scholarly journals Stromal Support of Metabolic Function through Mitochondrial Transfer in Multiple Myeloma

2019 ◽  
Vol 79 (9) ◽  
pp. 2102-2103 ◽  
Author(s):  
Lawrence H. Boise ◽  
Mala Shanmugam
Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3153-3153
Author(s):  
Christopher Richard Marlein ◽  
Rebecca H Horton ◽  
Rachel E Piddock ◽  
Jayna J Mistry ◽  
Charlotte Hellmich ◽  
...  

Abstract Background Multiple myeloma (MM) is malignancy highly reliant on its microenvironment. In this study, we investigated whether mitochondrial transfer occurred between bone marrow stromal cells (BMSC) and malignant plasma cells. We then used our observations as a platform to investigate the mechanisms controlling pro-tumoral mitochondrial transfer with a view to identifying druggable targets. Methods Primary MM cells were obtained from patients' bone marrow after informed consent and under approval from the United Kingdom Health Research Authority. Animal experiments were conducted under approvals from the UK Home Office and the University of East Anglia Animal Welfare and Ethics Review Board. Primary BMSC were also obtained from patient bone marrow, using adherence and characterised using flow cytometry. Mitochondrial transfer was assessed using two methods; a MitoTracker Green based staining of the BMSC (in-vitro), rLV.EF1.AcGFP-Mem9 labelling of the MM plasma membrane with MitoTracker CMXRos staining of the BMSC (in-vitro) and an in vivo MM NSG xenograft model. CD38 expression on MM cells was tested after ATRA treatment, using RT-qPCR and flow cytometry. Mitochondrial transfer levels were assessed when CD38 was over expressed using ATRA or inhibited using lentivirus targeted shRNA. Results We report that mitochondria are transferred from BMSC to MM cells. First, we cultured MM cells on MitoTracker Green labelled BMSC and found increased MitoTracker Green fluorescence in the MM cells. We then transduced MM with rLV.EF1.AcGFP-Mem9 lentivirus and stained BMSC with MitoTracker CMXRos and used wide field microscopy to show MM derived tunnelling nanotubles (TNT) formed between MM cells and BMSC, with red mitochondria located within the GFP-tagged TNT. Next, we engrafted the MM cell lines MM1S and U266 into NSG mouse, after isolation we detected the presence of mouse mitochondrial DNA in the purified MM population. Together, these data show that mitochondria are transferred from BMSC to MM cells. We next analysed OXPHOS levels in MM cells grown on BMSC, using the seahorse extracellular flux assay. We found that the MM cells had increased levels of OXPHOS after culture with BMSC, which was also the case for MM cell lines analysed after isolation from NSG mice, showing the micro-environment of MM can alter the metabolism of the malignant cell. To examine whether the mitochondrial transfer process was controlled by CD38, we knocked down CD38 in MM cells using lentiviral targeted shRNA. We found reduced levels of mitochondrial transfer in CD38KD MM cells, with a consequent reduction of OXPHOS in the malignant cells. Finally, as ATRA has previously been shown to increase CD38 expression in AML, we next quantified CD38 mRNA and surface glycoprotein level on malignant plasma cells with and without ATRA treatment. We found ATRA increased CD38 expression at the mRNA and protein levels and this resulted in an increase in mitochondrial transfer from BMSC to MM cells. Conclusion Here we show that CD38 mediated mitochondrial transfer in the MM micro-environment forms part of the malignant phenotype of multiple myeloma. This finding develops our understanding of the mechanisms which underpin the efficacy of CD38 directed therapy in MM. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 3950-3950
Author(s):  
Shannon M Matulis ◽  
Jonathan L. Kaufman ◽  
Sagar Lonial ◽  
Lawrence H. Boise

Abstract Abstract 3950 It is well established that the bone marrow microenvironment contributes to the growth and survival of multiple myeloma plasma cells, however the molecular mechanisms that control these changes within the myeloma cell have not been completely elucidated. We have recently demonstrated that myeloma cell lines can be dependent on the anti-apoptotic protein MCL-1 or can be co-dependent on MCL-1 and Bcl-2/xL. The primary distinction between MCL-1 dependence and co-dependence is the distribution of the pro-apoptotic BH3-only protein Bim. In MCL-1-dependent lines, Bim is primarily associated with MCL-1. In contrast in co-dependent lines Bim is either predominantly associated with Bcl-2/xL or when it is released from Bcl-2/xL it can not bind to MCL-1 because of the presence of the MCL-1 inhibitor, Noxa. This renders these cells sensitive to the Bcl-2/xL inhibitor ABT-737. We have confirmed these findings in freshly isolated myeloma cells and demonstrated that among the 4 patient samples tested, Bim was associated with both MCL-1 and Bcl-xL and the cells were sensitive to ABT-737. This suggests that co-dependence on MCL-1 and Bcl-xL may be a common phenomena in myeloma. We now demonstrate that in about 50% of patient samples tested, CD138 column-purified MM cells are more sensitive to ABT-737 than cells that were not purified following ficoll separation of the bone marrow aspirate. This suggests that stromal components in the marrow alter the dependence on Bcl-2 proteins resulting in a cell that is more MCL-1 dependent when in the presence of stroma. To directly test this possibility we determined the effect of co-culturing the co-dependent line, MM.1s with the human stromal line Hs-5 on ABT-737 (0.6 mM) sensitivity and Bim binding. MM1.s cells were incubated with Hs-5 cells for 30 min prior to addition of drug or were cultured in medium containing 50% conditioned medium (48 h growth) from Hs-5 cells to determine if any effects observed were due to a soluble factor. Following a 24 h culture, cells were subjected to CD138 column purification and apoptosis analyzed by Annexin V-FITC/PI flow cytometry. While mono-cultured MM1.s were very sensitive to ABT-737 (88.5% +/− 3.6% control apoptosis, mean +/− SE), the addition of Hs-5 cells or conditioned medium had a profound effect on cell survival resulting in significant resistance to Bcl-2/xL inhibition (23% +/− 4.4% and 24.5% +/− 2.3% control apoptosis respectively). To determine the molecular basis in this shift from co-dependence to MCL-1 dependence, western blot and co-immunoprecipitation analyses were performed. While neither co-culture nor conditioned medium had any significant effect on the expression of levels of Bim, MCL-1 or Bcl-xL, both conditions resulted in a shift of Bim binding from being equally split amongst MCL-1 and Bcl-xL to a binding pattern where Bim was preferentially bound to MCL-1. This is consistent with loss of sensitivity to ABT-737 and a shift to MCL-1 dependence. We next tested the effects of Hs5 co-culture and conditioned medium on other inducers of cell death. We focused on bortezomib (5 nM) and arsenic trioxide (2 mM) as both can induce the MCL-1 inhibitor Noxa, therefore sensitivity would not be expected to be significantly influenced by a change from co-dependence to MCL-1 dependence. Consistent with this possibility we found that bortezomib induced cell death was only modestly affected by co-culture (79% +/− 2.3% vs. 66.5% +/− 1.9% control apoptosis) and no protection was observed with Hs5 conditioned medium. Similar results were observed with arsenic trioxide (monoculture – 31% +/− 2.3%, Hs5 co-culture −15% +/− 1.0%, conditioned medium – 30% +/− 4.2% control apoptosis). Taken together these data demonstrate that a soluble factor produced by stromal support can alter Bim binding to anti-apoptotic Bcl-2 family members, rendering myeloma cells primarily dependent on MCL-1 and protecting them from cell death signals that function through inhibition of Bcl-2/xL. Moreover these data in part explain why bortezomib can overcome survival signaling provided by stromal support, as proteasome inhibition is capable of inducing the MCL-1 inhibitor Noxa. Disclosures: Kaufman: Millenium: Consultancy; Onyx Pharmaceuticals: Consultancy; Novartis: Consultancy; Keryx: Consultancy; Merck: Research Funding; Celgene: Research Funding. Boise:University of Chicago: Patents & Royalties.


2019 ◽  
Vol 5 (11) ◽  
pp. eaaw7215 ◽  
Author(s):  
Junjie Gao ◽  
An Qin ◽  
Delin Liu ◽  
Rui Ruan ◽  
Qiyang Wang ◽  
...  

Mitochondrial transfer plays a crucial role in the regulation of tissue homeostasis and resistance to cancer chemotherapy. Osteocytes have interconnecting dendritic networks and are a model to investigate its mechanism. We have demonstrated, in primary murine osteocytes with photoactivatable mitochondria (PhAM)floxed and in MLO-Y4 cells, mitochondrial transfer in the dendritic networks visualized by high-resolution confocal imaging. Normal osteocytes transferred mitochondria to adjacent metabolically stressed osteocytes and restored their metabolic function. The coordinated movement and transfer of mitochondria within the dendritic network rely on contact between the endoplasmic reticulum (ER) and mitochondria. Mitofusin 2 (Mfn2), a GTPase that tethers ER to mitochondria, predominantly mediates the transfer. A decline in Mfn2 expression with age occurs concomitantly with both impaired mitochondrial distribution and transfer in the osteocyte dendritic network. These data show a previously unknown function of ER-mitochondrial contact in mediating mitochondrial transfer and provide a mechanism to explain the homeostasis of osteocytes.


Cancers ◽  
2021 ◽  
Vol 13 (14) ◽  
pp. 3461
Author(s):  
Zsolt Matula ◽  
Gábor Mikala ◽  
Szilvia Lukácsi ◽  
János Matkó ◽  
Tamás Kovács ◽  
...  

Recently, it has become evident that mitochondrial transfer (MT) plays a crucial role in the acquisition of cancer drug resistance in many hematologic malignancies; however, for multiple myeloma, there is a need to generate novel data to better understand this mechanism. Here, we show that primary myeloma cells (MMs) respond to an increasing concentration of chemotherapeutic drugs with an increase in the acquisition of mitochondria from autologous bone marrow stromal cells (BM-MSCs), whereupon survival and adenosine triphosphate levels of MMs increase, while the mitochondrial superoxide levels decrease in MMs. These changes are proportional to the amount of incorporated BM-MSC-derived mitochondria and to the concentration of the used drug, but seem independent from the type and mechanism of action of chemotherapeutics. In parallel, BM-MSCs also incorporate an increasing amount of MM cell-derived mitochondria accompanied by an elevation of superoxide levels. Using the therapeutic antibodies Daratumumab, Isatuximab, or Elotuzumab, no similar effect was observed regarding the MT. Our research shows that MT occurs via tunneling nanotubes and partial cell fusion with extreme increases under the influence of chemotherapeutic drugs, but its inhibition is limited. However, the supportive effect of stromal cells can be effectively avoided by influencing the metabolism of myeloma cells with the concomitant use of chemotherapeutic agents and an inhibitor of oxidative phosphorylation.


2005 ◽  
Vol 41 ◽  
pp. 205-218
Author(s):  
Constantine S. Mitsiades ◽  
Nicholas Mitsiades ◽  
Teru Hideshima ◽  
Paul G. Richardson ◽  
Kenneth C. Anderson

The ubiquitin–proteasome pathway is a principle intracellular mechanism for controlled protein degradation and has recently emerged as an attractive target for anticancer therapies, because of the pleiotropic cell-cycle regulators and modulators of apoptosis that are controlled by proteasome function. In this chapter, we review the current state of the field of proteasome inhibitors and their prototypic member, bortezomib, which was recently approved by the U.S. Food and Drug Administration for the treatment of advanced multiple myeloma. Particular emphasis is placed on the pre-clinical research data that became the basis for eventual clinical applications of proteasome inhibitors, an overview of the clinical development of this exciting drug class in multiple myeloma, and a appraisal of possible uses in other haematological malignancies, such non-Hodgkin's lymphomas.


2000 ◽  
Vol 111 (4) ◽  
pp. 1118-1121 ◽  
Author(s):  
A. Bellahcene ◽  
I. Van Riet ◽  
C. de Greef ◽  
N. Antoine ◽  
M. F. Young ◽  
...  

2000 ◽  
Vol 110 (1) ◽  
pp. 240-241 ◽  
Author(s):  
Faith E. Davies ◽  
Andrew C. Rawstron ◽  
Roger G. Owen ◽  
Gareth J. Morgan
Keyword(s):  

Sign in / Sign up

Export Citation Format

Share Document