Increasing adiponectin via an apolipoprotein peptide mimetic, L-4F, increases bone formation in normal mice and prevents myeloma bone disease in vivo

Bone ◽  
2011 ◽  
Vol 48 ◽  
pp. S81
Author(s):  
J.A. Fowler ◽  
S.T. Lwin ◽  
G.R. Mundy ◽  
C.M. Edwards
Blood ◽  
2009 ◽  
Vol 114 (9) ◽  
pp. 1803-1812 ◽  
Author(s):  
Angela Pennisi ◽  
Wen Ling ◽  
Xin Li ◽  
Sharmin Khan ◽  
John D. Shaughnessy ◽  
...  

Myeloma bone disease is caused by uncoupling of osteoclastic bone resorption and osteoblastic bone formation. Bidirectional signaling between the cell-surface ligand ephrinB2 and its receptor, EphB4, is involved in the coupling of osteoblastogenesis and osteoclastogenesis and in angiogenesis. EphrinB2 and EphB4 expression in mesenchymal stem cells (MSCs) from myeloma patients and in bone cells in myelomatous bones was lower than in healthy counterparts. Wnt3a induced up-regulation of EphB4 in patient MSCs. Myeloma cells reduced expression of these genes in MSCs, whereas in vivo myeloma cell-conditioned media reduced EphB4 expression in bone. In osteoclast precursors, EphB4-Fc induced ephrinB2 phosphorylation with subsequent inhibition of NFATc1 and differentiation. In MSCs, EphB4-Fc did not induce ephrinB2 phosphorylation, whereas ephrinB2-Fc induced EphB4 phosphorylation and osteogenic differentiation. EphB4-Fc treatment of myelomatous SCID-hu mice inhibited myeloma growth, osteoclastosis, and angiogenesis and stimulated osteoblastogenesis and bone formation, whereas ephrinB2-Fc stimulated angiogenesis, osteoblastogenesis, and bone formation but had no effect on osteoclastogenesis and myeloma growth. These chimeric proteins had similar effects on normal bone. Myeloma cells expressed low to undetectable ephrinB2 and EphB4 and did not respond to the chimeric proteins. The ephrinB2/EphB4 axis is dysregulated in MM, and its activation by EphB4-Fc inhibits myeloma growth and bone disease.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3717-3717
Author(s):  
Xin Li ◽  
Wen Ling ◽  
Angela Pennisi ◽  
Yuping Wang ◽  
Sharmin Khan ◽  
...  

Abstract Abstract 3717 Human placenta has emerged as a valuable, uncontroversial source of transplantable cells for many cytotherapeutic purposes, including modulation of inflammation, bone repair, and cancer. Placenta-derived adherent cells (PDAC) are mesenchymal like adherent cells isolated from postpartum human placenta and capable of supporting bone formation in vivo. Multiple myeloma (MM) is closely associated with induction of bone disease and large lytic lesions, which are often not repaired and are usually the sites of relapses. The aim of the study was to evaluate the antimyeloma therapeutic potential, in vivo survival, and trafficking of PDAC in the SCID-rab model of MM-associated bone disease. SCID-rab system constructed by implanting a 4-weeks old rabbit bone into which primary human myeloma cells were directly injected (Yatta et al., Leukemia 2004; Yaccoby et al., Blood 2007). Bone disease was evaluated by measurements of bone mineral density (BMD) and X-rays. MM growth was determined by human immunoglobulin (hIg) ELISA and histologically. For in vivo tracking PDAC were transduced with a luciferase/GFP reporter in a lentiviral vector. SCID-rab mice engrafted with primary myeloma cells from 2 patients. Upon establishment of MM growth, PDAC (1×106 cells/bone) or vehicle were injected into the implanted myelomatous bone (Patient's 1, 5 mice/group; Patient's 2, 7 mice/group). While BMD of the implanted bones was significantly reduced in control hosts, intralesional PDAC cytotherapy significantly increased BMD of the implanted bones from pretreatment levels by >37% (p<0.01 versus control) and inhibited MM growth in the 2 sets of experiments (p<0.04). The bone anabolic effect of PDAC was associated with increased number of osteoblasts (p<0.003) and reduced number of osteoclasts (p<0.004). Intralesional PDAC cytotherapy also promoted bone formation in nonmyelomatous SCID-rab mice. Intralesional but not subcutaneous engraftment of PDAC inhibited bone disease and tumor growth in SCID-rab mice. In contrast to intra-bone injection in SCID-rab mice, intra-tumor injection of PDAC had no effect on subcutaneous growth of the H929 myeloma cell line in SCID mice (8 mice/group). Live-animal imaging revealed that the majority of PDAC disappeared from the injected bones within 4 weeks. To test their systemic behavior, PDAC were intravenously injected into 18 SCID-rab mice engrafted with H929 myeloma cells. The presences of PDAC in various organs were evaluated 1, 2 and 7 days after injection. Ex vivo bioluminescence analysis of the implanted myelomatous bones detected PDAC in two of five bones on day 1, in four of four bones on day 2, and in four of nine bones on day 7. Intravenously injected PDAC were also detected in lungs not in any other murine tissues. Immunohistochemical staining for GFP in myelomatous bone sections detected GFP-expressing PDAC in rabbit marrow areas infiltrated with myeloma cells, supporting bioluminescence analysis. Our study suggest that PDAC stimulate bone formation by acting as bystander cells that increase endogenous osteoblastogenesis and inhibit osteoclastogenesis, and that alteration of the bone marrow microenvironment by PDAC attenuates growth of MM. PDAC cytotherapy is a promising therapeutic approach for myeloma bone disease. Disclosures: Khan: Celgene Cellular Therapeutics: Research Funding. Heidaran:Celgene Cellular Therapeutics: Employment. Pal:Celgene Cellular Therapeutics: Employment. Zhang:Celgene Cellular Therapeutics: Employment. He:Celgene Cellular Therapeutics: Employment. Zeitlin:Celgene Cellular Therapeutics: Employment. Abbot:Celgene Cellular Therapeutics: Employment. Faleck:Celgene Cellular Therapeutics: Employment. Hariri:Celgene Cellular Therapeutics: Employment.


Blood ◽  
2007 ◽  
Vol 110 (4) ◽  
pp. 1098-1104 ◽  
Author(s):  
Evangelos Terpos ◽  
Orhan Sezer ◽  
Peter Croucher ◽  
Meletios-Athanassios Dimopoulos

AbstractBone disease is one of the most debilitating manifestations of multiple myeloma. A complex interdependence exists between myeloma bone disease and tumor growth, creating a vicious circle of extensive bone destruction and myeloma progression. Proteasome inhibitors have recently been shown to promote bone formation in vitro and in vivo. Preclinical studies have demonstrated that proteasome inhibitors, including bortezomib, which is the first-in-class such agent, stimulate osteoblast differentiation while inhibiting osteoclast formation and bone resorption. Clinical studies are confirming these observations. Bortezomib counteracts the abnormal balance of osteoclast regulators (receptor activator of nuclear factor-κB ligand and osteoprotegerin), leading to osteoclast inhibition and decreased bone destruction, as measured by a reduction in markers of bone resorption. In addition, bortezomib stimulates osteoblast function, possibly through the reduction of dickkopf-1, leading to increased bone formation, as indicated by the elevation in bone-specific alkaline phosphatase and osteocalcin. The effect of bortezomib on bone disease is thought to be direct and not only a consequence of the agent's antimyeloma properties, making it an attractive agent for further investigation, as it may combine potent antimyeloma activity with beneficial effects on bone. However, the clinical implication of these effects requires prospective studies with specific clinical end points.


Blood ◽  
2008 ◽  
Vol 111 (5) ◽  
pp. 2833-2842 ◽  
Author(s):  
Claire M. Edwards ◽  
James R. Edwards ◽  
Seint T. Lwin ◽  
Javier Esparza ◽  
Babatunde O. Oyajobi ◽  
...  

There is increasing evidence to suggest that the Wnt signaling pathway plays a critical role in the pathogenesis of myeloma bone disease. In the present study, we determined whether increasing Wnt signaling within the bone marrow microenvironment in myeloma counteracts development of osteolytic bone disease. C57BL/KaLwRij mice were inoculated intravenously with murine 5TGM1 myeloma cells, resulting in tumor growth in bone and development of myeloma bone disease. Lithium chloride (LiCl) treatment activated Wnt signaling in osteoblasts, inhibited myeloma bone disease, and decreased tumor burden in bone, but increased tumor growth when 5TGM1 cells were inoculated subcutaneously. Abrogation of β-catenin activity and disruption of Wnt signaling in 5TGM1 cells by stable overexpression of a dominant-negative TCF4 prevented the LiCl-induced increase in subcutaneous growth but had no effect on LiCl-induced reduction in tumor burden within bone or on osteolysis in myeloma-bearing mice. Together, these data highlight the importance of the local microenvironment in the effect of Wnt signaling on the development of myeloma bone disease and demonstrate that, despite a direct effect to increase tumor growth at extraosseous sites, increasing Wnt signaling in the bone marrow microenvironment can prevent the development of myeloma bone disease and inhibit myeloma growth within bone in vivo.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 3508-3508 ◽  
Author(s):  
Patrice Boissy ◽  
Thomas Lund ◽  
Thomas L. Andersen ◽  
Torben Plesner ◽  
Jean-Marie Delaisse

Abstract Multiple myeloma (MM) leads to high risk for bone pain and fractures. MM-induced bone disease is due to acute degradation of bone matrix by osteoclasts, and absence of repair by bone forming osteoblasts. It is currently treated with bisphosphonates, highly effective bone resorption inhibitors, which do not promote but rather inhibit bone formation and may cause renal damage and osteonecrosis of the jaw. Thus, it is important to reconsider the management of MM bone disease in long-term treatment. Recent preclinical studies reported that the proteasome inhibitor Bortezomib (V) used for the treatment of MM patients can stimulate bone formation, and that in MM patients treated with V, serum levels of bone formation markers are increased. The present study aims at investigating if V may inhibit osteoclast activity. Methods: Osteoclasts were differentiated from pure populations of blood derived CD14-positive monocytes cultured with M-CSF and RANKL for 6–7 days, and treated continuously with V at various concentrations. As prolonged inhibition of proteasome activity has been reported to be toxic for any cell type, and in vivo pharmacodynamic studies have shown V to be eliminated from the vascular compartment as soon as 30min after intravenous injection, displaying maximal inhibitory activity of the proteasome within 24 hours subsiding rapidly thereafter, V was also given intermittently, to mimick the in vivo situation. Osteoclast differentiation and activity were assessed by measuring Tartrate-Resistant Acid Phosphatase (TRACP) activity in the medium. Cell viability was determined with Celltiter Blue measuring metabolic activity. To extend our observations to the clinical situation, serum levels of CTX-I, a bone resorption marker, were measured during the 3 days following therapeutic V administration in a single patient. Results: A continuous treatment of cultures with V at 4 nM and higher concentrations proved to be highly toxic for differentiating osteoclasts but also monocytes. A 3-hour-pulse treatment with V followed by a 3-day culture in the absence of V, was not toxic neither to monocytes nor to osteoclasts, even at a concentration as high as 100 nM. This 3-hour pulse was however highly toxic for myeloma cells. Interestingly, a 3-hour pulse with 25 nM V induced a 50% inhibition of the resorptive activity of osteoclasts, as assessed by culturing them for 3 days on bone slices and measuring the formation of resorption pits. The release of TRACP in the medium was inhibited to a similar extent within the first 24 hours post-pulse, but tended to return to the control level during the next 2 days. This 3-hour pulse with 25 nM V inhibited strongly RANKL-induced translocation of NF-KB in the osteoclast nuclei, an event dependent on proteasome function and critical for osteoclastic activity. Serum CTX-I levels decreased during the first 48 hours after each V injection (n = 3), and tended to increase again after 72 hours suggesting a partial recovery of osteoclast activity between each administration. Conclusions: Our results suggest that Bortezomib temporarily inhibits osteoclast activity in vitro and in vivo. This effect is linked to RANKL-induced translocation of NF-KB in the osteoclast nuclei and proteasome function. Since recent reports suggested that formation of new bone requires at least a transient activity of osteoclasts transient inhibition of osteoclasts could be an advantage compared to the more persistent inhibition of osteoclast activity by bisphosphonate.


Cancers ◽  
2021 ◽  
Vol 13 (17) ◽  
pp. 4277
Author(s):  
Syed Hassan Mehdi ◽  
Carol A Morris ◽  
Jung Ae Lee ◽  
Donghoon Yoon

Multiple myeloma (MM) is a plasma cell malignancy that causes an accumulation of terminally differentiated monoclonal plasma cells in the bone marrow, accompanied by multiple myeloma bone disease (MMBD). MM animal models have been developed and enable to interrogate the mechanism of MM tumorigenesis. However, these models demonstrate little or no evidence of MMBD. We try to establish the MMBD model with severe bone lesions and easily accessible MM progression. 1 × 106 luciferase-expressing 5TGM1 cells were injected into 8–12 week-old NOD SCID gamma mouse (NSG) and C57BL/KaLwRij mouse via the tail vein. Myeloma progression was assessed weekly via in vivo bioluminescence (BL) imaging using IVIS-200. The spine and femur/tibia were extracted and scanned by the micro-computer tomography for bone histo-morphometric analyses at the postmortem. The median survivals were 56 days in NSG while 44.5 days in C57BL/KaLwRij agreed with the BL imaging results. Histomorphic and DEXA analyses demonstrated that NSG mice have severe bone resorption that occurred at the lumbar spine but no significance at the femur compared to C57BL/KaLwRij mice. Based on these, we conclude that the systemic 5TGM1 injected NSG mouse slowly progresses myeloma and develops more severe MMBD than the C57BL/KaLwRij model.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 844-844
Author(s):  
Angela Pennisi ◽  
Wen Ling ◽  
Xin Li ◽  
Jianmei Chen ◽  
Sharmin Khan ◽  
...  

Abstract Induction of osteolytic bone lesions in myeloma (MM) is caused by an uncoupling of osteoclastic bone resorption and osteoblastic bone formation. Recent studies indicate that in addition to role in cell adhesion, repulsion and neovascularization, bidirectional signaling between the cell surface molecules EphrinB2 and EphB4 also mediates the coupling between osteoblasts and osteoblasts. While mesenchymal stem cells (MSCs) and osteoblasts express the ligand EphrinB2 land its receptor, EphB4, osteoclasts and their precursors mainly express EphrinB2. Forward signaling in MSCs promotes osteogenic differentiation and reverse signaling in osteoclast precursors inhibits their differentiation. The aims of the study were to investigate whether the EphrinB2/Eph4 axis is dysregulated in MM osteoprogenitors and whether activation of this axis in myelomatous bone by EphrinB2-Fc or EphB4-Fc affects MM bone disease, angiogenesis and tumor growth. MSCs were generated from bone marrow of healthy donors (n=5) and patients with MM (n=13). Gene expression was determined by qRT-PCR. MSCs from MM patients had reduced expression of EphrinB2 (EFNB2) by 61±6% (p<0.02) and EphB4 by 60±10% (p<0.02) than expression levels of these molecules in MSCs from healthy donors. Expression of other EFN and EPH B genes were detected and similarly expressed in patients and donors MSCs. Differentiation of MSCs from MM patients into osteoblasts resulted in upregulation of EFNB2 and downregulation of EPHB4. MM cell lines and primary MM plasma cells expressed low to undetectable levels of this family of genes. We exploited our SCID-hu system for primary MM to study the consequences of activation of forward signaling by EphrinB2-Fc or reverse signaling by EphB4-Fc on MM-induced bone disease and MM growth. Twelve SCID-hu mice were engrafted with MM cells from a patient with active MM. Upon detection of MM growth (by human Ig ELISA) and bone disease (radiographically), hosts were locally treated with Fc (control), EphrinB2-Fc or EPHB4 (4 mice/group) for 4 weeks using Alzet pump that continually released 0.11 μg/hour of each compound. While in Fc-treated hosts BMD of the implanted bone was reduced by 8±3% from pretreatment levels, it was increased by EphrinB2-Fc and EPhB4-Fc by 15±8% (p<0.03 vs. Fc) and 2±1% (p<0.02 vs. Fc) from pretreatment levels, respectively. At experiment’s end levels of human Ig in mice sera were increased by 308±99% and 244±86% from pretreatment levels in Fc- and EphrinB2- Fc groups, respectively, while were reduced by 92±1% (p<0.02 vs. Fc) from pretreatment levels in EphB4-Fc group. In myelomatous bones, EphB4-Fc and EphrinB2-Fc increased the numbers of osteoblasts by >3 folds (p<0.004) while EphB4-Fc, but not EphrinB2-Fc, reduced osteoclast numbers by 5 folds (p<0.01 vs. Fc group). The numbers of CD34-reactive neovessels were reduced by 2 folds following treatment with EphB4-Fc (p<0.03) and were increased by 2.5 folds following treatment with EphrinB2-Fc (p<0.05). Our study suggests that downregulation of EphrinB2 and EhpB4 in MSCs from MM patients contributes to their impaired osteogenic differentiation and that treatment with EphrinB2-Fc or EphB4-Fc helps restore coupling of bone remodeling in myelomatous bones. The results also indicate that EphB4-Fc treatment is an effective approach to simultaneously inhibit MM and its associated bone disease.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 2961-2961 ◽  
Author(s):  
Silvia Colucci ◽  
Giacomina Brunetti ◽  
Angela Oranger ◽  
Giorgio Mori ◽  
Francesca Sardone ◽  
...  

Abstract Abstract 2961 Reduced osteoblast activity contributes to the development of multiple myeloma-bone disease. Wingless-type (Wnt) signalling pathway is critical in osteoblastogenesis, and it is negatively regulated by molecules such as frizzled-related proteins (sFRPs), Dickkopf proteins (DKKs) and sclerostin. Myeloma cells are known to induce inhibition of osteoblastogenesis through Wnt antagonists such as DKK-1 and sFRP-2 and -3 whereas the role of sclerostin, an osteocyte-expressed negative regulator of bone formation, has not been yet investigated. We provide novel evidence showing sclerostin expression by myeloma cells from patients with multiple myeloma-bone disease and human myeloma cell lines (HMCLs). By means of a co-culture system of bone marrow stromal cells (BMSCs) and HMCLs, we demonstrated that sclerostin expression by myeloma cells and HMCLs is responsible for reduced expression of major osteoblastic specific proteins namely bone-specific alkaline phosphatase, collagen-type I, bone sialoprotein II and osteocalcin as well as decreased mineralized nodule formation and attenuated expression of member of the AP-1 transcription factor family (i.e. Fra-1, Fra-2 and Jun-D). The addition of a neutralizing anti-sclerostin antibody to our co-culture system can restore the above parameters, through the intranuclear accumulation of β-catenin in BMSCs. On the other hand, we demonstrated that sclerostin is also involved in inducing increased receptor activator of nuclear factor-k B ligand (RANKL) and decreased osteoprotegerin (OPG) expression in osteoblasts, contributing to the enhanced osteoclast activity occurring in patients with multiple myeloma-bone disease. Our data suggest that myeloma cells contribute to the suppression of bone formation through sclerostin secretion. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3164-3164
Author(s):  
Fani Ziouti ◽  
Maximilian Rummler ◽  
Andreas Brandl ◽  
Andreas Beilhack ◽  
Maureen Lynch ◽  
...  

Abstract Osteolytic bone disease (BD) is a hallmark of multiple myeloma (MM) with tumor cells in the bone marrow shifting the balance of the bone remodeling process towards massive bone resorption. As a result, patients develop devastating osteolytic lesions that lead to non-healing bone fractures and pain, affecting life quality and mortality rates. Bones have the capacity to adapt mass and structure to mechanical stimuli, as dramatically seen in young tennis athletes with muscle-bone asymmetries in the playing arm. We have previously shown that tibial mechanical loading rescued bone loss in our murine MOPC315.BM MM model with an advanced osteolytic phenotype. Here, we hypothesize that mechanical strain (1) modulates the bone microenvironment and (2) has antitumor activity in mice. (1) We determined bone formation and bone resorption parameters by time-lapsed microCT analysis to show how skeletal mechanical stimuli control MM bone disease (MMBD) progression over time. (2) To monitor tumor progression, we used non-invasive bioluminescence imaging (BLI) and enzyme-linked immunosorbent assay (ELISA) for detection of MOPC315.BM specific immunoglobulin A (IgA) levels. In our in vivo loading study, we injected MOPC315.BM cells intratibially (i.t.) in BALB/c mice to establish MMBD (n=17) and used PBS-injected (n=13) as well as noninjected mice (n=8) as controls. Eight (MM), seven (PBS) and 8 (noninjected) mice received compressive tibial loading for three weeks while nine (MM) and six (PBS) mice served as nonloaded controls. The bone remodeling response to mechanical loading was investigated by longitudinal in vivo microCT imaging performed every 5 days (at day 13, 18, 23, 28, and 33 after i.t. injection). MicroCT images from day 33 were geometrically registered onto images of day 13 and resampled into the same coordinate system using Amira and scripts written in Matlab for post-processing. Normalized newly mineralized and eroded bone volume (MV/BV, EV/BV), normalized formed and eroded bone surface area (MS/BS, ES/BS), mineralized thickness (MTh) and eroded depth (ED) were quantified. ANOVA was performed to examine the effect of loading and injection. Loading significantly increased the periosteal MV/BV, periosteal and endosteal MS/BS as well as decreased the periosteal EV/BV and periosteal and endocortical ES/BS. Endosteal MV/BV or EV/BV were not affected, which may be due to differences in the local strain environment at the two surfaces. In addition, mechanical stimuli did not influence ED, but led to diminished periosteal EV/BV and periosteal ES/BS suggesting fewer resorption sites in tibiae subjected to loading. Injection significantly affected periosteal and endosteal bone formation and resorption (Fig.1). Significant increases in cortical bone mass of loaded MM mice were accompanied by decreases in tumor load as evidenced by MOPC315.BM specific IgA levels (Fig. 2A). Interestingly, quantification of tibial and whole body bioluminescence signal intensities revealed controlled tumor growth in the loaded left tibia and a further delay of tumor cell dissemination throughout body of MM mice (Fig. 2B). Our data provide evidence that skeletal mechanical stimuli have anti-myeloma effects and rescue osteolytic bone loss in MMBD. The anabolic response to mechanical loads outweighs the anti-resorptive effect of MM cells, suggesting a combination of loading with bone resorption inhibitors in future therapeutic strategies. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 812-812
Author(s):  
Claire M. Edwards ◽  
James R. Edwards ◽  
Seint T. Lwin ◽  
Gregory R. Mundy

Abstract Multiple myeloma is characterized by uncontrolled proliferation of myeloma cells within the bone marrow and the development of a severe osteolytic bone disease. In addition to a well characterized increase in osteoclastic bone resorption, myeloma bone disease is associated with a reduction in bone formation. Osteoblast differentiation and bone formation are regulated in vivo by canonical Wnt signaling and activation of β-catenin. Therefore increasing Wnt signaling in the bone microenvironment in multiple myeloma may prevent the development of myeloma bone disease. In support of this, we have previously demonstrated that activation of Wnt signaling with lithium chloride (LiCl) in the 5TGM1 murine model of myeloma reduces tumor burden and osteolytic bone disease. However, we also found that LiCl treatment increased subcutaneous (s.c.) tumor growth. This suggests that the reduction in tumor burden within the bone microenvironment may be an indirect effect mediated through the effects of LiCl to prevent myeloma bone disease. The aim of the current study was to determine the effect of specific molecular blockade of Wnt signaling in myeloma cells in vivo. 5TGM1-GFP myeloma cells were transfected by electroporation with either myc-tagged dominant negative TCF4 (DNTCF4) or pcDNA. Following stable selection by culture in G418, expression of DNTCF4 was confirmed by western blot for myc. No difference was found in the growth rates of 5TGM1-pcDNA or 5TGM1-DNTCF4 in vitro. Treatment with LiCl or Wnt3A had no significant effect on cell viability in vitro, but significantly increased β-catenin activity, as measured by TOPFLASH activity in 5TGM1-pcDNA cells. This increase was not observed in 5TGM1-DNTCF4, confirming that expression of DNTCF4 blocked Wnt signaling induced by LiCl in 5TGM1 myeloma cells. C57Bl/KaLwRij mice were inoculated with 5TGM1-pcDNA or 5TGM1-DNTCF4 cells by either intravenous (i.v.) or s.c. injection. Mice were treated from time of tumor cell inoculation with 200mg/kg/day LiCl or vehicle control (d.H20) by oral gavage for 28 days. I.v. inoculation of myeloma cells resulted in a significant increase in serum IgG2bκ concentrations and the proportion of GFP-positive cells in the bone marrow. A significant reduction in trabecular bone volume was also observed. MicroCT analysis of the tibia demonstrated that LiCl significantly increased trabecular bone volume in both 5TGM1-pcDNA and 5TGM1-DNTCF4 myeloma-bearing mice. LiCl significantly decreased serum IgG2bκ concentrations in both 5TGM1-pcDNA and 5TGM1-DNTCF4 myeloma-bearing mice, with a greater effect in 5TGM1-DNTCF4 myeloma-bearing mice. FACS analysis of GFP-positive cells demonstrated that LiCl significantly reduced tumor burden in the bone marrow in both 5TGM1-pcDNA and 5TGM1-DNTCF4 myeloma-bearing mice. However, following s.c inoculation, LiCl significantly increased s.c. tumor volume of 5TGM1-pcDNA tumors, but had no effect on 5TGM1-DNTCF4 s.c. tumor volume. Taken together these results demonstrate that the effect of increasing Wnt signaling in myeloma is dependent upon the microenvironment. By specific inhibition of β-catenin activity in myeloma cells combined with systemic stimulation of the Wnt signaling pathway, our results suggest that increasing Wnt signaling in myeloma in vivo has dual effects; firstly to enhance myeloma growth directly, and secondly to enhance osteoblast differentiation and thus indirectly reduce tumor burden in bone, highlighting the importance of the bone marrow microenvironment in regulating myeloma growth and survival.


Sign in / Sign up

Export Citation Format

Share Document