Network


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

Hotspot


Dive into the research topics where Qunchuan Zhuang is active.

Publication


Featured researches published by Qunchuan Zhuang.


International Journal of Oncology | 2012

Pien Tze Huang inhibits tumor cell proliferation and promotes apoptosis via suppressing the STAT3 pathway in a colorectal cancer mouse model.

Qunchuan Zhuang; Fei Hong; Aling Shen; Liangpu Zheng; Jianwei Zeng; Wei Lin; Youqin Chen; Thomas J. Sferra; Zhenfeng Hong; Jun Peng

Signal transducer and activator of transcription 3 (STAT3) plays a critical role in cell survival and proliferation. Constitutive activation of STAT3 is strongly correlated with pathogenesis of various types of malignant tumors including colorectal cancer (CRC), and therefore is a major focus in the development of anti-cancer agents. Pien Tze Huang (PZH), a well-known traditional Chinese formula prescribed already in the Ming Dynasty, has been demonstrated to be clinically effective in the treatment of CRC. However, the precise mechanism of its anti-cancer activity remains largely unknown. In the present study we evaluated the efficacy of PZH against tumor growth in vivo in the CRC mouse xenograft model, and investigated the underlying molecular mechanisms. We found that administration of PZH reduced tumor volume and tumor weight but had no effect on body weight gain in CRC mice, demonstrating that PZH can inhibit colon cancer growth in vivo without apparent adverse effect. We also observed that PZH treatment inhibited the phosphorylation level of STAT3 in tumor tissues. Consequently, the inhibitory effect of PZH on STAT3 activation resulted in the up-regulation of Bax/Bcl-2 ratio as well as down-regulation of Cyclin D1 and CDK4 expression, leading to the induction of apoptosis as well as the inhibition of cell proliferation. These results suggest that promotion of cancer cell apoptosis and inhibition of proliferation via suppression of STAT3 pathway might be one of the mechanisms by which PZH treats colorectal cancer.


Chinese Journal of Integrative Medicine | 2012

Effects of Pien Tze Huang (片仔癀) on angiogenesis in vivo and in vitro

A.-ling Shen; Fei Hong; Liya Liu; Jiumao Lin; Qunchuan Zhuang; Zhenfeng Hong; Jun Peng

ObjectiveTo investigate the anti-angiogenic effects of Pien Tze Huang (片仔癀, PZH) in vivo and in vitro.MethodsHuman umbilical vein endothelial cells (HUVECs) were treated with 0 mg/mL, 0.25 mg/mL, 0.5 mg/mL, and 1 mg/mL of PZH for 24 h, 48 h and 72 h, respectively. Chicken embryo chorioallantoic membrane (CAM) model was used to evaluate in vivo angiogenesis. An ECMatrix gel system was used to evaluate in vitro angiogenesis by examining the tube formation of HUVECs. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was performed to determine HUVEC viability. Cell density of HUVECs was observed by phasecontrast microscopy. HUVEC migration was determined by wound healing method. The mRNA and protein expression of vascular endothelial growth factor A (VEGF-A) and basic fibroblast growth factor (bFGF) in both HUVEC and human colon adenocarcinoma cells (HT-29) was examined by reverse transcription polymerase chain reaction (RT-PCR) and enzyme linked immune sorbent assay (ELISA), respectively.ResultsPZH treatment significantly reduced the total number of blood vessels compared with the untreated control in the chicken embryos and resulted in a significant decrease in capillary tube formation and cell density of HUVECs (P<0.05). In addition, treatment with 0.25–1 mg/mL of PZH for 24 h, 48 h, and 72 h respectively reduced cell viability by 9%–52%, 24%–87% or 25%–87%, compared with the untreated control cells (P<0.05). Moreover, PZH treatment decreased the migration of HUVECs. Furthermore, PZH dose-dependently suppressed the expression of VEGF-A and bFGF on both mRNA and protein levels (P<0.05).ConclusionPZH could inhibit angiogenesis in vivo in CAM model and in vitro on HUVECs, suggesting that inhibiting tumor angiogenesis might be one of the mechanisms by which PZH treats cancer.


Molecular Medicine Reports | 2011

Patrinia scabiosaefolia extract suppresses proliferation and promotes apoptosis by inhibiting the STAT3 pathway in human multiple myeloma cells.

Jun Peng; Youqin Chen; Jiumao Lin; Qunchuan Zhuang; Wei Xu; Zhenfeng Hong; Thomas J. Sferra

Signal transducer and activator of transcription 3 (STAT3) plays an important role in tumor cell survival and proliferation and thus has become a major focus in the development of anti-cancer therapies. Patrinia scabiosaefolia has been used for the treatment of various types of cancer. However, the precise mechanism of the anti-cancer activity of Patrinia scabiosaefolia remains unclear. In this study, we evaluated the effect of the ethanol extract of Patrinia scabiosaefolia (EEPS) on proliferation and apoptosis in human multiple myeloma U266 cells that persistently express phosphorylated STAT3, and investigated the possible molecular mechanisms mediating its biological effects. We found that EEPS inhibited the phosphorylation of STAT3 in U266 cells. Consequently, the inhibitory effect of EEPS on STAT3 activation resulted in the suppression of cell proliferation and the induction of cell apoptosis. Moreover, EEPS treatment inhibited the expression of cyclin D1 (a promoter of cell proliferation) and Bcl-2 (an inhibitor of apoptosis), two important target genes of the STAT3 signaling pathway. Our findings for the first time demonstrate that Patrinia scabiosaefolia inhibits proliferation and promotes the apoptosis of cancer cells via inhibition of the STAT3 pathway, which may in part explain its anti-cancer activity.


International Journal of Oncology | 2013

Total alkaloids of Rubus aleaefolius Poir inhibit hepatocellular carcinoma growth in vivo and in vitro via activation of mitochondrial-dependent apoptosis

Jinyan Zhao; Xuzheng Chen; Wei Lin; Guangwen Wu; Qunchuan Zhuang; Xiaoyong Zhong; Zhenfeng Hong; Jun Peng

The aim of this study was to evaluate the therapeutic efficacy of Rubus aleaefolius Poir total alkaloids (TARAP) against hepatocellular carcinoma growth in vivo and in vitro, and to investigate the possible molecular mechanisms mediating its biological activity. Nude mice were implanted with HepG2 human hepatocellular carcinoma cells and fed with vehicle (physiological saline) or 3 g/kg/d dose of TARAP, 5 days per week, for 21 days. The in vivo efficacy of TARAP against tumor growth was investigated by evaluating its effect on tumor volume and tumor weight in mice with HCC xenografts and its adverse effect was determined by measuring the body weight gain. The in vitro effect of TARAP on the viability of HepG2 cells was determined by MTT assay. HepG2 cell morphology was observed via phase-contrast microscopy. Apoptosis in tumor tissues or in HepG2 cells was analyzed by TUNEL assay or FACS analysis with Annexin V/PI, respectively. The loss of mitochondrial membrane potential in HepG2 cells was determined via JC-1 staining followed by FACS analysis. Activation of caspase-9 and -3 in HepG2 cells was examined by a colorimetric assay. The mRNA and protein expression of Bcl-2 and Bax in tumor tissues were measured by RT-PCR and immunohistochemistry. TARAP reduced tumor volume and tumor weight, but had no effect on the body weight gain in HCC mice. TARAP decreased the viability of HepG2 cells and induced cell morphological changes in vitro in a dose- and time-dependent manner. In addition, TARAP induced apoptosis both in tumor tissues and in HepG2 cells. Moreover, TARAP treatment resulted in the collapse of mitochondrial membrane potential in HepG2 cells, as well as the activation of caspase-9 and -3. Furthermore, administration of TARAP increased the pro-apoptotic Bax/Bcl-2 ratio in HCC mouse tumors, at both transcriptional and translational levels. TARAP inhibits hepatocellular carcinoma growth both in vivo and in vitro probably through the activation of mitochondrial-dependent apoptosis, which may, in part, explain its anticancer activity. These results suggest that total alkaloids in Rubus aleaefolius Poir may be a potential novel therapeutic agent for the treatment of hepatocellular carcinoma and other cancers.


International Journal of Oncology | 2015

Pien Tze Huang inhibits metastasis of human colorectal carcinoma cells via modulation of TGF-β1/ZEB/miR‑200 signaling network

Aling Shen; Wei Lin; Youqin Chen; Liya Liu; Hongwei Chen; Qunchuan Zhuang; Jiumao Lin; Thomas J. Sferra; Jun Peng

Tumor metastasis, a complex process involving the spread of malignant tumor cells from a primary tumor site to a distant organ, is a major cause of failure of cancer chemotherapy. Epithelial-mesenchymal transition (EMT) is a critical step for the initiation of cancer metastasis. The processes of EMT and metastasis are highly regulated by a double-negative feedback loop consisting of TGF-β1/ZEB pathway and miR-200 family, which therefore has become a promising target for cancer chemotherapy. Pien Tze Huang (PZH), a well-known traditional Chinese formula first prescribed in the Ming Dynasty, has been demonstrated to be clinically effective in the treatment of various types of human malignancy including colorectal cancer (CRC). Our published data proposed that PZH was able to induce apoptosis, inhibit cell proliferation and tumor angiogenesis, leading to the suppression of CRC growth in vitro and in vivo. To further elucidate the mode of action of PZH, in the present study we evaluated its effects on the metastatic capacities of human colorectal carcinoma HCT-8 cells and investigated the underlying molecular mechanisms. We found that PZH significantly inhibited the migration and invasion of HCT-8 cells in a dose-dependent manner. In addition, PZH treatment inhibited the expression of key mediators of TGF-β1 signaling, such as TGF-β1, Smad2/3 and Smad4. Moreover, PZH treatment suppressed the expression of ZEB1 and ZEB2, two critical target genes of TGF-β1 pathway, leading to a decrease in the expression of mesenchymal marker N-cadherin and an increased expression of epithelial marker E-cadherin. Furthermore, PZH treatment upregulated the expression of miR-200a, miR-200b and miR-200c. Collectively, our findings in this study suggest that PZH can inhibit metastasis of colorectal cancer cells via modulating TGF-β1/ZEB/miR-200 signaling network, which might be one of the mechanisms whereby PZH exerts its anticancer function.


Oncology Reports | 2014

Livistona chinensis seeds inhibit hepatocellular carcinoma angiogenesis in vivo via suppression of the Notch pathway

Wei Lin; Jinyan Zhao; Zhiyun Cao; Qunchuan Zhuang; Liangpu Zheng; Jianwei Zeng; Zhenfeng Hong; Jun Peng

Livistona chinensis seeds have been used for centuries to clinically treat various types of cancer. Our published data suggest that Livistona chinensis seeds are able to inhibit hepatocellular carcinoma (HCC) growth in vitro and in vivo via promotion of mitochondrial-dependent apoptosis. To further elucidate the molecular mechanisms of its antitumor activity, in the present study, we used an HCC xenograft mouse model to evaluate the effect of an ethanol extract of Livistona chinensis seeds (EELC) on tumor angiogenesis and on the activation of the Notch pathway. Intratumoral microvessel density (MVD) in HCC xenograft mouse tumors was evaluated via immunohistochemical (IHC) staining for CD31. The mRNA and protein expression of vascular endothelial growth factor A (VEGF-A), VEGFR-2, Notch, Dll4 and Jagged1 was evaluated using RT-PCR and IHC, respectively. We found that EELC profoundly reduced MVD in the HCC mouse tumors, demonstrating the in vivo inhibitory effect of EELC on tumor angiogenesis. In addition, EELC treatment reduced the expression of VEGF-A and VEGFR-2 in tumor tissues. Furthermore, EELC treatment inhibited the expression of Notch, Dll4 and Jagged1. Our findings suggest that Livistona chinensis seeds inhibit tumor angiogenesis through suppression of the Notch pathway.


Oncology Letters | 2013

Spica Prunellae extract inhibits the proliferation of human colon carcinoma cells via the regulation of the cell cycle

Wei Lin; Liangpu Zheng; Qunchuan Zhuang; Aling Shen; Liya Liu; Youqin Chen; Thomas J. Sferra; Jun Peng

Spica Prunellae has long been used as a significant component in numerous traditional Chinese medicine (TCM) formulas to clinically treat cancers. Previously, Spica Prunellae was shown to promote cancer cell apoptosis and inhibit angiogenesis in vivo and in vitro. To further elucidate the precise mechanism of its tumoricidal activity, the effect of the ethanol extract of Spica Prunellae (EESP) on the proliferation of human colon carcinoma HT-29 cells was elucidated and the underlying molecular mechanisms were investigated. The proliferation of HT-29 cells was evaluated using 3-(4, 5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and colony formation analyses. The cell cycle was determined using fluorescence-activated cell sorting (FACS) with propidium iodide (PI) staining. The mRNA and protein expression of cyclin-dependent kinase 4 (CDK4) and cyclin D1 was examined using RT-PCR and western blotting, respectively. EESP was observed to inhibit HT-29 viability and survival in a dose- and time-dependent manner. Furthermore, EESP treatment blocked G1/S cell cycle progression and reduced the expression of pro-proliferative cyclin D1 and CDK4 at the transcriptional and translational levels. Altogether, these data suggest that the inhibition of cell proliferation via G1/S cell cycle arrest may be one of the mechanisms through which Spica Prunellae treats cancer.


African Journal of Pharmacy and Pharmacology | 2011

Anti-angiogenic effect of Spica prunellae extract in vivo and in vitro

Wei Lin; Liangpu Zheng; Jinyan Zhao; Qunchuan Zhuang; Zhenfeng Hong; Wei Xu; Youqin Chen; Thomas J. Sferra; Jun Peng; Minhou Shangjie

1 Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350108, China. 2 Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350108, China. 3 Department of Pharmacology, Fujian University of Traditional Chinese Medicine, 1 Huatuo Road, Minhou Shangjie, Fuzhou, Fujian 350108, China. 4 Department of Pediatrics, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Boulevard, Oklahoma City, OK 73190, USA. 5 Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L. Young Boulevard, Oklahoma City, OK 73190, USA.


Oncology Reports | 2013

Total alkaloids of Rubus aleaefolius Poir. inhibit the STAT3 signaling pathway leading to suppression of proliferation and cell cycle arrest in a mouse model of hepatocellular carcinoma

Jinyan Zhao; Wei Lin; Zhiyun Cao; Liya Liu; Qunchuan Zhuang; Xiaoyong Zhong; Zhenfeng Hong; Jun Peng

Signal transducer and activator of transcription 3 (STAT3) plays a critical role in cell survival and proliferation and is constitutively activated in many types of human cancers including hepatocellular carcinoma (HCC). Therefore, it is a major focus in the development of anticancer agents. Rubus aleaefolius Poir. has been demonstrated to be effective in the treatment of HCC. However, the precise mechanism of its anticancer activity remains largely unknown. Using HepG2 cells and a HCC mouse xenograft model, in the present study we evaluated the effect of the total alkaloids of Rubus aleaefolius Poir. (TARAP) on tumor growth in vitro and in vivo and investigated the underlying molecular mechanisms. We found that TARAP inhibited the proliferation of HepG2 human HCC cells and blocked G1/S cell cycle progression. In addition, TARAP treatment suppressed STAT3 phosphorylation in tumor tissues. Consequently, the inhibitory effect of TARAP on STAT3 activation resulted in the inhibition of proliferation. Moreover, TARAP altered the expression of several important target genes of the STAT3 signaling pathway, such as decreased expression of cyclinD1, cyclinE, cyclin-dependent kinase (CDK) 4 and CDK2 as well as upregulated p21. These results suggest that suppression of the STAT3 signaling pathway leading to inhibition of proliferation and cell cycle arrest may be one of the mechanisms of the anticancer activity of TARAP against HCC.


Molecular Medicine Reports | 2015

Total alkaloids of Rubus alceifolius Poir inhibit tumor angiogenesis through suppression of the Notch signaling pathway in a mouse model of hepatocellular carcinoma.

Jinyan Zhao; Wei Lin; Zhiyun Cao; Qunchuan Zhuang; Liangpu Zheng; Jun Peng; Zhenfeng Hong

Angiogenesis, which has a critical role in human tumor growth and development, is tightly regulated by the Notch signaling pathway. Total alkaloids are active components of the plant Rubus alceifolius Poir, which is used for the treatment of various types of cancer. A previous study by our group showed that the total alkaloids of Rubus alceifolius Poir (TARAP) induced hepatocellular carcinoma (HCC) cell apoptosis through the activation of the mitochondria-dependent pathway in vitro and in vivo, as well as inhibited angiogenesis in a chick embryo chorioallantoic membrane model. In the present study, to further analyze the specific mechanisms underlying the antitumor activity of TARAP, a HCC xenograft mouse model was used to assess the effect of TARAP on angiogenesis in vivo. TARAP was found to suppress the expression of vascular endothelial growth factor (VEGF) A and VEGF receptor-2 in tumor tissues, which resulted in the inhibition of tumor angiogenesis. In addition, TARAP treatment was observed to inhibit the expression of Notch1, delta-like ligand 4 and jagged 1, which are key mediators of the Notch signaling pathway. The present study identified that the inhibition of tumor angiogenesis through the suppression of the Notch signaling pathway may be one of the mechanisms through which TARAP may be effective in the treatment of cancer.

Collaboration


Dive into the Qunchuan Zhuang's collaboration.

Top Co-Authors

Avatar

Jun Peng

Fujian University of Traditional Chinese Medicine

View shared research outputs
Top Co-Authors

Avatar

Wei Lin

Fujian University of Traditional Chinese Medicine

View shared research outputs
Top Co-Authors

Avatar

Zhenfeng Hong

Fujian University of Traditional Chinese Medicine

View shared research outputs
Top Co-Authors

Avatar

Jinyan Zhao

Fujian University of Traditional Chinese Medicine

View shared research outputs
Top Co-Authors

Avatar

Liangpu Zheng

Fujian University of Traditional Chinese Medicine

View shared research outputs
Top Co-Authors

Avatar

Zhiyun Cao

Fujian University of Traditional Chinese Medicine

View shared research outputs
Top Co-Authors

Avatar

Youqin Chen

University of Oklahoma

View shared research outputs
Top Co-Authors

Avatar

Jiumao Lin

Fujian University of Traditional Chinese Medicine

View shared research outputs
Top Co-Authors

Avatar

Thomas J. Sferra

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

Liya Liu

Fujian University of Traditional Chinese Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge