Network


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

Hotspot


Dive into the research topics where Shanliang Zhong is active.

Publication


Featured researches published by Shanliang Zhong.


PLOS ONE | 2014

Exosomes from Drug-Resistant Breast Cancer Cells Transmit Chemoresistance by a Horizontal Transfer of MicroRNAs

Wei-xian Chen; Xuemin Liu; Mengmeng Lv; Lin Chen; Jianhua Zhao; Shanliang Zhong; Minghua Ji; Qing Hu; Zhou Luo; Jianzhong Wu; Jinhai Tang

Adriamycin and docetaxel are two agents commonly used in treatment of breast cancer, but their efficacy is often limited by the emergence of chemoresistance. Recent studies indicate that exosomes act as vehicles for exchange of genetic cargo between heterogeneous populations of tumor cells, engendering a transmitted drug resistance for cancer development and progression. However, the specific contribution of breast cancer-derived exosomes is poorly understood. Here we reinforced others report that human breast cancer cell line MCF-7/S could acquire increased survival potential from its resistant variants MCF-7/Adr and MCF-7/Doc. Additionally, exosomes of the latter, A/exo and D/exo, significantly modulated the cell cycle distribution and drug-induced apoptosis with respect to S/exo. Exosomes pre-treated with RNase were unable to regulate cell cycle and apoptosis resistance, suggesting an RNA-dependent manner. Microarray and polymerase chain reaction for the miRNA expression profiles of A/exo, D/exo, and S/exo demonstrated that they loaded selective miRNA patterns. Following A/exo and D/exo transfer to recipient MCF-7/S, the same miRNAs were significantly increased in acquired cells. Target gene prediction and pathway analysis showed the involvement of miR-100, miR-222, and miR-30a in pathways implicated in cancer pathogenesis, membrane vesiculation and therapy failure. Furthermore, D/exo co-culture assays and miRNA mimics transfection experiments indicated that miR-222-rich D/exo could alter target gene expression in MCF-7/S. Our results suggest that drug-resistant breast cancer cells may spread resistance capacity to sensitive ones by releasing exosomes and that such effects could be partly attributed to the intercellular transfer of specific miRNAs.


Gene | 2013

MiR-222 and miR-29a contribute to the drug-resistance of breast cancer cells.

Shanliang Zhong; Wenjing Li; Zhiyuan Chen; Jinjin Xu; Jianhua Zhao

Adriamycin (Adr) and docetaxel (Doc) are two chemotherapeutic agents commonly used in the treatment of breast cancer. However, patients with breast cancer who are treated by the drugs often develop resistance to them and some other drugs. Recently studies have shown that microRNAs (miRNAs, miRs) play an important role in drug-resistance. In present study, miRNA expression profiles of MCF-7/S and its two resistant variant MCF-7/Adr and MCF-7/Doc cells were analyzed using microarray and the results were confirmed by real-time quantitative polymerase chain reaction. Here, 183 differentially expressed miRNAs were identified in the two resistant sublines compared to MCF-7/S. Then, five up-regulated miRNAs (miR-100, miR-29a, miR-196a, miR-222 and miR-30a) in both MCF-7/Adr and MCF-7/Doc were selected to explore their roles in acquisition of drug-resistance using transfection experiment. The results showed that miR-222 and miR-29a mimics and inhibitors had partially changed the drug-resistance of breast cancer cells, which was also confirmed by apoptosis assay. Western blot results suggested that miR-222 and -29a could regulate the expression of PTEN, maybe through which the two miRNAs conferred Adr and Doc resistance in MCF-7 cells. Finally, pathway mapping tools were employed to further analyze signaling pathways affected by the two miRNAs. In summary, this study demonstrates that altered miRNA expression pattern is involved in acquiring resistance to Adr and Doc in breast cancer MCF-7 cells, and that there are some miRNAs who displayed consistent up- or down-regulated expression changes in the two resistant sublines. The most importance is that we identify two miRNAs (miR-222 and miR-29a) involved in drug-resistance, at least in part via targeting PTEN.


Cancer Treatment Reviews | 2015

Statin use and mortality in cancer patients: Systematic review and meta-analysis of observational studies.

Shanliang Zhong; Xiaohui Zhang; Lin Chen; Tengfei Ma; Jinhai Tang; Jianhua Zhao

BACKGROUND Previous studies have examined the effect of statin use on the mortality in cancer patients, but the results are inconsistent. A meta-analysis was performed to assess the association with all available studies. METHODS Relevant studies were identified by searching PubMed and EMBASE to April 2015. We calculated the summary hazard ratios (HRs) and 95% confidence intervals (CIs) using random-effects models. We estimated combined HRs associated with defined increments of statin use, using random-effects meta-analysis and dose-response meta-regression models. RESULTS Thirty-nine cohort studies and two case-control studies involving 990,649 participants were included. The results showed that patients who used statins after diagnosis had a HR of 0.81 (95% CI: 0.72-0.91) for all-cause mortality compared to non-users. Those who used statin after diagnosis (vs. non-users) had a HR of 0.77 (95% CI: 0.66-0.88) for cancer-specific mortality. Prediagnostic exposure to statin was associated with both all-cause mortality (HR=0.79, 95% CI: 0.74-0.85) and cancer-specific mortality (HR=0.69, 95% CI: 0.60-0.79). Stratifying by cancer type, the three largest cancer-type subgroups were colorectal, prostate and breast cancer and all showed a benefit from statin use. HRs per 365 defined daily doses increment were 0.80 (95% CI: 0.69-0.92) for all-cause mortality and 0.77 (95% CI: 0.67-0.89) for cancer-specific mortality. A 1year increment in duration only conferred a borderline decreased risk of death. CONCLUSIONS In conclusion, the average effect of statin use, both postdiagnosis and prediagnosis, is beneficial for overall survival and cancer-specific survival.


Tumor Biology | 2014

Exosomes mediate drug resistance transfer in MCF-7 breast cancer cells and a probable mechanism is delivery of P-glycoprotein

Mengmeng Lv; Xingya Zhu; Wei-xian Chen; Shanliang Zhong; Qing Hu; Tengfei Ma; Jun Zhang; Lin Chen; Jinhai Tang; Jianhua Zhao

Acquired drug resistance is a major obstacle to chemotherapy of cancers. In this study, we aim to investigate the role of exosomes in drug-resistance transfer between breast cancer cells and detect the probable mechanism. A docetaxel-resistant variant of MCF-7 cell line (MCF-7/DOC) was established and then compared with the drug-sensitive variant (MCF-7/S). Exosomes were expelled from the cell supernatant using ultracentrifugation. Drug resistance was assessed by apoptosis assay and MTT examination. Expressions of P-glycoprotein (P-gp) were analyzed by flow cytometry. Stained exosomes were absorbed by receipt cells. MCF-7/S in the presence of exosomes extracted from the supernatant of MCF-7/DOC (DOC/exo) acquired drug resistance, while MCF-7/S exposed to their own exosomes (S/exo) did not. P-gp expression patterns of exosomes were similar as the originated cells. P-gp expression of MCF-7/S increased after incubation with DOC/exo and was affected by the amount of exosomes. Exosomes are effective in transferring drug resistance as well as P-gp from drug-resistant breast cancer cells to sensitive ones. The delivery of P-gp via exosomes may be a mechanism of exosome-mediated drug resistance transfer.


Tumor Biology | 2014

Exosomes from docetaxel-resistant breast cancer cells alter chemosensitivity by delivering microRNAs

Wei-xian Chen; Yan-qin Cai; Mengmeng Lv; Lin Chen; Shanliang Zhong; Tengfei Ma; Jianhua Zhao; Jinhai Tang

Breast cancer (BCa) remains chemo-unresponsive by inevitable progression of resistance to first-line treatment with docetaxel (doc). Emerging studies indicate that exosomes act as mediators of intercellular communication between heterogeneous populations of tumor cells, engendering a transmitted drug resistance for cancer development. Such modulatory effects have been related to the constant shuttle of biologically active molecules including microRNAs (miRNAs). Here, we aimed to investigate the relevance of exosome-mediated miRNA delivery in resistance transmission of BCa subpopulations. Using microarray and polymerase chain reaction, we found that exosomes from doc-resistant BCa cells (D/exo) loaded cellular miRNAs. Following D/exo transfer to the fluorescent sensitive cells (GFP-S), some miRNAs were significantly increased in recipient GFP-S. Target gene prediction and pathway analysis revealed the involvement of the top 20 most abundant miRNAs of D/exo in pathways implicated in therapy failure. Coculture assays showed that miRNA-containing D/exo increased the overall resistance of GFP-S to doc exposure. Moreover, D/exo was able to alter gene expression in GFP-S. Our results open up an intriguing possibility that drug-resistant BCa cells may spread chemoresistance to sensitive ones by releasing exosomes and that the effects could be partly attributed to the intercellular transfer of specific miRNAs.


Cancer Science | 2015

Exosomes in development, metastasis and drug resistance of breast cancer

Dan-dan Yu; Ying Wu; Hongyu Shen; Mengmeng Lv; Wei-xian Chen; Xiaohui Zhang; Shanliang Zhong; Jinhai Tang; Jianhua Zhao

Transport through the cell membrane can be divided into active, passive and vesicular types (exosomes). Exosomes are nano‐sized vesicles released by a variety of cells. Emerging evidence shows that exosomes play a critical role in cancers. Exosomes mediate communication between stroma and cancer cells through the transfer of nucleic acid and proteins. It is demonstrated that the contents and the quantity of exosomes will change after occurrence of cancers. Over the last decade, growing attention has been paid to the role of exosomes in the development of breast cancer, the most life‐threatening cancer in women. Breast cancer could induce salivary glands to secret specific exosomes, which could be used as biomarkers in the diagnosis of early breast cancer. Exosome‐delivered nucleic acid and proteins partly facilitate the tumorigenesis, metastasis and resistance of breast cancer. Exosomes could also transmit anti‐cancer drugs outside breast cancer cells, therefore leading to drug resistance. However, exosomes are effective tools for transportation of anti‐cancer drugs with lower immunogenicity and toxicity. This is a promising way to establish a drug delivery system.


Tumor Biology | 2014

MicroRNAs delivered by extracellular vesicles: an emerging resistance mechanism for breast cancer

Wei-xian Chen; Shanliang Zhong; Minghua Ji; Meng Pan; Qing Hu; Mengmeng Lv; Zhou Luo; Jianhua Zhao; Jinhai Tang

Resistance to chemotherapy and endocrine therapy as well as targeted drugs is a major problem in treatment of breast cancer. Over the last decades, emerging studies have revealed that extracellular vesicles, which are chronically released by breast cancer cells and surrounding stromal cells, influence the action of most commonly used therapeutics. Such modulatory effects have been related to the transport of biologically active molecules including proteins and functional microRNAs. In this review, we highlight recent studies regarding extracellular vesicle-mediated microRNA delivery in formatting drug resistance. We also suggest the use of extracellular vesicles as a promising method in antiresistance treatment.


Asian Pacific Journal of Cancer Prevention | 2014

Down-regulation of miRNA-452 is associated with adriamycin-resistance in breast cancer cells.

Qing Hu; Jian-Ping Gong; Jian Li; Shanliang Zhong; Wei-xian Chen; Jun-Ying Zhang; Tengfei Ma; Hao Ji; Mengmeng Lv; Jianhua Zhao; Jinhai Tang

Adriamycin (ADR) is an important chemotherapeutic agent frequently used in treatment of breast cancer. However, resistance to ADR results in treatment failure in many patients. Recent studies have indicated that microRNAs (miRNAs) may play an important role in such drug-resistance. In the present study, microRNA-452 (miR-452) was found to be significantly down-regulated in adriamycin-resistant MCF-7 cells (MCF-7/ADR) compared with the parental MCF-7 cells by miRNA microarray and real-time quantitative PCR (RT-qPCR). MiR-452 mimics and inhibitors partially changed the adriamycin-resistance of breast cancer cells, as also confirmed by apoptosis assay. In exploring the potential mechanisms of miR-452 in the adriamycin-resistance of breast cancer cells, bioinformatics analysis, RT-qPCR and Western blotting showed that dysregulation of miR-452 played an important role in the acquired adriamycin-resistance of breast cancer, maybe at least in part via targeting insulin-like growth factor-1 receptor (IGF-1R).


Tumor Biology | 2016

Exosomes decrease sensitivity of breast cancer cells to adriamycin by delivering microRNAs.

Ling Mao; Jian Li; Wei-xian Chen; Yan-qin Cai; Dan-dan Yu; Shanliang Zhong; Jianhua Zhao; Jianwei Zhou; Jinhai Tang

While adriamycin (adr) offers improvement in survival for breast cancer (BCa) patients, unfortunately, drug resistance is almost inevitable. Mounting evidence suggests that exosomes act as a vehicle for genetic cargo and constantly shuttle biologically active molecules including microRNAs (miRNAs) between heterogeneous populations of tumor cells, engendering a resistance-promoting niche for cancer progression. Our recent study showed that exosomes from docetaxel-resistance BCa cells could modulate chemosensitivity by delivering miRNAs. Herein, we expand on our previous finding and explore the relevance of exosome-mediated miRNA delivery in resistance transmission of adr-resistant BCa sublines. We now demonstrated the selective packing of miRNAs within the exosomes (A/exo) derived from adr-resistant BCa cells. The highly expressed miRNAs in A/exo were significantly increased in recipient fluorescent sensitive cells (GFP-S) after A/exo incorporation. Gene ontology analysis of predicted targets showed that the top 30 most abundant miRNAs in A/exo were involved in crucial biological processes. Moreover, A/exo not only loaded miRNAs for its production and release but also carried miRNAs associated with Wnt signaling pathway. Furthermore, A/exo co-culture assays indicated that miRNA-containing A/exo was able to increase the overall resistance of GFP-S to adr exposure and regulate gene levels in GFP-S. Our results reinforce our earlier reports that adr-resistant BCa cells could manipulate a more deleterious microenvironment and transmit resistance capacity through altering gene expressions in sensitive cells by transferring specific miRNAs contained within exosomes.


Gene | 2017

MiR-222 promotes drug-resistance of breast cancer cells to adriamycin via modulation of PTEN/Akt/FOXO1 pathway.

Hongyu Shen; Dan-dan Wang; Liangpeng Li; Sujin Yang; Xiu Chen; Siying Zhou; Shanliang Zhong; Jianhua Zhao; Jinhai Tang

BACKGROUND AND PURPOSE Acquisition of resistance to adriamycin (ADR) is one of the most important clinical obstacles in the treatment of breast cancer, but the molecular mechanisms underlying sensitivity to ADR remain elusive. In our previous study, through miRNA microarray and experiments, we have emphasized that miR-222 could promote the ADR-resistance in breast cancer cells. The aim of this study was to explore the possible mechanism by which miR-222 affects sensitivity to ADR. METHODS Through pathway enrichment analyses for miR-222, we found that PTEN/Akt/FOXO1 signaling pathway may be of importance. RT-qPCR analyses and western blot assays confirmed the relationship between miR-222 expression and target genes. Immunofluorescence further visually displayed the location of FOXO1. When blocking PTEN/Akt/FOXO1 signaling pathway, we demonstrated the effects of miR-222-mediated ADR resistance by MTT and apoptosis assays. RESULTS RT-qPCR and Western blot results showed that miR-222 expression was negatively correlated with FOXO1 expression. In addition, the subcellular translocation of FOXO1 due to the altered expression of miR-222 was observed from immunofluorescence. Moreover, upregulation of miR-222 expression in MCF-7/S cells is associated with decreased PTEN expression levels and increased phospho-Akt (p-Akt) expression. Conversely in MCF-7/ADR cells, inhibition of miR-222 resulted in increased PTEN expression and decreased p-Akt expression. For further validation, results of the present study also demonstrated that PTEN/Akt/FOXO1 signaling was responsible for the ADR-resistance of breast cancer cells since LY294002, an inhibitor of Akt signaling, partially increased the sensitivity of MCF-7/S cells to ADR. More importantly, we postulated that high expression of miR-222 is closely related to poor overall survival by TCGA database validation. CONCLUSIONS Taken together, these data elucidated that miR-222 mediated ADR-resistance of breast cancer cells partly through regulation of PTEN/Akt/FOXO1 signaling pathway and inhibition of miR-222 may improve the prognosis of breast cancer patients.

Collaboration


Dive into the Shanliang Zhong's collaboration.

Top Co-Authors

Avatar

Jianhua Zhao

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Jinhai Tang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Sujin Yang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiu Chen

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiaohui Zhang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Dan-dan Wang

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Wei-xian Chen

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Hongyu Shen

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Mengmeng Lv

Nanjing Medical University

View shared research outputs
Top Co-Authors

Avatar

Tengfei Ma

Nanjing Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge