reduced. Adenosine triphosphate (ATP) is the most important source of energy for intracellular reactions. Intracellular ATP levels drastically decreased. Because mitochondria generate ATP and participate in signal transduction and cellular pathology and cell death. The quantitative analysis of JC-1 stained cells changed mitochondrial membrane potential in cell death, which were induced by panobinostat and CB-839 on myeloma cells. Immunoblot analysis revealed that protein expression of aurora kinase A, aurora kinase B, HSP90 and cyclin B1 were reduced, and cleaved caspase 3 and γ- H2AX were increased by panobinostat and CB-839 treatment. GLS shRNA transfectant cells were inhibited cellular proliferation and sub-G1 phase was increased by cell cycle analysis. GLS shRNA transfectant cells were increased the sensitivity of panobinostat compared to control cells.

Conclusion:

The glutaminolysis is involved myeloma cell proliferation and GLS inhibitor is effective to myeloma cells and enhance cytotoxic effects of HDAC inhibitors. We also provide the promising clinical relevance as a candidate drug for treatment of myeloma patients.

Disclosures

No relevant conflicts of interest to declare.

Sign in via your Institution