Gene Modified Human Mesenchymal Stem Cells Expressing Osteoprotegerin Inhibit Osteoclast Activation and Increase Trabecular Bone Area in a Xenogeneic Murine Model of Myeloma.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 505-505
Author(s):  
Neil Rabin ◽  
Chara Kyriakou ◽  
Reuben Benjamin ◽  
Arnold Pizzey ◽  
Orla Gallagher ◽  
...  

Abstract Bone disease in multiple myeloma (MM) results from increased osteoclast (OCL) numbers and activity, which is associated with an increase in RANK Ligand and reduction in osteoprotegerin (OPG). Systemic administration of recombinant OPG reduces MM bone disease, but the short half life of OPG limits its usefulness. Gene modified mesenchymal stem cells (MSCs) offer a potential means of delivering stable expression of OPG in vivo to reduce OCL activation and bone destruction. Bone marrow derived human MSCs were transduced with a self-inactivating bicistronic lentiviral vector containing human OPG and GFP (MSCOPG). Control vector was identical except the OPG was cloned in reverse orientation (MSCGPO). Efficient transduction was demonstrated by high GFP expression (96% MSCOPG, 92% (MSCGPO). Stable transgene expression of human OPG (hOPG) occurred for beyond 20 passages in vitro, and hOPG was detected in vivo after tail vein administration of MSCOPG (2ng/mL hOPG detected in mouse serum 1 week after tail vein administration of 3 x 106 MSCOPG). Immunophenotype and differentiation potential of MSCs were maintained following transduction. A xenogeneic model of MM was developed. 1 x 107 KMS-12-BM cells injected tail vein into b2 m NOD/SCID mice leads to tumour infiltration in the bone marrow at 6 weeks, with varied tumour take between the bones examined. Using histomorphometric analysis trabecular bone area (TBA) was assessed as the proportion of trabecular bone in 0.5625 mm2 of marrow space 0.2 mm from growth plate. OCL were recorded as the proportion lining the endocortical surface (%OcPm). Reduction of trabecular bone in the tibia is related to the amount of tumour (KMS-12-BM tibia with >70% tumour mean TBA 0.7+/− 0.2 vs. KMS-12-BM tibia with <70% tumour mean 5.1+/− 0.8, p<0.01, which is similar to non diseased animals). All subsequent analysis were carried out on tibia with >70% tumour. There was no change in trabecular bone in the lumbar vertebrae. OCL were increased in the tibia and lumbar vertebrae of tumour bearing mice (PBS group mean %OcPm 0.9+/− 0.3 and 1.1+/− 0.4 vs. KMS-12-BM group mean 7.2+/− 3.2 and 7.5 +/− 2.2 in tibia and lumbar vertebrae respectively, p=0.01 in both groups). We hypothesised that MSCs expressing OPG will prevent the increase in OCL and subsequent loss of trabecular bone. Infusion of unmanipulated MSC or MSCGPO had no effect on %OcPm or TBA in diseased animals. 1 x106 MSCOPG or MSCGPO were injected by tail vein 2, 3 and 4 weeks after KMS-12-BM injection. Another group received KMS-12-BM alone. All mice were culled at 6 weeks. Trabecular bone was increased in the tibia of tumour bearing mice treated with MSCOPG (mean TBA 1.4 +/− 0.5) compared to control animals receiving MSCGPO or tumour alone (mean TBA 0.6 +/− 0.2), p=0.03, with a trend showing a reduction of OCL in the tibia of the MSCOPG group (mean %OcPm 2.6+/− 1.0) vs. control group (mean %OcPm 4.2+/− 1.5, NS). Importantly in the lumbar vertebrae, OCL were reduced in the MSCOPG group (mean %OcPm 1.9 +/− 0.4) compared to control animals (mean %OcPm 3.5+/− 0.5), p<0.01. Conclusion: MSCs gene modified with OPG are able to increase TBA in the tibia and reverse OCL activation in a xenogeneic model of MM. Gene modified MSCs may have future potential in treating MM induced bone disease.

2015 ◽  
Vol 137 (1) ◽  
Author(s):  
T. J. Vaughan ◽  
M. Voisin ◽  
G. L. Niebur ◽  
L. M. McNamara

Mechanical loading directs the differentiation of mesenchymal stem cells (MSCs) in vitro and it has been hypothesized that the mechanical environment plays a role in directing the cellular fate of MSCs in vivo. However, the complex multicellular composition of trabecular bone marrow means that the precise nature of mechanical stimulation that MSCs experience in their native environment is not fully understood. In this study, we developed a multiscale model that discretely represents the cellular constituents of trabecular bone marrow and applied this model to characterize mechanical stimulation of MCSs in vivo. We predicted that cell-level strains in certain locations of the trabecular marrow microenvironment were greater in magnitude (maximum ε12 = ∼24,000 με) than levels that have been found to result in osteogenic differentiation of MSCs in vitro (>8000 με), which may indicate that the native mechanical environment of MSCs could direct cellular fate in vivo. The results also showed that cell–cell adhesions could play an important role in mediating mechanical stimulation within the MSC population in vivo. The model was applied to investigate how changes that occur during osteoporosis affected mechanical stimulation in the cellular microenvironment of trabecular bone marrow. Specifically, a reduced bone volume (BV) resulted in an overall increase in bone deformation, leading to greater cell-level mechanical stimulation in trabecular bone marrow (maximum ε12 = ∼48,000 με). An increased marrow adipocyte content resulted in slightly lower levels of stimulation within the adjacent cell population due to a shielding effect caused by the more compliant behavior of adipocytes (maximum ε12 = ∼41,000 με). Despite this reduction, stimulation levels in trabecular bone marrow during osteoporosis remained much higher than those predicted to occur under healthy conditions. It was found that compensatory mechanobiological responses that occur during osteoporosis, such as increased trabecular stiffness and axial alignment of trabeculae, would be effective in returning MSC stimulation in trabecular marrow to normal levels. These results have provided novel insight into the mechanical stimulation of the trabecular marrow MSC population in both healthy and osteoporotic bone, and could inform the design three-dimensional (3D) in vitro bioreactor strategies techniques, which seek to emulate physiological conditions.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 510-510
Author(s):  
Alissa Huston ◽  
Ganwei Lu ◽  
Judy Anderson ◽  
Ken Patrene ◽  
Lily Hong ◽  
...  

Abstract MM is the most common cancer metastasizing to bone, with 85–90% of patients developing bone disease. It is unique from other forms of metastatic bone disease, as it is purely lytic. There are a multitude of factors involved in promoting bone disease in MM; however, what remains unanswered is why patients in complete remission from MM are unable to heal their bone lesions. We hypothesize that MM cells exert a permanent change in the bone marrow microenvironment, either through a change in the differentiation potential of mesenchymal stem cells or through stem cell depletion. To investigate this question we developed a new model of MM bone disease in which a herpes-simplex thymidine kinase gene (HSVtk), sensitive to ganciclovir (GCV), is expressed within a MM cell line and injected into the tibia of mice. This allows for eradication of the MM cells without the use of chemotherapy or immunotherapy, minimizes adverse effects on neighboring mesenchymal stem cells and directly involves the bone. The murine MM cell line (5TGM1) was infected, using a lentiviral system, with the tricistronic construct of HSVtk linked to green fluorescence protein (GFP) and blasticidin. Following blasticidin selection, 5TGM1tk cells stably expressing HSVtk and GFP were used for all experiments. GCV dosing curves to determine the dose resulting in 100% eradication were completed. The effects of GCV on both murine hematopoietic cells and mesenchymal stem cells induced to osteoblasts were completed using methylcellulose colony formation and alkaline phosphatase assays. Bystander effects of 5TGM1tk cells on murine hematopoietic cells and murine osteoblasts were completed using murine methylcellulose colony formation, alkaline phosphatase and colony-forming unit fibroblast (CFU-F) assays. In vivo data was obtained following intratibial injection of 5TGM1tk cells (1x105, 5x105, 1x106) or saline into NIH-III mice (3 mice per group, 12 mice total). Mice were evaluated at weekly intervals for tumor and lytic lesion development with plain radiographs and Micro QCT, and osteoblast activity evaluated by alkaline phosphatase analysis. No significant effects of GCV, at doses capable of eradicating the MM cell line (1ug/ml) were observed on murine hematopoietic cells or osteoblasts. No significant effect was observed on hematopoietic colony development when 5TGM1tk cells were cocultured with murine hematopoietic cells. A small bystander effect, approximately 30%, was identified when 5TGM1tk cells were cocultured with murine mesenchymal stem cells induced to osteoblasts in the presence of GCV. Dose-dependent CFU-F suppression was observed with increasing percentages of tumor cells (0–50%), however reversal of suppression was noted when cultured in the presence of GCV. 60% of the mice injected with 5TGM1tk cells developed tumor, and evidence of dose-dependent osteoblast suppression in vivo observed as measured by the alkaline phosphatase assay (p&lt;0.00001). The current model provides us with the capacity to specifically target MM cells without using chemotherapy or immunotherapy. It provides a model to study and dissect the interactions between MM cells and mesenchymal stem cells, within the bone marrow microenvironment, which lead to osteoblast suppression. This model should provide important insights into the mechanisms of osteoblast suppression in MM and help to identify targeted therapies that can reverse this process for patients.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Pegah Nammian ◽  
Seyedeh-Leili Asadi-Yousefabad ◽  
Sajad Daneshi ◽  
Mohammad Hasan Sheikhha ◽  
Seyed Mohammad Bagher Tabei ◽  
...  

Abstract Introduction Critical limb ischemia (CLI) is the most advanced form of peripheral arterial disease (PAD) characterized by ischemic rest pain and non-healing ulcers. Currently, the standard therapy for CLI is the surgical reconstruction and endovascular therapy or limb amputation for patients with no treatment options. Neovasculogenesis induced by mesenchymal stem cells (MSCs) therapy is a promising approach to improve CLI. Owing to their angiogenic and immunomodulatory potential, MSCs are perfect candidates for the treatment of CLI. The purpose of this study was to determine and compare the in vitro and in vivo effects of allogeneic bone marrow mesenchymal stem cells (BM-MSCs) and adipose tissue mesenchymal stem cells (AT-MSCs) on CLI treatment. Methods For the first step, BM-MSCs and AT-MSCs were isolated and characterized for the characteristic MSC phenotypes. Then, femoral artery ligation and total excision of the femoral artery were performed on C57BL/6 mice to create a CLI model. The cells were evaluated for their in vitro and in vivo biological characteristics for CLI cell therapy. In order to determine these characteristics, the following tests were performed: morphology, flow cytometry, differentiation to osteocyte and adipocyte, wound healing assay, and behavioral tests including Tarlov, Ischemia, Modified ischemia, Function and the grade of limb necrosis scores, donor cell survival assay, and histological analysis. Results Our cellular and functional tests indicated that during 28 days after cell transplantation, BM-MSCs had a great effect on endothelial cell migration, muscle restructure, functional improvements, and neovascularization in ischemic tissues compared with AT-MSCs and control groups. Conclusions Allogeneic BM-MSC transplantation resulted in a more effective recovery from critical limb ischemia compared to AT-MSCs transplantation. In fact, BM-MSC transplantation could be considered as a promising therapy for diseases with insufficient angiogenesis including hindlimb ischemia.


2018 ◽  
Vol 106 ◽  
pp. 1126-1134 ◽  
Author(s):  
Wenbo Zhang ◽  
Chao Huang ◽  
Aijun Sun ◽  
Liang Qiao ◽  
Xi Zhang ◽  
...  

2001 ◽  
Vol 169 (1) ◽  
pp. 12-20 ◽  
Author(s):  
Jizong Gao ◽  
James E. Dennis ◽  
Raymond F. Muzic ◽  
Magnus Lundberg ◽  
Arnold I. Caplan

2019 ◽  
Vol 7 (1) ◽  
pp. 362-372 ◽  
Author(s):  
Shuhao Liu ◽  
Yang Liu ◽  
Libo Jiang ◽  
Zheng Li ◽  
Soomin Lee ◽  
...  

BMP-2-induced migration of BMSCs can be inhibited by silencing CDC42 in vitro and in vivo.


2021 ◽  
Author(s):  
meng li ◽  
ning yang ◽  
li hao ◽  
wei zhou ◽  
lei li ◽  
...  

Abstract ObjectivesSteroid-induced osteoporosis (SIOP) is a secondary osteoporosis, which is a systemic bone disease characterized by low bone mass, bone microstructure damage, increased bone fragility, and easy fracture. However, the specific mechanism remains unclear. Glucocorticoid-induced death of osteoblasts and bone marrow mesenchymal stem cells (BMSCs) is an important factor in SIOP. Ferroptosis is an iron-dependent programmed cell death that differs from apoptosis, cell necrosis, and autophagy, which can be induced by many factors. Herein, we aimed to explore whether glucocorticoids (GCs) cause ferroptosis in BMSCs and determine possible treatment pathways and mechanisms of action. Melatonin (MT), a hormone secreted by the pineal gland, displays strong antioxidant abilities to scavenge free radicals and alleviates ferroptosis in many tissues and organs. MethodsIn this study, we used high-dose dexamethasone (DEX) to observe whether glucocorticoids induced ferroptosis in BMSCs. We then assessed whether MT can inhibit the ferroptotic pathway, thereby providing early protection against GC-induced SIOP, and investigated the signaling pathways involved.ResultsIn vitro experiments showed that MT intervention significantly improved GC-induced ferroptosis in BMSCs and significantly improved SIOP in vivo. Pathway analysis showed that MT improves ferroptosis by activating the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) axis. MT upregulates expression of PI3K, which is an important regulator of ferroptosis resistance. PI3K activators mimic the anti-ferroptosis effect of MT, but after blocking the PI3K pathway, the effect of MT is weakened. Obviously, MT can protect against SIOP induced by GC. Notably, even after GC-induced ferroptosis begins, MT can confer protection against SIOP. ConclusionOur research confirms that GC-induced ferroptosis is closely related to SIOP. Melatonin can inhibit ferroptosis by activating the PI3K-AKT-mTOR signaling pathway, thereby reducing the occurrence of steroid-induced osteoporosis. Therefore, MT may provide a novel strategy for preventing and treating SIOP.


2012 ◽  
Vol 315 (1) ◽  
pp. 28-37 ◽  
Author(s):  
Wei Zhu ◽  
Ling Huang ◽  
Yahong Li ◽  
Xu Zhang ◽  
Jianmei Gu ◽  
...  

2017 ◽  
Vol 357 (1) ◽  
pp. 25-32 ◽  
Author(s):  
Long-Wei Hu ◽  
Xiao Wang ◽  
Xin-Qun Jiang ◽  
Li-Qun Xu ◽  
Hong-Ya Pan

Sign in / Sign up

Export Citation Format

Share Document