Network


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

Hotspot


Dive into the research topics where Jingshu Geng is active.

Publication


Featured researches published by Jingshu Geng.


The Journal of Pathology | 2009

Reduced expression and novel splice variants of ING4 in human gastric adenocarcinoma

Ming Li; Yan Jin; Wenjing Sun; Yang Yu; Jing Bai; Dandan Tong; Jiping Qi; Jin-rong Du; Jingshu Geng; Qi Huang; Xiaoyi Huang; Yun Huang; Fei-fei Han; Xiangning Meng; Jesusa L. Rosales; Ki-Young Lee; Songbin Fu

ING4, a new member of the ING (inhibitor of growth) family of tumour suppressor genes, has been found to be deleted or down‐regulated in gliomas, breast tumours, and head and neck squamous cell carcinomas. The goal of the present study was to investigate whether the expression and alternative splicing of ING4 transcripts are involved in the initiation and progression of stomach adenocarcinoma. ING4 mRNA and protein expression was examined in gastric adenocarcinoma tissues and human gastric adenocarcinoma cell lines by RT‐PCR, real‐time RT‐PCR, tissue microarray immunohistochemistry, and western blot analysis. Alterations in ING4 transcripts were determined through sequence analysis of ING4 cDNA. Our data showed that ING4 mRNA and protein were dramatically reduced in stomach adenocarcinoma cell lines and tissues, and significantly less in female than in male patients. We also found that reduced ING4 mRNA expression correlated with the stage of the tumour. Interestingly, by sequence analysis, we discovered five novel aberrantly spliced variant forms of ING4_v1 and ING4_v2. These variants cause a codon frame‐shift and, eventually, deletion of the NLS or PHD domain contributing to the mislocalization of p53 and/or HAT/HDAC complexes and, subsequently, altered gene expression in gastric adenocarcinoma. These results suggest that attenuated and aberrant ING4 expression may be involved in the initiation and progression of stomach adenocarcinoma. Copyright


Journal of Clinical Laboratory Analysis | 2008

Elevated serum level and gene polymorphisms of TGF-β1 in gastric cancer

Xue Li; Zhichao Yue; Yun-Yan Zhang; Jing Bai; Xiangning Meng; Jingshu Geng; Songbin Fu

Transforming growth factor (TGF)‐β1, as a candidate tumor marker, is currently of interest. In this study, serum TGF‐β1 levels in gastric cancer (GC) patients and healthy volunteers were measured using enzyme‐linked immunosorbent assay (ELISA). In addition, single nucleotide polymorphisms (SNPs) of the TGF‐β1 gene at codon 10 and codon 25 were identified by means of amplification refractory mutation system–polymerase chain reaction (ARMS‐PCR) and sequence analysis. Our results indicated that serum concentrations of TGF‐β1 in GC patients were significantly higher than those in the control, and positively correlated with tumor mass, invasion, metastasis, and clinical stage. The serum TGF‐β1 levels of patients recovering from radical resection were markedly lower than those before surgery. Meanwhile, no deoxyribonucleic acid (DNA) sequence variation at codon 25 of the TGF‐β1 gene was found and a TGF‐β1 gene polymorphism at codon 10 did not show obvious correlations with either TGF‐β1 expression or clinicopathological parameters of GC. Our evidence suggested that serum concentration of TGF‐β1 might be a novel tumor marker for GC and the polymorphisms of TGF‐β1 gene did not play a role as a determinant of serum TGF‐β1 concentration or as a genetic risk factor in the gastric carcinogenesis and progression. J. Clin. Lab. Anal. 22:164–171, 2008.


Journal of Investigative Medicine | 2012

Differential expression of PAI-RBP1, C1orf142, and COTL1 in non-small cell lung cancer cell lines with different tumor metastatic potential.

Wenjing Sun; Changlong Guo; Xiangning Meng; Yang Yu; Yan Jin; Dandan Tong; Jingshu Geng; Qi Huang; Jiping Qi; An Liu; Rongwei Guan; Lidan Xu; Donglin Sun; Wei Ji; Peng Liu; Fangli Liu; Haiming Sun; Guohua Ji; Songbin Fu; Jing Bai

Human non–small cell lung cancer (NSCLC) is one of the most common malignancies in the modern world. Its recurrence is mainly due to its ability to invade and metastasize. However, the precise mechanism for tumor development and metastasis is still not fully understood. To shed light on the development of lung cancer, the human giant cell lung carcinoma cell lines 95D with high metastatic potential and 95C with low metastatic potential were selected in this study. The 2 cell lines originated from the same parental cell and share a similar genetic background. In the current study, we identified 3 differentially expressed proteins in 95C and 95D cell lines, namely, PAI-RBP1, C1orf142, and COTL1, by using 2-dimensional electrophoresis proteomics analysis. We found that PAI-RBP1 and C1orf142 expression levels were higher in 95D than in 95C cells, whereas COTL1 expression level was lower in 95D when compared to 95C cells. We also confirmed these results by reverse transcription–polymerase chain reaction and immunoblotting analyses. The messenger RNA and protein levels of PAI-RBP1 and C1orf142 were much higher in 95D than in 95C cells, and COTL1 expression level was lower in 95D than in 95C cells. The PAI-RBP1 expression was assessed by immunohistochemistry in 70 NSCLC and 7 normal lung tissue samples from patients. PAI-RBP1 expression level was higher in tumor tissues (positive staining in 87.1% of cases [61/70]) than in normal tissues (positive staining in 14.3% of cases [1/7]). In conclusion, by studying protein expression in NSCLC cell lines with high and low metastasis as well as in human lung cancer tissues, we have identified 3 proteins, namely, PAI-RBP1, C1orf142, and COTL1, which were differentially expressed in NSCLC cell lines with different metastatic potential. In addition, we also found that PAI-RBP1 might contribute to NSCLC development.


Molecules and Cells | 2011

Identification of Novel Subregions of LOH in Gastric Cancer and Analysis of the HIC1 and TOB1 Tumor Suppressor Genes in These Subregions

Jingcui Yu; Peng Liu; Xiaobo Cui; Yu Sui; Guohua Ji; Rongwei Guan; Donglin Sun; Wei Ji; Fangli Liu; An Liu; Yuzhen Zhao; Yang Yu; Yan Jin; Jing Bai; Jingshu Geng; Yingwei Xue; Jiping Qi; Ki-Young Lee; Songbin Fu

Previously, we identified 3 overlapping regions showing loss of heterozygosity (LOH, R1–R3 from 11 to 30 cM) on chromosome 17 in 45 primary gastric cancers (GCs). The data indicated the presence of tumor suppressor genes (TSGs) on chromosome 17 involved in GC. Among the putative TSGs in these regions, HIC1 (in SR1) and TOB1 (in SR3) remain to be examined in GC. By immunohistochemistry (IHC), methylation-specific PCR (MSP) and western blot, we evaluated the expression and regulation status for HIC1 and TOB1 protein in GC. We narrowed down the deletion intervals on chromosome 17 and defined five smaller LOH subregions, SR1–SR5 (0.54 to 3.42 cM), in GC. We found that HIC1 had downregulated expression in 86% (91/106) and was methylated in 87% (26/30) of primary GCs. Of the primary GCs showing downregulation of HIC1 protein, 75% (18/24) had methylated HIC1 gene. TOB1 was either absent or expressed at reduced levels in 75% (73/97) of the GC samples. In addition, a general reduction was found in total and the ratio of unphosphorylated to phosphorylated TOB1 protein levels in the differentiated GC cell lines. Further analysis revealed significant simultaneous downregulation of both HIC1 and TOB1 protein in GC tissue microarray samples (67%, 52/78) and in primary GCs (65%, 11/17). These results indicate that silencing of HIC1 and TOB1 expression is a common occurrence in GC and may contribute to the development and progression of the disease.


Pathobiology | 2009

RUNX3 Inhibits Cell Proliferation and Induces Apoptosis by TGF-β-Dependent and -Independent Mechanisms in Human Colon Carcinoma Cells

Dandan Tong; Ying Jiang; Ming Li; Dan Kong; Xiang-nin Meng; Yuzhen Zhao; Yan Jin; Jing Bai; Songbin Fu; Jingshu Geng

Background: Genes involved in the TGF-β signaling pathway are often altered in several types of cancers. The TGF-β-resistant human colon cancer cell line HT-29 has inactivated TβRII and deficient expression of RUNX3 and Smad4, which are involved in the TGF-β signaling pathway. Methods: Western blot and immunocytochemistry were performed to confirm gene expression, the MTT assay to detect cell growth, flow cytometry to investigate the cell cycle and the TUNEL to detect cell apoptosis. Results: In the absence of TGF-β, Bim was upregulated, cell growth was inhibited and apoptosis was induced. TGF-β treatment did not affect RUNX3 expression; however, the increase in Bim expression was significant and time dependent. Interestingly, Smad4 but not Smad2/3 was also upregulated upon exposure to TGF-β. This was not the case after TGF-β treatment of parent HT-29 cells. As expected, TGF-β further inhibited cell growth and induced apoptosis in HT-29/RUNX3+ cells. Conclusion: Our data demonstrate that RUNX3 is involved in TGF-β-dependent and -independent cell growth inhibition and apoptosis induction pathways.


Oncology Reports | 2014

S-allylmercaptocysteine promotes MAPK inhibitor-induced apoptosis by activating the TGF-β signaling pathway in cancer cells

Dandan Tong; Hui Qu; Xiangning Meng; Yang Jiang; Duanyang Liu; Shengqian Ye; He Chen; Yan Jin; Songbin Fu; Jingshu Geng

S-allylmercaptocysteine (SAMC), one of the water-soluble organosulfur garlic derivatives, can induce the apoptosis of many types of cancer cells through the MAPK signaling pathway. The TGF-β signaling pathway also plays a pivotal role in the process of oncogenesis, and has a certain crosstalk with the MAPK pathway. In the present study, hepatocellular carcinoma cell line HepG2 with an intact TGF-β signal and colon cancer cell line SW620 with an imperfect TGF-β signal were selected to ascertain whether SAMC induces the apoptosis of cancer cells by TGF-β signaling. In both cell lines treated with MAPK inhibitors and SAMC, an increased apoptosis rate was observed by electron microscopy, TUNEL and flow cytometric assays. Immunohistochemistry and western blot assays showed that SAMC induced the apoptosis of cancer cells by activating TGF-β1, TβRII, p-smad2/3, smad4 and smad7 signals, and promoting Bim expression while decreasing Bcl-2 expression and finally activating the mitochondrial apoptosis pathway proteins caspase-3 and caspase-9 in the HepG2 cell line. In contrast, in the SW620 cell line, the apoptosis induced by SAMC only affected TGF-β1 and smad7 signals, and promoted the expression of Bax and Bad and finally activated the mitochondrial apoptosis pathway protein caspase-9. When we compare the apoptosis rate in both cell lines, a significantly lower apoptosis rate was noted in the SW620 cell line than the rate noted in the HepG2 cell line. In summary, SAMC induces the apoptosis of cancer cells by activating the TGF-β signaling pathway, after MAPK signaling is inhibited.


Oncotarget | 2017

TRIB1 promotes colorectal cancer cell migration and invasion through activation MMP-2 via FAK/Src and ERK pathways

Yuhui Wang; Nan Wu; Bo Pang; Dandan Tong; Donglin Sun; Haiming Sun; Chunyu Zhang; Wenjing Sun; Xiangning Meng; Jing Bai; Feng Chen; Jingshu Geng; Songbin Fu; Yan Jin

Colorectal cancer (CRC) is the third most common cancer in the world and distant metastasis is the leading cause of death among CRC patients. However, the underlying mechanisms of distant metastasis remain largely unknown. Amplification of 8q24 is a common chromosomal abnormality in CRC. In the present study, a putative oncogene at 8q24, TRIB1, was characterized for its role in CRC metastasis and underlying molecular mechanisms. Higher expression of TRIB1 protein was detected in 58/83 (69.9%) of CRC tissues, compared with adjacent non-tumor tissues. Moreover, the expression of TRIB1 was significantly associated with distant metastasis (P=0.043) and advanced staging (P=0.008) in CRC tissues. TRIB1 overexpression was also correlated with poor prognosis in CRC patients as analyzed in PrognoScan database. In addition, elevated expression of TRIB1 promoted CRC cell motility and adhesive ability, while silencing of TRIB1 reduced those effects. Further study revealed that TRIB1-mediated migration and invasion of CRC cells required up-regulation of MMP-2 through the activation of FAK/Src and ERK pathway. Collectively, the results suggest that TRIB1 promotes CRC cell motility by activation MMP-2 via the FAK/Src and ERK pathways. It may provide a potential diagnostic and therapeutic target for CRC.


PLOS ONE | 2015

Ribosomal L22-like1 (RPL22L1) Promotes Ovarian Cancer Metastasis by Inducing Epithelial-to-Mesenchymal Transition.

Nan Wu; Jia Wei; Yuhui Wang; Jinyan Yan; Ying Qin; Dandan Tong; Bo Pang; Donglin Sun; Haiming Sun; Yang Yu; Wenjing Sun; Xiangning Meng; Chunyu Zhang; Jing Bai; Feng Chen; Jingshu Geng; Ki-Young Lee; Songbin Fu; Yan Jin

Double minute chromosomes (DMs) have important implications for cancer progression because oncogenes frequently amplified on them. We previously detected a functionally undefined gene amplified on DMs, Ribosomal L22-like1 (RPL22L1). The relationship between RPL22L1 and cancer progression is unknown. Here, RPL22L1 was characterized for its role in ovarian cancer (OC) metastasis and its underlying mechanism was examined. DNA copy number and mRNA expression of RPL22L1 in OC cells was analyzed using data obtained from The Cancer Genome Atlas and the Gene Expression Omnibus database. An immunohistochemical analysis of clinical OC specimens was performed and the relationships between expression level and clinicopathological factors were evaluated. Additionally, in vivo and in vitro assays were performed to understand the role of RPL22L1 in OC. RPL22L1 expression was higher in OC specimens than in normal tissues, and its expression level was highly positively correlated with invasion and lymph node metastasis (P < 0.05). RPL22L1 over-expression significantly enhanced intraperitoneal xenograft tumor development in nude mice and promoted invasion and migration in vitro. Additionally, RPL22L1 knockdown remarkably inhibited UACC-1598 cells invasion and migration. Further, RPL22L1 over-expression up-regulated the mesenchymal markers vimentin, fibronectin, and α-SMA, reduced expression of the epithelial markers E-cadherin, α-catenin, and β-catenin. RPL22L1 inhibition reduced expression of vimentin and N-cadherin. These results suggest that RPL22L1 induces epithelial-to-mesenchymal transition (EMT). Our data showed that the DMs amplified gene RPL22L1 is critical in maintaining the aggressive phenotype of OC and in triggering cell metastasis by inducing EMT. It could be employed as a novel prognostic marker and/or effective therapeutic target for OC.


Journal of International Medical Research | 2008

Human Gastric Adenocarcinoma Allelotype on Chromosomes 17 and 18

Jingcui Yu; H Zhou; Jing Bai; Yang Yu; Jingshu Geng; Jiping Qi; Songbin Fu

Allelic losses of multiple chromosome loci in gastric adenocarcinoma suggest that inactivation of tumour suppressor genes in these regions may be important for tumourigenesis. To define deletion intervals and find candidate tumour suppressor genes involved in gastric adenocarcinoma pathogenesis, a genome-wide search for loss of heterozygosity (LOH) was conducted in 45 patients with primary gastric adenocarcinoma. Investigations using 29 microsatellite markers spanning chromosomes 17 and 18 showed allelic deletion in 29 (64%) specimens at one or more loci. Five LOH overlap regions, three newly identified as deletion regions, were defined: RI, D17S831–D17S921 at 17p12-13.3; RII, D17S1868–D17S787 at 17q21.3-22; RIII, D17S785–D17S928 at 17q25.3; RIV, D18S61–D18S1161 at 18q22; and RV, D18S462–D18S70 at 18q22-q23. Eleven (24%) patients with chromosome 17 allelic loss also showed LOH on 18q, with at least one region of overlapping. LOH mapping showed allelic losses were widespread on both chromosomes and suggests the possibility that multiple tumour suppressor genes, including one or more that are unknown, might be inactivated in the aetiology of gastric adenocarcinoma.


Cancer Science | 2017

NUBPL , a novel metastasis-related gene, promotes colorectal carcinoma cell motility by inducing epithelial-mesenchymal transition

Yuhui Wang; Nan Wu; Donglin Sun; Haiming Sun; Dandan Tong; Duo Liu; Bo Pang; Su Li; Jia Wei; Jialin Dai; Yang Liu; Jing Bai; Jingshu Geng; Songbin Fu; Yan Jin

Nucleotide binding protein‐like, NUBPL, is an assembly factor for human mitochondrial complex I, which is the biggest member of the mitochondrial respiratory chain. However, the relationship between NUBPL and carcinoma progression remains unknown. In this study, NUBPL was characterized for its role in colorectal cancer (CRC) and the underlying molecular mechanisms. Data (n = 197) from the Oncomine database revealed that mRNA levels of NUBPL were remarkably overexpressed in CRC tissues compared with normal tissues. In addition, immunohistochemical analysis of 75 pairs of CRC and non‐tumor tissues showed that the expression level of NUBPL was significantly higher in CRC tissues, and its expression level was positively associated with lymph node metastasis (P = 0.028) and advanced staging (P = 0.030). Expression of NUBPL in metastatic lymph nodes of CRC patients was also detected by immunohistochemical staining and high expression levels of NUBPL were observed. Overexpression of NUBPL significantly promoted the migration and invasion ability of CRC cell lines SW480 and SW620, whereas knockdown of NUBPL lead to an opposite effect. Our further study found that NUBPL could induce epithelial–mesenchymal transition (EMT), characterized by downregulation of epithelial markers (E‐cadherin) and upregulation of mesenchymal markers (N‐cadherin and vimentin). Moreover, NUBPL was able to activate ERK, which is believed to promote EMT and tumor metastasis. Inhibition of ERK suppressed the NUBPL‐induced changes in EMT and cell motility. These data showed that NUBPL plays a vital role in CRC migration and invasion by inducing EMT and activating ERK. It might be a novel therapeutic target for CRC.

Collaboration


Dive into the Jingshu Geng's collaboration.

Top Co-Authors

Avatar

Songbin Fu

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Jing Bai

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Yan Jin

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Dandan Tong

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiangning Meng

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Yang Yu

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jiping Qi

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Wenjing Sun

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Donglin Sun

Harbin Medical University

View shared research outputs
Top Co-Authors

Avatar

Haiming Sun

Harbin Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge