scholarly journals Mesenchymal stem cells suppress leukemia via macrophage-mediated functional restoration of bone marrow microenvironment

Leukemia ◽  
2020 ◽  
Vol 34 (9) ◽  
pp. 2375-2383 ◽  
Author(s):  
Chengxiang Xia ◽  
Tongjie Wang ◽  
Hui Cheng ◽  
Yong Dong ◽  
Qitong Weng ◽  
...  
Author(s):  
Xiaofang Chen ◽  
Ningyu Li ◽  
Jianyu Weng ◽  
Xin Du

Myelodysplastic syndrome (MDS) is a group of clonal hematopoietic disorders related to hematopoietic stem and progenitor cell dysfunction. However, therapies that are currently used to target hematopoietic stem cells are not effective. These therapies are able to slow the evolution toward acute myeloid leukemia but cannot eradicate the disease. Mesenchymal stem cells (MSCs) have been identified as one of the main cellular components of the bone marrow microenvironment, which plays an indispensable role in normal hematopoiesis. When functional and regenerative capacities of aging MSCs are diminished, some enter replicative senescence, which promotes inflammation and disease progression. Recent studies that investigated the contribution of bone marrow microenvironment and MSCs to the initiation and progression of the disease have offered new insights into the MDS. This review presents the latest updates on the role of MSCs in the MDS and discusses potential targets for the treatment of MDS.


2020 ◽  
Vol 2020 ◽  
pp. 1-13
Author(s):  
Xiaoyan Zhang ◽  
Yazhi Yang ◽  
Yang Yang ◽  
Huijun Chen ◽  
Huaijun Tu ◽  
...  

Although major advances have been achieved in the treatment of chronic myeloid leukemia (CML) by using tyrosine kinase inhibitors, patients relapse after withdrawal and need long-term medication. This reflects the CML clones have not been eliminated completely. The precise mechanisms for the maintenance of CML cells are not yet fully understood. The bone marrow microenvironment constitutes the sanctuary for leukemic cells. Mesenchymal stem cells (MSC) are an important component of the bone marrow microenvironment (BM). It plays an important role in the development and drug resistance of CML. Accumulating evidence indicates that exosomes play a vital role in cell-to-cell communication. We successfully isolated and purified exosomes from human bone marrow microenvironment-derived mesenchymal stem cells (hBMMSC-Exo) by serial centrifugation. In the present study, we investigated the effect of hBMMSC-Exo on the proliferation, apoptosis, and drug resistance of CML cells. The results demonstrated that hBMMSC-Exo had the ability to inhibit the proliferation of CML cells in vitro via miR-15a and arrest cell cycle in the G0/G1 phase. However, the results obtained from BALB/c nu/nu mice studies apparently contradicted the in vitro results. In fact, hBMMSC-Exo increased tumor incidence and promoted tumor growth in vivo. Further study showed the antiapoptotic protein Bcl-2 expression increased, whereas the Caspase3 expression decreased. Moreover, the in vivo study in the xenograft tumor model showed that hBMMSC-Exo promoted the proliferation and decreased the sensitivity of CML cells to tyrosine kinase inhibitors, resulting in drug resistance. These results demonstrated that hBMMSC-Exo supported the maintenance of CML cells and drug resistance in BM by cell-extrinsic protective mechanisms. They also suggested that hBMMSC-Exo might be a potential target to overcome the microenvironment-mediated drug resistance.


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<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 ◽  
Author(s):  
Xin Sun ◽  
Liyi Huang ◽  
Chenying Fu ◽  
Lijuan Li ◽  
Lu Wang ◽  
...  

Abstract Background: Spinal cord injury (SCI) typically results in a devastating loss of neurological function below the level of injury. Although many strategies show considerable potential for SCI treatment, the therapeutic efficacy is limited. Here, we used a mouse model of thoracic contusive SCI to investigate whether the combination of bone marrow mesenchymal stem cells (BMMSCs) transplantation and exercise training has a synergistic effect on functional restoration. Methods: BMMSCs were injected directly into the contusion epicenter immediately after SCI, and the mice started treadmill training (TMT) 3 days after SCI. Locomotor function was evaluated by the Basso Mouse Scale (BMS), horizontal ladder test, and footprint analysis. Histological examination, transmission electron microscopy observation, immunofluorescence staining and western blotting were performed 8 weeks after SCI to further explore the potential mechanism of the synergistic repair effect. Results: The combination of BMMSCs transplantation and TMT showed the best therapeutic effect on motor function recovery compared with the other treatment groups. Further investigations revealed that the combination of BMMSCs transplantation and TMT markedly reduced fibrotic scar tissue, protected neurons, promoted axon and myelin regeneration, and increased synapse formation to a larger extent than either TMT or BMMSCs transplantation alone. Additionally, the synergistic effects of BMMSCs transplantation and TMT on SCI recovery occurred via activation of the PI3K/AKT/mTOR pathway. Conclusions: These findings suggest that BMMSCs transplantation combined with exercise training represents a promising combinatorial strategy to facilitate clinically meaningful recovery after SCI.


Sign in / Sign up

Export Citation Format

Share Document