Network


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

Hotspot


Dive into the research topics where Xia Xiao is active.

Publication


Featured researches published by Xia Xiao.


Scientific Reports | 2015

ROS-mediated iron overload injures the hematopoiesis of bone marrow by damaging hematopoietic stem/progenitor cells in mice

Xiao Chai; Deguan Li; Xiaoli Cao; Yuchen Zhang; Juan Mu; Wenyi Lu; Xia Xiao; Chengcheng Li; Juanxia Meng; Jie Chen; Qing Li; Jishi Wang; Aimin Meng; Mingfeng Zhao

Iron overload, caused by hereditary hemochromatosis or repeated blood transfusions in some diseases, such as beta thalassemia, bone marrow failure and myelodysplastic syndrome, can significantly induce injured bone marrow (BM) function as well as parenchyma organ dysfunctions. However, the effect of iron overload and its mechanism remain elusive. In this study, we investigated the effects of iron overload on the hematopoietic stem and progenitor cells (HSPCs) from a mouse model. Our results showed that iron overload markedly decreased the ratio and clonogenic function of murine HSPCs by the elevation of reactive oxygen species (ROS). This finding is supported by the results of NAC or DFX treatment, which reduced ROS level by inhibiting NOX4 and p38MAPK and improved the long-term and multi-lineage engrafment of iron overload HSCs after transplantation. Therefore, all of these data demonstrate that iron overload injures the hematopoiesis of BM by enhancing ROS through NOX4 and p38MAPK. This will be helpful for the treatment of iron overload in patients with hematopoietic dysfunction.


PLOS ONE | 2015

Effects of Iron Overload on the Bone Marrow Microenvironment in Mice

Yuchen Zhang; Wenjing Zhai; Mingfeng Zhao; Deguan Li; Xiao Chai; Xiaoli Cao; Juanxia Meng; Jie Chen; Xia Xiao; Qing Li; Juan Mu; Jichun Shen; Aimin Meng

Objective Using a mouse model, Iron Overload (IO) induced bone marrow microenvironment injury was investigated, focusing on the involvement of reactive oxygen species (ROS). Methods Mice were intraperitoneally injected with iron dextran (12.5, 25, or 50mg) every three days for two, four, and six week durations. Deferasirox(DFX)125mg/ml and N-acetyl-L-cysteine (NAC) 40mM were co-administered. Then, bone marrow derived mesenchymal stem cells (BM-MSCs) were isolated and assessed for proliferation and differentiation ability, as well as related gene changes. Immunohistochemical analysis assessed the expression of haematopoietic chemokines. Supporting functions of BM-MSCs were studied by co-culture system. Results In IO condition (25mg/ml for 4 weeks), BM-MSCs exhibited proliferation deficiencies and unbalanced osteogenic/adipogenic differentiation. The IO BM-MSCs showed a longer double time (2.07±0.14 days) than control (1.03±0.07 days) (P<0.05). The immunohistochemical analysis demonstrated that chemokine stromal cell-derived factor-1, stem cell factor -1, and vascular endothelial growth factor-1 expression were decreased. The co-cultured system demonstrated that bone marrow mononuclear cells (BMMNCs) co-cultured with IO BM-MSCs had decreased colony forming unit (CFU) count (p<0.01), which indicates IO could lead to decreased hematopoietic supporting functions of BM-MSCs. This effect was associated with elevated phosphatidylinositol 3 kinase (PI3K) and reduced of Forkhead box protein O3 (FOXO3) mRNA expression, which could induce the generation of ROS. Results also demonstrated that NAC or DFX treatment could partially attenuate cell injury and inhibit signaling pathway striggered by IO. Conclusion These results demonstrated that IO can impair the bone marrow microenvironment, including the quantity and quality of BM-MSCs.


Asian Pacific Journal of Cancer Prevention | 2013

Multiple Cytotoxic Factors Involved in IL-21 Enhanced Antitumor Function of CIK Cells Signaled through STAT-3 and STAT5b Pathways

Sajin Rajbhandary; Mingfeng Zhao; Nan Zhao; Wenyi Lu; Haibo Zhu; Xia Xiao; Qi Deng; Yuming Li

BACKGROUND/OBJECTIVES Maintenance of cellular function in culture is vital for transfer and development following adoptive immunotherapy. Dual properties of IL-21 in activating T cells and reducing activation induced cell death led us to explore the mechanism of action of IL-21 enhanced proliferation and cytotoxic potential of CIK cells. METHOD CIK cells cultured from PBMCs of healthy subjects were stimulated with IL-21 and cellular viability and cytotoxicity to K562 cells were measured. To elucidate the mechanism of action of IL-21, mRNA expression of cytotoxic factors was assessed by RT-PCR and protein expression of significantly important cytotoxic factors and cytokine secretion were determined through flow cytometry and ELISA. Western blotting was performed to check the involvement of the JAK/STAT pathway following stimulation. RESULTS We found that IL-21 did not enhance in vitro proliferation of CIK cells, but did increase the number of cells expressing the CD3+/ CD56+ phenotype. Cytotoxic potential was increased with corresponding increase in perforin (0.9831±0.1265 to 0.7592±0.1457), granzyme B (0.4084±0.1589 to 0.7319±0.1639) and FasL (0.4015±0.2842 to 0.7381±0.2568). Interferon gamma and TNF-alpha were noted to increase (25.8±6.1 ng/L to 56.0±2.3 ng/L; and 5.64±0.61 μg/L to 15.14±0.93 μg/L, respectively) while no significant differences were observed in the expression of granzyme A, TNF-alpha and NKG2D, and NKG2D. We further affirmed that IL-21 signals through the STAT-3 and STAT- 5b signaling pathway in the CIK cell pool. CONCLUSION IL-21 enhances cytotoxic potential of CIK cells through increasing expression of perforin, granzyme B, IFN-gamma and TNF-alpha. The effect is brought about by the activation of STAT-3 and STAT-5b proteins.


Oncology Letters | 2014

Effect of CIK on multidrug-resistance reversal and increasing the sensitivity of ADR in K562/ADR cells.

Lei Wang; Qi Deng; Jian Wang; Xue Bai; Xia Xiao; Hai‑Rong Lv; Ming‑Feng Zhao; Peng‑Jiang Liu

Leukemia is a leading cause of cancer-related mortality in children worldwide, and multidrug-resistance (MDR) is a main reason for tumor chemotherapy failure. The present study investigated the effects of ADR following incubation with cytokine-induced killer (CIK) cells on reversing MDR in K562/ADR cells. Mononuclear cells were isolated from the peripheral blood of healthy individuals and cultured in vitro in the presence of a combination of cytokines to generate CIK for K562/ADR cell treatment. A decreased level of P-glycoprotein expression and glutathione (GSH), an increased intracellular Rh-123 content, decreased mRNA and protein expression levels of MDR gene 1, MDR-associated protein 1, GSH S-transferase-π, B-cell lymphoma 2 and Survivin, and the decreased phosphorylation of AKT and the transcriptional activity of nuclear factor-κB and activator protein 1 were detected following ADR treatment in CIK co-cultured K562/ADR cells. Additionally, the level of ADR sensitivity and the apoptosis rate were increased in the CIK co-cultured K562/ADR cells. These results indicate that pre-treatment with CIK could reverse the MDR of K562/ADR cells, and that patients would be most likely to benefit from the combination of chemotherapy and CIK therapy.


Experimental and Therapeutic Medicine | 2015

Anti-CD20 antibody induces the improvement of cytokine-induced killer cell activity via the STAT and MAPK/ERK signaling pathways

Qi Deng; Xue Bai; Hai‑Rong Lv; Xia Xiao; Ming‑Feng Zhao; Yuming Li

There is a current requirement for novel therapeutic strategies for the treatment of hematopoietic tumors. Residual tumor cells are the main origin of tumor relapse. The aim of this study was to eliminate the residual tumor cells of hematopoietic tumors. Cytokine-induced killer (CIK) cells are used in immunotherapy to deplete the residual cells. However, it is necessary to increase the antitumor activity and clinical applicability of CIK cells. The present study investigated the antitumor activity of CIK cells to the SU-DHL2 human B-cell lymphoma and K562 human chronic myelogenous leukemia cell lines. CD3+CD56+ cells from healthy donors were expanded in culture with cytokines and anti-CD20 monoclonal antibody (mAb; rituximab) to generate CIK cells. A preliminary investigation of their mechanism was then performed. The increase in the cytotoxicity of the CIK cells induced by the anti-CD20 mAb was associated with an increase in the expression of cytotoxic factors. The expression of components of the signal transducer and activator of transcription (STAT) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling pathways was found to increase. Upregulation of the expression of STAT1, STAT3 and STAT5 is important as these co-stimulatory molecules enhance T-cell proliferation. Activation of the MAPK signaling pathway is a possible mechanism for the anti-apoptosis effect on the proliferation of CIK cells. In conclusion, anti-CD20 mAb may play an important role in the improvement of CIK-mediated cytotoxicity to tumor cells. These observations may aid in the improvement of the effects of immunotherapy in depleting the residual cells of hematopoietic tumors. Thus, the use of CIK cells cultured with anti-CD20 mAb could be a novel therapeutic strategy for the depletion of chemotherapy-resistant or residual cells in anaplastic large and B-cell lymphoma.


Transplantation Proceedings | 2018

Tacrolimus-induced Pain Syndrome After Bone Marrow Transplantation: A Case Report and Literature Review

Xiao-chen Wei; Ming-feng Zhao; Qing Li; Xia Xiao; Li-qin Zhu

BACKGROUND Calcineurin-inhibitor-induced pain syndrome (CIPS), a rare complication seen in patients with bone marrow transplants, is associated with the use of cyclosporine A (CsA) or tacrolimus (FK506). This case demonstrates the successful pain control of FK506-related CIPS in a 23-year-old male patient with previously reported characteristic clinical features of CIPS in addition to neuropathic symptoms and uncharacteristic imaging findings. On day 15 after the transplantation, the patient complained of severe pain in the lower limbs. Afterwards, the patient started to complain of pain on his hands and back too. During this period, FK506 levels ranged from 9.5 to 16.1 ng/mL. All laboratory exams were normal, except for an increased level of alkaline phosphatase (141 U/L). The pain was not ameliorated by various analgesic drugs. Although MRI done for our patient showed no typical radiological signs such as bone marrow edema, CIPS was suggested based on characteristic clinical features of CIPS. Of note, our patients pain had neuropathic pain-like characteristics, unlike the pain in previously reported patients with CIPS. CONCLUSION The patient was treated successfully by switching FK506 to CsA and administrating gabapentin and nifedipine. Heightened awareness of this complication after bone marrow transplants may be needed for hematologists, otherwise CIPS can result in catastrophic consequences.


Blood | 2010

Increased Intracellular Concentration of Reactive Oxygen Species Mediated the Deficient Hematopoiesis of Iron Overload Bone Marrow

Mingfeng Zhao; Fang Xie; Yuming Li; Juan Mu; Xia Xiao; Haibo Zhu; Xinnv Xu; Qi Deng; Pengjiang Liu; Li Geng; Zonghong Shao


Annals of Hematology | 2017

Reactive oxygen species mediated T lymphocyte abnormalities in an iron-overloaded mouse model and iron-overloaded patients with myelodysplastic syndromes

Jie Chen; Wenyi Lu; Mingfeng Zhao; Xiaoli Cao; Yanyu Jiang; Xin Jin; Ping Xu; Ting-ting Yuan; Yuchen Zhang; Xiao Chai; Juanxia Meng; Qing Li; Xia Xiao; Juan Mu; Deguan Li; Ai-ping Qi


Clinical Cancer Research | 2018

Plasma exchange can be an alternative therapeutic modality for severe cytokine release syndrome after chimeric antigen receptor-T cell infusion: a case report

Xia Xiao; Xiaoyuan He; Qing Li; Huan Zhang; Juanxia Meng; Yanyu Jiang; Qi Deng; Mingfeng Zhao


Blood | 2014

The Bone Marrow-Derived Mesenchymal Cells Were Injured By Iron Overload through Catalyzing the Production of Reactive Oxidative Stress in Mice

Yuchen Zhang; Mingfeng Zhao; Deguan Li; Xiaoli Cao; Jie Chen; Juan Mu; Qing Li; Xia Xiao; Xiao Chai; Aimin Meng

Collaboration


Dive into the Xia Xiao's collaboration.

Top Co-Authors

Avatar

Mingfeng Zhao

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Juan Mu

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiao Chai

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Juanxia Meng

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Wenyi Lu

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Deguan Li

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Yuchen Zhang

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Aimin Meng

Peking Union Medical College

View shared research outputs
Top Co-Authors

Avatar

Jie Chen

Tianjin Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaoli Cao

Tianjin Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge