Network


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

Hotspot


Dive into the research topics where Juanjuan Ou is active.

Publication


Featured researches published by Juanjuan Ou.


Nature | 2016

Follicular CXCR5-expressing CD8 + T cells curtail chronic viral infection

Ran He; Shiyue Hou; Cheng Liu; Anli Zhang; Qiang Bai; Miao Han; Yu Yang; Gang Wei; Ting Shen; Xinxin Yang; Lifan Xu; Xiangyu Chen; Yaxing Hao; Pengcheng Wang; Chuhong Zhu; Juanjuan Ou; Houjie Liang; Ting Ni; Xiaoyan Zhang; Xinyuan Zhou; Kai Deng; Yaokai Chen; Yadong Luo; Jianqing Xu; Hai Qi; Yuzhang Wu; Lilin Ye

During chronic viral infection, virus-specific CD8+ T cells become exhausted, exhibit poor effector function and lose memory potential. However, exhausted CD8+ T cells can still contain viral replication in chronic infections, although the mechanism of this containment is largely unknown. Here we show that a subset of exhausted CD8+ T cells expressing the chemokine receptor CXCR5 has a critical role in the control of viral replication in mice that were chronically infected with lymphocytic choriomeningitis virus (LCMV). These CXCR5+ CD8+ T cells were able to migrate into B-cell follicles, expressed lower levels of inhibitory receptors and exhibited more potent cytotoxicity than the CXCR5− subset. Furthermore, we identified the Id2–E2A signalling axis as an important regulator of the generation of this subset. In patients with HIV, we also identified a virus-specific CXCR5+ CD8+ T-cell subset, and its number was inversely correlated with viral load. The CXCR5+ subset showed greater therapeutic potential than the CXCR5− subset when adoptively transferred to chronically infected mice, and exhibited synergistic reduction of viral load when combined with anti-PD-L1 treatment. This study defines a unique subset of exhausted CD8+ T cells that has a pivotal role in the control of viral replication during chronic viral infection.


PLOS ONE | 2012

αV Integrin Induces Multicellular Radioresistance in Human Nasopharyngeal Carcinoma via Activating SAPK/JNK Pathway

Juanjuan Ou; Wei Luan; Jia Deng; Rina Sa; Houjie Liang

Background Tumor cells acquire the capacity of resistance to chemotherapy or radiotherapy via cell–matrix and cell–cell crosstalk. Integrins are the most important cell adhesion molecules, in which αV integrin mainly mediating the tight contact between tumor cells. Methodology/Principal Findings To investigate the role of αV integrin in multi-cellular radioresistance (MCR) of human nasopharyngeal carcinoma (NPC), we performed immunohistochemistry and Western blotting to find that the expression of αV integrin in the tumor tissue of radioresistant patients is much higher than that in radiosensitive patients. In vitro, we cultured human NPC cell line CNE-2 cells as multi-cellular spheroids (MCSs) or as monolayer cells (MCs), and found that the expression of αV integrin in MCSs is significantly higher than that in MCs. MTT, flow cytometry and clonogenic suvival assays showed that MCSs are less sensitive to X-ray irradiation than MCs while blocking of αV integrin in MCSs dramatically reversed their radioresistance. Furthermore, as detected by Western blotting, MCSs displayed sustained activation of the stress-activated protein kinase/c-Jun NH2-terminal kinase (SAPK/JNK) pathway in presence of irradiation. Blocking of αV integrin in MCSs decreased the expression of phosphorylated JNK. Additionally, blocking of SAPK/JNK signaling pathway synergistically induced apoptosis of MCSs exposed to irradiation by increasing the expression of cleaved caspase-3. In vivo, we found that irradiation combined with αV integrin blocking treatment significantly enhanced the radiosensitivity of NPC xenografts. Conclusions Our results indicate a novel role of αV integrin in multi-cellular radioresistance of NPCs.


Clinical Cancer Research | 2011

Endogenous Axon Guiding Chemorepulsant Semaphorin-3F Inhibits the Growth and Metastasis of Colorectal Carcinoma

Feng Wu; Qi Zhou; Jing Yang; Guangjie Duan; Juanjuan Ou; Rong Zhang; Feng Pan; Qiuping Peng; Hong Tan; Yi-Fang Ping; You-Hong Cui; Cheng Qian; Xiaochu Yan; Xiu-wu Bian

Purpose: To elucidate the role of Semaphorin-3F (SEMA3F), originally described as an axon guiding chemorepulsant implicated in nerve development, in the progression of colorectal carcinoma. Experimental Design: SEMA3F and its receptor NRP2 were examined in 72 cases of human colorectal carcinoma specimens and cell lines LoVo, SW480, and SW620 with immunohistochemistry and Western blotting. SEMA3F mRNA expression in the frozen tissue specimens and cell lines was examined with quantitative reverse transcriptase-PCR. Confocal laser scanning microscopy was used for detection of cellular localization of the proteins by immunofluorescent staining. MTT assay, flow cytometry, cell adhesion and migration, and xenografts were used to evaluate biological significance of SEMA3F. Results: SEMA3F was significantly reduced in colorectal carcinoma tissues and cell lines. Overexpression of SEMA3F resulted in reduced proliferation, adhesion to fibronectin, and migratory capability as well as reduced S-phase population and integrin αvβ3 expression of SW480 colon cancer cells. In addition, SEMA3F-overexpressing cells exhibited diminished tumorigenesis when transplanted orthotopically in nude mice and reduced liver metastases. Moreover, transfection of siRNA targeting SEMA3F in colon cancer cells increased their tumorigenicity in vivo. Conclusions: Endogenous SEMA3F acts as a suppressor of the growth and metastasis of human colorectal cancer cells. Clin Cancer Res; 17(9); 2702–11. ©2011 AACR.


Journal of Cellular Biochemistry | 2011

Endostatin suppresses colorectal tumor-induced lymphangiogenesis by inhibiting expression of fibronectin extra domain A and integrin α9.

Juanjuan Ou; Jianjun Li; Feng Pan; Ganfeng Xie; Qi Zhou; Haihui Huang; Houjie Liang

Endostatin is a natural occurring anti‐angiogenic peptide and has been shown to inhibit tumor lymphangiogenesis by suppressing the expression of tumor‐stimulating growth factors. We have previously shown that fibronectin alternative extra domain A (EDA) facilitates lymphangiogenesis of colorectal tumors. Since it is known that EDA interacts with integrin α9 in the lymphatic endothelial cells (LECs), we hypothesized that endostatin may target EDA‐integrin α9 pathway to inhibit colorectal tumor‐induced lymphangiogenesis. To test this hypothesis, we examined the effect of endostatin on EDA secreted by SW480 colorectal cancer cells and treated human LECs with different doses of endostatin in the presence of conditional medium from SW480 cells. We found that endostatin significantly reduced EDA secretion by SW480 cells and the expression of integrin α9 in LECs. Immunofluorescence studies showed that EDA and integrin α9 colocalized on the cell membrane of LECs and these colocalizations were dramatically reduced by endostatin. Co‐immunoprecipitation studies demonstrated that EDA interacted with integrin α9 in LECs, and showed that endostatin treatment inhibited the formation of EDA–integrin α9 complex in LECs. Furthermore, we found that the arrangement and polarity of LEC cytoskeletons were destroyed by endostatin substantially, leading to a reduced formation of tube‐like structures of LECs and a suppressed chemotaxis of LECs toward SW480 cells. Consistently, EDA and integrin α9 expressions as well as lymphangiogenesis were significantly suppressed by endostatin in colorectal cancer xenografts. In conclusion, our results suggest that endostatin reduces colorectal tumor‐induced lymphangiogenesis, at least in part, by inhibiting EDA‐integrin α9 pathway. J. Cell. Biochem. 112: 2106–2114, 2011.


Cancer Letters | 2015

Neuropilin-2 mediates lymphangiogenesis of colorectal carcinoma via a VEGFC/VEGFR3 independent signaling

Juanjuan Ou; Xing Wei; Yuan Peng; Lin Zha; Rongbin Zhou; Hang Shi; Qi Zhou; Houjie Liang

Lymphangiogenesis critically contributes to the lymphatic metastasis of colorectal carcinomas (CRCs), but the underlying mechanism of CRC lymphangiogenesis remains largely elusive. We have previously demonstrated that Semaphorin-3F (SEMA3F) is critically involved in CRC metastasis, and the receptor of SEMA3F, neuropilin-2 (NRP2), originally described as an axon guiding chemorepulsant implicated in nerve development, has been suggested in promoting lymphangiogenesis via acting as an obligate co-receptor of VEGFR3 cooperatively enhancing the activity of VEGF-C. Our present study revealed that in colorectal carcinomas, NRP2 expression levels of tumor-associated lymphatic endothelial cells (LECs) are significantly correlated with the density of tumor lymphatic vessels. In vitro, activation of NRP2 in LECs substantially facilitates their migration, sprouting, and tubulogenesis capacity via regulating the rearrangement of cytoskeleton polarity. In vivo model further showed that in the xenografts generated from SEMA3F knockdown CRC cells, NRP2 is substantially activated in tumor-associated LECs, resulting in a significantly increased tumor lymphangiogenesis. Further evidence demonstrated that CRC cell induces the activation of NRP2 in LECs to promote tumor lymphangiogenesis via integrinα9β1/FAK/Erk pathway independent VEGF-C/VEGFR3 signaling. Our study for the first time revealed the novel molecular mechanism of NRP2-mediated-lymphangiogenesis in CRCs, suggesting NRP2 as a potential therapeutic target in preventing lymphatic metastasis of CRCs.


Carcinogenesis | 2014

Endothelial cell-derived fibronectin extra domain A promotes colorectal cancer metastasis via inducing epithelial–mesenchymal transition

Juanjuan Ou; Yuan Peng; Jia Deng; Hongming Miao; Jie Zhou; Lin Zha; Rongbin Zhou; Liqing Yu; Hang Shi; Houjie Liang

Recent evidence has been suggesting the important roles of endothelial cells (ECs) involved in the pathogenesis of several cancers, including colorectal carcinomas (CRCs), but the underlying mechanism remains elusive. We have demonstrated previously that CRC-derived fibronectin extra domain A (EDA) promotes vasculogenesis, tumorigenesis and metastasis of CRCs. At the current study, we showed that EC-secreted EDA promotes the metastatic capacity CRC cells via inducing an epithelial-mesenchymal transition. In vitro and in vivo experiments showed that EC-secreted EDA, via the interaction with integrin α9β1 on neighboring CRC cells, leads to the activation of focal adhesion kinase as well as Rac signalings, thus strengthens the polarity of cytoskeleton and promotes the invasion capacity of CRC cells. Furthermore, Erk signaling pathway was revealed to critically mediate the effect of EC-derived EDA on CRC cells. Our findings reveal a novel oncogenic role of ECs in promoting CRC malignancy through secreting EDA.


Stem Cell Research | 2013

Fibronectin extra domain A (EDA) sustains CD133(+)/CD44(+) subpopulation of colorectal cancer cells.

Juanjuan Ou; Jia Deng; Xing Wei; Ganfeng Xie; Rongbin Zhou; Liqing Yu; Houjie Liang

Fibronectin is a major extracellular matrix glycoprotein with several alternatively spliced variants, including extra domain A (EDA), which was demonstrated to promote tumorigenesis via stimulating angiogenesis and lymphangiogenesis. Given that CD133(+)/CD44(+) cancer cells are critical in tumorigenesis of colorectal cancer (CRC), we hypothesize that fibronectin EDA may promote tumorigenesis by sustaining the properties of CD133(+)/CD44(+) colon cancer cells. We found that tumor tissue and serum EDA levels are substantially higher in advanced versus early stage human CRC. Additionally we showed that tumor tissue EDA levels are positively correlated with differentiation status and chemoresistance, and correlated with a poor prognosis of CRC patients. We also showed that in colon cancer cells SW480, CD133(+)/CD44(+) versus CD133(-)/CD44(-) cells express significantly elevated EDA receptor integrin α9β1. Silencing EDA in SW480 cells reduces spheroid formation and cells positive for CD133 or CD44, which is associated with reduced expressions of embryonic stem cell markers and increased expressions of differentiation markers. Blocking integrin α9β1 function strongly reversed the effect of EDA overexpression. We also provided evidence suggesting that EDA sustains Wnt/β-catenin signaling activity via activating integrin/FAK/ERK pathway. In xenograft models, EDA-silenced SW480 cells exhibit reduced tumorigenic and metastatic capacity. In conclusion, EDA is essential for the maintenance of the properties of CD133(+)/CD44(+) colon cancer cells.


PLOS ONE | 2012

The extra domain A of fibronectin increases VEGF-C expression in colorectal carcinoma involving the PI3K/AKT signaling pathway.

Lisha Xiang; Ganfeng Xie; Juanjuan Ou; Xing Wei; Feng Pan; Houjie Liang

The extra domain A (EDA)-containing fibronectin (EDA-FN), an alternatively spliced form of the extracellular matrix protein fibronectin, is predominantly expressed in various malignancies but not in normal tissues. In the present study, we investigated the potential pro-lymphangiogenesis effects of extra domain A (EDA)-mediated vascular endothelial growth factor-C (VEGF-C) secretion in colorectal carcinoma (CRC). We detected the expressions of EDA and VEGF-C in 52 human colorectal tumor tissues and their surrounding mucosae by immunohistochemical analysis, and further tested the correlation between the expressions of these two proteins in aforementioned CRC tissues. Both EDA and VEGF-C were abundantly expressed in the specimens of human CRC tissues. And VEGF-C was associated with increased expression of EDA in human CRC according to linear regression analysis. Besides, EDA expression was significantly correlated with lymph node metastasis, tumor differentiation and clinical stage by clinicopathological analysis of tissue microarrays containing tumor tissues of 115 CRC patients. Then, human CRC cell SW480 was transfected with lentivectors to elicit expression of shRNA against EDA (shRNA-EDA), and SW620 was transfected with a lentiviral vector to overexpress EDA (pGC-FU-EDA), respectively. We confirmed that VEGF-C was upregulated in EDA-overexpressed cells, and downregulated in shRNA-EDA cells. Moreover, a PI3K-dependent signaling pathway was found to be involved in EDA-mediated VEGF-C secretion. The in vivo result demonstrated that EDA could promote tumor growth and tumor-induced lymphangiogenesis in mouse xenograft models. Our findings provide evidence that EDA could play a role in tumor-induced lymphangiogenesis via upregulating autocrine secretion of VEGF-C in colorectal cancer, which is associated with the PI3K/Akt-dependent pathway.


Nature Communications | 2016

Macrophage ABHD5 promotes colorectal cancer growth by suppressing spermidine production by SRM

Hongming Miao; Juanjuan Ou; Yuan Peng; Xuan Zhang; Yujuan Chen; Lijun Hao; Ganfeng Xie; Zhe Wang; Xueli Pang; Zhihua Ruan; Jianjun Li; Liqing Yu; Bingzhong Xue; Hang Shi; Chunmeng Shi; Houjie Liang

Metabolic reprogramming in stromal cells plays an essential role in regulating tumour growth. The metabolic activities of tumour-associated macrophages (TAMs) in colorectal cancer (CRC) are incompletely characterized. Here, we identify TAM-derived factors and their roles in the development of CRC. We demonstrate that ABHD5, a lipolytic co-activator, is ectopically expressed in CRC-associated macrophages. We demonstrate in vitro and in mouse models that macrophage ABHD5 potentiates growth of CRC cells. Mechanistically, ABHD5 suppresses spermidine synthase (SRM)-dependent spermidine production in macrophages by inhibiting the reactive oxygen species-dependent expression of C/EBPɛ, which activates transcription of the srm gene. Notably, macrophage-specific ABHD5 transgene-induced CRC growth in mice can be prevented by an additional SRM transgene in macrophages. Altogether, our results show that the lipolytic factor ABHD5 suppresses SRM-dependent spermidine production in TAMs and potentiates the growth of CRC. The ABHD5/SRM/spermidine axis in TAMs might represent a potential target for therapy.


Journal of Pineal Research | 2017

Melatonin regulates PARP1 to control the senescence‐associated secretory phenotype (SASP) in human fetal lung fibroblast cells

Songtao Yu; Xiaojiao Wang; Peiliang Geng; Xudong Tang; Lisha Xiang; Xin Lu; Jianjun Li; Zhihua Ruan; Jianfang Chen; Ganfeng Xie; Zhe Wang; Juanjuan Ou; Yuan Peng; Xi Luo; Xuan Zhang; Yan Dong; Xueli Pang; Hongming Miao; Hongshan Chen; Houjie Liang

Cellular senescence is an important tumor‐suppressive mechanism. However, acquisition of a senescence‐associated secretory phenotype (SASP) in senescent cells has deleterious effects on the tissue microenvironment and, paradoxically, promotes tumor progression. In a drug screen, we identified melatonin as a novel SASP suppressor in human cells. Strikingly, melatonin blunts global SASP gene expression upon oncogene‐induced senescence (OIS). Moreover, poly(ADP‐ribose) polymerase‐1 (PARP‐1), a sensor of DNA damage, was identified as a new melatonin‐dependent regulator of SASP gene induction upon OIS. Here, we report two different but potentially coherent epigenetic strategies for melatonin regulation of SASP. The interaction between the telomeric repeat‐containing RNA (TERRA) and PARP‐1 stimulates the SASP, which was attenuated by 67.9% (illustrated by the case of IL8) by treatment with melatonin. Through binding to macroH2A1.1, PARP‐1 recruits CREB‐binding protein (CBP) to mediate acetylation of H2BK120, which positively regulates the expression of target SASP genes, and this process is interrupted by melatonin. Consequently, the findings provide novel insight into melatonins epigenetic role via modulating PARP‐1 in suppression of SASP gene expression in OIS‐induced senescent cells. Our studies identify melatonin as a novel anti‐SASP molecule, define PARP‐1 as a new target by which melatonin regulates SASP, and establish a new epigenetic paradigm for a pharmacological mechanism by which melatonin interrupts PARP‐1 interaction with the telomeric long noncoding RNA(lncRNA) or chromatin.

Collaboration


Dive into the Juanjuan Ou's collaboration.

Top Co-Authors

Avatar

Houjie Liang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Ganfeng Xie

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Jianjun Li

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Peiliang Geng

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Lisha Xiang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Ning Wang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Rina Sa

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yunmei Liao

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Chen Liu

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Hongming Miao

Third Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge