Network


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

Hotspot


Dive into the research topics where Yao Wang is active.

Publication


Featured researches published by Yao Wang.


Cancer Letters | 2013

Knockdown of Oct4 and Nanog expression inhibits the stemness of pancreatic cancer cells

Yuhua Lu; Hui Zhu; Haiyan Shan; Junjie Lu; Xu Chang; Xiaohong Li; Jingjing Lu; Xiangjun Fan; Shajun Zhu; Yao Wang; Qingsong Guo; Lei Wang; Yan Huang; Mingyan Zhu; Zhiwei Wang

Pancreatic cancer is notorious for its difficult diagnosis at early stage and poor recurrence-free prognosis. This study aimed to investigate the possible involvement of Oct4 and Nanog in pancreatic cancer. The high expressions of Oct4 and Nanog in human pancreatic cancer tissues were found to indicate a worse prognostic value of patients. The pancreatic cancer stem cells (PCSCs) that isolated from PANC-1 cell line by flow cytometry exhibited high expressions of Oct4 and Nanog. To investigate whether Oct4 and Nanog play crucial role in maintaining the stemness of PCSCs, double knockdown of Oct4 and Nanog demonstrated that Oct4 and Nanog significantly reduced proliferation, migration, invasion, chemoresistance, and tumorigenesis of PCSCs in vitro and in vivo. The altered expression of the genes related to pancreatic carcinogenesis, metastasis, drug resistance and epithelial-mesenchymal transdifferentiation (EMT) might affect the biological characteristics of PCSCs. Our results suggest that Oct4 and Nanog may serve as a potential marker of prognosis and a novel target of therapy for pancreatic cancer.


BMC Cancer | 2014

MiR-200a inhibits epithelial-mesenchymal transition of pancreatic cancer stem cell

Yuhua Lu; Jingjing Lu; Xiaohong Li; Hui Zhu; Xiangjun Fan; Shajun Zhu; Yao Wang; Qingsong Guo; Lei Wang; Yan Huang; Mingyan Zhu; Zhiwei Wang

BackgroundPancreatic cancer is one of the most aggressive cancers, and the aggressiveness of pancreatic cancer is in part due to its intrinsic and extrinsic drug resistance characteristics, which are also associated with the acquisition of epithelial-to-mesenchymal transition (EMT). Increasing evidence suggests that EMT-type cells share many biological characteristics with cancer stem-like cells. And miR-200 has been identified as a powerful regulator of EMT.MethodsCancer Stem Cells (CSCs) of human pancreatic cancer cell line PANC-1 were processed for CD24, CD44 and ESA multi-colorstaining, and sorted out on a BD FACS Aria II machine. RT-qPCR was performed using the miScript PCR Kit to assay the expression of miR-200 family. In order to find the role of miR-200a in the process of EMT, miR-200a mimic was transfected to CSCs.ResultsPancreatic cancer cells with EMT phenotype displayed stem-like cell features characterized by the expression of cell surface markers CD24, CD44 and epithelial-specific antigen (ESA), which was associated with decreased expression of miR-200a. Moreover, overexpression of miR-200a was resulted in down-regulation of N-cadherin, ZEB1 and vimentin, but up-regulation of E-cadherin. In addition, miR-200a overexpression inhibited cell migration and invasion in CSCs.ConclusionIn our study, we found that miR-200a played an important role in linking the characteristics of cancer stem-like cells with EMT-like cell signatures in pancreatic cancer. Selective elimination of cancer stem-like cells by reversing the EMT phenotype to mesenchymal-to-epithelial transition (MET) phenotype using novel agents would be useful for prevention and/or treatment of pancreatic cancer.


Artificial Organs | 2016

Decellularization and Recellularization of Rat Livers With Hepatocytes and Endothelial Progenitor Cells.

Pengcheng Zhou; Yan Huang; Yibing Guo; Lei Wang; Changchun Ling; Qingsong Guo; Yao Wang; Shajun Zhu; Xiangjun Fan; Mingyan Zhu; Hua Huang; Yuhua Lu; Zhiwei Wang

Whole-organ decellularization has been identified as a promising choice for tissue engineering. The aim of the present study was to engineer intact whole rat liver scaffolds and repopulate them with hepatocytes and endothelial progenitor cells (EPCs) in a bioreactor. Decellularized liver scaffolds were obtained by perfusing Triton X-100 with ammonium hydroxide. The architecture and composition of the original extracellular matrix were preserved, as confirmed by morphologic, histological, and immunolabeling methods. To determine biocompatibility, the scaffold was embedded in the subcutaneous adipose layer of the back of a heterologous animal to observe the infiltration of inflammatory cells. Hepatocytes were reseeded using a parenchymal injection method and cultured by continuous perfusion. EPCs were reseeded using a portal vein infusion method. Morphologic and functional examination showed that the hepatocytes and EPCs grew well in the scaffold. The present study describes an effective method of decellularization and recellularization of rat livers, providing the foundation for liver engineering and the development of bioartificial livers.


Diabetes Research and Clinical Practice | 2014

DIFFERENTIATION OF IPSCS INTO INSULIN-PRODUCING CELLS VIA ADENOVIRAL TRANSFECTION OF PDX-1, NEUROD1 AND MAFA

Lei Wang; Yan Huang; Qingsong Guo; Xiangjun Fan; Yuhua Lu; Shajun Zhu; Yao Wang; Xiangkun Bo; Xu Chang; Mingyan Zhu; Zhiwei Wang

AIMS The aim of this study was to evaluate the effect of PDX-1 (pancreatic and duodenal homeobox-1), NeuroD1 (neurogenic differentiation-1) and MafA (V-maf musculoaponeurotic fibrosarcoma oncogene homolog A) in the differentiation of induced pluripotent stem cells (iPSCs) into insulin-producing cells and to explore this new approach of cell transplantation therapy for type 1 diabetes in mice. METHODS iPSCs were infected with adenovirus (Ad-Mouse PDX-1-IRES-GFP, Ad-Mouse NeuroD1-IRES-GFP and Ad-Mouse Mafa-IRES-GFP) and then differentiated into insulin-producing cells in vitro. RT-PCR was applied to detect insulin gene expression, immunofluorescence to identify insulin protein, and mouse insulin enzyme-linked immunosorbent assay (ELISA) was used to evaluate the amount of insulin at different concentration of glucose. Insulin-producing cells were transplanted into the liver parenchyma of diabetic mice. Immunohistochemistry, intraperitoneal glucose tolerance test (IPGTT) and fasting blood glucose (FBG) were performed to assess the function of insulin-producing cells. RESULTS Insulin biosynthesis and secretion were induced in iPSCs and insulin-producing cells were responsive to glucose in a dose-dependent manner. Gene expression of the three-gene-modified embryoid bodies (EBs) was similar to the mouse pancreatic β cell line MIN6. Transplantation of insulin-producing cells into type I diabetic mice resulted in hyperglycemia reversal. CONCLUSIONS The insulin-producing cells we obtained from three-gene-modified EBs may be used as seed cells for tissue engineering and may represent a cell replacement strategy for the production of β cells for the treatment of type 1 diabetes.


BioMed Research International | 2017

Culture of iPSCs Derived Pancreatic β-Like Cells In Vitro Using Decellularized Pancreatic Scaffolds: A Preliminary Trial

Jian Wan; Yan Huang; Pengcheng Zhou; Yibing Guo; Cen Wu; Shajun Zhu; Yao Wang; Lei Wang; Yuhua Lu; Zhiwei Wang

Diabetes mellitus is a disease which has affected 415 million patients in 2015. In an effort to replace the significant demands on transplantation and morbidity associated with transplantation, the production of β-like cells differentiated from induced pluripotent stem cells (iPSCs) was evaluated. This approach is associated with promising decellularized scaffolds with natural extracellular matrix (ECM) and ideal cubic environment that will promote cell growth in vivo. Our efforts focused on combining decellularized rat pancreatic scaffolds with mouse GFP+-iPSCs-derived pancreatic β-like cells, to evaluate whether decellularized scaffolds could facilitate the growth and function of β-like cells. β-like cells were differentiated from GFP+-iPSCs and evaluated via cultivating in the dynamic circulation perfusion device. Our results demonstrated that decellularized pancreatic scaffolds display favorable biochemical properties. Furthermore, not only could the scaffolds support the survival of β-like cells, but they also accelerated the expression of the insulin as compared to plate-based cell culture. In conclusion, these results suggest that decellularized pancreatic scaffolds could provide a suitable platform for cellular activities of β-like cells including survival and insulin secretion. This study provides preliminary support for regenerating insulin-secreting organs from the decellularized scaffolds combined with iPSCs derived β-like cells as a potential clinical application.


Cell Transplantation | 2017

Transcriptome analysis of the induced pluripotent stem cell (iPSC)-derived pancreatic β-like cell differentiation.

Yan Huang; Jian Wan; Yibing Guo; Shajun Zhu; Yao Wang; Lei Wang; Qingsong Guo; Yuhua Lu; Zhiwei Wang

Diabetes affects millions of people worldwide, and β-cell replacement is one of the promising new strategies for treatment. Induced pluripotent stem cells (iPSCs) can differentiate into any cell type, including pancreatic β cells, providing a potential treatment for diabetes. However, the molecular mechanisms underlying the differentiation of iPSC-derived β cells have not yet been fully elucidated. Here, we generated pancreatic β-like cells from mouse iPSCs using a 3-step protocol and performed deep RNA sequencing to get a transcriptional landscape of iPSC-derived pancreatic β-like cells during the selective differentiation period. We then focused on the differentially expressed genes (DEGs) during the time course of the differentiation period, and these genes underwent Gene Ontology annotation and Kyoto Encyclopedia of Genes and Genomes pathway analysis. In addition, gene-act networks were constructed for these DEGs, and the expression of pivotal genes detected by quantitative real-time polymerase chain reaction was well correlated with RNA sequence (RNA-seq). Overall, our study provides valuable information regarding the transcriptome changes in β cells derived from iPSCs during differentiation, elucidates the biological process and pathways underlying β-cell differentiation, and promotes the identification and functional analysis of potential genes that could be used for improving functional β-cell generation from iPSCs.


Pediatric Transplantation | 2015

Controlling the blood glucose of type 1 diabetes mice by co-culturing MIN-6 β cells on 3D scaffold

Xiaoqi Yuan; Yan Huang; Yibing Guo; Lei Wang; Qingsong Guo; Tianxing Xu; Di Wu; Pengcheng Zhou; Shajun Zhu; Yao Wang; Xiangjun Fan; Mingyan Zhu; Yuhua Lu; Zhiwei Wang

T1D is an autoimmune disease, which may be caused by lack of insulin‐secreting β cells due to damage of autoimmune system. Living with T1D is a challenge for the child and the family; cell transplantation is a treatment option for diabetes in children. To establish a microenvironment suitable for cell growth and proliferation as well as for sustained cellular function, we used MIN‐6 β cells as seed cells and SF‐IV collagen as a 3D composite scaffold to construct artificial pancreas in this experiment. The cell viabilities were determined by MTT assay, and the response of cells to different glucose concentrations was observed by glucose stimulation test. Artificial pancreas was transplanted into the abdominal cavity of T1D mice, and the changes of blood glucose were monitored. After 10 days, insulin expression was detected by immunohistochemical method, and the claybank stained area showed effectiveness of insulin secretion. A series of experiments showed that implantation of 3D cell scaffold into the abdominal cavity can effectively control the blood glucose level of T1D mice. It also had longer‐lasting hypoglycemic effects than simple cell transplantation, which was expected to become a new method for the treatment of T1D.


Journal of Biomaterials Applications | 2018

Constructing heparin-modified pancreatic decellularized scaffold to improve its re-endothelialization:

Liancheng Xu; Yibing Guo; Yan Huang; Yicheng Xiong; Yang Xu; Xiaohong Li; Jingjing Lu; Lei Wang; Yao Wang; Yuhua Lu; Zhiwei Wang

Pancreas transplantation is considered as a promising therapeutic option with the potential to cure diabetes. However, efficacy of current clinical transplantation is limited by the donor organ. With regard to creating a functional pancreas-tissue equivalent for transplantation, vascularization remains a large obstacle. To enhance the angiogenic properties of pancreatic decellularized scaffold, surface modification of the vasculature was used to promote endothelialization efficiency. In this study, an endothelialized pancreatic decellularized scaffold was obtained through heparin modification under mild conditions. The immobilization of heparin was performed through 1-ethyl-3–(3-dimethylaminopropyl)-carbodiimide and N-Hydroxysuccinimide. The morphology, ultra-structure and porosity of the heparinized scaffold were characterized by toluidine blue staining, scanning electron microscope and infrared spectrum. The adhesion, proliferation and angiogenesis of human umbilical vein endothelial cells on heparin-pancreatic decellularized scaffold were also researched in vitro. In vivo transplantation was also performed to observe the location of human umbilical vein endothelial cells and the formation of new blood vessel, which exhibited significant differences with pancreatic decellularized scaffold group (p<0.05). These findings indicated that the endothelialized heparin-pancreatic decellularized scaffold may be used to solve the problem of blood supply and to support the function of insulin-secreting cells better after in vivo transplantation, and therefore, would be a potential candidate for pancreatic tissue engineering.


Experimental Cell Research | 2017

YB-1 expression promotes pancreatic cancer metastasis that is inhibited by microRNA-216a

Jingjing Lu; Xiaohong Li; Fei Wang; Yibing Guo; Yan Huang; Hui Zhu; Yao Wang; Yuhua Lu; Zhiwei Wang

ABSTRACT Pancreatic cancer is one of the most aggressive cancers. The vast majority of patients are diagnosed with advanced, unresectable disease because of early invasive growth and metastatic spread. The aim of this study was to examine YB‐1 expression in pancreatic cancer and determine its effects on cell invasion. YB‐1 is overexpressed in pancreatic cancer cell lines and patient tissue samples. In patient tissues, high YB‐1 levels correlated with perineural invasion. Silencing of YB‐1 significantly reduced cell invasion with decreased expression of MMPs in vitro. Furthermore, we found that the expression of YB‐1 was suppressed by miR‐216a via direct binding to the YB‐1 3’‐untranslated region. MiR‐216a and YB‐1 expression levels were inversely correlated in pancreatic cancer cell lines. In addition, ectopic expression of miR‐216a inhibited cell invasion in vitro. Taken together, our findings suggest that YB‐1 may play an important role in mediating metastatic behaviour and that repression of YB‐1 by miR‐216a could have a promising therapeutic potential to inhibit tumor metastasis in pancreatic cancer.


Oncology Reports | 2016

Salidroside induces apoptosis and autophagy in human colorectal cancer cells through inhibition of PI3K/Akt/mTOR pathway

Xiangjun Fan; Yao Wang; Lei Wang; Mingyan Zhu

Collaboration


Dive into the Yao Wang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge