Network


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

Hotspot


Dive into the research topics where Ti Wen is active.

Publication


Featured researches published by Ti Wen.


Molecular BioSystems | 2013

Glycoproteomic analysis of tissues from patients with colon cancer using lectin microarrays and nanoLC-MS/MS

Yangguang Li; Ti Wen; Minzhi Zhu; Lixin Li; Jun Wei; Xiaoli Wu; Mingzhou Guo; Shuangping Liu; Huiyuan Zhao; Siyuan Xia; Weili Huang; Puyue Wang; Zhenzhou Wu; Liqing Zhao; Wenqing Shui; Zheng Li; Zhinan Yin

The current study evaluated the glycoproteomic profile of tissues from colon cancer patients. The lectin microarray was first performed to compare the glycoprotein profiles between colon cancer and matched normal tissues. Level of N-acetylglucosamine (GlcNAc) that Solanum tuberosum lectin (STL) bound was found to be elevated in colon cancer, which was verified through lectin histochemistry. The subsequent glycoproteomic analysis based on STL enrichment of glycoproteins followed by label-free quantitative nano liquid chromatography-mass spectrometry/mass spectrometry (nanoLC-MS/MS) analysis identified 72 proteins in high confidence. Among these proteins, 17 were exclusively detected in cancer tissues, and 14 were significantly upregulated in tumor tissues. Annexin A1 and HSP90β were chosen for further investigation by immunoprecipitation coupled with lectin blots, western blots and tissue microarrays. Both Annexin A1 and HSP90β were GlcNAcylated, and their protein expressions were elevated in colon cancer, compared to normal tissues. Moreover, specific changes of GlcNAc abundances in Annexin A1 and HSP90β suggested that tumor-specific glycan patterns could serve as candidate biomarkers of colon cancer for distinguishing cancer patients from healthy individuals.


Oncology Reports | 2015

Silencing β-linked N-acetylglucosamine transferase induces apoptosis in human gastric cancer cells through PUMA and caspase-3 pathways

Ti Wen; Kezuo Hou; Zhi Li; Lu Li; Hanjie Yu; Yunpeng Liu; Yangguang Li; Zhinan Yin

β-linked N-acetylglucosamine (GlcNAc) is a monosaccharide that is catalyzed by O-GlcNAcylation transferase (OGT) to bind serine or threonine hydroxyl moieties of numerous nuclear and cytoplasmic proteins. Recent studies have shown that O-GlcNAcylation is elevated in various cancer types, which is associated with oncogenesis and tumor progression. However, whether OGT is expressed and/or plays a role in gastric cancer is unknown. In the present study, we used qPCR to determine that OGT mRNA levels are significantly elevated in gastric cancer tissues compared with that in corresponding adjacent tissues. In addition, in vivo silencing of OGT in nude mice suppressed tumor proliferation and decreased tumor burden. Furthermore, in vitro OGT knockdown induced more cell apoptosis through increasing PUMA and caspase-3 expression. We used a glycan-binding protein gene microarray to identify potential downstream target genes of OGT, and found that apoptosis-related genes such as galectin and HBEGF were decreased after OGT suppression, suggesting that OGT silencing induces apoptosis in gastric cancer tissues. We concluded that OGT plays a key role in gastric cancer proliferation and survival, and could be a potential target for therapy.


International Immunopharmacology | 2012

A novel tylophorine analog NK-007 ameliorates colitis through inhibition of innate immune response.

Ti Wen; Yangguang Li; Meng Wu; Xi Chen; Lin Han; Xiucong Bao; Ziwen Wang; Kailiang Wang; Yanna Hu; Xinglong Zhou; Zhenzhou Wu; Puyue Wang; Zhangyong Hong; Liqing Zhao; Qingmin Wang; Zhinan Yin

In this study, we synthesized (±)-tylophorine malate (NK-007), an analog of tylophorine (DCB3503), and analyzed its anti-inflammatory effect in vivo using a dextran sulfate sodium (DSS)-induced colitis model and an acetic acid-induced colitis model. As indicated by disease activity index (DAI) and degree of macroscopic colonic damage, NK-007 can significantly suppress colitis. To delineate the underlying mechanism, we have explored the influence of NK-007 on the production of TNF-α by murine primary bone marrow-derived dendritic cells (BMDCs) as well as monocyte/macrophage cell line Raw 264.7 triggered by lipopolysaccharide (LPS). For both types of innate immune cells, NK-007 showed a potent TNF-α inhibitory effect, and has in addition reduced the expression of IL-12 in BMDCs. Moreover, Raw cells treated with NK-007 also showed decreased phosphorylation of NF-κB, which may explain the protective immune-regulatory effect of NK-007 for experimental colitis.


Neoplasia | 2017

E3 Ubiquitin Ligase Cbl-b Prevents Tumor Metastasis by Maintaining the Epithelial Phenotype in Multiple Drug-Resistant Gastric and Breast Cancer Cells

Ling Xu; Ye Zhang; Xiujuan Qu; Xiaofang Che; Tianshu Guo; Ying Cai; Aodi Li; Danni Li; Ce Li; Ti Wen; Yibo Fan; Kezuo Hou; Yanju Ma; Xuejun Hu; Yunpeng Liu

Multiple drug resistance (MDR) and metastasis are two major factors that contribute to the failure of cancer treatment. However, the relationship between MDR and metastasis has not been characterized. Additionally, the role of the E3 ubiquitin ligase Cbl-b in metastasis of MDR gastric and breast cancer is not well known. In the present study, we found that MDR gastric and breast cancer cells possess a typical mesenchymal phenotype and enhanced cell migration capacity. Additionally, Cbl-b is poorly expressed in MDR gastric and breast cancer cells. In MDR gastric adenocarcinoma tissues, gastric cancer patients with low Cbl-b expression were more likely to have tumor invasion (P = .016) and lymph node metastasis (P = .007). Moreover, overexpression of Cbl-b reduced cell migration in MDR cell cultures both in vitro and in vivo. Cbl-b overexpression also prevented EMT by inducing ubiquitination and degradation of EGFR, leading to inhibition of the EGFR-ERK/Akt-miR-200c-ZEB1 axis. However, further overexpression of EGFR on a background of Cbl-b overexpression restored both the mesenchymal phenotype and cell migration capacity of MDR gastric and breast cancer cells. These results suggest that Cbl-b is an important factor for maintenance of the epithelial phenotype and inhibition of cell migration in MDR gastric and breast cancer cells.


FEBS Letters | 2018

E3 ubiquitin ligases Cbl‐b and c‐Cbl downregulate PD‐L1 in EGFR wild‐type non‐small cell lung cancer

Shuo Wang; Ling Xu; Xiaofang Che; Ce Li; Lu Xu; Kezuo Hou; Yibo Fan; Ti Wen; Xiujuan Qu; Yunpeng Liu

Anti‐PD‐1/PD‐L1 therapies have demonstrated prominent clinical effects in the treatment of non‐small cell lung cancer (NSCLC). However, limited understanding of the regulatory mechanisms of PD‐L1 has become one of the biggest challenges for further improving efficacy. In this study, we observed that in wild‐type EFGR cell lines A549 and H460, the ubiquitin ligases Cbl‐b and c‐Cbl inhibit PD‐L1 by inactivating STAT, AKT, and ERK signaling. MiR‐181a and miR‐940 were screened and validated to target Cbl‐b and c‐Cbl, respectively. Furthermore, in NSCLC tissues, the expression of Cbl‐b/c‐Cbl is negatively correlated with PD‐L1 expression. Taken together, these findings indicated a new regulatory mechanism for PD‐L1 in wild‐type EGFR NSCLC cell lines by Cbl‐b and c‐Cbl.


Translational Oncology | 2018

The Chemokine Receptor CXCR4 and c-MET Cooperatively Promote Epithelial-Mesenchymal Transition in Gastric Cancer Cells

Yu Cheng; Yongxi Song; Jinglei Qu; Xiaofang Che; Na Song; Yibo Fan; Ti Wen; Ling Xu; Jing Gong; Xiaoxun Wang; Chenlu Zhang; Xiujuan Qu; Yunpeng Liu

The C-X-C motif chemokine receptor 4 (CXCR4) pathway can promote tumor metastasis but is dependent on cross talk with other signaling pathways. The MET proto-oncogene (c-MET) participates in metastasis and is highly expressed in gastric cancer. However, the relationship between CXCR4 and c-MET signaling and their mechanisms of action in gastric cancer metastasis remain unclear. In this study, in vitro experiments demonstrated that C-X-C motif chemokine ligand 12 (CXCL12)/CXCR4 induces epithelial-mesenchymal transition (EMT) and promotes migration in gastric cancer cells, which is accompanied by c-MET activation. These phenomena were reversed by c-MET inhibition. Further investigation revealed that c-MET activation correlated with its interaction with caveolin 1 in lipid rafts, induced by CXCL12. In clinical samples, we observed a significant positive association between CXCR4 expression and c-MET phosphorylation (r = 0.259, P = .005). Moreover, samples expressing both receptors were found to indicate significantly poorer patient prognosis (P < .001). These results suggest that CXCL12 induces EMT at least partially through cross talk between CXCR4 and c-MET signaling. In addition, changes in these pathways could have clinical importance for the treatment of gastric cancer.


BMC Cancer | 2018

C-Cbl reverses HER2-mediated tamoxifen resistance in human breast cancer cells

Wei Li; Ling Xu; Xiaofang Che; Haizhou Li; Ye Zhang; Na Song; Ti Wen; Kezuo Hou; Yi Yang; Lu Zhou; Xing Xin; Lu Xu; Xue Zeng; Sha Shi; Yunpeng Liu; Xiujuan Qu; Yuee Teng

BackgroundTamoxifen is a frontline therapy for estrogen receptor (ER)-positive breast cancer in premenopausal women. However, many patients develop resistance to tamoxifen, and the mechanism underlying tamoxifen resistance is not well understood. Here we examined whether ER-c-Src-HER2 complex formation is involved in tamoxifen resistance.MethodsMTT and colony formation assays were used to measure cell viability and proliferation. Western blot was used to detect protein expression and protein complex formations were detected by immunoprecipitation and immunofluorescence. SiRNA was used to examine the function of HER2 in of BT474 cells. An in vivo xenograft animal model was established to examine the role of c-Cbl in tumor growth.ResultsMTT and colony formation assay showed that BT474 cells are resistant to tamoxifen and T47D cells are sensitive to tamoxifen. Immunoprecipitation experiments revealed ER-c-Src-HER2 complex formation in BT474 cells but not in T47D cells. However, ER-c-Src-HER2 complex formation was detected after overexpressing HER2 in T47D cells and these cells were more resistant to tamoxifen. HER2 knockdown by siRNA in BT474 cells reduced ER-c-Src-HER2 complex formation and reversed tamoxifen resistance. ER-c-Src-HER2 complex formation was also disrupted and tamoxifen resistance was reversed in BT474 cells by the c-Src inhibitor PP2 and HER2 antibody trastuzumab. Nystatin, a lipid raft inhibitor, reduced ER-c-Src-HER2 complex formation and partially reversed tamoxifen resistance. ER-c-Src-HER2 complex formation was disrupted by overexpression of c-Cbl but not by the c-Cbl ubiquitin ligase mutant. In addition, c-Cbl could reverse tamoxifen resistance in BT474 cells, but the ubiquitin ligase mutant had no effect. The effect of c-Cbl was validated in BT474 tumor-bearing nude mice in vivo. Immunofluorescence also revealed ER-c-Src-HER2 complex formation was reduced in tumor tissues of nude mice with c-Cbl overexpression.ConclusionsOur results suggested that c-Cbl can reverse tamoxifen resistance in HER2-overexpressing breast cancer cells by inhibiting the formation of the ER-c-Src-HER2 complex.


BMC Cancer | 2018

CXCL9/10/11, a regulator of PD-L1 expression in gastric cancer

Chenlu Zhang; Zhi Li; Ling Xu; Xiaofang Che; Ti Wen; Yibo Fan; Ce Li; Shuo Wang; Yu Cheng; Xiaoxun Wang; Xiujuan Qu; Yunpeng Liu

BackgroundProgrammed death-ligand 1 (PD-L1) is an immunosuppressor that plays an important role in cancer treatments. Although majority of the studies demonstrated that PD-L1 expression was regulated by cellular intrinsic and extrinsic controls, and IFN-γ was a key molecule of extrinsic control, other studies imply that other cytokines play important roles in PD-L1 expression. In this study, we investigated the regulation of PD-L1 by chemokine signaling pathway in gastric cancer (GC) cells.MethodsBioinformatics was used to explore the PD-L1-related genes in GC and propose a hypothesis. PD-L1 and CXCR3 expression were detected by western blot in SGC7901 and MKN74 cell lines. Meanwhile, PD-L1 and CXCR3 expressions were immunohistochemically assessed for their relevance. Moreover, PD-L1, pSTAT3 and pAkt were detected after treatment with CXCL9/10/11. Furthermore,PD-L1, pSTAT3 and pAkt were evaluated after blocking chemokine signaling in SGC7901 cells.ResultsBased on online database analysis, CXCL9/10/11-CXCR3 is proposed to upregulate PD-L1 expression by activating the STAT and PI3K-Akt pathways. This hypothesis was confirmed by in vitro and vivo experiments. CXCR3 and PD-L1 were expressed in GC cell lines and tissues, and the expression of CXCR3 and PD-L1 was positively related. PD-L1 was upregulated after treatment with CXCL9/10/11, accompanied by activation of STAT3 and Akt. After blocking chemokine signaling, upregulation of PD-L1 and activation of STAT3 and Akt were diminished.ConclusionsCXCL9/10/11-CXCR3 upregulated the expression of PD-L1 by activating the STAT and PI3K-Akt signaling pathways in GC cells. There was a significant positive correlation between the expression of PD-L1 and CXCR3 in gastric cancer patient tissues.


BMC Cancer | 2018

MicroRNA-29b-2-5p inhibits cell proliferation by directly targeting Cbl-b in pancreatic ductal adenocarcinoma

Ce Li; Qian Dong; Xiaofang Che; Ling Xu; Zhi Li; Yibo Fan; Kezuo Hou; Shuo Wang; Jinglei Qu; Lu Xu; Ti Wen; Xianghong Yang; Xiujuan Qu; Yunpeng Liu

BackgroundMicroRNAs can be used in the prognosis of malignancies; however, their regulatory mechanisms are unknown, especially in pancreatic ductal adenocarcinoma (PDAC).MethodsIn 120 PDAC specimens, miRNA levels were assessed by quantitative real time polymerase chain reaction (qRT-PCR). Then, the role of miR-29b-2-5p in cell proliferation was evaluated both in vitro (Trypan blue staining and cell cycle analysis in the two PDAC cell lines SW1990 and Capan-2) and in vivo using a xenograft mouse model. Next, bioinformatics methods, a luciferase reporter assay, Western blot, and immunohistochemistry (IHC) were applied to assess the biological effects of Cbl-b inhibition by miR-29b-2-5p. Moreover, the relationship between Cbl-b and p53 was evaluated by immunoprecipitation (IP), Western blot, and immunofluorescence.ResultsFrom the 120 PDAC patients who underwent surgical resection, ten patients with longest survival and ten with shortest survival were selected. We found that high miR-29b-2-5p expression was associated with good prognosis (p = 0.02). The validation cohort confirmed miR-29b-2-5p as an independent prognostic factor in PDAC (n = 100, 95% CI = 0.305–0.756, p = 0.002). Furthermore, miR-29b-2-5p inhibited cell proliferation, induced cell cycle arrest, and promoted apoptosis both in vivo and in vitro. Interestingly, miR-29b-2-5p directly bound the Cbl-b gene, down-regulating its expression and reducing Cbl-b-mediated degradation of p53. Meanwhile, miR-29b-2-5p expression was negatively correlated with Cbl-b in PDAC tissues (r = − 0.33, p = 0.001).ConclusionsTaken together, these findings indicated that miR-29b-2-5p improves prognosis in PDAC by targeting Cbl-b to promote p53 expression, and would constitute an important prognostic factor in PDAC.


Cancer Research | 2017

Abstract 4916: E3 ubiquitin ligase Cbl-b prevents tumor metastasis by maintaining the epithelial phenotype in multiple drug-resistant gastric and breast cancer cells

Ling Xu; Xiujuan Qu; Ye Zhang; Xiaofang Che; Tianshu Guo; Ying Cai; Aodi Li; Danni Li; Ce Li; Ti Wen; Yibo Fan; Kezuo Hou; Yanju Ma; Xuejun Hu; Yunpeng Liu

Background: Multiple drug resistance (MDR) and metastasis are two major unfavorable factors causing failure of cancer treatment. However, the relationship between MDR and metastasis is not characterized. Additionally, the role of the E3 ubiquitin ligase Cbl-b in metastasis of MDR gastric and breast cancer is little known. Methods: The effect of Cbl-b on tumor invasion and metastasis was analyzed in P-gp-positive gastric adenocarcinoma tissues. Transfections of Cbl-b WT and epidermal growth factor receptor (EGFR) WT plasmid were used to assess Cbl-b and EGFR involvement in epithelial-mesenchymal transition (EMT) and tumor metastasis. Results: MDR gastric and breast cancer cells possess a typical mesenchymal phenotype and an enhanced cell migration capacity. Additionally, Cbl-b is poorly expressed in MDR gastric and breast cancer cells. We performed immunohistochemical analysis on P-gp-positive gastric adenocarcinoma tissues and identified associations between Cbl-b expression and clinical pathological parameters. Gastric cancer patients with low Cbl-b expression were more likely to have tumor invasion (P=0.016) and lymph node metastasis (P=0.007). Moreover, over-expression of Cbl-b reduced cell migration in MDR cell cultures both in vitro and in vivo. Cbl-b over-expression also prevented EMT by inducing ubiquitination and degradation of EGFR, leading to inhibition of the EGFR- ERK/Akt-miR-200c-ZEB1 axis. However, further over-expression of EGFR on a background of Cbl-b over-expression restored both the mesenchymal phenotype and the cell migration capacity of MDR gastric and breast cancer cells. Conclusions: These results suggest that Cbl-b is an important factor for maintenance of the epithelial phenotype and inhibition of cell migration in MDR gastric and breast cancer cells. Note: This abstract was not presented at the meeting. Citation Format: Ling Xu, Xiujuan Qu, Ye Zhang, Xiaofang Che, Tianshu Guo, Ying Cai, Aodi Li, Danni Li, Ce Li, Ti Wen, Yibo Fan, Kezuo Hou, Yanju Ma, Xuejun Hu, Yunpeng Liu. E3 ubiquitin ligase Cbl-b prevents tumor metastasis by maintaining the epithelial phenotype in multiple drug-resistant gastric and breast cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4916. doi:10.1158/1538-7445.AM2017-4916

Collaboration


Dive into the Ti Wen'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