Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Yanyong Yang is active.

Publication


Featured researches published by Yanyong Yang.


Journal of Cellular Biochemistry | 2013

miR‐200c enhances radiosensitivity of human breast cancer cells

Jing Lin; Cong Liu; Fu Gao; R.E.J. Mitchel; Luqian Zhao; Yanyong Yang; Jixiao Lei; Jianming Cai

Due to the intrinsic resistance of many tumors to radiotherapy, current methods to improve the survival of cancer patients largely depend on increasing tumor radiosensitivity. It is well‐known that miR‐200c inhibits epithelial–mesenchymal transition (EMT), and enhances cancer cell chemosensitivity. We sought to clarify the effects of miR‐200c on the radiosensitization of human breast cancer cells. In this study, we found that low levels of miR‐200c expression correlated with radiotolerance in breast cancer cells. miR‐200c overexpression could increase radiosensitivity in breast cancer cells by inhibiting cell proliferation, and by increasing apoptosis and DNA double‐strand breaks. Additionally, we found that miR‐200c directly targeted TANK‐binding kinase 1 (TBK1). However, overexpression of TBK1 partially rescued miR‐200c mediated apoptosis induced by ionizing radiation. In summary, miR‐200c can be a potential target for enhancing the effect of radiation treatment on breast cancer cells. J. Cell. Biochem. 114: 606–615, 2013.


Laboratory Investigation | 2014

Inhibition of TBK1 attenuates radiation-induced epithelial-mesenchymal transition of A549 human lung cancer cells via activation of GSK-3β and repression of ZEB1.

Wen Liu; Yijuan Huang; Cong Liu; Yanyong Yang; Hu Liu; Jianguo Cui; Ying Cheng; Fu Gao; Jianming Cai; Bailong Li

Radiotherapy is an effective treatment method for lung cancer, particularly when the disease is at an advanced stage. However, previous researchers have observed that the majority of patients with conventional radiation therapy develop distant metastases and succumb to the disease. Thus, identifying and understanding novel pathways for the development of new therapeutic targets is a major goal in research on pulmonary neoplasms. Recent studies suggest that epithelial–mesenchymal transition (EMT) is the most important contributor to cancer metastasis. Induction of this complex process requires endogenously produced microRNAs; specifically, downregulation of the miRNA-200c causes an induction of EMT. We recently identified the tank-binding kinase-1 (TBK1) as a downstream effector of the miR-200c-driven pathway, but the biological function of TBK1 in EMT remains unknown. In this study, we tested whether TBK1 has a role in radiation-induced EMT and identified associated potential mechanisms. Human alveolar type II epithelial carcinoma A549 cells were irradiated with 60Co γ-rays. Western blotting revealed a time- and dose-dependent decrease in E-cadherin with a concomitant increase in vimentin after radiation, suggesting that the epithelial cells acquired a mesenchymal-like morphology. TBK1 siRNA significantly inhibited radiation-induced suppression of the epithelial marker E-cadherin and upregulation of the mesenchymal marker vimentin. The invasion and migratory potential of lung cancer cells upon radiation treatment was also reduced by TBK1 knockdown. Furthermore, radiation-induced EMT attenuated by TBK1 depletion was partially dependent on transcriptional factor ZEB1 expression. Finally, we found glycogen synthase kinase-3β (GSK-3β) is involved in regulation of radiation-induced EMT by TBK1. Thus, our findings reveal that TBK1 signaling regulates radiation-induced EMT by controlling GSK-3β phosphorylation and ZEB1 expression. TBK1 may therefore constitute a useful target for treatment of radiotherapy-induced metastasis diseases.


Medical Science Monitor | 2012

Hydrogen-rich saline protects immunocytes from radiation-induced apoptosis.

Yanyong Yang; Bailong Li; Cong Liu; Yunhai Chuai; Jixiao Lei; Fu Gao; Jianguo Cui; Ding Hui Sun; Ying Cheng; Chuanfeng Zhou; Jianming Cai

Summary Background Radiation often causes depletion of immunocytes in tissues and blood, which results in immunosuppression. Molecular hydrogen (H2) has been shown in recent studies to have potential as a safe and effective radioprotective agent through scavenging free radicals. This study was designed to test the hypothesis that H2 could protect immunocytes from ionizing radiation (IR). Material/Methods H2 was dissolved in physiological saline or medium using an apparatus produced by our department. A 2-[6-(4′-hydroxy) phenoxy-3H-xanthen-3-on-9-yl] benzoate (HPF) probe was used to detect intracellular hydroxyl radicals (•OH). Cell apoptosis was evaluated by annexin V-FITC and Propidium iodide (PI) staining as well as the caspase 3 activity. Finally, we examined the hematological changes using an automatic Sysmex XE 2100 hematology analyzer. Results We demonstrated H2-rich medium pretreatment reduced •OH level in AHH-1 cells. We also showed H2 reduced radiation-induced apoptosis in thymocytes and splenocytes in living mice. Radiation-induced caspase 3 activation was also attenuated by H2 treatment. Finally, we found that H2 rescued the radiation-caused depletion of white blood cells (WBC) and platelets (PLT). Conclusions This study suggests that H2 protected the immune system and alleviated the hematological injury induced by IR.


Cellular Physiology and Biochemistry | 2013

GSK-3β Inhibition Attenuates LPS-Induced Death but Aggravates Radiation-Induced Death via Down-Regulation of IL-6

Bailong Li; Chaoxiong Zhang; Feng He; Wen Liu; Yanyong Yang; Hu Liu; Xin Liu; Jie Wang; Lin Zhang; Bo Deng; Fu Gao; Jianguo Cui; Cong Liu; Jianming Cai

Background: Exposure of high dose ionizing radiation is lethal. Signal pathways involved in radiation biology reaction still remain illdefined. Lipopolysaccharides (LPS), the ligands of Toll-like receptor 4(TLR4), could elicit strong immune responses. Glycogen synthase kinase-3β(GSK-3β) promotes the production of inflammatory molecules and cell migration. Inhibition of GSK-3β provides protection against inflammation in animal models. The aim of the study was to investigate role of GSK-3β in LPS shock and ionizing radiation. Methods: WT or IL-6-/-mice or cells were pretreated with SB216763, a GSK-3β inhibitor, and survival of the mice was determined. Cell viability was assayed by Cell Counting Kit. Apoptosis was assayed by Annexin V-PI double staining. Serum concentrations of IL-6 and TNF-α were determined by ELISA. Results: SB216763 attenuated LPS induced mice or cell death but aggravated radiation induced mice or cell death. SB216763 reduced IL-6, but not TNF-α levels in vivo. IL-6-/- mice were more resistant to LPS-induced death but less resistant to radiation-induced death than wild type mice. Conclusions: Inhibition of GSK-3β conferred resistance to LPS shock but fostered death induced by ionizing radiation. Inhibition of GSK-3β was effective by reducing IL-6.


Journal of Cellular and Molecular Medicine | 2014

Protective effect of hydrogen-rich saline against radiation-induced immune dysfunction.

Sanhu Zhao; Yanyong Yang; Wen Liu; Zhiqiang Xuan; Shouming Wu; Shunfei Yu; Ke Mei; Yijuan Huang; Pei Zhang; Jianming Cai; Jin Ni; Yaoxian Zhao

Recent studies showed that hydrogen can be used as an effective radioprotective agent through scavenging free radicals. This study was undertaken to evaluate the radioprotective effects of hydrogen on immune system in mice. H2 was dissolved in physiological saline using an apparatus produced by our department. Spleen index and histological analysis were used to evaluate the splenic structural damage. Spleen superoxide dismutase, GSH, MDA were measured to appraise the antioxidant capacity and a DCF assay for the measurement of radical oxygen species. Cell apoptosis was evaluated by an Annexin V‐FITC and propidium iodide staining method as well as the apoptotic proteins such as Bcl‐2, Bax, caspase‐3 and c‐caspase‐3. CD4+ and CD8+ T cells subtypes were detected by flow cytometry with FITC‐labelled antimouse CD4 and PE antimouse CD8 staining. Real‐time PCR was utilized to determine the CD4+ T cell subtypes and related cytokines. Our study demonstrated that pre‐treatment with H2 could increase the spleen index and attenuate the radiation damage on splenic structure. Radical oxygen species level was also reduced by H2 treatment. H2 also inhibited radiation‐induced apoptosis in splenocytes and down‐regulated pro‐apoptotic proteins in living mice. Radiation‐induced imbalance of T cells was attenuated by H2. Finally, we found that H2 could regulate the polarization of CD4+ T cells and the level of related cytokines. This study suggests H2 as an effective radioprotective agent on immune system by scavenging reactive oxygen species.


International Journal of Radiation Biology | 2013

Molecular hydrogen protects human lymphocyte AHH-1 cells against 12C6+ heavy ion radiation.

Yanyong Yang; Fu Gao; Hong Zhang; Yijuan Hunag; Pei Zhang; Cong Liu; Bailong Li; Jianming Cai

Abstract Purpose: To investigate the potential protective role of molecular hydrogen (H2) against 12C6+ heavy ion radiation, which is a major hazard for space travel and has been also widely used in heavy ion radiotherapy. Materials and methods: H2 was dissolved in Roswell Park Memorial Institute (RPMI) 1640 medium under high pressure (0.4 Mpa) to a saturated level by using an apparatus produced by our department. A 2-[6-(4′-hydroxy) phenoxy-3H-xanthen-3-on-9-yl] benzoate (HPF) probe and a 2′,7′-Dichlorodihydrofluorescein diacetate (H2DCFH-DA) fluorescent dye were used to measure the intracellular reactive oxygen species (ROS) level. Cell apoptosis were determined by double-staining with Annexin V-fluorescein isothiocyanate (Annexin V-FITC) and propidium iodide (PI) as well as a Hoechst 33342 staining method alternatively. Subsequently, cell cycle analysis was performed using a PI staining method and the expression of apoptotic protein was examined by Western blot. Results: In this study, we demonstrated H2 reduced ROS level in Human lymphocyte AHH-1 cells as well as in the radiolysis of water. Our data also showed H2 attenuated 12C6+ radiation- induced cell apoptosis and also alleviated radiation-induced G2/M cell cycle arrest. Heavy ion radiation-induced Caspase 3 activation was also inhibited by H2 treatment. Conclusion: In conclusion, these data showed that H2 attenuated 12C6+ radiation-induced cell apoptosis through reducing the ROS level and modulating apoptotic molecules, thus indicating the potential of H2 as a safe and effective radioprotectant.


Journal of Cellular and Molecular Medicine | 2014

Grape seed pro-anthocyanidins ameliorates radiation-induced lung injury.

Yijuan Huang; Wen Liu; Hu Liu; Yanyong Yang; Jianguo Cui; Pei Zhang; Hainan Zhao; Feng He; Ying Cheng; Jin Ni; Jianming Cai; Bailong Li; Fu Gao

Radiation‐induced lung injury (RILI) is a potentially fatal and dose‐limiting complication of thoracic radiotherapy. This study was to investigate the protective effects of grape seed pro‐anthocyanidins (GSPs), an efficient antioxidant and anti‐carcinogenic agent, on RILI. In our study, it was demonstrated that acute and late RILI was ameliorated after GSPs treatment possibly through suppressing TGF‐β1/Smad3/Snail signalling pathway and modulating the levels of cytokines (interferon‐γ, IL‐4 and IL‐13) derived from Th1/Th2 cells. In addition, a sustained high level of PGE2 was also maintained by GSPs treatment to limited fibroblast functions. As shown by electron spin resonance spectrometry, GSPs could scavenge hydroxyl radical (•OH) in a dose‐dependent manner, which might account for the mitigation of lipid peroxidation and consequent apoptosis of lung cells. In vitro, GSPs radiosensitized lung cancer cell A549 while mitigating radiation injury on normal alveolar epithelial cell RLE‐6TN. In conclusion, the results showed that GSPs protects mice from RILI through scavenging free radicals and modulating RILI‐associated cytokines, suggesting GSPs as a novel protective agent in RILI.


Cellular Physiology and Biochemistry | 2016

Protective Effects of Myrtol Standardized Against Radiation-Induced Lung Injury.

Deyun Zhao; Hong-jin Qu; Jiaming Guo; Hainan Zhao; Yanyong Yang; Pei Zhang; Kun Cao; Xiao Lei; Jianguo Cui; Cong Liu; Jianming Cai; Fu Gao; Bailong Li

Background/Aims: As a major complication after thoracic radiotherapy, radiation-induced lung injury (RILI) has great impact on long term quality of life and could result in fatal respiratory insufficiency The present study was aimed to evaluate the effects of Myrtol standardized on RILI, and to investigate the underlying mechanism. Methods: A mouse model of radiation-induced lung injury was generated by using thoracic irradiation with a single dose of 16Gy. Mice were orally administrated with Myrtol (25 mg/kg/day) for 4 weeks after irradiation, while prednisone (5 mg/kg/day) was used as a positive control. After then, the body weight and lung coefficient were calculated. The severity of fibrosis was evaluated by observing pulmonary sections after radiation and collagen content in lung tissues was calculated following the hydroxyproline (HYP) assay. Pathological changes were observed in all the groups by using HE staining and Masson staining. The serum levels of TGF-β1, TNF-α, IL-1β, IL-6, and PGE2 were also measured with an ELISA assay. Western blot assay was used to measure the impact of Myrtol on AKT and its downstream signaling pathway, including MMP-2 and MMP-9. The levels of Vimentin and α-SMA were evaluated with an immunofluorescence assay. Results: Treatment with Myrtol standardized, but not prednisone, reduced lung coefficient and collagen deposition in lung tissues, while attenuated histological damages induced by irradiation. Myrtol standardized also reduced the production of MDA, while increased the level of SOD. It was also observed that Myrtol standardized inhibited TGF-β1 and a series of pro-inflammatory cytokines including TNF-α, IL-1β, IL-6, PGE2. While in prednisone group, even though the early pneumonitis was ameliorated, the collagen disposition remained unchanged in latter times. Immunofluorescence analysis also revealed elevation of vimentin and α-SMA in the alveoli after a single dose of 16Gy. Conclusion: The present results suggest Myrtol standardized as an effective agent for attenuating the lung injury induced by irradiation.


Cellular Physiology and Biochemistry | 2013

Therapeutic Effects of Hydrogen-Rich Solution on Aplastic Anemia in Vivo

Sanhu Zhao; Ke Mei; Liren Qian; Yanyong Yang; Wen Liu; Yijuan Huang; Chao Zhang; Xunjun Sun; Cong Liu; Bailong Li; Fu Gao; Jianming Cai; Jin Ni

Background: Aplasitc anemia (AA) is a bone marrow failure syndrome characterized by an immune-mediated destruction of hematopoietic stem cells. Though clinical symptoms could be ameliorated by bone marrow transplantation and/or immunosuppressive therapy, frequent recurrence and especially evolution of clonal hematologic diseases remains problematic clinically. Cytokines such as interferon-γ (INF-γ), tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) secreted by autologous T cells are closely related with the development of AA. Hydrogen-rich solution was reported to inhibit the levels of cytokines including INF-γ, TNF-α and IL-6 in vivo in recent studies. This study was to investigate the potential therapeutic effects of hydrogen-rich solution on AA in vivo. Methods: AA model was determined in vivo by mice and body weights of the mice were used as the basic physiological index. Peripheral blood cells were calculated to evaluate the hematologic recovery degree. Bone marrow nucleated cells (BMNCs), tissue histology, as well as CFU-S and CFU-GM forming units were used to evaluate the recovery of bone marrow microenvironment. The ratio of CD4+ and CD8+ cells were examined along with cytokine levels in serum to determine the efficacy of H2-rich solution on the affected immunological functions. Results: Body weight and number of peripheral blood cells were significantly improved for mice in the H2-rich solution treated groups as compared with those with AA. The number of BMNCs and CFUs increased markedly and the bone marrow microenvironment was also improved significantly. The experimental group restrained the cell apoptosis, relieved hyperemia and accelerated tissue repair. The number of CD4+ and CD8+ cells as well as the ratio of CD4/CD8 increased to normal gradually, while the levels of TNF-α, IFN-γ, and IL-6 in serum decreased after H2-rich solution treatment. Conclusion: Our study firstly showed that hydrogen-rich solution accelerated the recovery of either hematological or immunological recovery on aplastic anemia mice. This finding suggests hydrogen-rich solution as a potential clinical therapeutic agent for AA.


Cellular Physiology and Biochemistry | 2016

Radioprotective Effects of Heat-Killed Mycobacterium Tuberculosis in Cultured Cells and Radiosensitive Tissues

Yuanyuan Chen; Yang Xu; Jicong Du; Jiaming Guo; Xiao Lei; Jianguo Cui; Cong Liu; Ying Cheng; Bailong Li; Fu Gao; Jintao Ju; Jianming Cai; Yanyong Yang

Background: Exposure to ionizing radiation (IR) often causes severe damage to radiosensitive tissues, which limits the use of radiotherapy in cancer patients. Novel safe and effective radioprotectant is urgently required. It has been reported toll like receptor 2 (TLR2) plays a critical role in radioresistance. In this study, we demonstrated the protective effects of Heat-Killed Mycobacterium tuberculosis (HKMT), a potent TLR2 agonist, against IR. Methods: Cell survival and apoptosis were determined by CCK-8 assay and Annexin V assay, respectively. An immunofluorescence staining assay was used to detect the translocation of nuclear faktor-kappa beta (NF-kB) p65. Tissue damage was evaluated by Haematoxilin-Eosin (HE) staining assay. We also used a flow cytometry assay to measure the number of nucleated cells and CD34+ hemopoietic stem cells in bone marrow. A western blot assay was used to detect the changes of proteins involving TLR signaling pathway. Results: We found that HKMT increased cell viability and inhibited cell apoptosis after irradiation. HKMT induced NF-kB translocation and activated Erk1/2, p38 signaling pathway. HKMT also protected bone marrow and testis from destruction. Radiation-induced decreases of nucleated cells and CD34+ hemopoietic stem cells in bone marrow were also inhibited by HKMT treatment. We found that radiation caused increase of inflammatory cytokines was also suppressed by HKMT. Conclusion: Our data showed that HKMT exhibited radioprotective effects in vivo and in vitro through activating NF-kB and MAPK signaling pathway, suggesting a potential of HKMT as novel radioprotector.

Collaboration


Dive into the Yanyong Yang's collaboration.

Top Co-Authors

Avatar

Fu Gao

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jianming Cai

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Bailong Li

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jianguo Cui

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Pei Zhang

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Hu Liu

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Ying Cheng

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jiaming Guo

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Wen Liu

Second Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge