Network


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

Hotspot


Dive into the research topics where Yi-Ping Hu is active.

Publication


Featured researches published by Yi-Ping Hu.


Nature | 2011

Induction of functional hepatocyte-like cells from mouse fibroblasts by defined factors

Pengyu Huang; Zhiying He; Shuyi Ji; Huawang Sun; Dao Xiang; Changcheng Liu; Yi-Ping Hu; Xin Wang; Lijian Hui

The generation of functional hepatocytes independent of donor liver organs is of great therapeutic interest with regard to regenerative medicine and possible cures for liver disease. Induced hepatic differentiation has been achieved previously using embryonic stem cells or induced pluripotent stem cells. Particularly, hepatocytes generated from a patient’s own induced pluripotent stem cells could theoretically avoid immunological rejection. However, the induction of hepatocytes from induced pluripotent stem cells is a complicated process that would probably be replaced with the arrival of improved technology. Overexpression of lineage-specific transcription factors directly converts terminally differentiated cells into some other lineages, including neurons, cardiomyocytes and blood progenitors; however, it remains unclear whether these lineage-converted cells could repair damaged tissues in vivo. Here we demonstrate the direct induction of functional hepatocyte-like (iHep) cells from mouse tail-tip fibroblasts by transduction of Gata4, Hnf1α and Foxa3, and inactivation of p19Arf. iHep cells show typical epithelial morphology, express hepatic genes and acquire hepatocyte functions. Notably, transplanted iHep cells repopulate the livers of fumarylacetoacetate-hydrolase-deficient (Fah−/−) mice and rescue almost half of recipients from death by restoring liver functions. Our study provides a novel strategy to generate functional hepatocyte-like cells for the purpose of liver engineering and regenerative medicine.


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.


Gastroenterology | 2010

Hepatoblast-Like Progenitor Cells Derived From Embryonic Stem Cells Can Repopulate Livers of Mice

Fuming Li; Pingyu Liu; Changcheng Liu; Dao Xiang; Li Deng; Wenlin Li; Kirk J. Wangensteen; Jianguo Song; Yue Ma; Lijian Hui; Lixin Wei; Lingsong Li; Xiaoyan Ding; Yi-Ping Hu; Zhiying He; Xin Wang

BACKGROUND & AIMS Hepatocyte-like cells can be derived from pluripotent stem cells such as embryonic stem (ES) cells, but ES cell-derived hepatic cells with extensive capacity to repopulate liver have not been identified. We aimed to identify and purify ES cell-derived hepatoblast-like progenitor cells and to explore their capacity for liver repopulation in mice after in vitro expansion. METHODS Unmanipulated mouse ES cells were cultured under defined conditions and allowed to undergo stepwise hepatic differentiation. The derived hepatic cells were examined by morphologic, fluorescence-activated cell sorting, gene expression, and clonal expansion analyses. The capacities of ES cell-derived hepatic progenitor cells to repopulate liver were investigated in mice that were deficient in fumarylacetoacetate hydrolase (Fah) (a model of liver injury). RESULTS Mouse ES cells were induced to differentiate into a population that contained hepatic progenitor cells; this population included cells that expressed epithelial cell adhesion molecule (EpCAM) but did not express c-Kit. Clonal hepatic progenitors that arose from single c-Kit(-)EpCAM(+) cells could undergo long-term expansion and maintain hepatoblast-like characteristics. Enriched c-Kit(-)EpCAM(+) cells and clonally expanded hepatic progenitor cells repopulated the livers of Fah-deficient mice without inducing tumorigenesis. CONCLUSIONS ES cell-derived c-Kit(-)EpCAM(+) cells contain a population of hepatoblast-like progenitor cells that can repopulate livers of mice.


Cell Death and Disease | 2013

Cuprous oxide nanoparticles inhibit the growth and metastasis of melanoma by targeting mitochondria

Yan Wang; Yang F; Zhang Hx; Zi Xy; Pan Xh; Fei Chen; Luo Wd; Li Jx; Zhu Hy; Yi-Ping Hu

Metal and its oxide nanoparticles show ideal pharmacological activity, especially in anti-tumor therapy. Our previous study demonstrated that cuprous oxide nanoparticles (CONPs) selectively induce apoptosis of tumor cells in vitro. To explore the anti-tumor properties of CONPs in vivo, we used the particles to treat mouse subcutaneous melanoma and metastatic lung tumors, based on B16-F10 mouse melanoma cells, by intratumoral and systemic injections, respectively. The results showed that CONPs significantly reduced the growth of melanoma, inhibited the metastasis of B16-F10 cells and increased the survival rate of tumor-bearing mice. Importantly, the results also indicated that CONPs were rapidly cleared from the organs and that these particles exhibited little systemic toxicity. Furthermore, we observed that CONPs targeted the mitochondria, which resulted in the release of cytochrome C from the mitochondria and the activation of caspase-3 and caspase-9 after the CONPs entered the cells. In conclusion, CONPs can induce the apoptosis of cancer cells through a mitochondrion-mediated apoptosis pathway, which raises the possibility that CONPs could be used to cure melanoma and other cancers.


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.


American Journal of Pathology | 2010

Liver Xeno-Repopulation with Human Hepatocytes in Fah−/−Rag2−/− Mice after Pharmacological Immunosuppression

Zhiying He; Haibin Zhang; Xin Zhang; Dong-Fu Xie; Yixin Chen; Kirk J. Wangensteen; Stephen C. Ekker; Meri T. Firpo; Changcheng Liu; Dao Xiang; Lijian Hui; Guangshun Yang; Xiaoyan Ding; Yi-Ping Hu; Xin Wang

Functional human hepatocytes xeno-engrafted in mouse liver can be used as a model system to study hepatitis virus infection and vaccine efficacy. Significant liver xeno-repopulation has been reported in two kinds of genetically modified mice that have both immune deficiency and liver injury-induced donor hepatocyte selection: the uPA/SCID mice and Fah(-/-) Rag2(-/-)Il2rg(-/-) mice. The lack of hardy breeding and the overly elaborated technique in these two models may hinder the potential future application of these models to hepatitis virus infection and vaccination studies. Improving the transplantation protocol for liver xeno-repopulation from human hepatocytes will increase the model efficiency and application. In this study, we successfully apply immunosuppressive drug treatments of anti-asialo GM1 and FK506 in Fah(-/-)Rag2(-/-) mice, resulting in significant liver xeno-repopulation from human hepatocytes and human fetal liver cells. This methodology decreases the risk of animal mortality during breeding and surgery. When infected with hepatitis B virus (HBV) sera, Fah(-/-)Rag2(-/-) mice with liver xeno-repopulation from human hepatocytes accumulate significant levels of HBV DNA and HBV proteins. Our new protocol for humanized liver could be applied in the study of human hepatitis virus infection in vivo, as well as the pharmacokinetics and efficacy of potential vaccines.


Journal of Cellular Biochemistry | 2011

Combined activin A/LiCl/Noggin treatment improves production of mouse embryonic stem cell‐derived definitive endoderm cells

Fuming Li; Zhiying He; Yangfang Li; Pingyu Liu; Fei Chen; Min-Jun Wang; Hai-Ying Zhu; Xiaoyan Ding; Kirk J. Wangensteen; Yi-Ping Hu; Xin Wang

Induction of definitive endoderm (DE) cells is a prerequisite for the whole process of embryonic stem (ES) cells differentiating into hepatic or pancreatic progenitor cells. We have established an efficient method to induce mouse ES cell‐derived DE cells in suspension embryonic body (EB) culture. Similar to previous studies, mouse ES cell‐derived DE cells, which were defined as Cxcr4+c‐Kit+, Cxcr4+E‐cadherin+ cells or Cxcr4+PDGFRa− cells, could be induced in the serum‐free EBs at Day 4 of induction. The activations of Wnt, Nodal, and FGF signaling pathways in differentiating EBs promoted DE cell differentiation, while activation of BMP4 signaling inhibited the process. In the present study, we found that chemical activation of canonical Wnt signaling pathway by LiCl could synergize with Activin A‐mediated Nodal signaling pathway to promote induction of DE cells, and inhibition of Bmp4 signaling by Noggin along with Activin A/LiCl further improved the efficiency of DE cell differentiation. The derived DE cells were proved for their capacities to become hepatic progenitor cells or pancreatic progenitor cells. In conclusion, we significantly improved the efficiency of generating mouse ES cell‐derived DE cells by combined Activin A/LiCl/Noggin treatment. Our work will be greatly helpful to generate ES cell‐derived hepatic cells and ES cell‐derived pancreatic cells for future regenerative medicine. J. Cell. Biochem. 112: 1022–1034, 2011.


Journal of Biological Chemistry | 2010

H3K27 Trimethylation is an Early Epigenetic Event of p16INK4a Silencing for Regaining Tumorigenesis in Fusion Reprogrammed Hepatoma Cells

Jia-Yi Yao; Lei Zhang; Xin Zhang; Zhiying He; Yue Ma; Lijian Hui; Xin Wang; Yi-Ping Hu

Stable epigenetic silencing of p16INK4a is a common event in hepatocellular carcinoma (HCC) cells, which is associated with abnormal cell proliferation and liberation from cell cycle arrest. Understanding the early epigenetic events in silencing p16INK4a expression may illuminate a prognostic strategy to block HCC development. Toward this end, we created a reprogram cell model by the fusion mouse HCC cells with mouse embryonic stem cells, in which the ES-Hepa hybrids forfeited HCC cell characteristics along with reactivation of the silenced p16INK4a. HCC characteristics, in terms of gene expression pattern and tumorigenic potential, was restored upon induced differentiation of these reprogrammed ES-Hepa hybrids. The histone methylation pattern relative to p16INK4a silencing during differentiation of the ES-Hepa hybrids was analyzed. H3K27 trimethylation at the p16INK4a promoter region, occurring in the early onset of p16INK4a silencing, was followed by H3K9 dimethylation at later stages. During the induced differentiation of the ES-Hepa hybrids, H3K4 di- and trimethylations were maintained at high levels during the silencing of p16INK4a, strongly suggesting that H3K4 methylation events did not cause the silencing of p16INK4a. Our results suggested that the enrichment of H3K27 trimethylation, independent of H3K9 dimethylation, trimethylation, and DNA methylation, was an early event in the silencing of p16INK4a during the tumor development. This unique chromatin pattern may be a heritable marker of epigenetic regulation for p16INK4a silencing during the developmental process of hepatocellular carcinogenesis.


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)

Collaboration


Dive into the Yi-Ping Hu's collaboration.

Top Co-Authors

Avatar

Zhiying He

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xin Wang

Inner Mongolia University

View shared research outputs
Top Co-Authors

Avatar

Changcheng Liu

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

Jian-Xiu Li

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

Dao Xiang

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

Hai-Ying Zhu

Second Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaoyan Ding

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge