Network


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

Hotspot


Dive into the research topics where Ning Luo is active.

Publication


Featured researches published by Ning Luo.


Oncotarget | 2015

A long noncoding RNA AB073614 promotes tumorigenesis and predicts poor prognosis in ovarian cancer

Zhongping Cheng; Jing Guo; Li Chen; Ning Luo; Weihong Yang; Xiaoyan Qu

Long noncoding RNA (lncRNA) profiles in ovarian cancer (OC) remain largely unknown. In the present study, we screened AB073614 as a new candidate lncRNA which promotes development of OC, in two independent datasets (GSE18521 and GSE38666) from the Gene Expression Omnibus (GEO). The level of AB073614 was then detected in 75 paired OC tissues and adjacent normal tissues by qRT-PCR. Results showed that AB073614 expression was significantly up-regulated in 85.3% (64/75) cancerous tissues compared with normal counterparts (P < 0.01). Further, the 5-year overall survival (OS) in OC patients with high expression of AB073614 was inferior to that with low expression (17.2 months vs 30.0 months, P = 0.0025). To investigate the functional role of AB073614, AB073614 siRNA was transfected into OC cell lines. Knockdown of AB073614 expression significantly inhibited cell proliferation and invasion, resulted in cell arrest in G1 phase of cell cycle and a dramatic increase of apoptosis, both in HO-8910 and OVCAR3 cells. In vivo experiment also revealed that knockdown AB073614 inhibited OVCAR3 cells proliferation. Finally, western blot assays indicated that lncRNA AB073614 may exert its function by targeting ERK1/2 and AKT-mediated signaling pathway. In conclusion, our study suggests that lncRNA AB073614 acts as a functional oncogene in OC development.


Scientific Reports | 2016

LPS/TLR4-mediated stromal cells acquire an invasive phenotype and are implicated in the pathogenesis of adenomyosis

Jing Guo; Li Chen; Ning Luo; Caixia Li; Rong Chen; Xiaoyan Qu; Mingmin Liu; Le Kang; Zhongping Cheng

The present study tested whether the LPS/TLR4 signal pathway in endometrial stromal cells is essential for the pathogenesis of adenomyosis. We tested the expression of TLR4, MD2 in the endometrium without adenomyosis (CE), the eutopic endometrium with adenomyosis (EuE) and the ectopic endometrium with adenomyosis (EE). We isolated the stromal cells from CE, EuE and EE (CESC, EuESC, EESC), treated with lipopolysaccharide (LPS) and TLR4 antagonist and detected the cell viability. And we also measured the key protein of the TLR4 signal pathway and inflammatory proliferation and invasive growth of experimental cells. We found that the viability of experimental cells treated with LPS was significantly greater than that of the non-treated cells, blocked by the TLR4 antagonist VIPER. TLR4 signal pathway and inflammatory proliferation and invasive growth of experimental cells stimulated by LPS, and it was inhibited by VIPER. This study suggested that stromal cells were activated by the TLR4 signalling pathway, which processed the cellular inflammatory proliferation and invasive growth involved in the pathogenesis of adenomyosis.


Translational Research | 2014

Estrogen-mediated activation of fibroblasts and its effects on the fibroid cell proliferation

Ning Luo; Qiyu Guan; Lihua Zheng; Xiaoyan Qu; Hong Dai; Zhongping Cheng

In this study, we explored the role of estrogen-mediated activation of stromal fibroblasts in the pathogenesis of uterine fibroid in patients. We isolated uterine fibroids and surrounding smooth muscle from patients and separated fibroblasts using immunomagnetic beads. We also measured the expression levels of estrogen receptors in fibroblasts and examined cell proliferation, expressions of fibroblast activation protein (FAP), extracellular matrix (ECM) (fibronectin, laminin, collagen I), growth factors (transforming growth factor-β, insulin-like growth factor-1), and cell proliferation pathway stimulated by estrogen. We also silenced the expression of FAP by RNA interference and analyzed the expression levels of these markers before and after E2 stimulation. Finally, we also investigated the effect of activated fibroblast supernatant on cell proliferation of fibroblasts, smooth muscle cells, and fibroid cells. We found that fibroblasts in uterine fibroid were activated, and the expression levels of estrogen receptors from fibroid cells were higher than those from smooth muscle cells. After estrogen stimulation, the proliferation activity of fibroblast was enhanced, and the expression of FAP, ECM, and growth factors was increased; the signaling pathway involved in cell proliferation was also activated. Interestingly, the activated fibroblast supernatant stimulation can promote cell proliferation. Silencing of FAP expression could inhibit the E2-mediated biological effects. In conclusion, estrogen promotes proliferation of uterine fibroids through the activation of fibroblasts, thus, activated fibroblasts may play an important role in the pathogenesis of uterine fibroids, which could be targeted in future for the treatment of uterine fibroid.


Cell Death and Disease | 2016

ARHGAP10, downregulated in ovarian cancer, suppresses tumorigenicity of ovarian cancer cells.

Ning Luo; J Guo; L Chen; Weihong Yang; X Qu; Zhongping Cheng

Rho GTPase-activating proteins (RhoGAPs) are implicated in the development and progression of ovarian cancer. ARHGAP10 is a member of RhoGAP proteins and inactivates Cdc42 by converting GTP-bound form to GDP-bound form. Here, we aimed to evaluate ARHGAP10 expression profile and functions in ovarian cancer. The decreased expression of ARHGAP10 was found in 77.3% (58/75) of ovarian cancer tissues, compared with their non-tumorous counterparts. Furthermore, overall survival in ovarian cancer patients with higher expression of ARHGAP10 was longer than those with lower expression. Ectopic expression of ARHGAP10 in two ovarian cancer cell lines with lower expression of ARHGAP10 (A2780 and HO-8910) dramatically suppressed cell proliferation in vitro. In nude mice, its stable overexpression significantly inhibited the tumorigenicity of A2780 cells. We further demonstrated that overexpression of ARHGAP10 significantly inhibited cell adhesion, migration and invasion, resulted in cell arrest in G1 phase of cell cycle and a significant increase of apoptosis. Moreover, ARHGAP10 interacted with Cdc42 and overexpression of ARHGAP10 inhibited the activity of Cdc42 in A2780 cells. Gene set enrichment analysis on The Cancer Genome Atlas dataset showed that KEGG cell cycle, replication and base excision repair (BER) pathways were correlatively with the ARHGAP10 expression, which was further confirmed in ovarian cancer cells by western blotting. Hence, ARHGAP10 may serve as a tumor suppressor through inactivating Cdc42, as well as inhibiting cell cycle, replication and BER pathways. Our data suggest an important role of ARHGAP10 in the molecular etiology of cancer and implicate the potential application of ARHGAP10 in cancer therapy.


Oncology Reports | 2015

Inhibition of TMEM45A suppresses proliferation, induces cell cycle arrest and reduces cell invasion in human ovarian cancer cells

Jing Guo; Li Chen; Ning Luo; Weihong Yang; Xiaoyan Qu; Zhongping Cheng

The association of TMEM45A with various cancers has been recently reported. However, the biological function of TMEM45A in ovarian cancer remains unclear. The present study aimed to elucidate the role of TMEM45A in regulating the biological behavior of ovarian cancer cells. We compared the expression of TMEM45A between ovarian cancer tissues and normal tissues based on RNA-sequencing data of the ovarian cancer cohort from The Cancer Genome Atlas (TCGA) project and our real-time PCR data from 25 pairs of ovarian cancer and their matched non-cancerous tissue samples. The expression of TMEM45A was then suppressed in two ovarian cancer cell lines, HO-8910 and A2780, by RNA interference. Cell proliferation, cell cycle distribution, adhesion and invasive ability were then detected using the Cell Counting Kit-8 assay (CCK-8), propidium iodide (PI) staining, and cell adhesion and Transwell assays, respectively. In addition, the mRNA and protein levels of transforming growth factor-β (TGF-β1 and TGF-β2), Ras homolog family member A (RhoA) and Rho-associated kinase 2 (ROCK2) were detected with real-time PCR and western blotting, respectively. TCGA data and our real-time PCR results demonstrated the overexpression of TMEM45A in ovarian cancer. Silencing of TMEM45A significantly inhibited cell proliferation and significantly increased the cell population in the G1 phase. Moreover, knockdown of TMEM45A also inhibited cell adhesion as well as cell invasion. More importantly, suppression of TMEM45A notably downregulated the expression of TGF-β1, TGF-β2, RhoA and ROCK2. In conclusion, TMEM45A may function as an oncogene for ovarian cancer, and inhibition of TMEM45A may be a therapeutic strategy for ovarian cancer.


Molecular Carcinogenesis | 2016

Knockdown of EHF inhibited the proliferation, invasion and tumorigenesis of ovarian cancer cells.

Zhongping Cheng; Jing Guo; Li Chen; Ning Luo; Weihong Yang; Xiaoyan Qu

Ovarian cancer is the most lethal gynecologic malignancy worldwide. ETS homologous factor (EHF), a member of E26 transformation specific (ETS) transcription factors, has been reported overexpressed in ovarian cancer. However, the molecular mechanism underlying the biological function of EHF in ovarian cancer is still unclear. Here, we found that EHF was elevated in ovarian cancer tissues compared with non‐tumorous tissues. Moreover, high EHF expression level was correlated with short survival time of patients with ovarian cancer. Knockdown of EHF in ovarian cancer cells, SKOV3 and OVCAR3, significantly inhibited cell proliferation and increased cells population in G1 phase. The proteins promoting cell cycles (Cyclin B1, Cyclin D1, and PCNA) were down‐regulated and the protein negatively regulating cell cycle progression (P21) was up‐regulated after EHF knockdown. Moreover, inhibition of EHF in ovarian cancer cells dramatically induced cell apoptosis, but impaired cell adhesion and cell invasion. Furthermore, phosphorylation levels of ERK and AKT were notably reduced in EHF knockdown cells. Finally, in vivo data showed that knockdown of EHF inhibited tumor growth in nude mice. Our data indicates that EHF could be a potential prognosis marker for ovarian cancer and work as an oncogene by targeting ERK and AKT signaling, which can serve as a new target for ovarian cancer treatment.


Journal of Experimental & Clinical Cancer Research | 2015

Overexpression of TMEM158 contributes to ovarian carcinogenesis

Zhongping Cheng; Jing Guo; Li Chen; Ning Luo; Weihong Yang; Xiaoyan Qu

BackgroundTransmembrane protein 158 (TMEM158) is a recently identified upregulated gene during Ras-induced senescence. Its association with various cancers has been recently reported. However, the expression and biological function of TMEM158 in ovarian cancer is still unclear. This study was aimed to elucidate the roles of TMEM158 in cell proliferation, adhesion and cell invasion of ovarian cancer cells.MethodsWe analyzed TMEM158 mRNA level in ovarian cancer tissues and adjacent no-tumorous tissues by real-time PCR. We then suppressed TMEM158 expression of ovarian cancer cells by RNA interference and examined the effects of TMEM158 knockdown on cancerous transformation of ovarian cancer cells.ResultsThe RNA-sequencing data of the ovarian cancer cohort from The Cancer Genome Atlas project (TCGA) and our real-time PCR data showed that TMEM158 was overexpressed in ovarian cancer. Knockdown of TMEM158 by RNA interference in ovarian cancer cells significantly inhibited cell proliferation, which may be due to the increase of G1-phase arrest. Silencing of TMEM158 also inhibited cell adhesion, cell invasion as well as tumorigenicity in nude mice. Moreover, knockdown of TMEM158 notably repressed cell adhesion via down-regulating the expression intercellular adhesion molecule1 (ICAM1) and vascular cell adhesion molecule1 (VCAM1). Transforming Growth Factor-β (TGF-β) signaling pathway was also remarkably impaired by TMEM158 silencing.ConclusionsOur data suggests that TMEM158 may work as an oncogene for ovarian cancer and that inhibition of TMEM158 may be a therapeutic strategy for ovarian cancer.


Scientific Reports | 2017

The Expression of Toll-like receptors in eutopic and ectopic endometrium and its implication in the inflammatory pathogenesis of adenomyosis.

Caixia Jiang; Chao Liu; Jing Guo; Li Chen; Ning Luo; Xiaoyan Qu; Weihong Yang; Qing Ren; Zhongping Cheng

In this study, we investigated the expression profiles of Toll-like receptors(TLRs) in eutopic endometrium(EU) and ectopic endometrium(EC) and its implication in the inflammatory pathogenesis of adenomyosis. Thirty adenomyosis patients who underwent laparoscopy were recruited in this study. We tested the mRNA and protein expression of TLRs, and the mRNA expression of IL-6 and IL-8 in EU and EC of adenomyosis patients, and control endometrium without adenomyosis(CE). We found that the mRNA expression of IL-6 and IL-8 in EU was significantly higher than that in CE, and was the highest in EC (P < 0.01). The mRNA and protein expression of TLRs were higher in EU, with the expression of TLR1-6, 8 and 9 being significantly higher in EU than in CE, and were the highest in EC (except TLR6) (P < 0.05 or P < 0.01). Pearson correlation analysis showed that the expression of TLR1, 2, 4, 5 and 9 in EU and EC was positively correlated with that of IL-6 and IL-8 (P < 0.00139). This study suggested that adenomyosis was a state of inflammatory pathology. High expression of TLRs in EU and EC were positively correlated with IL-6 and IL-8, which may be involved in the inflammatory pathogenesis of adenomyosis.


Cell Death and Disease | 2018

TRIM52 plays an oncogenic role in ovarian cancer associated with NF-kB pathway

Weihong Yang; Li Liu; Caixia Li; Ning Luo; Rong Chen; Li Li; Fudong Yu; Zhongping Cheng

Emerging evidence suggests that the members of the tripartite motif (TRIM) family play a crucial role in cancer development and progression. The purpose of the study was to explore TRIM52s role in tumorigenesis and its potential molecular mechanism in ovarian cancer. The study demonstrated that knockdown of TRIM52 in SKOV3 and CAOV3 cells inhibited ovarian cancer cell invasion, migration, and proliferation, and induced cell apoptosis. On the contrary, overexpression of TRIM52 in HO8910 cells showed contrary results. Further, overexpression of TRIM52 enhanced the expression of phosphorylated IKKβ and IKBα proteins and nuclear protein P65, which implied the activation of NF-kB signal pathway. Knockdown of TRIM52 downregulated the mRNA and protein levels of NF-kB signal downstream effectors of the NF-kB pathway, including MMP9, Bcl2, IL8, and TNFα, but upregulated caspase-3 expression. These results suggested that activation of the NF-kB pathway is involved in TRIM52-mediated regulation in ovarian cancer. The nude mice study further confirmed that knockdown of TRIM52 blocked tumor growth, inhibited cell proliferation, and promoted cell apoptosis. Our data strongly suggested that TRIM52 plays an oncogenic role in ovarian cancer development associated with the NF-kB signal pathway and may be a potential target for cancer therapy.


PLOS ONE | 2017

Eutopic/ectopic endometrial apoptosis initiated by bilateral uterine artery occlusion: A new therapeutic mechanism for uterus-sparing surgery in adenomyosis

Li Chen; Caixia Li; Jing Guo; Ning Luo; Xiaoyan Qu; Le Kang; Mingmin Liu; Zhongping Cheng

The objective of the present study was to investigate differences in the expression of apoptosis-related factors in the eutopic and ectopic endometrium (EuE/EE) in women with adenomyosis before and after laparoscopic bilateral uterine artery occlusion (LUAO). Ten patients with uterine adenomyosis who received LUAO were selected as the research subjects, from whom EuE and EE tissues were obtained before and after LUAO and detected for the expression of apoptosis-related molecules in EuE and EE by PT-PCR and Western blot, and changes in the mitochondrial structure by electron microscopy. Normal endometrial stromal cells (NESC), and EuE/EE stromal cells in women with adenomyosis were cultured in a 1% O2, 5% CO2 incubator to establish a physical anoxia state in an in vitro stromal cell model. The expression of apoptosis-related molecules was observed at 0, 6, 12, 24 and 48h of hypoxic. The results showed that the expression of apoptosis-related factors in EuE and EE were increased significantly after LUAO and under hypoxic conditions in vitro, suggesting that transient ischemia and hypoxia were involved in the apoptosis of adenomysis lesions, and that uterine artery occlusion could remove adenomyosis lesions on tissue/cell level by cytoreduction, thus reaching the goal of treating adenomyosis effectively.

Collaboration


Dive into the Ning Luo'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