Multiple myeloma (MM) remains incurable for the vast majority of patients due to emergence of drug resistant clones and mutations inducing drug resistant relapses. This is despite the fact that new therapies have greatly improved progression-free and overall survival for patients with standard risk myeloma. We recently found that the transcriptional repressor GFI1 is increased in bone marrow stromal cells of MM patients (MM-BMSC) where it causes prolonged suppression of osteoblast differentiation, and in CD138+ cells from MM patients, where GFI1 levels significantly correlate with disease progression. We also found that GFI1 overexpression (o/e) enhances MM cell growth and partially confers resistance to proteasome inhibitors in vitro as well as enhances tumor growth and osteoclastogenesis in vivo.

Although the mechanisms responsible for these GFI1 effects in p53wt MM cells were p53-dependent, we found that GFI1 is also essential for MM cell survival regardless of their p53 status. The p53-independent mechanisms responsible for Gfi1 effects on MM cells growth and survival of are unknown. Sphingolipids are bioactive lipids that can control MM cell growth and survival. The balance between the levels of Sphingosine-1-phosphate (S1P) and its metabolic precursors ceramide (Cer) and sphingosine (SPH) form a rheostat that determines whether a cell proliferates or dies. We hypothesized that GFI1 represses SGPP1, the enzyme responsible for degrading S1P via salvage and recycling of sphingosine into long-chain ceramides. This repression changes the intracellular sphingolipid profile (Cer/S1P/SPH ratio) to maintain c-Myc upregulation in a protein phosphatase 2 (PP2A)-dependent manner, thus promoting growth and survival of MM cells.

To test this hypothesis we measured S1P, SPH and Cer levels by mass spectrometry (LC-MS/MS). LC-MS/MS evaluation showed that bone marrow plasma of MM patients has significant higher levels of S1P when compared to normal donors. Moreover, intracellular S1P levels of MM.1S GFI1 o/e cells were also significantly higher as compared to those of MM.1S empty vector controls. Knock-down (KD) of Gfi1 in MM.1S cells strikingly increased SGPP1 and decreased SphK1 (the enzyme which catalyzes S1P production) mRNA levels, while GFI1 o/e cells had the opposite effect. We found that CD138+ cells isolated from MM patients expressed elevated levels of SphK1 mRNA compared to MGUS patients, and that SphK1 protein levels directly correlate with GFI1 levels in MM patient CD138+ cells and cell lines (r= 0.527). We also detected an indirect correlation (r= -0.961) between GFI1 and SGPP1 mRNA levels in five different MM cell lines. These results indicate a GFI1-dependent imbalance of the enzymes regulating S1P production.

Further, KD GFI1 and SphK1 inhibition (5 μM SK1I) had a profound inhibitory effect on c-Myc protein levels and induced caspase 3 activation as detected by Western blotting, while GFI1 o/e cells had significant higher levels of c-Myc and were more resistant to SK1I treatment. Exogenous ceramide (10 μM Cer 16:0) treatment or SphK1 inhibition (5 μM SK1I), both treatments known to trigger intracellular ceramide production, significantly inhibited MM cell viability (measured by AlamarBlue), regardless of their p53 status (MM1.S p53 +/+ and KMS-11 p53 -/-). This inhibition of MM viability was GFI1-dependent, as GFI1 o/e cells were significantly more resistant to ceramide-induced cell death, which was PP2A dependent, as PP2A inhibition with okadaic acid (OA) restored it. MM.1S cells with KD of GFI1 exhibited significantly higher PP2A activity then control cells, supporting our observation that c-Myc modulation by GFI1 is PP2A-dependent. c-Myc protein levels were significantly decreased in MM.1S control cells treated with ceramide and rescued by OA pre-treatment; thus mimicking the effects of changing GFI1 levels and I2PP2A (the PP2A endogenous inhibitor) and confirms that PP2A mediates the effects of GFI1 on c-Myc.

Taken together, our results show that GFI1 acts as a key regulator of MM growth and survival, at least partially through modulation of SGPP1. Therefore, targeting lipid metabolism to modulate the levels of specific bioactive lipid components that can modify cancer cell fate may provide a new and attractive therapeutic approach for MM.

Disclosures

Roodman:Amgen: Membership on an entity's Board of Directors or advisory committees.

Author notes

*

Asterisk with author names denotes non-ASH members.

Sign in via your Institution