Network


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

Hotspot


Dive into the research topics where Chao-Chi Ho is active.

Publication


Featured researches published by Chao-Chi Ho.


American Journal of Pathology | 2002

Up-regulated caveolin-1 accentuates the metastasis capability of lung adenocarcinoma by inducing filopodia formation

Chao-Chi Ho; Pei-Hsin Huang; Hsin-Yi Huang; Yen-Ho Chen; Pan-Chyr Yang; Su-Ming Hsu

Caveolin-1, a 21- to 24-kd integral membrane protein, is primarily implicated as a tumor suppressor gene. Transformed cells normally contain reduced or no caveolin-1. Re-expression of caveolin-1 is found in advanced human and mouse prostate adenocarcinomas. To explore its potential role in tumorigenesis and tumor progression of human lung cancers, we used the well-characterized cell line (CL) series of lung adenocarcinoma cells with increasing cellular invasiveness to show that expression of caveolin-1 mRNA and protein was up-regulated with enhanced invasion/metastatic capability of CL cells. Reintroducing the caveolin-1 gene into the less invasive, caveolin-1-negative CL cells enhanced their invasive capability at least by twofold, as revealed by an in vitro chamber invasion assay. Thus, a correlation exists for both constitutive and induced expression of caveolin-1 in CL cells. Immunohistochemical examination of caveolin-1 was performed in 95 specimens obtained retrospectively from patients who had lung adenocarcinoma either with (35 patients) or without (60 patients) ipsilateral hilar/peribronchial tumor-metastasized lymph nodes. Caveolin-1 immunoreactivity was either totally absent or just barely detectable in a few lung adenocarcinoma cells from cases diagnosed as lung adenocarcinoma without regional lymph node metastasis. In contrast, increased caveolin-1 immunoreactivity both in number and intensity was detected in primary lung adenocarcinoma cells as well as in cancer cells that metastasized to regional lymph nodes from the cases diagnosed as advanced lung adenocarcinoma with nodal metastases. Multivariate analysis considering caveolin-1 immunoreactivity in addition to the established prognostic parameters such as pT stage, pN in these patients confirmed that caveolin-1 is an independent functional predictor of poor survival. We further revealed that up-regulated caveolin-1 in CL cells is necessary for mediating filopodia formation, which may enhance the invasive ability of lung adenocarcinoma cells.


Cancer Research | 2008

Curcumin inhibits lung cancer cell invasion and metastasis through the tumor suppressor HLJ1.

Huei-Wen Chen; Jen-Yi Lee; Ji-Ying Huang; Chi-Chung Wang; Wan-Jiun Chen; Sheng-Fang Su; Chia-Wen Huang; Chao-Chi Ho; Jeremy J.W. Chen; Meng-Feng Tsai; Sung-Liang Yu; Pan-Chyr Yang

Curcumin (diferuloylmethane) is an active component of the spice turmeric and has a diversity of antitumor activities. In this study, we found that curcumin can inhibit cancer cell invasion and metastasis through activation of the tumor suppressor DnaJ-like heat shock protein 40 (HLJ1). Human lung adenocarcinoma cells (CL1-5) treated with curcumin (1-20 mumol/L) showed a concentration-dependent reduction in cell migration, invasion, and metastatic ability, and this was associated with increased HLJ1 expression. Knockdown of HLJ1 expression by siRNA was able to reverse the curcumin-induced anti-invasive and antimetastasis effects in vitro and in vivo. The HLJ1 promoter and enhancer in a luciferase reporter assay revealed that curcumin transcriptionally up-regulates HLJ1 expression through an activator protein (AP-1) site within the HLJ1 enhancer. JunD, one of the AP-1 components, was significantly up-regulated by curcumin (1-20 mumol/L) in a concentration- and time-dependent manner. Knockdown of JunD expression could partially reduce the curcumin-induced HLJ1 activation and diminish the anti-invasive effect of curcumin, indicating that JunD would seem to be involved in curcumin-induced HLJ1 expression. Curcumin was able to induce c-Jun NH(2)-kinase (JNK) phosphorylation, whereas the JNK inhibitor (SP-600125) could attenuate curcumin-induced JunD and HLJ1 expression. Activation of HLJ1 by curcumin further leads to up-regulation of E-cadherin and a suppression of cancer cell invasion. Our results show that curcumin induces HLJ1, through activation of the JNK/JunD pathway, and inhibits lung cancer cell invasion and metastasis by modulating E-cadherin expression. This is a novel mechanism and supports the application of curcumin in anti-cancer metastasis therapy.


Nature Communications | 2014

Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling

Wan-Jiun Chen; Chao-Chi Ho; Yih-Leong Chang; Hsuan-Yu Chen; Chih-An Lin; Thai-Yen Ling; Sung-Liang Yu; Shinsheng Yuan; Yu-Ju Louisa Chen; Chien-Yu Lin; Szu-Hua Pan; Han-Yi Elizabeth Chou; Yu-Ju Chen; Gee-Chen Chang; Wen-Cheng Chu; Yee-Ming Lee; Jen-Yi Lee; Pei-Jung Lee; Ker-Chau Li; Huei-Wen Chen; Pan-Chyr Yang

Cancer stem cells (CSCs) are a promising target for treating cancer, yet how CSC plasticity is maintained in vivo is unclear and is difficult to study in vitro. Here we establish a sustainable primary culture of Oct3/4(+)/Nanog(+) lung CSCs fed with CD90(+) cancer-associated fibroblasts (CAFs) to further advance our knowledge of preserving stem cells in the tumour microenvironment. Using transcriptomics we identify the paracrine network by which CAFs enrich CSCs through de-differentiation and reacquisition of stem cell-like properties. Specifically, we find that IGF1R signalling activation in cancer cells in the presence of CAFs expressing IGF-II can induce Nanog expression and promote stemness. Moreover, this paracrine signalling predicts overall and relapse-free survival in stage I non-small cell lung cancer (NSCLC) patients. IGF-II/IGF1R signalling blockade inhibits Nanog expression and attenuates cancer stem cell features. Our data demonstrate that CAFs constitute a supporting niche for cancer stemness, and targeting this paracrine signalling may present a new therapeutic strategy for NSCLC.


American Journal of Respiratory and Critical Care Medicine | 2012

Trifluoperazine, an Antipsychotic Agent, Inhibits Cancer Stem Cell Growth and Overcomes Drug Resistance of Lung Cancer

Chi-Tai Yeh; Alexander T H Wu; Peter Mu-Hsin Chang; Kuan-Yu Chen; Chia Ning Yang; Shuenn Chen Yang; Chao-Chi Ho; Chun Chi Chen; Yu Lun Kuo; Pei Ying Lee; Yu-Wen Liu; Chueh Chuan Yen; Michael Hsiao; Pei Jung Lu; Jin Mei Lai; Liang Shun Wang; Chih Hsiung Wu; Jeng Fong Chiou; Pan-Chyr Yang; Chi-Ying F. Huang

RATIONALE Cancer stem cell (CSC) theory has drawn much attention, with evidence supporting the contribution of stem cells to tumor initiation, relapse, and therapy resistance. OBJECTIVES To screen drugs that target CSCs to improve the current treatment outcome and overcome drug resistance in patients with lung cancer. METHODS We used publicly available embryonic stem cell and CSC-associated gene signatures to query the Connectivity Map for potential drugs that can, at least in part, reverse the gene expression profile of CSCs. High scores were noted for several phenothiazine-like antipsychotic drugs, including trifluoperazine. We then treated lung CSCs with different EGFR mutation status with trifluoperazine to examine its anti-CSC properties. Lung CSCs resistant to epidermal growth factor receptor-tyrosine kinase inhibitor or cisplatin were treated with trifluoperazine plus gefitinib or trifluoperazine plus cisplatin. Animal models were used for in vivo validation of the anti-CSC effect and synergistic effect of trifluoperazine with gefitinib. MEASUREMENTS AND MAIN RESULTS We demonstrated that trifluoperazine inhibited CSC tumor spheroid formation and down-regulated the expression of CSC markers (CD44/CD133). Trifluoperazine inhibited Wnt/β-catenin signaling in gefitinib-resistant lung cancer spheroids. The combination of trifluoperazine with either gefitinib or cisplatin overcame drug resistance in lung CSCs. Trifluoperazine inhibited the tumor growth and enhanced the inhibitory activity of gefitinib in lung cancer metastatic and orthotopic CSC animal models. CONCLUSIONS Using in silico drug screening by Connectivity Map followed by empirical validations, we repurposed an existing phenothiazine-like antipsychotic drug, trifluoperazine, as a potential anti-CSC agent that could overcome epidermal growth factor receptor-tyrosine kinase inhibitor and chemotherapy resistance.


Journal of Thoracic Oncology | 2010

Good Response to Gefitinib in Lung Adenocarcinoma Harboring Coexisting EML4-ALK Fusion Gene and EGFR Mutation

Yao-Wen Kuo; Shang-Gin Wu; Chao-Chi Ho; Jin-Yuan Shih

A 72-year-old female nonsmoker presented with dry cough and progressive exertional dyspnea for 4 months. Chest radiography disclosed multiple nodular opacities in bilateral lung fields and right pleural effusion. A computed tomography (CT) of the chest after the drainage of pleural effusion (Figure 1A) showed an irregular mass at right upper lung. Serum carcinoembryonic antigen (CEA) level was 442.1 ng/ml. The cytology of echoguided lung aspiration and right pleural effusion both yielded adenocarcinoma. The bronchial biopsy specimens showed neoplastic glands with immunoreactivity to thyroid transcription factor-1 protein and cytokeratin 7. Stage IV lung adenocarcinoma with metastasis to brain and bone was diagnosed. A repeated chest CT (Figure 1B) 92 days after gefitinib treatment showed dramatic shrinkage of right upper lobe mass. The CEA declined to 87.6 ng/ml 196 days after gefitinib treatment. The RNA extracted from cancer cells of echo-guided lung aspiration at the diagnosis of lung cancer was assessed for epidermal growth factor receptor (EGFR) mutation and echinoderm microtubule-associated protein-like 4 (EML4)-anaplastic lymphoma kinase (ALK) fusion by reverse transcriptionpolymerase chain reaction and sequencing assays as previously described.1,2 Besides a variant 1 of EML4-ALK translocation, a mutant form of EGFR del 2235–2249 (del E746-A750) in exon 19 was identified (Figure 2). After 232 days of gefitinib treatment, the patient showed disease progression. The trend of lung cancer treatment has been toward personalized therapy. Identifying molecular genetic marker to guide treatment has been successfully demonstrated in the treatment of lung adenocarcinoma in East Asia. Patients with the characteristics of being female, nonsmoker or former light smoker, previously untreated, and advanced stage, and especially the presence of mutation of the EGFR gene, benefited much from EGFR tyrosine kinase inhibitor (TKI) compared with conventional cytotoxic chemotherapy.3 Recently, the EML4-ALK, a fusion gene first described in 2007, possesses oncogenic activity that can be blocked by ALK kinase inhibitor and represents a new therapeutic target in non-small cell lung cancer (NSCLC). EML4-ALK has been identified in 0.4 to 13.5% of unselected NSCLC patients.4 This subset of patients are often light or never smokers and mostly with adenocarcinoma,4 similar to those with EGFR mutations. The EML4-ALK fusions are almost in mutual exclusion to EGFR mutation,4 with


Laboratory Investigation | 2001

Co-Expression of VEGF-C and Its Receptors, VEGFR-2 and VEGFR-3, in Endothelial Cells of Lymphangioma. Implication in Autocrine or Paracrine Regulation of Lymphangioma

Hsin-Yi Huang; Chao-Chi Ho; Pei-Hsin Huang; Su-Ming Hsu

Lymphangioma has long been thought of as congenital malformations resulting from the failure of lymphatic vessels communicating with the venous system in the fetal period. Alternatively, it is proposed to be true neoplasm originated from the transformation of lymphatic endothelia. To extend the molecular basis of the pathogenesis of lymphangioma, we have characterized the expression of vascular endothelial growth factor (VEGF) and VEGF receptors (VEGFR) in 29 cases of lymphangioma by RNA in situ hybridization. Endothelial cells of lymphangioma co-express transcripts of VEGF-C and its receptors VEGFR-3 (Flt4) and VEGFR-2 (Flk1), which are not detectable in the adjacent connective tissue. In contrast, there is little or no expression of VEGF-C, VEGFR-3, and VEGFR-2 mRNA in endothelial cells of hemangiomas, angiosarcomas, or normal lymphatic vessels of the small or large intestines. The results suggest that VEGF-C and its receptors may take active parts in the formation of lymphangioma by autocrine or paracrine regulation.


Oncogene | 2006

C2GnT-M is downregulated in colorectal cancer and its re-expression causes growth inhibition of colon cancer cells

Min-Chuan Huang; Hsuan Yu Chen; Hung-Tu Huang; John Huang; Jin-Tung Liang; Tang-Long Shen; Neng-Yu Lin; Chao-Chi Ho; Cho Im; Szu-Chun Hsu

Changes in carbohydrates on the cell surface are associated with tumor malignancy. The mucin-type core 2 β-1,6-N-acetylglucosaminyltransferase (C2GnT-M) is highly expressed in the gastrointestinal tract and catalyses the formation of core 2, core 4, and blood group I branches on O-glycans. In the present study, we evaluated the role of C2GnT-M in colorectal cancer. C2GnT-M downexpression was observed in 73.6% of the primary tumors from colorectal cancer patients (39 of 53) analysed by cancer profiling array. Consistently, the majority of colon cancer cell lines and primary colon tumors expressed lower levels of C2GnT-M than did normal colon tissues by RT–PCR. HCT116 cells stably transfected with C2GnT-M inhibited expression of the core 1 structure, Galβ1,3GalNAcα1-Ser/Thr, on the cell surface. Moreover, C2GnT-M expression suppressed cell adhesion, motility, and invasion as well as colony formation ability. The growth of C2GnT-M-transfected HCT116 and SW480 cells was dramatically suppressed, and the cell death induced by C2GnT-M was demonstrated by an increase in the annexin V-positive cells. Interestingly, C2GnT-M inhibited cell adhesion to collagen IV and fibronectin, and decreased tyrosine phosphorylation of paxillin, indicating that the changes in cancer behavior may be partly mediated by integrin-signaling pathways. Tumor growth in vivo was also significantly suppressed by C2GnT-M in the xenografts of nude mice. These results demonstrate that C2GnT-M is frequently downregulated in colorectal cancer and suppresses colon cancer cell growth.


Scientific Reports | 2015

Opposite Effects of M1 and M2 Macrophage Subtypes on Lung Cancer Progression.

Ang Yuan; Yi-Jing Hsiao; Hsuan-Yu Chen; Huei-Wen Chen; Chao-Chi Ho; Yu-Yun Chen; Yi-Chia Liu; Tsai-Hsia Hong; Sung-Liang Yu; Jeremy J.W. Chen; Pan-Chyr Yang

Macrophages in a tumor microenvironment have been characterized as M1- and M2-polarized subtypes. Here, we discovered the different macrophages’ impacts on lung cancer cell A549. The M2a/M2c subtypes promoted A549 invasion and xenograft tumor growth. The M1 subtype suppressed angiogenesis. M1 enhanced the sensitivity of A549 to cisplatin and decreased the tube formation activity and cell viability of A549 cells by inducing apoptosis and senescence. Different macrophage subtypes regulated genes involved in the immune response, cytoskeletal remodeling, coagulation, cell adhesion, and apoptosis pathways in A549 cells, which was a pattern that correlated with the altered behaviors of the A549 cells. Furthermore, we found that the identified M1/M2 gene signatures were significantly correlated with the extended overall survival of lung cancer patients. These results suggest that M1/M2 gene expression signature may be used as a prognostic indicator for lung cancer patients, and M1/M2 polarization may be a target of investigation of immune-modulating therapies for lung cancer in the future.


Breast Cancer Research | 2011

miRNA-34b as a tumor suppressor in estrogen-dependent growth of breast cancer cells

Yee Ming Lee; Jen Yi Lee; Chao-Chi Ho; Qi Sheng Hong; Sung-Liang Yu; Chii Ruey Tzeng; Pan-Chyr Yang; Huei-Wen Chen

IntroductionEstrogen is involved in several physiological and pathological processes through estrogen receptor (ER)-mediated transcriptional gene regulation. miRNAs (miRs), which are noncoding RNA genes, may respond to estrogen and serve as posttranscriptional regulators in tumorigenic progression, especially in breast cancer; however, only limited information about this possibility is available. In the present study, we identified the estrogen-regulated miR-34b and investigated its functional role in breast cancer progression.MethodsEstrogen-regulated miRNAs were identified by using a TaqMan low density array. Our in vivo Tet-On system orthotopic model revealed the tumor-suppressive ability of miR-34b. Luciferase reporter assays and chromatin immunoprecipitation assay demonstrated miR-34b were regulated by p53-ER interaction.ResultsIn this study, we identified one such estrogen downregulated miRNA, miR-34b, as an oncosuppressor that targets cyclin D1 and Jagged-1 (JAG1) in an ER+/wild-type p53 breast cancer cell line (MCF-7), as well as in ovarian and endometrial cells, but not in ER-negative or mutant p53 breast cancer cell lines (T47D, MBA-MB-361 and MDA-MB-435). There is a negative association between ERα and miR-34b expression levels in ER+ breast cancer patients. Tet-On induction of miR-34b can cause inhibition of tumor growth and cell proliferation. Also, the overexpression of miR-34b inhibited ER+ breast tumor growth in an orthotopic mammary fat pad xenograft mouse model. Further validation indicated that estrogens inhibition of miR-34b expression was mediated by interactions between ERα and p53, not by DNA methylation regulation. The xenoestrogens diethylstilbestrol and zeranol also showed similar estrogenic effects by inhibiting miR-34b expression and by restoring the protein levels of the miR-34b targets cyclin D1 and JAG1 in MCF-7 cells.ConclusionsThese findings reveal that miR-34b is an oncosuppressor miRNA requiring both ER+ and wild-type p53 phenotypes in breast cancer cells. These results improve our ability to develop new therapeutic strategies to target the complex estrogenic pathway in human breast cancer progression through miRNA regulation.


European Journal of Cardio-Thoracic Surgery | 2011

A rare constellation of empyema, lung abscess, and mediastinal abscess as a complication of endobronchial ultrasound-guided transbronchial needle aspiration

Chun-Ta Huang; Chung-Yu Chen; Chao-Chi Ho; Chong-Jen Yu

The introduction of endobronchial ultrasound-guided transbronchial needle aspiration (EBUS-TBNA) brought about significant advancement in the field of bronchoscopy. The major indications for EBUS-TBNA are lung cancer staging and diagnosis of mediastinal lymphadenopathy. This procedure is minimally invasive and cost saving, and no complications have been described in large-scale studies. In this report, we present a case of empyema, lung abscess, and mediastinal abscess that developed in a patient undergoing EBUS-TBNA; the patient subsequently recovered uneventfully after aggressive surgical debridement and antimicrobial therapy.

Collaboration


Dive into the Chao-Chi Ho's collaboration.

Top Co-Authors

Avatar

Chong-Jen Yu

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Pan-Chyr Yang

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Jin-Yuan Shih

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Wei-Yu Liao

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kuan-Yu Chen

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Huei-Wen Chen

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Sung-Liang Yu

National Taiwan University

View shared research outputs
Top Co-Authors

Avatar

Chia-Chi Lin

National Taiwan University

View shared research outputs
Researchain Logo
Decentralizing Knowledge