Network


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

Hotspot


Dive into the research topics where Zheng-Jun Zhou is active.

Publication


Featured researches published by Zheng-Jun Zhou.


Hepatology | 2012

Overexpression of CXCL5 mediates neutrophil infiltration and indicates poor prognosis for hepatocellular carcinoma

Shao-Lai Zhou; Zhi Dai; Zheng-Jun Zhou; Xiao-Ying Wang; Guo-Huan Yang; Zheng Wang; Xiao-Wu Huang; Jia Fan; Jian Zhou

CXCL5 (epithelial neutrophil‐activating peptide‐78) is a member of a proangiogenic subgroup of the CXC‐type chemokine family of small, secreted proteins. Recently, evidence that CXCL5 is involved in carcinogenesis and cancer progression has emerged. To investigate the role of CXCL5 in tumor growth, invasion, and prognosis of hepatocellular carcinoma (HCC), we examined CXCL5 messenger RNA (mRNA) and protein levels in HCC cell lines with various metastatic potentials and in three independent cohorts of 919 HCC patients. We found that CXCL5 expression was increased in the highly metastatic HCC cell lines and in tumor tissues from patients with recurrent HCC compared to controls. CXCL5 activated the PI3K‐Akt and ERK1/2 signaling pathways in HCC cells and promoted proliferation, migration, and invasion. Furthermore, we found that CXCL5 had a direct chemoattractant effect on neutrophils in vitro. In animal studies, the up‐regulation of CXCL5 in HCC cells promoted tumor growth, lung metastasis, and intratumoral neutrophil infiltration. Conversely, down‐regulation of CXCL5 in HCC cells reduced tumor growth, metastasis, and intratumoral neutrophil infiltration. Immunohistochemical analysis in HCC samples showed that overexpression of CXCL5 was well correlated with intratumoral neutrophil infiltration, shorter overall survival, and tumor recurrence. Multivariate analysis revealed that CXCL5 overexpression alone, or combined with the presence of intratumoral neutrophils, was an independent prognostic indicator for overall survival and cumulative recurrence. Conclusion: CXCL5 promotes HCC cell proliferation, invasion, and intratumoral neutrophil infiltration. CXCL5 overexpression, alone or combined with intratumoral neutrophil presence, is a novel prognostic predictor, and CXCL5 is a potential therapeutic target for HCC. (HEPATOLOGY 2012;56:2242–2254)


International Journal of Cancer | 2013

Overexpression of HnRNP A1 promotes tumor invasion through regulating CD44v6 and indicates poor prognosis for hepatocellular carcinoma

Zheng-Jun Zhou; Zhi Dai; Shao-Lai Zhou; Xiu-Tao Fu; Yi-Ming Zhao; Ying-Hong Shi; Jian Zhou; Jia Fan

Heterogeneous ribonucleoprotein (hnRNP) A1 is a member of the A/B subfamily of ubiquitously expressed hnRNPs, which have a wide variety of functions in gene expression and signal transduction. To investigate the biological function and clinical significance of hnRNP A1 in hepatocellular carcinoma (HCC), we measured hnRNP A1 expression in four HCC cell lines and two independent cohorts of HCC patients. We found that hnRNP A1 was overexpressed in the highly metastatic HCC cell lines and in tumor tissues of patients with recurrent HCC. Knockdown of hnRNP A1 in highly metastatic HCC cells caused a significant decrease in cell invasion, while upregulation of hnRNP A1 in poorly metastatic HCC cells led to a significant increase in their invasive capacity. We found that this effect may occur through the regulation of CD44v6 expression by hnRNP A1 in HCC cells. Both quantitative reverse transcription‐polymerase chain reaction (qRT‐RCR) and immunohistochemistry revealed that hnRNP A1 was upregulated in HCC tissues and coincided with overexpression of CD44v6. HCC patients with high hnRNP A1 tended to have higher levels of CD44v6, shorter overall survival (OS) and higher rates of tumor recurrence. Multivariate analyses revealed that hnRNP A1 alone or in combination with CD44v6 were independent prognostic indicators for OS and time to recurrence and have potential as therapeutic targets. In conclusion, overexpression of hnRNP A1 promotes HCC invasion by regulating the level of CD44v6 and indicates a poor prognosis for HCC patients after curative resection.


Cancer Letters | 2015

CXCR2/CXCL5 axis contributes to epithelial–mesenchymal transition of HCC cells through activating PI3K/Akt/GSK-3β/Snail signaling

Shao-Lai Zhou; Zheng-Jun Zhou; Zhi-Qiang Hu; Xun Li; Xiaowu Huang; Zheng Wang; Jia Fan; Zhi Dai; Jian Zhou

Upregulation of CXCR2 in tumor cells has been documented in several types of cancer. As one of its ligands, CXCL5 is associated with neutrophil infiltration and poor prognosis in hepatocellular carcinoma (HCC). However, little is known about the role of the CXCR2/CXCL5 axis in the invasion and metastasis of HCC cells. In this study, we examined CXCR2 expression in human HCC cell lines and in three independent cohorts of HCC patients. The molecular effects of high expression levels of CXCR2 and CXCL5 in HCC cells were determined using qRT-PCR, western blot analysis, immunofluorescence, matrigel invasion assay, and xenograft mouse models. We found that high levels of CXCR2 correlated with progression and poor prognosis in human HCC. CXCR2/CXCL5 together promoted cell spreading by inducing the epithelial-mesenchymal transition (EMT) through activation of the PI3K/Akt/GSK-3β/Snail signaling pathway. In clinical HCC samples, high expression of both CXCR2 and CXCL5 showed a significant correlation with the activation of PI3K/Akt/GSK-3β/Snail signaling and EMT phenotype. In conclusion, our data showed that the CXCR2/CXCL5 axis contributes to EMT of HCC cells through activating PI3K/Akt/GSK-3β/Snail signaling, and it may serve as a potential therapeutic target.


Carcinogenesis | 2014

CXCL5 contributes to tumor metastasis and recurrence of intrahepatic cholangiocarcinoma by recruiting infiltrative intratumoral neutrophils.

Shao-Lai Zhou; Zhi Dai; Zheng-Jun Zhou; Qing Chen; Zheng Wang; Yong-Sheng Xiao; Zhi-Qiang Hu; Xiao-Yong Huang; Guo-Huan Yang; Ying-Hong Shi; Shuang-Jian Qiu; Jia Fan; Jian Zhou

CXCL5 is a member of the CXC-type chemokine family that may play a role in carcinogenesis and cancer progression. This study investigates the biological function and clinical significance of CXCL5 in intrahepatic cholangiocarcinoma (ICC). We demonstrated that CXCL5 was overexpressed in ICC cell lines and tumor samples compared with paired normal tissues. CXCL5 had a direct chemoattractant effect on neutrophils in vitro through PI3K-Akt and extracellular signal-regulated kinase 1/2 signaling pathways. In animal studies, CXCL5 promoted tumor growth and metastasis without altering in vitro proliferative and invasive ability of ICC cells, and this effect was mediated by the recruitment of intratumoral infiltrative neutrophils by tumor-derived CXCL5. Immunohistochemical analysis of ICC samples showed that overexpression of CXCL5 correlated strongly with intratumoral neutrophil infiltration, shorter overall survival and high tumor recurrence. Multivariate analysis revealed that CXCL5 overexpression alone, or combined with the presence of intratumoral neutrophils, was an independent prognostic indicator for ICC. In conclusion, our data showed that CXCL5 promotes ICC growth and metastasis by recruiting intratumoral neutrophils. CXCL5 alone or combined with intratumoral neutrophils is a novel prognostic predictor for ICC patients and a potential therapeutic target.


International Journal of Oncology | 2015

Macrophage-secreted IL-8 induces epithelial-mesenchymal transition in hepatocellular carcinoma cells by activating the JAK2/STAT3/Snail pathway.

Xiu-Tao Fu; Zhi Dai; Kang Song; Zhuojun Zhang; Zheng-Jun Zhou; Shao-Lai Zhou; Yi-Ming Zhao; Yong-Sheng Xiao; Qi-Man Sun; Zhen-Bin Ding; Jia Fan

Macrophages are a major component of the leukocyte infiltrate of tumors and play a pivotal role in the progression of hepatocellular carcinoma (HCC). However, the molecular mechanisms by which macrophages promote HCC invasion are poorly understood. The present study was undertaken to investigate the relationship between macrophages and epithelial-mesenchymal transition (EMT) of HCC. Double-staining immunohistochemistry was used to observe the association between macrophages and EMT markers in clinical HCC samples and it showed that EMT primarily occurred at the edge of the tumor nest, in which infiltrating macrophages were always observed. This indicated that CD68 which is a marker of macrophages, was correlated with EMT marker levels. In addition, after being cultured with macrophages for 24 h, the ability of HCC cells to migrate and invade increased, Snail and N-Cadherin expression was upregulated, and E-Cadherin was downregulated. An antibody array assay was applied to analyze the supernatant of these cultures and it demonstrated IL-8 increased significantly in the macrophage co-culture system. Finally, the role of macrophage-derived IL-8 in the invasion of HCC cells was assayed, and downstream signaling pathways were also investigated. We found that IL-8: i) may induce EMT and promote HCC cell migration and invasion and ii) is associated with the JAK2/STAT3/Snail signaling pathway. Taking together, these findings revealed that macrophages that have infiltrated tumors may induce epithelial-mesenchymal transition of HCC cells via the IL-8 activated JAK2/STAT3/Snail pathway. Thus, this may offer a potential target for developing new HCC therapies.


Hepatology | 2016

miR‐28‐5p‐IL‐34‐Macrophage Feedback Loop Modulates Hepatocellular Carcinoma Metastasis

Shao-Lai Zhou; Zhi-Qiang Hu; Zheng-Jun Zhou; Zhi Dai; Zheng Wang; Ya Cao; Jia Fan; Xiaowu Huang; Jian Zhou

MicroRNAs (miRNAs) play a critical role in regulation of tumor metastasis. However, the role of these molecules in hepatocellular carcinoma (HCC) has not been fully elucidated. In this study, we employed miRNA‐sequencing and identified 22 miRNAs involved in HCC metastasis. One of these, miR‐28‐5p, was down‐regulated in HCCs. This down‐regulation correlated with tumor metastasis, recurrence, and poor survival. Biofunctional investigations revealed that miR‐28‐5p deficiency promoted tumor growth and metastasis in nude mice without altering the in vitro biological characteristics of HCC cells. Through gene expression profiles and bioinformatics analysis, we identified interleukin‐34 (IL‐34) as a direct target of miR‐28‐5p, and the effects of miR‐28‐5p deficiency on HCC growth and metastasis was dependent on IL‐34‐mediated tumor‐associated macrophage (TAM) infiltration. Moreover, we found that TAMs induced by miR‐28‐5p‐IL‐34 signaling inhibit miR‐28‐5p expression on HCC cells by transforming growth factor beta 1, resulting in an miR‐28‐5p‐IL‐34‐macrophage‐positive feedback loop. In clinical HCC samples, miR‐28‐5p levels were inversely correlated with IL‐34 expression and the number of TAMs. Patients with low miR‐28‐5p expression, high IL‐34 levels, and high numbers of TAMs had a poor prognosis with shorter overall survival and time to recurrence. Conclusion: A miR‐28‐5p‐IL‐34‐macrophage feedback loop modulates HCC metastasis and serves as a novel prognostic factor as well as a therapeutic target for HCC. (Hepatology 2016;63:1560‐1575)


Cancer Research | 2014

HNRNPAB Induces Epithelial–Mesenchymal Transition and Promotes Metastasis of Hepatocellular Carcinoma by Transcriptionally Activating SNAIL

Zheng-Jun Zhou; Zhi Dai; Shao-Lai Zhou; Zhi-Qiang Hu; Qing Chen; Yi-Ming Zhao; Ying-Hong Shi; Qiang Gao; Wei-Zhong Wu; Shuang-Jian Qiu; Jian Zhou; Jia Fan

Expression of heterogeneous nuclear ribonucleoprotein AB (HNRNPAB) has been reported to be dysregulated in tumors, but its specific contributions to tumor formation and progression are not fully understood. Here, we demonstrate that HNRNPAB is overexpressed in highly metastatic cells and tumor tissues from patients with hepatocellular carcinoma (HCC) with recurrence. We found that HNRNPAB overexpression promoted epithelial-mesenchymal transition (EMT) in a manner associated with HCC metastasis in vitro and in vivo. RNA interference-mediated silencing of the EMT factor SNAIL attenuated HNRNPAB-enhanced cell invasion in vitro and lung metastasis in vivo. Mechanistically, HNRNPAB acted to transactivate SNAIL1 transcription, which in turn inhibited transcription of the pivotal SNAIL target gene E-cadherin. Overexpression of HNRNPAB in HCC samples correlated with higher SNAIL levels, shorter overall survival, and higher tumor recurrence. HNRNPAB overexpression, alone or in combination with SNAIL, was found to be a significant independent risk factor for recurrence and survival after curative resection. In conclusion, our findings define HNRNPAB as an activator of EMT and metastasis in HCC that predicts poor clinical outcomes.


The Journal of Pathology | 2014

Capn4 contributes to tumour growth and metastasis of hepatocellular carcinoma by activation of the FAK–Src signalling pathways

Zhi Dai; Shao-Lai Zhou; Zheng-Jun Zhou; Dou-Sheng Bai; Xiao-Yu Xu; Xiu-Tao Fu; Qing Chen; Yi-Ming Zhao; Kai Zhu; Lei Yu; Guo-Huan Yang; Zheng Wang; Wei-Zhong Wu; Jian Zhou; Jia Fan

Calpain small subunit 1 (Capn4) has been identified as a major gene that promotes metastasis of hepatocellular carcinoma (HCC). However, the mechanism by which Capn4 promotes progression of HCC is not understood. In this study, we found that Capn4 expression was increased in highly metastatic HCC cell lines and in tumour tissue from HCC patients compared to healthy patient tissue. Over‐expression of Capn4 in HCC cells enhanced tumour cell growth in vitro and increased invasiveness, tumourigenicity and lung metastasis in vivo. Protein microarray analyses showed that expression of multiple proteins was regulated by Capn4. Interestingly, Capn4 was found to physically associate with FAK and promoted hyperactivity of the FAK–Src signalling pathway via increased phosphorylation of specific tyrosine residues of FAK, Src and p130Cas. Knock‐down of Capn4 expression suppressed the malignant behaviour of HCC cells and inhibited the FAK–Src signalling pathway. Furthermore, Capn4‐mediated invasion and metastasis of HCC cells required up‐regulation of matrix metalloproteinase‐2 (MMP2) through activation of this signalling pathway. Our clinical data revealed that Capn4 expression correlated well with the levels of phospho‐FAK, and over‐expression of both Capn4 and phospho‐FAK correlates with the poorest survival outcomes in HCC. In conclusion, our data showed that Capn4 can contribute to HCC growth and metastasis via activation of the FAK–Src signalling pathway and MMP2. Copyright


Transplantation Proceedings | 2011

Tacrolimus Enhances the Invasion Potential of Hepatocellular Carcinoma Cells and Promotes Lymphatic Metastasis in a Rat Model of Hepatocellular Carcinoma: Involvement of Vascular Endothelial Growth Factor-C

Shao-Lai Zhou; C. Tan; Zhi Dai; Huaqi Zhu; M. Xu; Zheng-Jun Zhou; Wei Wang; Y. Zhao; X. Fu; J. Zhou; Jin-Hu Fan

PURPOSE The purpose of this study was to elucidate the effect of tacrolimus treatment on tumor growth and metastasis of hepatocellular carcinoma (HCC). METHODS The effect of tacrolimus was investigated on tumor growth and lymph node metastasis in a rat model of HCC. Angiogenesis and lymphangiogenesis were assessed by CD31 and vascular endothelial growth factor receptor 3 (VEGFR-3) immunostaining. Cell proliferation and invasion were monitored in vitro using the Cell Counting Kit-8 (CCK8) and Matrigel Invasion Chambers, respectively. Levels of vascular endothelial growth factor-C (VEGF-C) expression were measured using quantitative real-time polymerase chain reaction (qRT-PCR), immunohistochemisty and Western blots. RESULTS Tacrolimus had no effect on the proliferation of HCC in vitro or in vivo. Treatment with tacrolimus resulted in a dose-dependent increase in the invasive potential of HCC cells in vitro, in the density of peritumoral lymphatic vessels, and in the number and volume of metastatic lymph nodes in August Copenhagen Irish (ACI) rats. qRT-PCR, immunohistochemisty, and Western blot revealed that tacrolimus increased the levels of expression of VEGF-C in HCC. CONCLUSIONS Tacrolimus enhanced the invasive potential of HCC cells, facilitating HCC lymphangiogenesis and promoting lymphatic metastasis in a rat model of HCC. This influence may be the result of an increase in VEGF-C expression by HCC cells. Targeting the VEGF-C/Fms-related tyrosine kinase 4 axis may be a novel treatment for HCC patients after liver transplantation.


Oncotarget | 2016

Overexpression of semaphorin 3A promotes tumor progression and predicts poor prognosis in hepatocellular carcinoma after curative resection

Zhi-Qiang Hu; Shao-Lai Zhou; Zheng-Jun Zhou; Chubin Luo; Er-Bao Chen; Hao Zhan; Peng-Cheng Wang; Zhi Dai; Jian Zhou; Jia Fan; Xiaowu Huang

The semaphorins were originally identified as having roles as guidance cues during neural development. Class 3 semaphorins are involved in cancer progression. However, the roles of class 3 semaphorins in hepatocellular carcinoma (HCC) are unknown. We examined the expression levels of class 3 semaphorins in HCC cell lines with different metastatic potential and in carcinoma tissue samples. The results indicated that Semaphorin 3A expression was up-regulated in metastatic cell lines and in samples from patients with tumor recurrence. Cell functional studies revealed that Semaphorin 3A promoted HCC cell proliferation, migration, and invasion. Animal studies indicated that Semaphorin 3A overexpression enhanced tumor growth and lung metastasis. Semaphorin 3A also acted as a chemoattractant involved in direct recruitment of macrophages in vitro, and facilitated tumor-associated macrophage (TAM) infiltration in vivo. Multivariate analysis revealed that Semaphorin 3A expression alone, or combined with the number of TAMs, can be an independent predictor for overall survival time and time to recurrence. Overall, the results suggested that Semaphorin 3A increased TAM infiltration and promoted HCC progression. Semaphorin 3A expression alone, or combined with the number of TAMs, is a new prognostic factor and potential target for the treatment of HCC.

Collaboration


Dive into the Zheng-Jun Zhou'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