Network


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

Hotspot


Dive into the research topics where Yinji Jin is active.

Publication


Featured researches published by Yinji Jin.


Oncotarget | 2016

Competing endogenous RNA networks in human cancer: hypothesis, validation, and perspectives.

Chao Yang; Di Wu; Lin Gao; Xi Liu; Yinji Jin; D. Wang; Tianzhen Wang; Xiaobo Li

Non-coding RNAs represent a majority of the human transcriptome. However, less is known about the functions and regulatory mechanisms of most non-coding species. Moreover, little is known about the potential non-coding functions of coding RNAs. The competing endogenous RNAs (ceRNAs) hypothesis is proposed recently. This hypothesis describes potential communication networks among all transcript RNA species mediated by miRNAs and miRNA-recognizing elements (MREs) within RNA transcripts. Here we review the evolution of the ceRNA hypothesis, summarize the validation experiments and discusses the significance and perspectives of this hypothesis in human cancer.


Translational Research | 2015

Ubiquitin-specific peptidase 22 overexpression may promote cancer progression and poor prognosis in human gastric carcinoma

Yan He; Yinji Jin; Yuhua Zhang; Hongxue Meng; Baoshan Zhao; Yang Jiang; Ji-Wei Zhu; Guan-Ying Liang; Dan Kong; Xiaoming Jin

Ubiquitin-specific peptidase 22 (USP22) was recently identified as a new tumor cell marker, and previous studies demonstrated its expression in a variety of tumors and its correlation with tumor progression. Because tumor progression plays an important role in cancer, researchers are paying more attention to the correlation between USP22 expression and metastatic potential, resistance to chemotherapy, and patient prognosis. This study showed that USP22 is highly expressed in gastric cancer tissues, and significant differences in USP22 expression (P < 0.01) were identified between different types of gastric cancer (the highest expression was found in poorly differentiated adenocarcinomas). In addition USP22 expression was found to be correlated with the promotion of cancer evolution, tumor invasion, and lymph node metastasis. The C-myc protein was also shown to have synergistic effects with USP22 in gastric cancer tissue. On the basis of the results, USP22 expression may play an important role in gastric carcinoma tissue, particularly in precancerous lesions during the gastric cancer evolution process.


Oncotarget | 2016

Comparison of the expression and function of Lin28A and Lin28B in colon cancer

Tianzhen Wang; Yan He; Yuanyuan Zhu; Mingwei Chen; Mingjiao Weng; Chao Yang; Yan Zhang; Ning Ning; Ran Zhao; Weiwei Yang; Yinji Jin; Jing Li; Riju James RajkumarEzakiel Redpath; Lei Zhang; Xiaoming Jin; Zhaohua Zhong; Fengmin Zhang; Yunwei Wei; Guomin Shen; D. Wang; Ying Liu; Guangyu Wang; Xiaobo Li

Lin28A and Lin28B are highly conserved RNA binding proteins with similar structure and functions. Recent studies demonstrated that both of them act as oncogenes and promote cancer progression. However, few researches compared the expression and functions of both oncogenes in human malignant tumors at same time. Additionally, although the expression and role of Lin28B in colon cancer is frequently reported, the expression and functions of Lin28A in colon cancer are largely unknown. In this study, we have systematically evaluated the expressional pattern, mutation status and correlation of both Lin28A and Lin28B in colon cancer tissues for the first time, and compared the roles of Lin28A and Lin28B in the proliferation, migration, invasion and apoptosis of colon cancer cells in vitro. We have showed that they are co-expressed and have functional similarities, however, the molecular mechanisms underlying their similar functions may not be identical. This study contributes to clarify the similarities and differences of Lin28A and Lin28B in colon cancer progression.


Tumor Biology | 2017

Necroptosis in cancer: An angel or a demon?:

Tianzhen Wang; Yinji Jin; Weiwei Yang; Lei Zhang; Xiaoming Jin; Xi Liu; Yan He; Xiaobo Li

In the past few decades, apoptosis has been regarded as the only form of programmed cell death. However, the traditional view has been challenged by the identification of several forms of regulated necrosis, including necroptosis. Necroptosis is typified by a necrotic cell death morphology and is controlled by RIP1, RIP3, and mixed lineage kinase domain–like protein. The physiological role of necroptosis is to serve as a “fail-safe” form of cell death for cells that fail to undergo apoptosis during embryonic development and disease defense. Currently, established studies have indicated that necroptosis is involved in cancer initiation and progression. Although elevated necroptosis contributes to cancer cell death, extensive cell death also increases the risk of proliferation and metastasis of the surviving cells by inducing the generation reactive oxygen species, activation of inflammation, and suppression of the immune response. Thus, questions regarding the overall impact of necroptosis on cancer remain open. In this review, we introduce the basic knowledge regarding necroptosis, summarize its dual effects on cancer progression, and analyze its advantages and disadvantages in clinical applications.


BioMed Research International | 2016

EF24 Suppresses Invasion and Migration of Hepatocellular Carcinoma Cells In Vitro via Inhibiting the Phosphorylation of Src

Ran Zhao; Lamtin Tin; Yuhua Zhang; Yiqi Wu; Yinji Jin; Xiaoming Jin; Fengmin Zhang; Xiaobo Li

Diphenyl difluoroketone (EF24), a curcumin analog, is a promising anticancer compound that exerts its effects by inhibiting cell proliferation and inducing apoptosis. However, the efficacy of EF24 against cancer metastasis, particularly in hepatocellular carcinoma (HCC), remains elusive. In this study, the effect of EF24 on HCCLM-3 and HepG2 cell migration and invasion was detected by wound healing and transwell assay, respectively. The results revealed that EF24 suppressed the migration and invasion of both HCCLM-3 and HepG2 cells. Furthermore, EF24 treatment decreased the formation of filopodia on the cell surface and inhibited the phosphorylation of Src in both cell lines, which may help contribute towards understanding the mechanism underlying the suppressive effect of EF24 on HCC migration and invasion. Additionally, the expression of total- and phosphorylated-Src in primary HCC tissues and their paired lymph node metastatic tissues was detected, and phosphorylated-Src was found to be associated with HCC lymph node metastasis. The results of this study suggest that Src is a novel and promising therapeutic target in HCC and provide evidence to support the hypothesis that EF24 may be a useful therapeutic agent for the treatment of HCC.


International Journal of Infectious Diseases | 2014

High load hepatitis B virus replication inhibits hepatocellular carcinoma cell metastasis through regulation of epithelial-mesenchymal transition.

Tianzhen Wang; Yinji Jin; Ran Zhao; Yiqi Wu; Yuhua Zhang; Di Wu; Dan Kong; Xiaoming Jin; Fengmin Zhang

OBJECTIVES The aims of this study were to investigate the effect of hepatitis B virus (HBV) replication on the metastatic ability of hepatocellular carcinoma (HCC) cells and to explore a potential mechanism from the perspective of epithelial-mesenchymal transition (EMT). METHODS Two short-interfering RNAs (siRNAs) against the HBV S gene were used to inhibit HBV replication in HepG2.2.15 cells. To evaluate the level of HBV replication and interference efficiency, HBV antigen and HBV DNA were detected by ELISA and quantitative PCR (Q-PCR). Invasion and metastatic abilities were compared between different groups by wound healing and trans-well assays. Immunofluorescent staining and Western blotting were utilized to detect EMT markers. RESULTS Both siRNAs effectively inhibited HBV replication in HepG2.2.15 cells. Compared to control HepG2.2.15 cells, cells transfected with the siRNAs showed characteristics of the mesenchymal phenotype and augmented their ability to invade and metastasize. Inhibition of HBV replication suppressed E-cadherin and induced a switch to vimentin expression. Western blots confirmed the decrease in E-cadherin expression. The level of E-cadherin expression was also lower in HepG2 cells than in HepG2.2.15 cells. CONCLUSIONS siRNAs were able to effectively inhibit HBV replication in vitro. A high load of HBV replication may inhibit the invasion and metastatic ability of HCC cells by reversing the EMT process.


Oncology Letters | 2015

MicroRNA-181a enhances the chemotherapeutic sensitivity of chronic myeloid leukemia to imatinib

Guangyu Wang; Ran Zhao; Xingsheng Zhao; Xi Chen; D. Wang; Yinji Jin; Xi Liu; Ci Zhao; Yuanyuan Zhu; Chengcheng Ren; Minghui Li; Xiaoming Jin; Fengmin Zhang; Zhaohua Zhong; Tianzhen Wang; Xiaobo Li

MicroRNA-181 (miR-181) has been recently demonstrated to participate in the differentiation and development of immune cells, including natural killer cells and B and T lymphocytes, and myeloid linages, including erythroid and megakaryocytic cells. The aberrant expression of miR-181, particularly low expression levels, has been observed in a number of leukemia types, including B-cell chronic lymphocytic leukemia and cytogenetically abnormal acute myeloid leukemia. However, the expression and function of miR-181 in chronic myeloid leukemia (CML) remains unknown. In the present study, the aberrant expression of miR-181a was analyzed in a patient with CML and in the CML K562 cell line. In addition, the function and potential mechanisms of miR-181a in K562 cells with regard to their chemotherapeutic sensitivity to imatinib were investigated. The expression levels of miR-181a were significantly reduced in the patient with CML and in the CML K562 cell line. Furthermore, the overexpression of miR-181a in the K562 cells enhanced the chemotherapeutic sensitivity of these cells to imatinib. The potential mechanism mediating these effects may be associated with the capacity of miR-181a to inhibit cell growth and/or to induce cells apoptosis and differentiation in K562 cells. The results of the present study suggested that miR-181a may be a target for the treatment of CML and a useful indicator of the therapeutic sensitivity of CML to imatinib.


Virology Journal | 2017

Hepatitis B virus x protein induces epithelial-mesenchymal transition of hepatocellular carcinoma cells by regulating long non-coding RNA

Yinji Jin; Di Wu; Weiwei Yang; Mingjiao Weng; Yafei Li; Xuefei Wang; Xiao Zhang; Xiaoming Jin; Tianzhen Wang

BackgroundIt has been widely accepted that hepatitis B virus X protein (HBx) plays an important role in hepatocellular carcinoma (HCC). This study aimed to explore the function of long non-coding RNAs (lncRNAs) in the epithelial-mesenchymal transition (EMT) induced by HBx.MethodsThe association between HBx and EMT markers was detected using immunohistochemistry in HCC tissues. The effect of HBx on HCC EMT was assessed through morphological analysis, transwell assay, metastatic in vivo study and detection of EMT markers. LncRNA microarray was used to screen the differently expressed lncRNAs. Small interfering RNA and Western blot were used to analyse the function and mechanism of the locked lncRNA.ResultsHBx was negatively correlated with the epithelial marker E-cadherin but positively correlated with the mesenchymal marker vimentin in HCC tissues. HBx induced the mesenchymal phenotype and improved the metastatic ability of HCC cells. Meanwhile, HBx down-regulated E-cadherin, whereas it up-regulated vimentin. In HCC cells, HBx altered the expression of 2002 lncRNAs by more than 2-fold. One of them was ZEB2-AS1. Inhibition of ZEB2-AS1 can compensate for the EMT phenotype and reverse the expression of EMT markers regulated by HBx. Additionally, HBx affected the Wnt signalling pathway.ConclusionsHBx promotes HCC cell metastasis by inducing EMT, which is at least partly mediated by lncRNAs.


International Journal of Molecular Sciences | 2017

Plasma Gelsolin Induced Glomerular Fibrosis via the TGF-β1/Smads Signal Transduction Pathway in IgA Nephropathy

Lei Zhang; Changsong Han; Fei Ye; Yan He; Yinji Jin; Tianzhen Wang; Yiqi Wu; Yang Jiang; Fengmin Zhang; Xiaoming Jin

Glomerular fibrosis has been shown to be closely related to the progression and prognosis of IgA nephropathy (IgAN). However, mechanism underlying IgAN glomerular fibrosis remains unclear. Recently, our study showed that plasma gelsolin (pGSN) was decreased in the serum of an IgAN mouse model and that pGSN deposition was found in the glomeruli. Another cytokine, TGF-β1, which is closely related to glomerular fibrosis, was also found to be highly expressed in the glomeruli. In the present study, we report that pGSN induces glomerular fibrosis through the TGF-β1/Smads signal transduction pathway. This is supported by the following findings: human mesangial cells (HMCs) show remarkable morphological changes and proliferation in response to co-stimulation with pGSN and polymeric IgA1 (pIgA1) from IgAN patients compared to other controls. Moreover, ELISA assays showed that more TGF-β1 secretion was found in HMCs supernatants in the co-stimulation group. Further experiments showed increased TGF-β1, Smad3, p-Smad2/3, Smad4, and collagen 1 and decreased Smad7 expression in the co-stimulation group. Our present study implied that the synergistic effect of pGSN and pIgA induced glomerular fibrosis via the TGF-β1/Smads signal transduction pathway. This might be a potential mechanism for the glomerular fibrosis observed in IgAN patients.


Molecular Medicine Reports | 2015

Adipose differentiation-related protein is not involved in hypoxia inducible factor-1-induced lipid accumulation under hypoxia

Guomin Shen; Ning Ning; Xingsheng Zhao; Xi Liu; Guangyu Wang; Tianzhen Wang; Ran Zhao; Chao Yang; Dongmei Wang; Pingyuan Gong; Yan Shen; Yongjian Sun; Xiao Zhao; Yinji Jin; Weiwei Yang; Yan He; Lei Zhang; Xiaoming Jin; Xiaobo Li

Increasing evidence has showed that hypoxia inducible factor-1 (HIF1) has an important role in hypoxia-induced lipid accumulation, a common feature of solid tumors; however, its role remains to be fully elucidated. Adipose differentiation-related protein (ADRP), a structural protein of lipid droplets, is found to be upregulated under hypoxic conditions. In the present study, an MCF7 breast cancer cell line was used to study the role of ADRP in hypoxia-induced lipid accumulation. It was demonstrated that hypoxia induced the gene expression of ADRP in a HIF1-dependent manner. Increases in the mRNA and protein levels of ADRP was accompanied by increased HIF1A activity. In addition, a significant decrease in the mRNA and protein levels of ADRP were detected in presence of siRNA targeting HIF1A. Using a dual-luciferase reporting experiment and chromatin immunoprecipitation assay, the present study demonstrated that ADRP is a direct target gene of HIF1, and identified a functional hypoxia response element localized 33 bp upstream of the transcriptional start site of the ADRP gene. Furthermore, the present study demonstrated the role of ADRP in low density liporotein (LDL) and very-LDL uptake-induced lipid accumulation under hypoxia. The knockdown of ADRP did not reduce HIF1-induced lipid accumulation under hypoxia. Together, these results showed that ADRP may be not involved in HIF1-induced lipid accumulation.

Collaboration


Dive into the Yinji Jin's collaboration.

Top Co-Authors

Avatar

Xiaoming Jin

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Tianzhen Wang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Fengmin Zhang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaobo Li

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Ran Zhao

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Yan He

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Lei Zhang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Weiwei Yang

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Yiqi Wu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Yuhua Zhang

Harbin Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge