Network


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

Hotspot


Dive into the research topics where Licun Wu is active.

Publication


Featured researches published by Licun Wu.


PLOS ONE | 2014

Metabolomic Heterogeneity of Pulmonary Arterial Hypertension

Yidan Zhao; Jenny Peng; Catherine Lu; Michael Hsin; Marco Mura; Licun Wu; Lei Chu; Ricardo Zamel; T.N. Machuca; Thomas K. Waddell; Mingyao Liu; Shaf Keshavjee; John Granton; Marc de Perrot

Although multiple gene and protein expression have been extensively profiled in human pulmonary arterial hypertension (PAH), the mechanism for the development and progression of pulmonary hypertension remains elusive. Analysis of the global metabolomic heterogeneity within the pulmonary vascular system leads to a better understanding of disease progression. Using a combination of high-throughput liquid-and-gas-chromatography-based mass spectrometry, we showed unbiased metabolomic profiles of disrupted glycolysis, increased TCA cycle, and fatty acid metabolites with altered oxidation pathways in the human PAH lung. The results suggest that PAH has specific metabolic pathways contributing to increased ATP synthesis for the vascular remodeling process in severe pulmonary hypertension. These identified metabolites may serve as potential biomarkers for the diagnosis of PAH. By profiling metabolomic alterations of the PAH lung, we reveal new pathogenic mechanisms of PAH, opening an avenue of exploration for therapeutics that target metabolic pathway alterations in the progression of PAH.


Molecular Cancer Therapeutics | 2012

CTLA-4 Blockade Expands Infiltrating T Cells and Inhibits Cancer Cell Repopulation during the Intervals of Chemotherapy in Murine Mesothelioma

Licun Wu; Z. Yun; Tetsuzo Tagawa; Katrina Rey-McIntyre; Marc de Perrot

Cancer immunotherapy has shown promising results when combined with chemotherapy. Blocking CTLA-4 signaling by monoclonal antibody between cycles of chemotherapy may inhibit cancer cell repopulation and enhance the antitumoral immune reaction, thus improve the efficacy of chemotherapy in mesothelioma. The impact of CTLA-4 blockade on the early stage of tumor development was evaluated in a subcutaneous murine mesothelioma model. CTLA-4 blocking antibody was administered following each cycle of chemotherapy, and monotherapy was included as controls. Antitumor effect was evaluated by tumor growth delay and survival of the animals. Tumor cell repopulation was quantified by bromodeoxyuridine incorporation and Ki67 by immunohistochemistry and/or flow cytometry. In vitro cell killing was determined by classic chromium-released assay, and reverse transcription PCR (RT-PCR) was carried out to determine the gene expression of associated cytokines. Anti-CTLA-4 monoclonal antibody was able to inhibit tumor growth at early stage of tumor development. Antitumor effect was achieved by administration of CTLA-4 blockade between cycles of chemotherapy. Tumor cell repopulation during the intervals of cisplatin was inhibited by CTLA-4 blockade. Anti-CTLA-4 therapy gave rise to an increased number of CD4 and CD8 T cells infiltrating the tumor. RT-PCR showed that the gene expression of interleukin IL-2, IFN-γ, granzyme B, and perforin increased in the tumor milieu. Blockade of CTLA-4 signaling showed effective anticancer effect, correlating with inhibiting cancer cell repopulation between cycles of chemotherapy and upregulating tumor-infiltrating T lymphocytes, cytokines, and cytolytic enzymes in a murine mesothelioma model. Mol Cancer Ther; 11(8); 1809–19. ©2012 AACR.


Journal of Immunology | 2010

Synergistic Antitumor Effects of Regulatory T Cell Blockade Combined with Pemetrexed in Murine Malignant Mesothelioma

Masaki Anraku; Tetsuzo Tagawa; Licun Wu; Zhihong Yun; Shaf Keshavjee; Li Zhang; Michael R. Johnston; Marc de Perrot

CD4+CD25+ regulatory T cells (Tregs) can promote the growth of some tumors, but it is unknown whether this is true for all tumors, including malignant pleural mesothelioma. We have previously shown that the existence of Tregs was associated with poor survival in patients with malignant pleural mesothelioma. In this study, using an intrathoracic murine model of malignant mesothelioma (MM), we provide evidence suggesting that Treg blockade could enhance survival when combined with pemetrexed in established tumor. AC29 murine MM cells were injected into the right pleural cavity of CBA mice for tumor development. Four days after the tumor injection, tumor-bearing mice were then treated with pemetrexed alone, Treg blockade alone, or a combination of pemetrexed and Treg blockade. We observed a synergistic antitumor effect of Treg blockade combined with pemetrexed resulting in prolonged survival. The combination of Treg blockade and pemetrexed was associated with decreased tumor-infiltrating Tregs, increased IL-2 production, dendritic cell maturation, and increased CD3+CD8+IFN-γ+ tumor-infiltrating T cells when compared with mice treated with pemetrexed alone or Treg blockade alone. The survival benefit was abrogated if anti-CD8 mAb was administered simultaneously. Likewise, the survival benefit resulting from the combined Treg blockade with pemetrexed was not observed when immunodeficient mice were used. Therefore, this study suggests that Treg blockade combined with pemetrexed can suppress mesothelioma growth in established tumor in vivo through an immune-mediated process. This study also validates a new intrathoracic tumor model of pleural effusion to explore the role of antitumor immunity in murine MM.


Journal of Thoracic Oncology | 2011

Tumor cell repopulation between cycles of chemotherapy is inhibited by regulatory T-cell depletion in a murine mesothelioma model

Licun Wu; Z. Yun; Tetsuzo Tagawa; Katrina Rey-McIntyre; Masaki Anraku; Marc de Perrot

Introduction: Malignant pleural mesothelioma is a highly aggressive cancer with poor prognosis. We have previously demonstrated that regulatory T cells (Treg) depletion can impact tumor microenvironment when combined with chemotherapy. The aim of this study is to analyze the impact of Treg depletion on tumor cell repopulation during cycles of chemotherapy in a murine mesothelioma model. Methods: Tumor-bearing mice were treated with chemotherapy once weekly to mimic clinical settings and with PC61 to cause Treg depletion after each cycle of chemotherapy. Tumor cell repopulation was evaluated by BrdU labeling index with immunohistochemistry and flow cytometry, and Ki67 gene expression was determined by real-time reverse-transcribed polymerase chain reaction. The proportion of CD4+ CD25+Foxp3+ Tregs, CD4+, and CD8+ T cells in the tumor, spleen, draining lymph node, and peripheral blood from tumor-bearing mice was determined by using flow cytometry, and gene expression of activated T-cell-related cytokines was quantified by enzyme-linked immunosorbent assay and reverse-transcribed polymerase chain reaction. Results: Tumor growth delay was achieved by cisplatin followed by PC61 or cyclophosphamide. The BrdU labeling index indicated that tumor cell repopulation between cycles of cisplatin was significantly inhibited by PC61. The CD4+CD25+Foxp3+ Tregs in tumor and lymphoid organs were almost completely depleted, whereas the CD4+ or CD8+ T cells did not change. PC61 after chemotherapy resulted in an increase of gene expression of interferon-&ggr;, granzyme B, perforin, and IP-10, thus leading to tumor cell lysis in cytotoxic lymphocyte assay. Nevertheless, cell killing induced by cyclophosphamide combined with cisplatin was due to cytotoxicity rather than specific immune response. Conclusion: Treg depletion between cycles of chemotherapy could improve the outcome of mesothelioma. Nevertheless, this effect seems limited, and more effective approaches need to be developed.


In Vitro Cellular & Developmental Biology – Animal | 2015

Establishment of immortalized murine mesothelial cells and a novel mesothelioma cell line

Walter Blum; László Pecze; Emanuela Felley-Bosco; Janine Worthmüller-Rodriguez; Licun Wu; Bart Vrugt; Marc de Perrot; Beat Schwaller

Mesothelial cells are susceptible to asbestos fiber-induced cytotoxicity and on longer time scales to transformation; the resulting mesothelioma is a highly aggressive neoplasm that is considered as incurable at the present time Zucali et al. (Cancer Treatment Reviews 37:543–558, 2011). Only few murine cell culture models of immortalized mesothelial cells and mesothelioma cell lines exist to date. We generated SV40-immortalized cell lines derived from wild-type (WT) and neurofibromatosis 2 (merlin) heterozygote (Nf2+/−) mice, both on a commonly used genetic background, C57Bl/6J. All immortalized mesothelial clones consistently grow in DMEM supplemented with fetal bovine serum. Cells can be passaged for more than 40 times without any signs of morphological changes or a decrease in proliferation rate. The tumor suppressor gene NF2 is one of the most frequently mutated genes in human mesothelioma, but its detailed function is still unknown. Thus, these genotypically distinct cell lines likely relevant for malignant mesothelioma formation are expected to serve as useful in vitro models, in particular to compare with in vivo studies in mice of the same genotype. Furthermore, we generated a novel murine mesothelioma cell line RN5 originating from an Nf2+/− mouse subjected to repeated crocidolite exposure. RN5 cells are highly tumorigenic.


PLOS ONE | 2015

A Biochemical Approach to Understand the Pathogenesis of Advanced Pulmonary Arterial Hypertension: Metabolomic Profiles of Arginine, Sphingosine-1-Phosphate, and Heme of Human Lung.

Yidan D. Zhao; Lei Chu; Kathleen Lin; Elise Granton; Li Yin; Jenny Peng; Michael Hsin; Licun Wu; Amy Yu; Thomas K. Waddell; Shaf Keshavjee; John Granton; Marc de Perrot

Pulmonary arterial hypertension (PAH) is a vascular disease characterized by persistent precapillary pulmonary hypertension (PH), leading to progressive right heart failure and premature death. The pathological mechanisms underlying this condition remain elusive. Analysis of global metabolomics from lung tissue of patients with PAH (n = 8) and control lung tissue (n = 8) leads to a better understanding of disease progression. Using a combination of high-throughput liquid-and-gas-chromatography-based mass spectrometry, we showed unbiased metabolomic profiles of disrupted arginine pathways with increased Nitric oxide (NO) and decreased arginine. Our results also showed specific metabolic pathways and genetic profiles with increased Sphingosine-1-phosphate (S1P) metabolites as well as increased Heme metabolites with altered oxidative pathways in the advanced stage of the human PAH lung. The results suggest that PAH has specific metabolic pathways contributing to the vascular remodeling in severe pulmonary hypertension. Profiling metabolomic alterations of the PAH lung has provided a new understanding of the pathogenic mechanisms of PAH, which benefits therapeutic targeting to specific metabolic pathways involved in the progression of PAH.


Metabolomics | 2014

De novo synthesize of bile acids in pulmonary arterial hypertension lung

Yidan D. Zhao; Hana Z. H. Yun; Jenny Peng; Li Yin; Lei Chu; Licun Wu; Ryan Michalek; Mingyao Liu; Shaf Keshavjee; Thomas K. Waddell; John Granton; Marc de Perrot

Although multiple, complex molecular studies have been done for understanding the development and progression of pulmonary hypertension (PAH), little is known about the metabolic heterogeneity of PAH. Using a combination of high-throughput liquid-and-gas-chromatography-based mass spectrometry, we found bile acid metabolites, which are normally product derivatives of the liver and gallbladder, were highly increased in the PAH lung. Microarray showed that the gene encoding cytochrome P450 7B1 (CYP7B1), an isozyme for bile acid synthesis, was highly expressed in the PAH lung compared with the control. CYP7B1 protein was found to be primarily localized on pulmonary vascular endothelial cells suggesting de novo bile acid synthesis may be involved in the development of PAH. Here, by profiling the metabolomic heterogeneity of the PAH lung, we reveal a newly discovered pathogenesis mechanism of PAH.


BMJ Open Respiratory Research | 2017

Metabolic heterogeneity of idiopathic pulmonary fibrosis: a metabolomic study

Yidan D. Zhao; Li Yin; Stephen L. Archer; Catherine Lu; George Zhao; Yan Yao; Licun Wu; Michael Hsin; Thomas K. Waddell; Shaf Keshavjee; John Granton; Marc de Perrot

Introduction Idiopathic pulmonary fibrosis (IPF) is a chronic and fatal disease of unknown cause characterised by progressive fibrotic formation in lung tissue. We hypothesise that disrupted metabolic pathways in IPF contribute to disease pathogenesis. Methods Metabolomics of human IPF was performed using mass spectroscopy (IPF lung=8; donor lung=8). Gene expression of key metabolic enzymes was measured using microarrays. Of the 108 metabolites whose levels were found altered, 48 were significantly increased, whereas 60 were significantly decreased in IPF samples compared with normal controls. Results Specific metabolic pathways mediating the IPF remodelling were found with a downregulated sphingolipid metabolic pathway but an upregulated arginine pathway in IPF. In addition, disrupted glycolysis, mitochondrial beta-oxidation and tricarboxylic acid cycle, altered bile acid, haem and glutamate/aspartate metabolism were found in IPF samples compared with control. Conclusions Our results show alterations in metabolic pathways for energy consumption during lung structural remodelling, which may contribute to IPF pathogenesis. We believe that this is the first report of simultaneously and systemically measuring changes of metabolites involving nine metabolic pathways in human severe IPF lungs. The measurement of the metabolites may serve in the future diagnosis and prognosis of IPF.


Lancet Oncology | 2017

Radiotherapy for the treatment of malignant pleural mesothelioma

Marc de Perrot; Licun Wu; Matthew Wu; B.C. John Cho

Malignant pleural mesothelioma is an aggressive disease that continues to be associated with poor outcomes. Although, traditionally this disease is considered to be resistant to radiotherapy, more recent evidence suggests that radiotherapy can produce positive outcomes. Over the past 15 years, the development of new, highly conformal radiotherapy techniques, such as intensity-modulated radiation therapy (IMRT), has enabled investigators to optimise the delivery of high-dose radiotherapy to the whole of the hemithorax. Prospective single-arm trials have shown that the median survival of patients who have completed high-dose hemithoracic radiotherapy after extrapleural pneumonectomy could reach 23·9-39·4 months independent of the chemotherapeutic response, suggesting that IMRT could potentially have an intrinsic benefit to this subset of patients. These observations have led to a change in practice, with the introduction of adjuvant pleural IMRT after pleurectomy-decortication and the development of induction-accelerated hemithoracic IMRT followed by extrapleural pneumonectomy. This Review focuses on recent observations on the role of radiotherapy in the treatment of malignant pleural mesothelioma, with particular emphasis on the results of clinical trials that evaluate the role of high-dose hemithoracic radiotherapy.


Clinical Cancer Research | 2017

Patient-Derived Xenograft Establishment from Human Malignant Pleural Mesothelioma.

Licun Wu; Ghassan Allo; Thomas John; Ming Li; Tetsuzo Tagawa; Isabelle Opitz; Masaki Anraku; Z. Yun; Melania Pintilie; Bethany Pitcher; Geoffrey Liu; Ron Feld; Michael R. Johnston; Marc de Perrot; Ming-Sound Tsao

Purpose: Malignant pleural mesothelioma (MPM) is a rare but aggressive disease with few therapeutic options. The tumor–stromal interface is important in MPM, but this is lost in cell lines, the main model used for preclinical studies. We sought to characterize MPM patient-derived xenografts (PDX) to determine their suitability as preclinical models and whether tumors that engraft reflect a more aggressive biological phenotype. Experimental Design: Fresh tumors were harvested from extrapleural pneumonectomy, decortication, or biopsy samples of 50 MPM patients and implanted subcutaneously into immunodeficient mice and serially passaged for up to five generations. We correlated selected mesothelioma biomarkers between PDX and patient tumors, and PDX establishment with the clinical pathologic features of the patients, including their survival. DNA of nine PDXs was profiled using the OncoScan FFPE Express platform. Ten PDXs were treated with cisplatin and pemetrexed. Results: A PDX was formed in 20 of 50 (40%) tumors implanted. Histologically, PDX models closely resembled the parent tumor. PDX models formed despite preoperative chemotherapy and radiotherapy. In multivariable analysis, patients whose tumors formed a PDX had significantly poorer survival when the model was adjusted for preoperative treatment (HR, 2.46; 95% confidence interval, 1.1–5.52; P = 0.028). Among 10 models treated with cisplatin, seven demonstrated growth inhibition. Genomic abnormalities seen in nine PDX models were similar to that previously reported. Conclusions: Patients whose tumors form PDX models have poorer clinical outcomes. MPM PDX tumors closely resemble the genotype and phenotype of parent tumors, making them valuable models for preclinical studies. Clin Cancer Res; 23(4); 1060–7. ©2016 AACR.

Collaboration


Dive into the Licun Wu's collaboration.

Top Co-Authors

Avatar

Marc de Perrot

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Y. Zhao

University of Toronto

View shared research outputs
Top Co-Authors

Avatar

Z. Yun

University Health Network

View shared research outputs
Top Co-Authors

Avatar

Shaf Keshavjee

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jenny Peng

University Health Network

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge