Network


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

Hotspot


Dive into the research topics where Jianda Zhou is active.

Publication


Featured researches published by Jianda Zhou.


Journal of Cancer | 2017

Role of tumor microenvironment in tumorigenesis

Maonan Wang; Jingzhou Zhao; Lishen Zhang; Fang Wei; Yu Lian; Yingfeng Wu; Zhaojian Gong; Shanshan Zhang; Jianda Zhou; Ke Cao; Li X; Wei Xiong; Guiyuan Li; Zhaoyang Zeng; Can Guo

Tumorigenesis is a complex and dynamic process, consisting of three stages: initiation, progression, and metastasis. Tumors are encircled by extracellular matrix (ECM) and stromal cells, and the physiological state of the tumor microenvironment (TME) is closely connected to every step of tumorigenesis. Evidence suggests that the vital components of the TME are fibroblasts and myofibroblasts, neuroendocrine cells, adipose cells, immune and inflammatory cells, the blood and lymphatic vascular networks, and ECM. This manuscript, based on the current studies of the TME, offers a more comprehensive overview of the primary functions of each component of the TME in cancer initiation, progression, and invasion. The manuscript also includes primary therapeutic targeting markers for each player, which may be helpful in treating tumors.


Experimental and Therapeutic Medicine | 2013

Saikosaponin A mediates the inflammatory response by inhibiting the MAPK and NF-κB pathways in LPS-stimulated RAW 264.7 cells

Jie Zhu; Chengqun Luo; Ping Wang; Quanyong He; Jianda Zhou; Hao Peng

Saikosaponin A (SSA) is a major triterpenoid saponin isolated from Radix bupleuri (RB), a widely used Chinese traditional medicine to treat various inflammation-related diseases. The aim of this study was to investigate the anti-inflammatory activity, as well as the molecular mechanism of SSA in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. In this study, we demonstrated that SSA markedly inhibits the expression of certain immune-related cytotoxic factors, including cyclooxygenase-2 (COX-2) and inducible nitric-oxide synthase (iNOS), as well as pro-inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6. It also significantly upregulates the expression of IL-10, an important anti-inflammatory cytokine, suggesting its anti-inflammatory activity in LPS-stimulated macrophages. We further demonstrated that SSA inhibits the activation of the nuclear factor-κB (NF-κB) signaling pathway by suppressing the phosphorylation of inhibitory NF-κB inhibitor α (IκBα) and thus holding p65 NF-κB in the cytoplasm to prevent its translocation to the nucleus. In addition, SSA also inhibits the mitogen-activated protein kinase (MAPK) signaling pathway by downregulating the phosphorylation of p38 MAPK, c-Jun N-terminal kinase (c-JNK) and extracellular signal-regulated kinase (ERK), the three key components of the MAPK family. In conclusion, our study demonstrates that SSA has an anti-inflammatory effect by regulating inflammatory mediators and suppressing the MAPK and NF-κB signaling pathways in LPS-stimulated RAW 264.7 cells.


Cancer Biology & Therapy | 2015

miR-33a functions as a tumor suppressor in melanoma by targeting HIF-1α

Jianda Zhou; Dan Xu; Huiqing Xie; Rui Liu; Jingjing Li; Shaohua Wang; Xiang Chen; Juan Su; Xiao Zhou; Kun Xia; Quanyong He; Jia Chen; Wei Xiong; Peiguo Cao; Ke Cao

Background: Our previous findings showed that miR-33 expressed abnormally in clinical specimens of melanoma, but the exact molecular mechanism has not been elucidated. Object: To determine miR-33s roles in melanoma and confirm whether HIF-1α is a direct target gene of miR-33a. Methods: First miR-33a/b expression levels were detected in HM, WM35, WM451, A375 and SK-MEL-1. Then lentiviral vectors were constructed to intervene miR-33a expression in melanoma cells. Cell proliferation, invasion and metastasis were detected. A375 cells mice model was performed to test the tumorigenesis of melanoma in vivo. Finally the dual reporter gene assay was carried out to confirm whether HIF-1α is a direct target gene of miR-33a. Results: MiR-33a/b exhibited a lower expression in WM35, WM451, A375 and SK-MEL-1 of the metastatic skin melanoma cell lines than that in HM. Then inhibition of miR-33a expression in WM35 and WM451 cell lines could promote cell proliferation, invasion and metastasis. Conversely, increased expression of miR-33a in A375 cells could inhibit cellproliferation, invasion and metastasis. In vivo tests also confirmed that overexpression of miR-33a in A375 cells significantly inhibited melanoma tumorigenesis. Finally, we confirmed that HIF-1α is a direct target gene of miR-33a. Conclusion: The newly identified miR-33a/HIF-1α axis might provide a new strategy for the treatment of melanoma.


Oncogene | 2010

The tumor suppressor ING3 is degraded by SCF(Skp2)-mediated ubiquitin-proteasome system.

Gong Chen; Yuji Wang; M Garate; Jianda Zhou; Guiyuan Li

The inhibitor of growth family member 3 (ING3) has been shown to modulate transcription, cell cycle control and apoptosis. We previously reported that nuclear ING3 expression was remarkably reduced in melanomas, which correlated with a poorer patient survival, suggesting that decreased ING3 expression may be associated with melanoma progression. However, the mechanism of diminished ING3 expression in melanoma is not clear. Here we show that ING3 level was decreased in metastatic melanoma cells because of a rapid degradation. Furthermore, we showed that ING3 undergoes degradation through the ubiquitin–proteasome pathway. ING3 physically interacts with subunits of E3 ligase Skp1-Cullin-F-box protein complex (SCF complex). Knockdown of F-box protein S-phase kinase-associated protein 2 (Skp2) reduces the ubiquitination of ING3 and significantly stabilizes ING3 in melanoma cells. In addition, lysine 96 residue is essential for ING3 ubiquitination as its mutation to arginine dramatically abrogated ING3 degradation. Disruption of ING3 degradation stimulated ING3-induced G1 cell-cycle arrest and enhanced ultraviolet-induced apoptosis. Taken together, our data show that ING3 is degraded by the ubiquitin–proteasome pathway through the SCFSkp2 complex and interruption of ING3 degradation enhances the tumor-suppressive function of ING3, which provides a potential cancer therapeutic approach by interfering ING3 degradation.


Journal of Dermatological Science | 2016

Analysis of lncRNAs expression in UVB-induced stress responses of melanocytes

Qinghai Zeng; Qi Wang; Xiang Chen; Kun Xia; Jintian Tang; Xiao Zhou; Yan Cheng; Yong Chen; Lihua Huang; Hong Xiang; Ke Cao; Jianda Zhou

BACKGROUND Long non-coding RNAs (lncRNAs) have close relationships with oxidative stress, nutritional deficiency, DNA damage and other types of cellular stress responses. Previous studies have demonstrated that some non-coding RNAs in melanocytes such as microRNAs can change and contribute to the synthesis of melanin or the development of melanoma after stimulation with UV. However, as an important component of non-coding RNAs, it is unclear what changes occur in lncRNAs during UV-induced stress responses in melanocytes. OBJECTIVE To explore changes in the expression of long non-coding RNAs (lncRNAs) in melanocytes following UVB-induced stress, and to explore if lncRNAs are involved in the synthesis of melanin. METHODS Primary melanocytes were irradiated by 20mJ/cm(2) UVB. The MTT method was used to detect cell proliferation. Quantitative real-time PCR was carried out to analyze expression of tyrosinase (TYR) and lncRNAs. Dopa colorimetry was performed to analyze TYR activity. The expression profile of lncRNAs and mRNAs were confirmed using an Agilent Human lncRNA 4×180K chip. Intracellular ROS levels were detected by flow cytometry. ROS scavenger (NAC) was employed to inhibit the ROS level. TYR mRNA expression and activity were re-analysed after transfecting of lnc-CD1D-2:1 siRNA and lnc-SGCG-5:4 siRNA in UVB-irradiated melanocytes to confirm the roles of the two lncRNAs in the synthesis of melanin. phospho-ERK, phospho-p38, and phospho-JNK expressions were detected by Western Blot. RESULTS Cell proliferation of the 20mJ/cm(2) UVB-irradiated melanocytes decreased to 91% of that of the control cells. Twenty-four hours after irradiation with 20mJ/cm(2) UVB, TYR mRNA expression and activity of the irradiated cells were significantly increased relative to the control group. Chip detection data showed that after irradiation with 20mJ/cm(2) UVB, the expression of 807 lncRNAs and 69 stress response-related genes had changed by more than two-fold. Expression levels of Lnc-GKN2-1:1, lnc-CD1D-2:1, and lnc-SGCG-5:4 and ROS content were significantly increased after UVB irradiation. NAC reduced UVB-induced ROS generation and inhibited UVB-induced upregulation of lnc-GKN2-1:1 and lnc-CD1D-2:1. Lnc-CD1D-2:1 siRNA significantly suppressed the UVB-induced TYR mRNA expression and tyrosinase activation. Lnc-CD1D-2:1 siRNA inhibited UVB-induced p38 phosphorylation. CONCLUSIONS LncRNAs in melanocytes undergo significant changes following irradiation with 20mJ/cm(2) UVB, suggestting that lncRNAs participate in the UVB-induced stress response. Some lncRNAs expression changes induced by UVB are dependent on ROS generation. ROS-mediated production of lnc-CD1D-2:1 may be involved in the melanogenesis induced by UVB.


Oncotarget | 2016

Sirt3-mediated mitophagy protects tumor cells against apoptosis under hypoxia

Aimin Qiao; Kuansong Wang; Yunsheng Yuan; Yi-di Guan; Xingcong Ren; Lanya Li; Xisha Chen; Feng Li; Alex F. Chen; Jianda Zhou; Jin-Ming Yang; Yan Cheng

Sirt3, a mitochondrial deacetylase, participates in the regulation of multiple cellular processes through its effect on protein acetylation. The objective of this study was to explore the role of Sirt3 in the mitochondrial autophagy (mitophagy), a process of the specific autophagic elimination of damaged mitochondria. We found that silencing of Sirt3 expression in human glioma cells by RNA interference blunted the hypoxia-induced the localization of LC3 on the mitochondria, and the degradation of mitochondria. These results suggest an important involvement of this protein deacetylase in the induction of mitophagy in cancer cells subjected to hypoxia. Further, we demonstrated that Sirt3 activated the hypoxia-induced mitophagy by increasing the interaction of VDAC1 with Parkin. In the cells subjected to hypoxia, inhibition of Sirt3-mediated mitophagy further decreased the mitochondrial membrane potential, and increased the accumulation of ROS that triggers the degradation of anti-apoptotic proteins Mcl-1 and survivin through the proteasomal pathway. Silencing of Sirt3 expression also promoted apoptosis, and enhanced the sensitivity of cancer cells to hypoxia. The regulatory role of Sirt3 in autophagy and apoptosis was also observed in human breast cancer cells. The results of the current study reveal Sirt3 as a novel regulator coupling mitophagy and apoptosis, two important cellular processes that determine cellular survival and death.


Geriatrics & Gerontology International | 2016

Depression in left-behind elderly in rural China: Prevalence and associated factors

Guoping He; Jianfei Xie; Jianda Zhou; Zhu-qing Zhong; Chun-xiang Qin; Siqing Ding

Several studies have reported the prevalence of depressive symptoms in the urban population of China, but no study reports the prevalence of depression in rural left‐behind elderly. The present study investigated the prevalence of depression and the associated factors that influence depression in the left‐behind elderly population in a rural area of China.


Oncology Reports | 2015

microRNA-195 functions as a tumor suppressor by inhibiting CBX4 in hepatocellular carcinoma

Changli Zheng; Jingjing Li; Qi Wang; Wei Liu; Jianda Zhou; Rui Liu; Qinghai Zeng; Xiaowei Peng; Chenghui Huang; Peiguo Cao; Ke Cao

MicroRNA-195 (miR-195) plays important roles in tumor metastasis and angiogenesis, yet its function and mechanism of action in hepatocellular carcinoma (HCC) remain to be elucidated. In this study, we aimed to confirm whether chromobox homolog 4 (CBX4) is a direct target gene of miR-195 and determine the functions of miR-195 through the CBX4 pathway. miR-195 expression was slightly lower in the HCC tissues compared with that in the adjacent normal tissues. In addition, western blotting and qRT-RCR results showed that both CBX4 mRNA and protein levels were downregulated upon miR-195 overexpression. Luciferase reporter assays revealed that CBX4 is a direct target gene of miR-195. Furthermore, overexpression of CBX4 significantly restored the proliferative, invasive and migratory capacities of the HepG2 cells. Finally, in vivo experiments confirmed that high expression of CBX4 in HepG2 cells promoted tumor growth. In conclusion, our study demonstrated that miR-195 acts as a tumor suppressor by directly targeting CBX4 in HCC. This finding suggests a potential novel strategy for therapeutic interventions of this disease.


Cancer Biology & Therapy | 2014

MiR-20a inhibits cutaneous squamous cell carcinoma metastasis and proliferation by directly targeting LIMK1

Jianda Zhou; Rui Liu; Chengqun Luo; Xiao Zhou; Kun Xia; Xiang Chen; Ming Zhou; Qiong Zou; Peiguo Cao; Ke Cao

Background MicroRNA-20a (miR-20a) plays a key role in tumorigenesis and progression. But its function is reverse in different kinds of malignant tumor, and its role and mechanism in cutaneous squamous cell carcinoma (CSCC) remains unclear. Object To determine the miR-20a’s roles in CSCC and confirm whether LIMK1 is a direct target gene of miR-20a. Methods First miR-20a and LIMK1 expression levels were detected in six pairs of CSCC tissues and corresponding normal skin by qRT-PCR. Then MTT assays and colony formation assays were performed to evaluate the impact of miR-20a on cell proliferation. In addition, scratch migration assays and transwell invasion assays were performed to check miR-20a’s effect on cell metastasis. Since LIMK1 (LIM kinase-1) was predicted as a target gene of miR-20a, the changes of LIMK1 protein and mRNA were measured by western blot and qRT-RCR methods after miR-20a overexpression. Moreover the dual reporter gene assay was performed to confirm whether LIMK1 is a direct target gene of miR-20a. Finally LIMK1 mRNA and miR-20a in other 30 cases of CSCC pathological specimens were determined and a correlation analysis was evaluated. Results The miR-20a significantly low-expressed in CSCC tissues compared with that in matched normal tissues while LIMK1 has a relative higher expression. MiR-20a inhibited A431 and SCL-1 proliferation and metastasis. Both of LIMK1 protein and mRNA levels were downregulated after miR-20a overexpression. The dual reporter gene assays revealed that LIMK1 is a direct target gene of miR-20a. Furthermore, qRT-PCR results of LIMK1 mRNA and miR-20a in 30 cases of CSCC pathological specimens showed miR-20a is inversely correlated with LIMK1 expression. Conclusion Our study demonstrated that miR-20a is involved in the tumor inhibition of CSCC by directly targeting LIMK1 gene. This finding provides potential novel strategies for therapeutic interventions of CSCC.


Oncogene | 2017

DEPTOR is a direct NOTCH1 target that promotes cell proliferation and survival in T-cell leukemia

Yunping Hu; Hexiu Su; C Liu; Zhenning Wang; L Huang; Qianfei Wang; Shujun Liu; Sai-Juan Chen; Jianda Zhou; P Li; Zhijian J. Chen; Hao Liu; Guoliang Qing

Aberrant activation of NOTCH1 signaling plays a vital role in the pathogenesis of T-cell acute lymphoblastic leukemia (T-ALL). Yet the molecular events downstream of NOTCH1 that drive T-cell leukemogenesis remain incompletely understood. Starting from genome-wide gene-expression profiling to seek important NOTCH1 transcriptional targets, we identified DEP-domain containing mTOR-interacting protein (DEPTOR), which was previously shown to be important in multiple myeloma but remains functionally unclear in other hematological malignancies. Mechanistically, we demonstrated NOTCH1 directly bound to and activated the human DEPTOR promoter in T-ALL cells. DEPTOR depletion abolished cellular proliferation, attenuated glycolytic metabolism and enhanced cell death, while ectopically expressed DEPTOR significantly promoted cell growth and glycolysis. We further showed that DEPTOR depletion inhibited while its overexpression enhanced AKT activation in T-ALL cells. Importantly, AKT inhibition completely abrogated DEPTOR-mediated cell growth advantages. Moreover, DEPTOR depletion in a human T-ALL xenograft model significantly delayed T-ALL onset and caused a substantial decrease of AKT activation in leukemic blasts. We thus reveal a novel mechanism involved in NOTCH1-driven leukemogenesis, identifying the transcriptional control of DEPTOR and its regulation of AKT as additional key elements of the leukemogenic program activated by NOTCH1.

Collaboration


Dive into the Jianda Zhou's collaboration.

Top Co-Authors

Avatar

Ke Cao

Central South University

View shared research outputs
Top Co-Authors

Avatar

Shaohua Wang

Central South University

View shared research outputs
Top Co-Authors

Avatar

Jia Chen

Central South University

View shared research outputs
Top Co-Authors

Avatar

Chengqun Luo

Central South University

View shared research outputs
Top Co-Authors

Avatar

Peiguo Cao

Central South University

View shared research outputs
Top Co-Authors

Avatar

Jingjing Li

Central South University

View shared research outputs
Top Co-Authors

Avatar

Xiang Chen

Central South University

View shared research outputs
Top Co-Authors

Avatar

Quanyong He

Central South University

View shared research outputs
Top Co-Authors

Avatar

Huiqing Xie

Central South University

View shared research outputs
Top Co-Authors

Avatar

Qinghai Zeng

Central South University

View shared research outputs
Researchain Logo
Decentralizing Knowledge