Network


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

Hotspot


Dive into the research topics where Nanping Wu is active.

Publication


Featured researches published by Nanping Wu.


Clinical Cancer Research | 2012

Integrative Survival-Based Molecular Profiling of Human Pancreatic Cancer

Timothy R. Donahue; Linh M. Tran; Reginald Hill; Yunfeng Li; Anne N. Kovochich; Joseph Hargan Calvopina; Sanjeet Patel; Nanping Wu; Antreas Hindoyan; James J. Farrell; Xinmin Li; David W. Dawson; Hong Wu

Purpose: To carry out an integrative profile of human pancreatic ductal adenocarcinoma (PDAC) to identify prognosis-significant genes and their related pathways. Experimental Design: A concordant survival-based whole genome in silico array analysis of DNA copy number, and mRNA and miRNA expression in 25 early-stage PDAC was carried out. A novel composite score simultaneously integrated gene expression with regulatory mechanisms to identify the signature genes with the most levels of prognosis-significant evidence. The predominant signaling pathways were determined via a pathway-based approach. Independent patient cohorts (n = 148 and 42) were then used as in vitro validation of the array findings. Results: The composite score identified 171 genes in which expressions were able to define two prognosis subgroups (P = 3.8e-5). Eighty-eight percent (151 of 171) of the genes were regulated by prognosis-significant miRNAs. The phosphoinositide 3-kinase/AKT pathway and SRC signaling were densely populated by prognosis-significant genes and driven by genomic amplification of SRC and miRNA regulation of p85α and CBL. On tissue microarray validation (n = 148), p85α protein expression was associated with improved survival for all patients (P = 0.02), and activated P-SRC (Y418) was associated shorter survival for patients with low-grade histology tumors (P = 0.04). Interacting P-SRC and p85α revealed that they define two distinct PDAC patient subgroups (P = 0.0066). Furthering the importance of these pathways, CBL protein expression was associated with improved survival (P = 0.03) on a separate cohort (n = 42). Conclusions: These pathways and related genes may represent putative clinical biomarkers and possible targets of individualized therapy in the distinct patient subgroups they define. Clin Cancer Res; 18(5); 1352–63. ©2012 AACR.


The American Journal of Gastroenterology | 2013

Endoscopically Acquired Pancreatic Cyst Fluid MicroRNA 21 and 221 Are Associated With Invasive Cancer

James J. Farrell; Paul A. Toste; Nanping Wu; Luyi Li; Jonathan L. Wong; Linh M. Tran; Xiaoyang Wu; Xinmin Li; David W. Dawson; Hong Wu; Timothy R. Donahue

OBJECTIVES:Pancreatic cysts are a group of lesions with heterogeneous malignant potential. Currently, there are no reliable biomarkers to aid in cyst diagnosis and classification. The objective of this study was to identify potential microRNA (miR) biomarkers in endoscopically acquired pancreatic cyst fluid that could be used to distinguish between benign, premalignant, and malignant cysts.METHODS:A list of candidate miRs was developed using a whole-genome expression array analysis of pancreatic cancer (pancreatic ductal adenocarcinoma) and nonmalignant samples overlapped with existing literature and predicted gene targets. Endoscopically acquired pancreatic cyst fluid samples were obtained from a group of 38 patients who underwent cyst fluid aspiration and surgical resection. Selected miR expression levels in cyst fluid samples were assessed by quantitative real-time-PCR. Additionally, in situ hybridization (ISH) on corresponding cyst tissue samples was performed to identify the source and validate the expression level of fluid miRs.RESULTS:Of the six miRs that were profiled in the study, two showed differential expression in malignant cysts. miR-221 was expressed at significantly higher levels in malignant cysts compared with benign or premalignant cysts (P=0.05). miR-21 was also expressed at significantly higher levels in malignant cysts (P<0.01). Additionally, the expression of miR-21 was significantly higher in premalignant cysts than benign cysts (P=0.03). The differential expression of miR-21 among cyst categories was confirmed by ISH.CONCLUSIONS:In this small single-center study, miRs are potential pancreatic cyst fluid diagnostic biomarkers. In particular, miR-21 is identified as a candidate biomarker to distinguish between benign, premalignant, and malignant cysts. Additionally miR-221 may be of use in the identification of more advanced malignant disease.


Molecular Cancer Therapeutics | 2014

The CREB binding protein inhibitor ICG-001 suppresses pancreatic cancer growth

Michael D. Arensman; Donatello Telesca; Anna R. Lay; Kathleen M. Kershaw; Nanping Wu; Timothy R. Donahue; David W. Dawson

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer due in part to a lack of highly robust cytotoxic or molecular-based therapies. Recent studies investigating ligand-mediated Wnt/β-catenin signaling have highlighted its importance in pancreatic cancer initiation and progression, as well as its potential as a therapeutic target in PDAC. The small-molecule ICG-001 binds cAMP-responsive element binding (CREB)-binding protein (CBP) to disrupt its interaction with β-catenin and inhibit CBP function as a coactivator of Wnt/β-catenin–mediated transcription. Given its ability to inhibit Wnt/β-catenin–mediated transcription in vitro and in vivo, as well as its efficacy in preclinical models of colorectal cancer and other Wnt-driven diseases, we examined ICG-001 and its potential role as a therapeutic in PDAC. ICG-001 alone significantly inhibited anchorage-dependent and -independent growth of multiple PDAC lines, and augmented in vitro growth inhibition when used in combination with gemcitabine. ICG-001 had only variable modest effects on PDAC apoptosis and instead mediated PDAC growth inhibition primarily through robust induction of G1 cell-cycle arrest. These effects, however, seemed decoupled from its inhibition of Wnt/β-catenin–mediated transcription. DNA microarrays performed on PDAC cells in the context of ICG-001 treatment revealed ICG-001 altered the expression of several genes with well-established roles in DNA replication and cell-cycle progression, including direct actions on SKP2 and CDKN1A. ICG-001 also significantly prolonged survival in an in vivo orthotopic xenograft model of PDAC, indicating ICG-001 or derived compounds that disrupt CBP activity are potentially useful small-molecule therapeutics for pancreatic cancer. Mol Cancer Ther; 13(10); 2303–14. ©2014 AACR.


Journal of Clinical Investigation | 2017

Tumor-penetrating peptide enhances transcytosis of silicasome-based chemotherapy for pancreatic cancer

Xiangsheng Liu; Paulina Lin; Ian Perrett; Joshua Lin; Yu-Pei Liao; Chong Hyun Chang; Jinhong Jiang; Nanping Wu; Timothy R. Donahue; Zev A. Wainberg; Andre E. Nel; Huan Meng

Pancreatic ductal adenocarcinoma (PDAC) is almost uniformly fatal; however, some improvement in overall survival has been achieved with the introduction of nanocarriers that deliver irinotecan or paclitaxel. Although it is generally assumed that nanocarriers rely principally on abnormal leaky vasculature for tumor access, a transcytosis transport pathway that is regulated by neuropilin-1 (NRP-1) has recently been reported. NRP-1–mediated transport can be triggered by the cyclic tumor-penetrating peptide iRGD. In a KRAS-induced orthotopic PDAC model, coadministration of iRGD enhanced the uptake of an irinotecan-loaded silicasome carrier that comprises lipid bilayer–coated mesoporous silica nanoparticles (MSNPs); this uptake resulted in enhanced survival and markedly reduced metastasis. Further, ultrastructural imaging of the treated tumors revealed that iRGD coadministration induced a vesicular transport pathway that carried Au-labeled silicacomes from the blood vessel lumen to a perinuclear site within cancer cells. iRGD-mediated enhancement of silicasome uptake was also observed in patient-derived xenografts, commensurate with the level of NRP-1 expression on tumor blood vessels. These results demonstrate that iRGD enhances the efficacy of irinotecan-loaded silicasome–based therapy and may be a suitable adjuvant in nanoparticle-based treatments for PDAC.


Journal of Surgical Research | 2015

p85α is a microRNA target and affects chemosensitivity in pancreatic cancer.

Paul A. Toste; Luyi Li; Brian E. Kadera; Andrew H. Nguyen; Linh M. Tran; Nanping Wu; David L. Madnick; Sanjeet Patel; David W. Dawson; Timothy R. Donahue

BACKGROUND We previously identified a correlation between increased expression of the phosphoinositide 3-kinase (PI3K) regulatory subunit p85α and improved survival in human pancreatic ductal adenocarcinoma (PDAC). The purpose of this study was to investigate the impact of changes in p85α expression on response to chemotherapy and the regulation of p85α by microRNA-21 (miR-21). MATERIALS AND METHODS PDAC tumor cells overexpressing p85α were generated by viral transduction, and the effect of p85α overexpression on sensitivity to gemcitabine was tested by MTT assay. Primary human PDAC tumors were stained for p85α and miR-21 via immunohistochemistry and in situ hybridization, respectively. Additionally, PDAC cells were treated with miR-21 mimic, and changes in p85α and phospho-AKT were assessed by Western blot. Finally, a luciferase reporter assay system was used to test direct regulation of p85α by miR-21. RESULTS Higher p85α expression resulted in increased sensitivity to gemcitabine (P < 0.01), which correlated with decreased PI3K-AKT activation. Human tumors demonstrated an inverse correlation between miR-21 and p85α expression levels (r = -0.353, P < 0.001). In vitro, overexpression of miR-21 resulted in decreased levels of p85α and increased phosphorylation of AKT. Luciferase reporter assays confirmed the direct regulation of p85α by miR-21 (P < 0.01). CONCLUSIONS Our results demonstrate that p85α expression is a determinant of chemosensitivity in PDAC. Additionally, we provide novel evidence that miR-21 can influence PI3K-AKT signaling via its direct regulation of p85α. These data provide insight into potential mechanisms for the known relationship between increased p85α expression and improved survival in PDAC.


Molecular Cancer Research | 2016

Chemotherapy-Induced Inflammatory Gene Signature and Protumorigenic Phenotype in Pancreatic CAFs via Stress-Associated MAPK.

Paul A. Toste; Andrew H. Nguyen; Brian E. Kadera; Mindy Duong; Nanping Wu; Irmina Gawlas; Linh M. Tran; Mihir Bikhchandani; Luyi Li; Sanjeet Patel; David W. Dawson; Timothy R. Donahue

Pancreatic ductal adenocarcinoma (PDAC) has a characteristically dense stroma comprised predominantly of cancer-associated fibroblasts (CAF). CAFs promote tumor growth, metastasis, and treatment resistance. This study aimed to investigate the molecular changes and functional consequences associated with chemotherapy treatment of PDAC CAFs. Chemoresistant immortalized CAFs (R-CAF) were generated by continuous incubation in gemcitabine. Gene expression differences between treatment-naïve CAFs (N-CAF) and R-CAFs were compared by array analysis. Functionally, tumor cells (TC) were exposed to N-CAF– or R-CAF–conditioned media and assayed for migration, invasion, and viability in vitro. Furthermore, a coinjection (TC and CAF) model was used to compare tumor growth in vivo. R-CAFs increased TC viability, migration, and invasion compared with N-CAFs. In vivo, TCs coinjected with R-CAFs grew larger than those accompanied by N-CAFs. Genomic analysis demonstrated that R-CAFs had increased expression of various inflammatory mediators, similar to the previously described senescence-associated secretory phenotype (SASP). In addition, SASP mediators were found to be upregulated in response to short duration treatment with gemcitabine in both immortalized and primary CAFs. Inhibition of stress-associated MAPK signaling (P38 MAPK or JNK) attenuated SASP induction as well as the tumor-supportive functions of chemotherapy-treated CAFs in vitro and in vivo. These results identify a negative consequence of chemotherapy on the PDAC microenvironment that could be targeted to improve the efficacy of current therapeutic regimens. Implications: Chemotherapy treatment of pancreatic cancer–associated fibroblasts results in a proinflammatory response driven by stress-associated MAPK signaling that enhances tumor cell growth and invasiveness. Mol Cancer Res; 14(5); 437–47. ©2016 AACR.


Integrative Biology | 2016

Stiffness of pancreatic cancer cells is associated with increased invasive potential

Angelyn V. Nguyen; Kendra D. Nyberg; Michael B. Scott; Alia M. Welsh; Andrew H. Nguyen; Nanping Wu; Sophia V. Hohlbauch; Nicholas A. Geisse; Ewan A. Gibb; A. Gordon Robertson; Timothy R. Donahue; Amy C. Rowat

Metastasis is a fundamentally physical process in which cells are required to deform through narrow gaps as they invade surrounding tissues and transit to distant sites. In many cancers, more invasive cells are more deformable than less invasive cells, but the extent to which mechanical phenotype, or mechanotype, can predict disease aggressiveness in pancreatic ductal adenocarcinoma (PDAC) remains unclear. Here we investigate the invasive potential and mechanical properties of immortalized PDAC cell lines derived from primary tumors and a secondary metastatic site, as well as noncancerous pancreatic ductal cells. To investigate how invasive behavior is associated with cell mechanotype, we flow cells through micron-scale pores using parallel microfiltration and microfluidic deformability cytometry; these results show that the ability of PDAC cells to passively transit through pores is only weakly correlated with their invasive potential. We also measure the Youngs modulus of pancreatic ductal cells using atomic force microscopy, which reveals that there is a strong association between cell stiffness and invasive potential in PDAC cells. To determine the molecular origins of the variability in mechanotype across our PDAC cell lines, we analyze RNAseq data for genes that are known to regulate cell mechanotype. Our results show that vimentin, actin, and lamin A are among the most differentially expressed mechanoregulating genes across our panel of PDAC cell lines, as well as a cohort of 38 additional PDAC cell lines. We confirm levels of these proteins across our cell panel using immunoblotting, and find that levels of lamin A increase with both invasive potential and Youngs modulus. Taken together, we find that stiffer PDAC cells are more invasive than more compliant cells, which challenges the paradigm that decreased cell stiffness is a hallmark of metastatic potential.


Clinical Cancer Research | 2015

Low expression of the E3 ubiquitin ligase CBL confers chemoresistance in human pancreatic cancer and is targeted by epidermal growth factor receptor inhibition.

Brian E. Kadera; Paul A. Toste; Nanping Wu; Luyi Li; Andrew H. Nguyen; David W. Dawson; Timothy R. Donahue

Purpose: Expression of CBL, an ubiquitin ligase, is decreased in 60% of human pancreatic ductal adenocarcinomas (PDAC) and is associated with shorter overall survival. We sought to determine how low CBL directly contributes to clinically more aggressive PDAC. Experimental Design: Human PDACs were stained for CBL, pEGFR, and EGFR. CBL-low was modeled in PDAC cells (Panc-1, L3.6pl, and AsPC-1) via transient transfection (siRNA) or stable knockdown (shRNA). Cell viability and apoptosis were measured by MTT assays and FACS. Immunoblot and a phospho-receptor tyrosine kinase (pRTK) array were used to probe signal transduction. NOD-scid-IL2Rγnull mice were subcutaneously implanted with PDAC or PDACCBL-low cells on opposite flanks and treated with gemcitabine ± erlotinib for ≥4 weeks. Results: There was an inverse correlation between CBL and pEGFR protein expression in 12 of 15 tumors. CBL knockdown increased PDAC resistance to gemcitabine and 5-fluorouracil (5-FU) by upregulating pEGFR (Y1068), pERK, and pAKT. A pRTK array of PDACCBL-low cells revealed additional activated tyrosine kinases but all to a much lower magnitude than EGFR. Increased chemoresistance from low CBL was abrogated by the EGFR inhibitor erlotinib both in vitro and in vivo. Erlotinib+gemcitabine–treated PDACCBL-low cells exhibited greater apoptosis by cleaved PARP, caspase-3, and Annexin V/PI. Conclusions: Low CBL causes chemoresistance in PDAC via stress-induced EGFR activation that can be effectively abrogated by EGFR inhibition. These results suggest that dysregulation of ubiquitination is a key mechanism of EGFR hyperactivation in PDAC and that low CBL may define PDAC tumors likely to respond to erlotinib treatment. Clin Cancer Res; 21(1); 157–65. ©2014 AACR.


Oncotarget | 2017

Histone deacetylase inhibitors provoke a tumor supportive phenotype in pancreatic cancer associated fibroblasts

Andrew H. Nguyen; Irmina A. Elliott; Nanping Wu; Cynthia Matsumura; Maria Vogelauer; Narsis Attar; Amanda M. Dann; Razmik Ghukasyan; Paul A. Toste; Sanjeet Patel; Jennifer L. Williams; Luyi Li; David W. Dawson; Caius G. Radu; Siavash K. Kurdistani; Timothy R. Donahue

Although histone deacetylase inhibitors (HDACi) are a promising class of anti-cancer drugs, thus far, they have been unsuccessful in early phase clinical trials for pancreatic ductal adenocarcinoma (PDAC). One potential reason for their poor efficacy is the tumor stroma, where cancer-associated fibroblasts (CAFs) are a prominent cell type and a source of resistance to cancer therapies. Here, we demonstrate that stromal fibroblasts contribute to the poor efficacy of HDACis in PDAC. HDACi-treated fibroblasts show increased biological aggressiveness and are characterized by increased secretion of pro-inflammatory tumor-supportive cytokines and chemokines. We find that HDAC2 binds to the enhancer and promoter regions of pro-inflammatory genes specifically in CAFs and in silico analysis identified AP-1 to be the most frequently associated transcription factor bound in these regions. Pharmacologic inhibition of pathways upstream of AP-1 suppresses the HDACi-induced inflammatory gene expression and tumor-supportive responses in fibroblasts. Our findings demonstrate that the combination of HDACis with chemical inhibitors of the AP-1 signaling pathway attenuate the inflammatory phenotype of fibroblasts and may improve the efficacy of HDACi in PDAC and, potentially, in other solid tumors rich in stroma.


Cancer Research | 2015

Abstract B18: MicroRNA-21 is involved in pancreatic cancer fibroblast tumor supportiveness

Andrew H. Nguyen; Luyi Li; Paul A. Toste; Nanping Wu; Timothy R. Donahue

Background: Pancreatic ductal adenocarcinoma (PDAC) is characterized by poor survival despite current medical and surgical therapies and has a propensity towards subclinical metastasis even with resectable disease. The tumor microenvironment consists of a network of supporting cells, including tumor-associated fibroblasts (TAFs), which promote tumor invasion and metastasis, and contribute to PDAC’s extremely poor prognosis. MicroRNAs (miRs) have recently emerged as critical players in post-transcriptional regulation of pathways in cancer biology. In this study, we identified miRs in fibroblasts induced by PDAC tumor cells (TCs). Further, we study the stable suppression of one of these miRs in TAFs and the effects upon fibroblast migration and invasion, as well as tumor cell proliferation. Methods: Primary pancreatic fibroblasts were cultured with MIA PaCa-2 TC conditioned media and RNA was analyzed by the NanoString MicroRNA Expression Assay. Stable miR knockdown (KD) was achieved with a stably expressed shRNA anti-microRNA by lentiviral vector. Migration was determined by an 18 hour scratch assay and 24 hour invasion by modified Boyden chamber. TC proliferation was measured by MTT assay. In vivo tumor growth was evaluated by subcutaneous TC and TAF co-implantation in NSG mice. Results: In primary pancreatic fibroblasts, MIA PaCa-2 tumor cells induced 62 miRs >50% increase or 30% decrease in expression. The most prevalent induced miR with 2-fold increase in expression was miR-21. miR-21 KD in TAFs was achieved by introducing stable expression of anti-miR-21, which resulted in a 75% decrease in expression (P 50% decreased ability to migrate (p Conclusions: Pancreatic cancer TCs can induce tumor supportive miRs in TAFs. miR-21 KD in PDAC TAFs results in decreased TAF migration and invasion, as well as reduced ability to support in vitro and in vivo tumor growth. Citation Format: Andrew Hieu Nguyen, Luyi Li, Paul Toste, Nanping Wu, Timothy R. Donahue. MicroRNA-21 is involved in pancreatic cancer fibroblast tumor supportiveness. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Innovations in Research and Treatment; May 18-21, 2014; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2015;75(13 Suppl):Abstract nr B18.

Collaboration


Dive into the Nanping Wu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luyi Li

University of California

View shared research outputs
Top Co-Authors

Avatar

Paul A. Toste

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Linh M. Tran

University of California

View shared research outputs
Top Co-Authors

Avatar

Sanjeet Patel

University of California

View shared research outputs
Top Co-Authors

Avatar

Hong Wu

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge