Abstract

Simple SummaryOsteolytic bone lesions represent an important clinical feature of multiple myeloma (MM). MM cells metabolize very high amounts of glutamine (Gln) and significantly lower Gln in the bone marrow. In this contribution we demonstrate that MM-dependent Gln depletion impairs the differentiation of bone marrow mesenchymal stromal cells into osteoblasts, the cells that form new bone tissue. We also found that osteoblast differentiation is associated with increased expression of glutaminase, the main enzyme that metabolizes Gln, SNAT2, a transporter able to accumulate Gln into the cells, and asparagine synthetase, the enzyme that uses Gln to obtain asparagine (Asn). Asn rescued osteoblast differentiation of Gln-starved mesenchymal stromal cells. These results demonstrate that MM cells impair osteoblast differentiation, hindering mesenchymal Asn synthesis through Gln depletion. Besides providing a metabolic mechanism underlying osteolytic lesions in MM, these results suggest that Asn supplementation may prevent bone disease in MM patients.Multiple myeloma (MM) cells consume huge amounts of glutamine and, as a consequence, the amino acid concentration is lower-than-normal in the bone marrow (BM) of MM patients. Here we show that MM-dependent glutamine depletion induces glutamine synthetase in stromal cells, as demonstrated in BM biopsies of MM patients, and reproduced in vitro by co-culturing human mesenchymal stromal cells (MSCs) with MM cells. Moreover, glutamine depletion hinders osteoblast differentiation of MSCs, which is also severely blunted by the spent, low-glutamine medium of MM cells, and rescued by glutamine restitution. Glutaminase and the concentrative glutamine transporter SNAT2 are induced during osteoblastogenesis in vivo and in vitro, and both needed for MSCs differentiation, pointing to enhanced the requirement for the amino acid. Osteoblastogenesis also triggers the induction of glutamine-dependent asparagine synthetase (ASNS), and, among non-essential amino acids, asparagine rescues differentiation of glutamine-starved MSCs, by restoring the transcriptional profiles of differentiating MSCs altered by glutamine starvation. Thus, reduced asparagine availability provides a mechanistic link between MM-dependent Gln depletion in BM and impairment of osteoblast differentiation. Inhibition of Gln metabolism in MM cells and supplementation of asparagine to stromal cells may, therefore, constitute novel approaches to prevent osteolytic lesions in MM.

Highlights

  • Metabolic alterations of cancer cells, aimed at sustaining their uncontrolled growth, can alter the biochemical features of the tumor microenvironment and affect the metabolic behavior of other cell populations

  • MM cells metabolize very high amounts of glutamine (Gln) and significantly lower Gln in the bone marrow. In this contribution we demonstrate that MM-dependent Gln depletion impairs the differentiation of bone marrow mesenchymal stromal cells into osteoblasts, the cells that form new bone tissue

  • Co-culture with any of the four human myeloma cell lines (HMCLs) tested caused a marked induction of glutamine synthetase (GS) in hTERT-mesenchymal stromal cells (MSCs) (Figure 1D,E)

Read more

Summary

Introduction

Metabolic alterations of cancer cells, aimed at sustaining their uncontrolled growth, can alter the biochemical features of the tumor microenvironment and affect the metabolic behavior of other cell populations. In murine models, bone mass formation is sustained by Gln metabolism [16], the inhibition of which decreases OB differentiation of human mesenchymal stromal cells (MSCs) [17] Overall, these data suggest a possible involvement of defects of Gln metabolism in MM-induced osteolysis, but little information is available from the studies on which Gln-dependent pathway may be important for OB differentiation. Gln is both a direct precursor of asparagine (Asn), through the operation of asparagine synthetase (ASNS), and of glutamate (Glu), through GLS; in turn, Glu provides carbon moieties for the synthesis of many other non-essential amino acids and of glutathione Based on this literature evidence, we investigate how Gln shortage may be involved in the pathogenesis of OB suppression in MM and identify Asn availability as a link between Gln and the osteogenic differentiation process

Results
Gln Deprivation Hinders the Expression of OB Markers in MSCs
OB Differentiation Is Associated with the Induction of the SNAT2 Transporter
Asn Content Increases during OB Differentiation of MSCs
High by HMCLs causes
Gln depletion
Amino Acid Dependence of OB Differentiation of Primary BM MSCs
Discussions
Patients
Primary MSCs
Co-Culture Experiments
OB Differentiation Experiments
ASNS Knockout in hTERT-MSCs
AminoAacid Uptake
4.11. Gene Expression Profiles Analysis
Conclusions
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