scholarly journals Graphene-modified CePO4 nanorods effectively treat breast cancer-induced bone metastases and regulate macrophage polarization to improve osteo-inductive ability

2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Yu-Wei Ge ◽  
Xiao-Liang Liu ◽  
De-gang Yu ◽  
Zhen-An Zhu ◽  
Qin-Fei Ke ◽  
...  

Abstract Background Breast cancer bone metastasis has become one of the most common complications; however, it may cause cancer recurrence and bone nonunion, as well as local bone defects. Methods Herein, In vitro, we verified the effect of bioscaffold materials on cell proliferation and apoptosis through a CCK8 trial, staining of live/dead cells, and flow cytometry. We used immunofluorescence technology and flow cytometry to verify whether bioscaffold materials regulate macrophage polarization, and we used ALP staining, alizarin red staining and PCR to verify whether bioscaffold material promotes bone regeneration. In vivo, we once again studied the effect of bioscaffold materials on tumors by measuring tumor volume in mice, Tunel staining, and caspase-3 immunofluorescence. We also constructed a mouse skull ultimate defect model to verify the effect on bone regeneration. Results Graphene oxide (GO) nanoparticles, hydrated CePO4 nanorods and bioactive chitosan (CS) are combined to form a bioactive multifunctional CePO4/CS/GO scaffold, with characteristics such as photothermal therapy to kill tumors, macrophage polarization to promote blood vessel formation, and induction of bone formation. CePO4/CS/GO scaffold activates the caspase-3 proteasein local tumor cells, thereby lysing the DNA between nucleosomes and causing apoptosis. On the one hand, the as-released Ce3+ ions promote M2 polarization of macrophages, which secretes vascular endothelial growth factor (VEGF) and Arginase-1 (Arg-1), which promotes angiogenesis. On the other hand, the as-released Ce3+ ions also activated the BMP-2/Smad signaling pathway which facilitated bone tissue regeneration. Conclusion The multifunctional CePO4/CS/GO scaffolds may become a promising platform for therapy of breast cancer bone metastases.

2020 ◽  
Author(s):  
Yuwei Ge ◽  
Xiao-Liang Liu ◽  
De-gang Yu ◽  
Zhen-An Zhu ◽  
Qin-Fei Ke ◽  
...  

Abstract Background: Breast cancer bone metastasis has become one of the most common complications; however, it may cause cancer recurrence and bone nonunion, as well as local bone defects. Methods: Herein, In vitro, we verified the effect of bioscaffold materials on cell proliferation and apoptosis through a CCK8 trial, staining of live/dead cells, and flow cytometry. We used immunofluorescence technology and flow cytometry to verify whether bioscaffold materials regulate macrophage polarization, and we used ALP staining, alizarin red staining and PCR to verify whether bioscaffold material promotes bone regeneration. In vivo, we once again studied the effect of bioscaffold materials on tumors by measuring tumor volume in mice, Tunel staining, and caspase-3 immunofluorescence. We also constructed a mouse skull ultimate defect model to verify the effect on bone regeneration. Results: Graphene oxide (GO) nanoparticles, hydrated CePO4 nanorods and bioactive chitosan (CS) are combined to form a bioactive multifunctional CePO4/CS/GO scaffold, with characteristics such as photothermal therapy to kill tumors, macrophage polarization to promote blood vessel formation, and induction of bone formation. CePO4/CS/GO scaffold activates the caspase-3 proteasein local tumor cells, thereby lysing the DNA between nucleosomes and causing apoptosis. On the one hand, the as-released Ce3+ ions promote M2 polarization of macrophages, which secretes vascular endothelial growth factor (VEGF) and Arginase-1 (Arg-1), which promotes angiogenesis. On the other hand, the as-released Ce3+ ions also activated the BMP-2/Smad signaling pathway which facilitated bone tissue regeneration. Conclusion: the multifunctional CePO4/CS/GO scaffolds may become a promising platform for therapy of breast cancer bone metastases.


2020 ◽  
Author(s):  
Yuwei Ge ◽  
Xiao-Liang Liu ◽  
De-gang Yu ◽  
Zhen-An Zhu ◽  
Qin-Fei Ke ◽  
...  

Abstract Background Breast cancer bone metastasis has become one of the most common complications; however, it may cause cancer recurrence and bone nonunion and local bone defects.Methods Herein, In vitro, we verified the effect of bioscaffold materials on cell proliferation and apoptosis through CCK8 experiment, staining of living dead cells, and flow cytometry. We used immunofluorescence technology and flow cytometry to verified whether bioscaffold materials regulated the polarization of macrophages, and we use ALP staining, alizarin red staining and PCR to verify whether the bioscaffold material promotes bone regeneration. In vivo, we once again studied the effect of bioscaffold materials on tumors by measuring tumor volume in mice, Tunel staining, and caspase-3 immunofluorescence. We also constructed a mouse skull ultimate defect model to verify the effect on bone regeneration.Results Graphene oxide (GO) nanoparticles, hydrated CePO4 nanorods and bioactive chitosan (CS) are combined to form a bioactive multifunctional CePO4/CS/GO scaffolds, which has the following characteristics such as photothermal therapy to kill tumors, macrophage polarization promotes blood vessel formation and induces bone formation. The CePO4/CS/GO scaffold activates the caspase-3 proteasein local tumor cells, thereby lysing DNA between nucleosomes and causing apoptosis. On the one hand, the as-released Ce3+ ions promote the M2 polarization of macrophages, which secretes vascular endothelial growth factor (VEGF) and Arginase-1 (Arg-1), which promotes angiogenesis. On the other hand, the as-released Ce3+ ions also activated BMP-2/Smad signaling pathway that facilitated bone tissue regeneration.Conclusion the multifunctional CePO4/CS/GO scaffolds may become a promising platform for therapy of breast cancer bone metastases.


2020 ◽  
Author(s):  
Yuwei Ge ◽  
Xiao-Liang Liu ◽  
De-gang Yu ◽  
Zhen-An Zhu ◽  
Qin-Fei Ke ◽  
...  

Abstract Background: Breast cancer bone metastasis has become one of the most common complications; however, it may cause cancer recurrence and bone nonunion and local bone defects.Methods: Herein, In vitro, we verified the effect of bioscaffold materials on cell proliferation and apoptosis through CCK8 experiment, staining of living dead cells, and flow cytometry. We used immunofluorescence technology and flow cytometry to verified whether bioscaffold materials regulated the polarization of macrophages, and we use ALP staining, alizarin red staining and PCR to verify whether the bioscaffold material promotes bone regeneration. In vivo, we once again studied the effect of bioscaffold materials on tumors by measuring tumor volume in mice, Tunel staining, and caspase-3 immunofluorescence. We also constructed a mouse skull ultimate defect model to verify the effect on bone regeneration.Results: Graphene oxide (GO) nanoparticles, hydrated CePO4 nanorods and bioactive chitosan (CS) are combined to form a bioactive multifunctional CePO4/CS/GO scaffolds, which has the following characteristics such as photothermal therapy to kill tumors, macrophage polarization promotes blood vessel formation and induces bone formation. The CePO4/CS/GO scaffold activates the caspase-3 proteasein local tumor cells, thereby lysing DNA between nucleosomes and causing apoptosis. On the one hand, the as-released Ce3+ ions promote the M2 polarization of macrophages, which secretes vascular endothelial growth factor (VEGF) and Arginase-1 (Arg-1), which promotes angiogenesis. On the other hand, the as-released Ce3+ ions also activated BMP-2/Smad signaling pathway that facilitated bone tissue regeneration.Conclusion: the multifunctional CePO4/CS/GO scaffolds may become a promising platform for therapy of breast cancer bone metastases.


2021 ◽  
Vol 11 (4) ◽  
pp. 778-785
Author(s):  
Xiaolin Chen ◽  
Yan Wang ◽  
Sunlu Jiang

Our study investigates the effect of high expression of Sirt2 in MSCs (MSCs-Sirt2) on Her-2 breast cancer cell proliferation. A mouse subcutaneous xenograft tumor model was established and MSCssirt2 analysis was performed on nude mice. TUNEL staining, flow cytometry, western-blot, real-time PCR and immunohistochemistry were used to detect cancer cell apoptosis. The number of NK cells infiltrated by flow cytometry detected the tumor tissue of tumor-bearing mice, and its killing activity on tumor-bearing mice was detected by isotope labeling and release method. The levels of TNF-α, IFN-γ, IL-8, IL-6 and IL-10 were detected by ELISA. Caspase-3 level was decreased in the MSCs group (P <0.01) while increased in the MSCs-sirt2 group (P <0.001). However, PCNA expression showed an opposite profile in the Her-2 group and MSCs-sirt2 group compared to Caspase-3 level (P <0.01). The tumor volume and weight in the MSCs-sirt2 group was significantly reduced (P < 0.01), while increased in the MSCs group significantly (P < 0.05). The number of Ki-67-positive tumor cells in MSCs-sirt2 group was significantly reduced (P <0.01) and increased in MSCs group (P < 0.001) with oppositive number of TUNEL-positive tumor cells in the MSCs-sirt2 group and MSCs group (P <0.01). IFN-γ level showed an upward trend (P <0.001). The NK cell toxicity of MSCs-Sirt2 group was significantly higher (P <0.001). MSCs-Sirt2 has an inhibitory effect on Her-2 breast cancer cell growth by enhancing the local inflammatory response of NK cells.


2021 ◽  
Vol 22 (1) ◽  
Author(s):  
Atena Daneshforouz ◽  
Samad Nazemi ◽  
Omid Gholami ◽  
Marzieh Kafami ◽  
Bahareh Amin

Abstract Background Despite significant advancements in breast cancer therapy, novel drugs with lower side effects are still being demanded. In this regard, we investigated the anti-cancer features of verbascoside in 4 T1 mouse mammary tumor cell. Methods First, MTT assay was performed with various concentrations (ranging between 5 to 200 μM) of verbascoside and IC50 was calculated. Then the expression of Bax, Bcl-2, and caspase-3 was evaluated in treated 4 T1 cells. In addition, we investigated the expression of TLR4, MyD88, and NF-κB to ascertain the underlying mechanism of the anti-proliferative feature of verbascoside. Also, flow cytometry followed by double PI and Annexin V was conducted to confirm the apoptosis-inducing effect of verbascoside. Results Our results from MTT assay showed verbascoside inhibits proliferation of 4 T1 cancer cells (IC50 117 μM) while is safe for normal HEK293T cells. By qRT-PCR, we observed that verbascoside treatment (100, 117 and, 130 μM) increases the expression of caspase-3 and Bax while reduces the expression of Bcl-2. Also, verbascoside (100, 117 and, 130 μM) increased the expression of TLR4 only at 130 μM dose and the expression of MyD88 whereas reduced the expression of NF-κB at mRNA level. Flow cytometry analysis also confirmed verbascoside induces apoptosis in 4 T1 cells at 117 μM. Conclusion Taken together, our data showed verbascoside is a safe natural compound for normal cells while has apoptosis-inducing feature through TLR4 axis on 4 T1 cells.


2020 ◽  
Author(s):  
Feng Wang ◽  
Lingchi Kong ◽  
Wenbo Wang ◽  
Li Shi ◽  
Mengwei Wang ◽  
...  

Abstract Background Both advanced glycation end products (AGEs) and AGE-mediated M1 macrophage polarization contribute to bone marrow mesenchymal stem cell (BMSC) dysfunction, leading to impaired bone regeneration in type 1 diabetes mellitus (T1DM). Adrenomedullin 2 (ADM2), an endogenous bioactive peptide belonging to the calcitonin gene-related peptide family, exhibits various biological activities associated with the inhibition of inflammation and reduction of insulin resistance. However, the effects and underlying mechanisms of ADM2 in AGE-induced macrophage M1 polarization, BMSC dysfunction, and impaired bone regeneration remain poorly understood. Methods The polarization of bone marrow-derived macrophages was verified by flow cytometry analysis. In addition, alkaline phosphatase (ALP) staining, ALP activity detection, and alizarin red staining were performed to assess the osteogenesis of BMSCs. Quantitative real-time polymerase chain reaction, enzyme-linked immunosorbent assay, western blotting, and immunofluorescence staining were used to assess polarization markers, PPARγ/IκBα/NF-κB signaling, and osteogenic markers. In vivo, a distraction osteogenesis (DO) rat model with T1DM was established, and the tibia samples were collected at different time points for radiological, biomechanical, and histological analyses, to verify the effects of ADM2 in terms of bone regeneration and M2 polarization under diabetic conditions. Results ADM2 treatment reversed the M1 macrophage polarization induced by AGEs towards the M2 phenotype, which was partially achieved by the PPARγ-mediated inhibition of NF-κB signaling. The PPARγ inhibitor GW9662 significantly attenuated the effects of ADM2. Besides, ADM2 treatment improved the AGE-impaired osteogenic potential of BMSCs in vitro. Furthermore, ADM2 accelerated bone regeneration, as revealed by improved radiological and histological manifestations and biomechanical parameters, accompanied by improved M2 macrophage polarization in diabetic DO rats, and these effects were partially blocked by GW9662 administration. Conclusions These results indicate that ADM2 enhances diabetic bone regeneration during DO, by attenuating AGE-induced imbalance in macrophage polarization, partly through PPARγ/IκBα/NF-κB signaling, and improving AGE-impaired osteogenic differentiation of BMSCs simultaneously. These findings reveal that ADM2 may serve as a potential bioactive factor for promoting bone regeneration under diabetic conditions, and imply that management of inflammation and osteogenesis, in parallel, might be a promising therapeutic strategy for diabetic patients during DO treatment.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Feng Wang ◽  
Lingchi Kong ◽  
Wenbo Wang ◽  
Li Shi ◽  
Mengwei Wang ◽  
...  

Abstract Background Both advanced glycation end products (AGEs) and AGE-mediated M1 macrophage polarization contribute to bone marrow mesenchymal stem cell (BMSC) dysfunction, leading to impaired bone regeneration in type 1 diabetes mellitus (T1DM). Adrenomedullin 2 (ADM2), an endogenous bioactive peptide belonging to the calcitonin gene-related peptide family, exhibits various biological activities associated with the inhibition of inflammation and reduction of insulin resistance. However, the effects and underlying mechanisms of ADM2 in AGE-induced macrophage M1 polarization, BMSC dysfunction, and impaired bone regeneration remain poorly understood. Methods The polarization of bone marrow-derived macrophages was verified using flow cytometry analysis. Alkaline phosphatase (ALP) staining, ALP activity detection, and alizarin red staining were performed to assess the osteogenesis of BMSCs. Quantitative real-time polymerase chain reaction, enzyme-linked immunosorbent assay, western blotting, and immunofluorescence staining were used to assess polarization markers, nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, and osteogenic markers. In vivo, a distraction osteogenesis (DO) rat model with T1DM was established, and tibia samples were collected at different time points for radiological, biomechanical, and histological analyses, to verify the effects of ADM2 on bone regeneration and M2 polarization under diabetic conditions. Results ADM2 treatment reversed AGE-induced M1 macrophage polarization towards the M2 phenotype, which was partially achieved by the peroxisome proliferator-activated receptor γ (PPARγ)-mediated inhibition of NF-κB signaling. The PPARγ inhibitor GW9662 significantly attenuated the effects of ADM2. Besides, ADM2 treatment improved the AGE-impaired osteogenic potential of BMSCs in vitro. Furthermore, ADM2 accelerated bone regeneration, as revealed by improved radiological and histological manifestations and biomechanical parameters, accompanied by improved M2 macrophage polarization in diabetic DO rats, and these effects were partially blocked by GW9662 administration. Conclusions These results indicate that ADM2 enhances diabetic bone regeneration during DO, by attenuating AGE-induced imbalances in macrophage polarization, partly through PPARγ/NF-κB signaling, and improving AGE-impaired osteogenic differentiation of BMSCs simultaneously. These findings reveal that ADM2 may serve as a potential bioactive factor for promoting bone regeneration under diabetic conditions, and imply that management of inflammation and osteogenesis, in parallel, may present a promising therapeutic strategy for diabetic patients during DO treatment.


Author(s):  
Masoumeh Dolati ◽  
Farzaneh Tafvizi ◽  
Masoud Salehipour ◽  
Tahereh Komeili Movahed ◽  
Parvaneh Jafari

Background and Objectives: Secondary metabolites in the supernatants of probiotic microorganisms have shown antican- cer effects. The present study was aimed to investigate the cytotoxicity of Bacillus coagulans supernatants and their role in apoptosis induction in MCF7 cancer cells. Materials and Methods: The inhibition of MCF7 cancer cells by Bacillus coagulans supernatants was assessed by MTT assay at three exposure times of 24, 48, and 72 h. Apoptosis induction was explored by flow cytometry while the expression levels of bax, caspase 3, caspase 9, and bcl2 were examined by real-time PCR and compared with normal HFF cells. Results: Bacillus coagulans supernatants exhibited inhibitory effects on MCF7 cells in a concentration-dependent and time-dependent manner; while lower cytotoxic effects were observed in normal HFF cells. The increase in the expression of bax, caspase 3, and caspase 9 genes and the decrease in the anti-apoptotic gene of bcl2, along with the flow cytometry results, confirmed the induction of apoptosis in the cancer cells. Conclusion: Regarding the cytotoxic influence of Bacillus coagulans supernatants against breast cancer cells, this bacterium can be considered as a potential candidate for a novel therapeutic strategy with lower side effects which of course requires further investigations.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Yu-Wei Ge ◽  
Xiao-Liang Liu ◽  
De-gang Yu ◽  
Zhen-An Zhu ◽  
Qin-Fei Ke ◽  
...  

An amendment to this paper has been published and can be accessed via the original article.


Sign in / Sign up

Export Citation Format

Share Document