Network


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

Hotspot


Dive into the research topics where Jian-Xiu Li is active.

Publication


Featured researches published by Jian-Xiu Li.


Cell Death & Differentiation | 2012

Mesenchymal stem cells: a double-edged sword in regulating immune responses

W Li; Guosheng Ren; Yin Huang; Juanjuan Su; Yanyan Han; Jian-Xiu Li; Xiaodong Chen; Qing Chen; Peishun Shou; Liying Zhang; Zengrong Yuan; Arthur I. Roberts; Songtao Shi; Anh D. Le; Yufang Shi

Mesenchymal stem cells (MSCs) have been employed successfully to treat various immune disorders in animal models and clinical settings. Our previous studies have shown that MSCs can become highly immunosuppressive upon stimulation by inflammatory cytokines, an effect exerted through the concerted action of chemokines and nitric oxide (NO). Here, we show that MSCs can also enhance immune responses. This immune-promoting effect occurred when proinflammatory cytokines were inadequate to elicit sufficient NO production. When inducible nitric oxide synthase (iNOS) production was inhibited or genetically ablated, MSCs strongly enhance T-cell proliferation in vitro and the delayed-type hypersensitivity response in vivo. Furthermore, iNOS−/− MSCs significantly inhibited melanoma growth. It is likely that in the absence of NO, chemokines act to promote immune responses. Indeed, in CCR5−/−CXCR3−/− mice, the immune-promoting effect of iNOS−/− MSCs is greatly diminished. Thus, NO acts as a switch in MSC-mediated immunomodulation. More importantly, the dual effect on immune reactions was also observed in human MSCs, in which indoleamine 2,3-dioxygenase (IDO) acts as a switch. This study provides novel information about the pathophysiological roles of MSCs.


Stem Cells | 2006

Isolation and Characterization of Bipotent Liver Progenitor Cells from Adult Mouse

Wenlin Li; Juan Su; Yu-Cheng Yao; Xin-Rong Tao; Yong‐Bi Yan; Hong-Yu Yu; Xin-Min Wang; Jian-Xiu Li; Yong‐Ji Yang; Joseph T.Y. Lau; Yi-Ping Hu

Liver progenitor cells have drawn a great deal of attention both for their therapeutic potential and for their usefulness in exploring the molecular events surrounding liver development and regeneration. Despite the intensive studies on liver progenitors from rats, equivalent progenitor cells derived from mice are relatively rare. We used retrosine treatment followed by partial hepatectomy to elicit liver progenitors in mice. From these animals showing prominent ductular reactions, mouse‐derived liver progenitor cell lines (LEPCs) were isolated by single‐cell cloning. Phenotypic and lineage profiling of the LEPC clones were performed using immunochemistry, reverse transcription–polymerase chain reaction, and a dual‐color system comprising the reporter EGFP under the control of the cytokeratin 19 promoter and the DsRed reporter under the control of the albumin promoter. LEPCs expressed liver progenitor cell markers. LEPCs also expressed some markers shared by bone marrow‐derived hematopoietic stem cells c‐Kit and Thy‐1 but not CD34 and CD45. When cultured as aggregates in Matrigel, LEPCs differentiated into hepatocyte upon treatment with 50 ng/ml epithelial growth factor or differentiated into biliary lineage cells upon treatment with 20 ng/ml hepatocyte growth factor. In the presence of 2% dimethyl sulfoxide and 2% Matrigel, LEPCs acquired predominantly bile lineage phenotypes, with occasional patches of cells exhibiting hepatocyte phenotypes. Upon transplantation into CCl4‐injured‐liver, LEPCs engrafted into liver parenchyma and differentiated into hepatocytes. Considering the amenability of the mouse to genetic manipulation, these mouse‐derived LEPCs may be useful tools as in vitro models to study molecular events in liver development and regeneration and can shed light in studying the therapy potential of liver stem cells.


Cell Death & Differentiation | 2014

Phylogenetic distinction of iNOS and IDO function in mesenchymal stem cell-mediated immunosuppression in mammalian species

Juanjuan Su; Xiangmei Chen; Yin Huang; W Li; Jian-Xiu Li; Gang Cao; Liying Zhang; F Li; Arthur I. Roberts; H Kang; Pengfei Yu; Guosheng Ren; Weizhi Ji; Ying Wang; Yufang Shi

Mammalian mesenchymal stem cells (MSCs) have been shown to be strongly immunosuppressive in both animal disease models and human clinical trials. We have reported that the key molecule mediating immunosuppression by MSCs is species dependent: indoleamine 2,3-dioxygenase (IDO) in human and inducible nitric oxide synthase (iNOS) in mouse. In the present study, we isolated MSCs from several mammalian species, each of a different genus, and investigated the involvement of IDO and iNOS during MSC-mediated immunosuppression. The characterization of MSCs from different species was by adherence to tissue culture plastic, morphology, specific marker expression, and differentiation potential. On the basis of the inducibility of IDO and iNOS by inflammatory cytokines in MSCs, the tested mammalian species fall into two distinct groups: IDO utilizers and iNOS utilizers. MSCs from monkey, pig, and human employ IDO to suppress immune responses, whereas MSCs from mouse, rat, rabbit, and hamster utilize iNOS. Interestingly, based on the limited number of species tested, the iNOS-utilizing species all belong to the phylogenetic clade, Glires. Although the evolutionary significance of this divergence is not known, we believe that this study provides critical guidance for choosing appropriate animal models for preclinical studies of MSCs.


International Journal of Nanomedicine | 2012

Cuprous oxide nanoparticles selectively induce apoptosis of tumor cells

Ye Wang; Juan Su; Hongxia Zhang; Xin-Rong Zhang; Hai-Ying Zhu; Jian-Xiu Li; Meng Yin; Feng Yang; Yi-Ping Hu

In the rapid development of nanoscience and nanotechnology, many researchers have discovered that metal oxide nanoparticles have very useful pharmacological effects. Cuprous oxide nanoparticles (CONPs) can selectively induce apoptosis and suppress the proliferation of tumor cells, showing great potential as a clinical cancer therapy. Treatment with CONPs caused a G1/G0 cell cycle arrest in tumor cells. Furthermore, CONPs enclosed in vesicles entered, or were taken up by mitochondria, which damaged their membranes, thereby inducing apoptosis. CONPs can also produce reactive oxygen species (ROS) and initiate lipid peroxidation of the liposomal membrane, thereby regulating many signaling pathways and influencing the vital movements of cells. Our results demonstrate that CONPs have selective cytotoxicity towards tumor cells, and indicate that CONPs might be a potential nanomedicine for cancer therapy.


Journal of Cellular Biochemistry | 2009

Clonal mesenchymal stem cells derived from human bone marrow can differentiate into hepatocyte-like cells in injured livers of SCID mice.

Xin-Rong Tao; Wenlin Li; Juan Su; Cai-Xia Jin; Xin-Min Wang; Jian-Xiu Li; Jun-Kai Hu; Zhen-Hua Xiang; Joseph T.Y. Lau; Yi-Ping Hu

There is increasing evidence that human mesenchymal stem cells (hMSCs) can be a valuable, transplantable source of hepatocytes. Most of the hMSCs preparations used in these studies were likely heterogeneous cell populations, isolated by adherence to plastic surfaces or by density gradient centrifugation. Therefore, the participation of other unknown trace cell populations cannot be rigorously discounted. Here we report the isolation and establishment of a cloned human MSC line (chMSC) from human bone marrow primary culture, through which we confirmed the hepatic differentiation capability of authentic hMSCs. chMSCs expressed markers of mesenchymal cells, but not markers of hematopoietic stem cells. In vitro, chMSCs can differentiate into either mesenchymal cells or cells exhibiting hepatocyte‐like phenotypes. When transplanted intrasplentically into carbon tetrachloride‐injured livers of SCID mice, EGFP‐tagged chMSCs engrafted into the host liver parenchyma, exhibited typical hepatocyte morphology, form a three‐dimensional architecture, and differentiate into hepatocyte‐like cells expressing human albumin and α‐1‐anti‐trypsin. By confocal microscopy, ultrafine intercellular nanotubular structures were visible between adjacent transplanted and host hepatocytes. We postulate that these structures may assist in the phenotype conversion of chMSCs, possibly by exchange of cytoplasmic components between native hepatocytes and transplanted cells. Thus, a clonal pure population of hMSCs, which can be expanded in culture, may have potential as a cellular source for substitution damaged cells in hepatic injury. J. Cell. Biochem. 108: 693–704, 2009.


Hepatology | 2014

Reversal of hepatocyte senescence after continuous in vivo cell proliferation

Min-Jun Wang; Fei Chen; Jian-Xiu Li; Changcheng Liu; Haibin Zhang; Yong Xia; Bing Yu; Pu You; Dao Xiang; Lian Lu; Hao Yao; Uyunbilig Borjigin; Guangshun Yang; Kirk J. Wangensteen; Zhiying He; Xin Wang; Yi-Ping Hu

A better understanding of hepatocyte senescence could be used to treat age‐dependent disease processes of the liver. Whether continuously proliferating hepatocytes could avoid or reverse senescence has not yet been fully elucidated. We confirmed that the livers of aged mice accumulated senescent and polyploid hepatocytes, which is associated with accumulation of DNA damage and activation of p53‐p21 and p16ink4a‐pRB pathways. Induction of multiple rounds continuous cell division is hard to apply in any animal model. Taking advantage of serial hepatocyte transplantation assays in the fumarylacetoacetate hydrolase‐deficient (Fah−/−) mouse, we studied the senescence of hepatocytes that had undergone continuous cell proliferation over a long time period, up to 12 rounds of serial transplantations. We demonstrated that the continuously proliferating hepatocytes avoided senescence and always maintained a youthful state. The reactivation of telomerase in hepatocytes after serial transplantation correlated with reversal of senescence. Moreover, senescent hepatocytes harvested from aged mice became rejuvenated upon serial transplantation, with full restoration of proliferative capacity. The same findings were also true for human hepatocytes. After serial transplantation, the high initial proportion of octoploid hepatocytes decreased to match the low level of youthful liver. Conclusion: These findings suggest that the hepatocyte “ploidy conveyer” is regulated differently during aging and regeneration. The findings of reversal of hepatocyte senescence could enable future studies on liver aging and cell therapy. (Hepatology 2014;60:349–361)


European Journal of Immunology | 2006

Long-term persistence of hepatitis B surface antigen and antibody induced by DNA-mediated immunization results in liver and kidney lesions in mice.

Yu-Cheng Yao; Hai-Ying Zhu; Jun Xiong; Xiu-Ju Wu; Nan Zhang; Yue Ba; Wenlin Li; Xin-Ming Wang; Jian-Xiu Li; Hong-Yu Yu; Xu-Ting Ye; Joseph T.Y. Lau; Yi-Ping Hu

DNA‐mediated immunization has been recognized as a new approach for prevention and treatment of hepatitis B virus (HBV) infection. However, the side effects of this approach have not been well described. Here we report that DNA‐mediated immunization by intramuscular injection of plasmid DNA encoding HBV surface antigen (HBsAg) induced long‐term persistence of HBsAg and HBsAg‐specific antibody (anti‐HBs) in the sera of the immunized BALB/c mice and resulted in liver and kidney lesions. The lesions persisted for 6 months after injection. Lesions were also found in normal mice injected with the sera from immunized mice, and in HBV‐transgenic mice injected with anti‐HBs antibody, or sera from immunized mice. Furthermore, lesions were accompanied by deposition of circulating immune complex (CIC) of HBsAg and anti‐HBs antibody in the damaged organs. These results indicate that long‐term persistence of HBsAg and anti‐HBs in the immunized mice can result in deposited CIC in liver and kidney, and in development of lesions. The use of DNA containing mammalian replication origins, such as the plasmids used in this study, is not appropriate for human vaccines due to safety concerns relating to persistence of DNA; nevertheless, the safety of DNA‐mediated immunization protocols still needs to be carefully evaluated before practical application.See accompanying commentary: http://dx.doi.org/10.1002/eji.200635986


Journal of Cellular Biochemistry | 2009

Oncoprotein BMI-1 induces the malignant transformation of HaCaT cells

Qian Wang; Wenlin Li; Pu You; Juan Su; Ming-Hua Zhu; Dong-Fu Xie; Hai-Yin Zhu; Zhiying He; Jian-Xiu Li; Xiaoyan Ding; Xin Wang; Yi-Ping Hu

BMI‐1 (B‐cell‐specific Moloney murine leukemia virus integration site 1), a novel oncogene, has attracted much attention in recent years for its involvement in the initiation of a variety of tumors. Recent evidence showed that BMI‐1 was highly expressed in neoplastic skin lesions. However, whether dysregulated BMI‐1 expression is causal for the transformation of skin cells remains unknown. In this study, we stably expressed BMI‐1 in a human keratinocyte cell line, HaCaT. The expression of wild‐type BMI‐1 induced the malignant transformation of HaCaT cells in vitro. More importantly, we found that expression of BMI‐1 promoted formation of squamous cell carcinomas in vivo. Furthermore, we showed that BMI‐1 expression led to the downregulation of tumore suppressors, such as p16INK4a and p14ARF, cell adhesion molecules, such as E‐Cadherin, and differentiation related factor, such as KRT6. Therefore, our findings demonstrated that dysregulated BMI‐1 could indeed lead to keratinocytes transformation and tumorigenesis, potentially through promoting cell cycle progression and increasing cell mobility. J. Cell. Biochem. 106: 16–24, 2009.


Science China-life Sciences | 2007

Murine fertilized ovum, blastomere and morula cells lacking SP phenotype

YiXin Xu; Zhiying He; Hai-Ying Zhu; XueSong Chen; Jian-Xiu Li; Hongxia Zhang; Xinghua Pan; Yi-Ping Hu

In the field of stem cell research, SP (side population) phenotype is used to define the property that cells maintain a high efflux capability for some fluorescent dye, such as Hoechst 33342. Recently, many researches proposed that SP phenotype is a phenotype shared by some stem cells and some progenitor cells, and that SP phenotype is regarded as a candidate purification marker for stem cells. In this research, murine fertilized ova (including conjugate and single nucleus fertilized ova), 2-cell stage and 8-cell stage blastomeres, morulas and blastocysts were isolated and directly stained by Hoechst 33342 dye. The results show that fertilized ovum, blastomere and morula cells do not demonstrate any ability to efflux the dye. However, the inner cell mass (ICM) cells of blastocyst exhibit SP phenotype, which is consistent with the result of embryonic stem cells (ESCs) in vitro. These results indicate that the SP phenotype of ICM-derived ESCs is an intrinsic property and independent of the culture condition in vitro, and that SP phenotype is one of the characteristics of at least some pluripotent stem cells, but is not shared by totipotent stem cells. In addition, the result that the SP phenotype of ICM cells disappeared when the inhibitor verapamil was added into medium implies that the SP phenotype is directly associated with ABCG2. These results suggest that not all the stem cells demonstrate SP phenotype, and that SP phenotype might act as a purification marker for partial stem cells such as some pluripotent embryonic stem cells and multipotent adult stem cells, but not for all stem cells exampled by the totipotent stem cells in the very early stage of mouse embryos.


Cancer management and research | 2018

Liver-enriched activator protein 1 as an isoform of CCAAT/enhancer-binding protein beta suppresses stem cell features of hepatocellular carcinoma

Li-Hua Yang; Ye Wang; Shi Qiao; Min-Jun Wang; Fei Chen; Jian-Xiu Li; Haibin Zhang; Bing Yu; Yi-Ping Hu

Purpose Liver cancer stem cells (CSCs) are known to be associated with the development, survival, proliferation, metastasis, and recurrence of liver tumors. The aim of this study was to investigate the association of liver-enriched activator protein 1 (LAP1) with hepatocellular carcinoma (HCC) and liver CSCs (LCSCs) and explore the impact of LAP1 on LCSCs. Materials and methods Differences in LAP1 expression in liver cancer tissues versus matched para-tumoral liver tissues and LCSCs versus non-CSCs were analyzed by Western blotting, real-time polymerase chain reaction, immunohistochemistry, and flow cytometry. The effect of LAP1 on liver cancer cells was evaluated by the expression of CSC markers, oncosphere formation, proliferation, migration, and invasion in vitro. Cell cycle distribution and the number of apoptotic cells were analyzed to assess cell cycle and cell apoptosis. Furthermore, a mouse subcutaneous tumor implant model was established to explore the role of LAP1 in the development of HCC in vivo. Finally, the expression of CSC markers in paraffin-embedded sections was evaluated by immunofluorescence. Results LAP1 was weakly expressed in HCC tumors and cell lines and even weaker in LCSCs. LAP1 inhibited the expression of stem cell–associated genes and reduced the abilities of oncosphere formation, proliferation, migration, and invasion in vitro. Cell cycle assay revealed that LAP1 induced G1/G0 arrest. Furthermore, LAP1 decreased subcutaneous tumor-formation ability and the expression of CSC markers and Ki67 in vivo. Conclusion LAP1 suppressed the stem cell features of HCC, indicating that it possessed an antitumor effect in liver cancer, both in vitro and in vivo; therefore, LAP1 may prove to be a potential target in liver CSC-targeted therapy.

Collaboration


Dive into the Jian-Xiu Li's collaboration.

Top Co-Authors

Avatar

Yi-Ping Hu

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Wenlin Li

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhiying He

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Bing Yu

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Fei Chen

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Hai-Ying Zhu

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Hong-Yu Yu

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Juan Su

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Min-Jun Wang

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yu-Cheng Yao

Second Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge