Network


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

Hotspot


Dive into the research topics where ng Li is active.

Publication


Featured researches published by ng Li.


Gastroenterology | 2014

Antibody Against CD44s Inhibits Pancreatic Tumor Initiation and Postradiation Recurrence in Mice

Ling Li; Xinbao Hao; Jun Qin; Wenhua Tang; Fengtian He; Amber Smith; Min Zhang; Diane M. Simeone; Xiaotan T. Qiao; Zhi Nan Chen; Theodore S. Lawrence; Liang Xu

BACKGROUND & AIMSnCD44s is a surface marker of tumor-initiating cells (TICs); high tumor levels correlate with metastasis and recurrence, as well as poor outcomes for patients. Monoclonal antibodies against CD44s might eliminate TICs with minimal toxicity. This strategy is unclear for treatment of pancreatic cancer, and little is known about how anti-CD44s affect pancreatic cancer initiation or recurrence after radiotherapy.nnnMETHODSnOne hundred ninety-two pairs of human pancreatic adenocarcinoma and adjacent nontumor pancreatic tissues were collected from patients undergoing surgery. We measured CD44s levels in tissue samples and pancreatic cancer cell lines by immunohistochemistry, real-time polymerase chain reaction, and immunoblot; levels were correlated with patient survival times. We studied the effects of anti-CD44s in mice with human pancreatic tumor xenografts and used flow cytometry to determine the effects on TICs. Changes in CD44s signaling were examined by real-time polymerase chain reaction, immunoblot, reporter assay, and inxa0vitro tumorsphere formation assays.nnnRESULTSnLevels of CD44s were significantly higher in pancreatic cancer than adjacent nontumor tissues. Patients whose tumors expressed high levels of CD44s had a median survival of 10 months compared with >43 months for those with low levels. Anti-CD44s reduced growth, metastasis, and postradiation recurrence of pancreatic xenograft tumors in mice. The antibody reduced the number of TICs in cultured pancreatic cancer cells and xenograft tumors, as well as their tumorigenicity. In cultured pancreatic cancer cell lines, anti-CD44s down-regulated the stem cell self-renewal genes Nanog, Sox-2, and Rex-1 and inhibited signal transducer and activator of transcription 3-mediated cell proliferation and survival signaling.nnnCONCLUSIONSnThe TIC marker CD44s is up-regulated in human pancreatic tumors and associated with patient survival time. CD44s is required for initiation, growth, metastasis, and postradiation recurrence of xenograft tumors in mice. Anti-CD44s eliminated bulk tumor cells as well as TICs from the tumors. Strategies to target CD44s cab be developed to block pancreatic tumor formation and post-radiotherapy recurrence in patients.


Clinical Cancer Research | 2013

HAb18G/CD147 Promotes pSTAT3-Mediated Pancreatic Cancer Development via CD44s

Ling Li; Wenhua Tang; Xiaoqing Wu; David Karnak; Xiaojie Meng; Rachel Thompson; Xinbao Hao; Yongmin Li; Xiaotan T. Qiao; Jiayuh Lin; James R. Fuchs; Diane M. Simeone; Zhi Nan Chen; Theodore S. Lawrence; Liang Xu

Purpose: Signal transducer and activator of transcription 3 (STAT3) plays a critical role in initiation and progression of pancreatic cancer. However, therapeutically targeting STAT3 has failed clinically. We previously identified HAb18G/CD147 as an effective target for cancer treatment. In this study, we aimed to investigate the potential role of HAb18G/CD147 in STAT3-involved pancreatic tumorigenesis in vitro and in vivo. Experimental Design: The expression of HAb18G/CD147, pSTAT3, and CD44s was determined in tissue microarrays. The tumorigenic function and molecular signaling mechanism of HAb18G/CD147 were assessed by in vitro cellular and clonogenic growth, reporter assay, immunoblot assay, immunofluorescence staining, immunoprecipitation, and in vivo tumor formation using loss or gain-of-function strategies. Results: Highly expressed HAb18G/CD147 promoted cellular and clonogenic growth in vitro and tumorigenicity in vivo. Cyclophilin A (CyPA), a ligand of CD147, stimulated STAT3 phosphorylation and its downstream genes cyclin D1/survivin through HAb18G/CD147-dependent mechanisms. HAb18G/CD147 was associated and colocalized with cancer stem cell marker CD44s in lipid rafts. The inhibitors of STAT3 and survivin, as well as CD44s neutralizing antibodies suppressed the HAb18G/CD147-induced cell growth. High HAb18G/CD147 expression in pancreatic cancer was significantly correlated with the poor tumor differentiation, and the high coexpression of HAb18G/CD147-CD44s-STAT3 associated with poor survival of patients with pancreatic cancer. Conclusions: We identified HAb18G/CD147 as a novel upstream activator of STAT3, which interacts with CD44s and plays a critical role in the development of pancreatic cancer. The data suggest that HAb18G/CD147 could be a promising therapeutic target for highly aggressive pancreatic cancer and a surrogate marker in the STAT3-targeted molecular therapies. Clin Cancer Res; 19(24); 6703–15. ©2013 AACR.


Oncotarget | 2016

Overcoming chemo/radio-resistance of pancreatic cancer by inhibiting STAT3 signaling

Xiaoqing Wu; Wenhua Tang; Rebecca T. Marquez; Ke Li; Chad Highfill; Fengtian He; Jiqin Lian; Jiayuh Lin; James R. Fuchs; Min Ji; Ling Li; Liang Xu

Chemo/radio-therapy resistance to the deadly pancreatic cancer is mainly due to the failure to kill pancreatic cancer stem cells (CSCs). Signal transducer and activator of transcription 3 (STAT3) is activated in pancreatic CSCs and, therefore, may be a valid target for overcoming therapeutic resistance. Here we investigated the potential of STAT3 inhibition in sensitizing pancreatic cancer to chemo/radio-therapy. We found that the levels of nuclear pSTAT3 in pancreatic cancer correlated with advanced tumor grade and poor patient outcome. Liposomal delivery of a STAT3 inhibitor FLLL32 (Lip-FLLL32) inhibited STAT3 phosphorylation and STAT3 target genes in pancreatic cancer cells and tumors. Consequently, Lip-FLLL32 suppressed pancreatic cancer cell growth, and exhibited synergetic effects with gemcitabine and radiation treatment in vitro and in vivo. Furthermore, Lip-FLLL32 reduced ALDH1-positive CSC population and modulated several potential stem cell markers. These results demonstrate that Lip-FLLL32 suppresses pancreatic tumor growth and sensitizes pancreatic cancer cells to radiotherapy through inhibition of CSCs in a STAT3-dependent manner. By targeting pancreatic CSCs, Lip-FLLL32 provides a novel strategy for pancreatic cancer therapy via overcoming radioresistance.


Oncotarget | 2016

A novel small-molecule compound targeting CD147 inhibits the motility and invasion of hepatocellular carcinoma cells.

Zhi-Guang Fu; Wang L; Hong-Yong Cui; Jian-long Peng; Shi-Jie Wang; Jie-jie Geng; Ji-de Liu; Fei Feng; Fei Song; Ling Li; Ping Zhu; Jian-Li Jiang; Zhi-Nan Chen

CD147, a type I transmembrane glycoprotein, is highly expressed in various cancer types and plays important roles in tumor progression, especially by promoting the motility and invasion of hepatocellular carcinoma (HCC) cells. These crucial roles make CD147 an attractive target for therapeutic intervention in HCC, but no small-molecule inhibitors of CD147 have been developed to date. To identify a candidate inhibitor, we used a pharmacophore model derived from the structure of CD147 to virtually screen over 300,000 compounds. The 100 highest-ranked compounds were subjected to biological assays, and the most potent one, dubbed AC-73 (ID number: AN-465/42834501), was studied further. We confirmed that AC-73 targeted CD147 and further demonstrated it can specifically disrupt CD147 dimerization. Moreover, molecular docking and mutagenesis experiments showed that the possible binding sites of AC-73 on CD147 included Glu64 and Glu73 in the N-terminal IgC2 domain, which two residues are located in the dimer interface of CD147. Functional assays revealed that AC-73 inhibited the motility and invasion of typical HCC cells, but not HCC cells that lacked the CD147 gene, demonstrating on-target action. Further, AC-73 reduced HCC metastasis by suppressing matrix metalloproteinase (MMP)-2 via down-regulation of the CD147/ERK1/2/signal transducer and activator of transcription 3 (STAT3) signaling pathway. Finally, AC-73 attenuated progression in an orthotopic nude mouse model of liver metastasis, suggesting that AC-73 or its derivatives have potential for use in HCC intervention. We conclude that the novel small-molecule inhibitor AC-73 inhibits HCC mobility and invasion, probably by disrupting CD147 dimerization and thereby mainly suppressing the CD147/ERK1/2/STAT3/MMP-2 pathways, which are crucial for cancer progression.


Oncotarget | 2016

Gemcitabine enhances cell invasion via activating HAb18G/CD147-EGFR-pSTAT3 signaling

Bao-Qing Xu; Zhi-Guang Fu; Yao Meng; Xiaoqing Wu; Bo Wu; Liang Xu; Jian-Li Jiang; Ling Li; Zhi-Nan Chen

Pancreatic cancer, one of the most lethal cancers, has very poor 5-year survival partly due to gemcitabine resistance. Recently, it was reported that chemotherapeutic agents may act as stressors to induce adaptive responses and to promote chemoresistance in cancer cells. During long-term drug treatment, the minority of cancer cells survive and acquire an epithelial-mesenchymal transition phenotype with increased chemo-resistance and metastasis. However, the short-term response of most cancer cells remains unclear. This study aimed to investigate the short-term response of pancreatic cancer cells to gemcitabine stress and to explore the corresponding mechanism. Our results showed that gemcitabine treatment for 24 hours enhanced pancreatic cancer cell invasion. In gemcitabine-treated cells, HAb18G/CD147 was up-regulated; and HAb18G/CD147 down-regulation or inhibition attenuated gemcitabine-enhanced invasion. Mechanistically, HAb18G/CD147 promoted gemcitabine-enhanced invasion by activating the EGFR (epidermal growth factor receptor)-STAT3 (signal transducer and activator of transcription 3) signaling pathway. Inhibition of EGFR-STAT3 signaling counteracted gemcitabine-enhanced invasion, and which relied on HAb18G/CD147 levels. In pancreatic cancer tissues, EGFR was highly expressed and positively correlated with HAb18G/CD147. These data indicate that pancreatic cancer cells enhance cell invasion via activating HAb18G/CD147-EGFR-pSTAT3 signaling. Our findings suggest that inhibiting HAb18G/CD147 is a potential strategy for overcoming drug stress-associated resistance in pancreatic cancer.


Cancer Research | 2010

Abstract 2084: Tumor suppressor microRNA miR-34 inhibits human pancreatic cancer stem cells

Wenhua Tang; Qing Ji; Xinbao Hao; Ling Li; DeSano Jeffrey; Eric R. Fearon; Theodore S. Lawrence; Liang Xu

Proceedings: AACR 101st Annual Meeting 2010‐‐ Apr 17‐21, 2010; Washington, DCnnMicroRNAs (miRNAs) have been implicated in cancer initiation and progression via their ability to affect expression of genes and proteins that regulate cell proliferation and/or cell death. Transcription of the three miRNA miR-34 family members was recently found to be directly regulated by p53. We show that miR-34 directly regulates Notch and Bcl-2, suggesting the possibility of a role for miR-34 in the maintenance and survival of cancer stem cells.nnWe examined the roles of miR-34 in p53-mutant human pancreatic cancer cell lines and the potential link to cancer stem cells. Restoration of miR-34 expression in the cancer cells by miR-34 mimics or lentiviral miR-34 downregulated Bcl-2 and Notch1/2. miR-34 restoration significantly inhibited clonogenic cell growth and invasion, induced apoptosis and G1 and G2/M arrest in cell cycle, and sensitized the cancer cells to chemotherapy and radiation. We identified that CD44+/CD133+ cancer cells are enriched with tumorsphere-forming and tumor-initiating cells or cancer stem/progenitor cells with high levels of Notch/Bcl-2 and loss of miR-34. More significantly, miR-34 restoration led to an 87% reduction of the tumor-initiating cell population, accompanied by significant inhibition of tumorsphere growth in vitro and tumor-initiation in vivo. Our results demonstrate that miR-34 may restore, at least in part, the tumor suppressing function of the p53 in p53-deficient human pancreatic cancer. Our data support that miR-34 may be involved in cancer stem cell self-renewal, potentially via the direct modulation of downstream targets Bcl-2 and Notch, implying that miR-34 may play an important role in cancer stem cell self-renewal and/or cell fate determination. Currently, we are exploring the nanoparticle-targeted delivery of miR-34 to cancer stem cells as a novel molecular therapy for human pancreatic cancer, based on our patented nanobiotechnology which has been approved by FDA for clinical trials. Taken together, our results demonstrate that restoration of miR-34 may hold significant promise as a novel molecular therapy for human pancreatic/gastric cancer with loss of p53-miR34, potentially via inhibiting cancer stem cells. The project is funded by NIH grants CA121830, CA128220 and CA134655 (to L. Xu.).nnCitation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 2084.


Translational Andrology and Urology | 2014

AB109. Concurrent CD44s and STAT3 expression in human clear cell renal cellular carcinoma and its impact on survival

Jun Qin; Bo Yang; Weijun Qin; Guojun Wu; Chen Shao; Bao-Qin Xu; Jianlin Yuan; Ling Li

Although CD44 was overexpressed and considered as a useful prognostic marker in renal cell carcinoma, the prognostic role of CD44s in clear cell renal cell carcinoma (ccRCC) remains controversial. Moreover, the correlation and prognostic significance of CD44s and its downstream signaling target pSTAT3 are unclear in ccRCC. In this study, 75 pairs of carcinoma and paired adjacent non-tumor renal tissue samples were collected from patients with localized ccRCC who underwent a nephrectomy. The expression levels of CD44s and pSTAT3 were analyzed using immunohistochemistry. Correlations between CD44s/pSTAT3 expression and clinical and pathological characteristics were determined using χ test, Kaplan-Meier analysis and Cox’s proportional hazards model. We found that CD44s is highly expressed in 46.67% of tumor tissues, and its high expression was significantly associated with high tumor grade (P<0.001), large tumor size (P=0.009) and advanced T stage (P=0.004). A strong correlation exists between high expression of CD44s and pSTAT3 (r=0.4013, P=0.004). The joint over expression of CD44s and pSTAT3 was present in 42.66% of tumor specimens and had an additive negative impact on overall survival. Patients with CD44spSTAT3 expression had significantly poor survival as compared to patients with CD44spSTAT3 tumor expression (P=0.024), though the concurrent overexpression of CD44s and pSTAT3 was not an independent prognostic factor for overall survival. Our data indicate that expression of both CD44s and pSTAT3 in ccRCC is associated with advanced tumor stage and patient survival. The conclusions from this study may improve the prediction of ccRCC prognosis information when CD44s and pSTAT3 expression are evaluated together with classical clinicopathological parameters.


Molecular Cancer Therapeutics | 2009

Abstract A53: Tumor suppressor microRNA miR‐34 inhibits human pancreatic and gastric cancer stem cells

Liang Xu; Qing Ji; Xinbao Hao; Wenhua Tang; Ling Li; Jeffrey T. DeSano; Eric R. Fearon; Theodore S. Lawrence

MicroRNAs (miRNAs) have been implicated in cancer initiation and progression via their ability to affect expression of genes and proteins that regulate cell proliferation and/or cell death. Transcription of the three miRNA miR‐34 family members was recently found to be directly regulated by p53. Among the target proteins regulated by miR‐34 are Notch and Bcl‐2, suggesting the possibility of a role for miR‐34 in the maintenance and survival of cancer stem cells. We examined the roles of miR‐34 in p53‐mutant human pancreatic and gastric cancer cell lines and the potential link to cancer stem cells. Restoration of miR‐34 expression in the cancer cells by miR‐34 mimics or lentiviral miR‐34 downregulated Bcl‐2 and Notch1/2. miR‐34 restoration significantly inhibited clonogenic cell growth and invasion, induced apoptosis and G1 and G2/M arrest in cell cycle, and sensitized the cancer cells to chemotherapy and radiation. We identified that CD44+/CD133+ cancer cells are enriched with tumorsphere‐forming and tumor‐initiating cells or cancer stem/progenitor cells with high levels of Notch/Bcl‐2 and loss of miR‐34. More significantly, miR‐34 restoration led to a significant reduction of the tumor‐initiating cell population, accompanied by significant inhibition of tumorsphere growth in vitro and tumor formation in vivo. Our results demonstrate that miR‐34 may restore, at least in part, the tumor suppressing function of the p53 in p53‐deficient human pancreatic and gastric cancer cells. Our data support that miR‐34 may be involved in cancer stem cell self‐renewal, potentially via the direct modulation of downstream targets Bcl‐2 and Notch, implying that miR‐34 may play an important role in cancer stem cell self‐renewal and/or cell fate determination. Currently, we are exploring the nanoparticle‐targeted delivery of miR‐34 to cancer stem cells as a novel molecular therapy for human pancreatic/gastric cancer, based on our patented nanobiotechnology which has been approved by FDA for clinical trials. Taken together, our results demonstrate that restoration of miR‐34 may hold significant promise as a novel molecular therapy for human pancreatic/gastric cancer with loss of p53‐miR34, potentially via inhibiting cancer stem cells. The project is funded by NIH grants CA121830, CA128220 and CA134655 (to L. Xu.). Citation Information: Mol Cancer Ther 2009;8(12 Suppl):A53.


International Journal of Clinical and Experimental Pathology | 2014

Concurrent CD44s and STAT3 expression in human clear cell renal cellular carcinoma and its impact on survival.

Jun Qin; Bo Yang; Bao-Qin Xu; Amber Smithc; Liang Xu; Jianlin Yuan; Ling Li


International Journal of Clinical and Experimental Medicine | 2015

Cytoplasmic EpCAM over-expression is associated with favorable clinical outcomes in pancreatic cancer patients with Hepatitis B virus negative infection

Yao Meng; Bao-Qing Xu; Zhi-Guang Fu; Bo Wu; Bo Xu; Zhi-Nan Chen; Ling Li

Collaboration


Dive into the ng Li's collaboration.

Top Co-Authors

Avatar

Liang Xu

University of Kansas

View shared research outputs
Top Co-Authors

Avatar

Wenhua Tang

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xinbao Hao

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jianlin Yuan

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhi-Guang Fu

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhi-Nan Chen

Fourth Military Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge