53 results on '"Chun A. Changou"'
Search Results
2. Supplementary Movie S2 from Dual Inhibition of PIK3C3 and FGFR as a New Therapeutic Approach to Treat Bladder Cancer
- Author
-
Yun Yen, Jing-Ping Liou, Yun-Ru Liu, Yan-Ni Lo, Yu-Chen Lin, Hao-Ching Wang, Kai-Cheng Hsu, Cheng-Ying Chu, Yu-Ching Lee, Tsung-Han Hsieh, Chun A. Changou, and Chun-Han Chen
- Abstract
Effect of MPT0L145 on endosomal functions.
- Published
- 2023
3. Supplementary data from Dual Inhibition of PIK3C3 and FGFR as a New Therapeutic Approach to Treat Bladder Cancer
- Author
-
Yun Yen, Jing-Ping Liou, Yun-Ru Liu, Yan-Ni Lo, Yu-Chen Lin, Hao-Ching Wang, Kai-Cheng Hsu, Cheng-Ying Chu, Yu-Ching Lee, Tsung-Han Hsieh, Chun A. Changou, and Chun-Han Chen
- Abstract
Supplementary Fig. S1, Time-dependent effects of BGJ398 on autophagy in bladder cancer cells. Supplementary Figure. S2. In vitro inhibitory effects of MPT0L145 and SAR405 on PIK3C3 (VPS34). Supplementary Figure. S3. A and B, The potentiation of MPT0L145-increased cytotoxicity by PIK3C3 knockdown in Panc1 and A2780 cells. C and D, ROS inhibitors partially reversed MPT0L145-decreased cell viability. Supplementary Figure S4. Validation of ATG5-knockout (K.O.) RT-112 cells. Supplementary Fig. S5. A, Effect of specific CSF1R inhibitor, BLZ945, on the viability in RT-112 and RT-4 cells. B, Dual inhibition of FGFR3 signaling and autophagy by MPT0L145. C, Effect of MPT0L145 on necrosis in bladder cancer cells.
- Published
- 2023
4. Supplementary Table from Dual Inhibition of PIK3C3 and FGFR as a New Therapeutic Approach to Treat Bladder Cancer
- Author
-
Yun Yen, Jing-Ping Liou, Yun-Ru Liu, Yan-Ni Lo, Yu-Chen Lin, Hao-Ching Wang, Kai-Cheng Hsu, Cheng-Ying Chu, Yu-Ching Lee, Tsung-Han Hsieh, Chun A. Changou, and Chun-Han Chen
- Abstract
Supplementary Table S1. In vitro inhibitory effects of MPT0L145 on a panel of lipid kinases. Supplementary Table S2. Binding kinetics and affinity of MPT0L145 to wildtype and mutant PIK3C3.
- Published
- 2023
5. Supplementary Movie S3 from Dual Inhibition of PIK3C3 and FGFR as a New Therapeutic Approach to Treat Bladder Cancer
- Author
-
Yun Yen, Jing-Ping Liou, Yun-Ru Liu, Yan-Ni Lo, Yu-Chen Lin, Hao-Ching Wang, Kai-Cheng Hsu, Cheng-Ying Chu, Yu-Ching Lee, Tsung-Han Hsieh, Chun A. Changou, and Chun-Han Chen
- Abstract
Effect of MPT0L145 on endosomal functions.
- Published
- 2023
6. Data from Dual Inhibition of PIK3C3 and FGFR as a New Therapeutic Approach to Treat Bladder Cancer
- Author
-
Yun Yen, Jing-Ping Liou, Yun-Ru Liu, Yan-Ni Lo, Yu-Chen Lin, Hao-Ching Wang, Kai-Cheng Hsu, Cheng-Ying Chu, Yu-Ching Lee, Tsung-Han Hsieh, Chun A. Changou, and Chun-Han Chen
- Abstract
Purpose: MPT0L145 has been developed as a FGFR inhibitor exhibiting significant anti-bladder cancer activity in vitro and in vivo via promoting autophagy-dependent cell death. Here, we aim to elucidate the underlying mechanisms.Experimental Design: Autophagy flux, morphology, and intracellular organelles were evaluated by Western blotting, transmission electron microscope, and fluorescence microscope. Molecular docking and surface plasmon resonance assay were performed to identify drug–protein interaction. Lentiviral delivery of cDNA or shRNA and CRISPR/Cas9-mediated genome editing was used to modulate gene expression. Mitochondrial oxygen consumption rate was measured by a Seahorse XFe24 extracellular flux analyzer, and ROS level was measured by flow cytometry.Results: MPT0L145 persistently increased incomplete autophagy and phase-lucent vacuoles at the perinuclear region, which were identified as enlarged and alkalinized late-endosomes. Screening of a panel of lipid kinases revealed that MPT0L145 strongly inhibits PIK3C3 with a Kd value of 0.53 nmol/L. Ectopic expression of PIK3C3 reversed MPT0L145-increased cell death and incomplete autophagy. Four residues (Y670, F684, I760, D761) at the ATP-binding site of PIK3C3 are important for the binding of MPT0L145. In addition, MPT0L145 promotes mitochondrial dysfunction, ROS production, and DNA damage, which may in part, contribute to cell death. ATG5-knockout rescued MPT0L145-induced cell death, suggesting simultaneous induction of autophagy is crucial to its anticancer activity. Finally, our data demonstrated that MPT0L145 is able to overcome cisplatin resistance in bladder cancer cells.Conclusions: MPT0L145 is a first-in-class PIK3C3/FGFR inhibitor, providing an innovative strategy to design new compounds that increase autophagy, but simultaneously perturb its process to promote bladder cancer cell death. Clin Cancer Res; 24(5); 1176–89. ©2017 AACR.
- Published
- 2023
7. Thyroid Hormone Induces Oral Cancer Growth via the PD-L1-Dependent Signaling Pathway
- Author
-
Kuan-Wei Su, Hung-Yun Lin, Hsien-Chung Chiu, Shin-Yu Shen, Chun A. ChangOu, Dana R. Crawford, Yu-Chen S. H. Yang, Ya-Jung Shih, Zi-Lin Li, Haw-Ming Huang, Jaqueline Whang-Peng, Yih Ho, and Kuan Wang
- Subjects
STAT3 Transcription Factor ,thyroxine ,oral cancer ,PD-L1 ,β-catenin ,Thyroid Hormones ,Thyroxine ,Humans ,Nuclear Proteins ,Mouth Neoplasms ,General Medicine ,RNA, Small Interfering ,Integrin alphaVbeta3 ,B7-H1 Antigen ,beta Catenin ,Signal Transduction - Abstract
Oral cancer is a fatal disease, and its incidence in Taiwan is increasing. Thyroid hormone as L-thyroxine (T4) stimulates cancer cell proliferation via a receptor on integrin αvβ3 of plasma membranes. It also induces the expression of programmed death-ligand 1 (PD-L1) and cell proliferation in cancer cells. Thyroid hormone also activates β-catenin-dependent cell proliferation in cancer cells. However, the relationship between PD-L1 and cancer proliferation is not fully understood. In the current study, we investigated the role of inducible thyroid hormone-induced PD-L1-regulated gene expression and proliferation in oral cancer cells. Thyroxine bound to integrin αvβ3 to induce PD-L1 expressions via activation of ERK1/2 and signal transducer and activator of transcription 3 (STAT3). Inactivated STAT3 inhibited PD-L1 expression and nuclear PD-L1 accumulation. Inhibition of PD-L1 expression reduced β-catenin accumulation. Furthermore, nuclear PD-L1 formed a complex with nuclear proteins such as p300. Suppression PD-L1 expression by shRNA blocked not only expression of PD-L1 and β-catenin but also signal transduction, proliferative gene expressions, and cancer cell growth. In summary, thyroxine via integrin αvβ3 activated ERK1/2 and STAT3 to stimulate the PD-L1-dependent and β-catenin-related growth in oral cancer cells.
- Published
- 2022
- Full Text
- View/download PDF
8. Bridging Size and Charge Effects of Mesoporous Silica Nanoparticles for Crossing the Blood–Brain Barrier
- Author
-
Yi-Ping, Chen, Chih-Ming, Chou, Tsu-Yuan, Chang, Hao, Ting, Julien, Dembélé, You-Tai, Chu, Tsang-Pai, Liu, Chun A, Changou, Chien-Wei, Liu, and Chien-Tsu, Chen
- Subjects
General Chemistry - Abstract
The blood–brain barrier (BBB) is a highly selective cellular barrier that tightly controls the microenvironment of the central nervous system to restrict the passage of substances, which is a primary challenge in delivering therapeutic drugs to treat brain diseases. This study aimed to develop simple surface modifications of mesoporous silica nanoparticles (MSNs) without external stimuli or receptor protein conjugation, which exhibited a critical surface charge and size allowing them to cross the BBB. A series of MSNs with various charges and two different sizes of 50 and 200 nm were synthesized, which showed a uniform mesoporous structure with various surface zeta potentials ranging from +42.3 to −51.6 mV. Confocal microscopic results showed that 50 nm of strongly negatively charged N4-RMSN50@PEG/THPMP (∼−40 mV) could be significantly observed outside blood vessels of the brain in Tg(zfli1:EGFP) transgenic zebrafish embryos superior to the other negatively charged MSNs. However, very few positively charged MSNs were found in the brain, indicating that negatively charged MSNs could successfully penetrate the BBB. The data were confirmed by high-resolution images of 3D deconvoluted confocal microscopy and two-photon microscopy and zebrafish brain tissue sections. In addition, while increasing the size to 200 nm but maintaining the similar negative charge (∼40 mV), MSNs could not be detected in the brain of zebrafish, suggesting that transport across the BBB based on MSNs occurred in charge- and size-dependent manners. No obvious cytotoxicity was observed in the CTX-TNA2 astrocyte cell line and U87-MG glioma cell line treated with MSNs. After doxorubicin (Dox) loading, N4-RMSN50@PEG/THPMP/Dox enabled drug delivery and pH-responsive release. The toxicity assay showed that N4-RMSN50@PEG/THPMP could reduce Dox release, resulting in the increase of the survival rate in zebrafish. Flow cytometry demonstrated N4-RMSN50@PEG/THPMP had few cellular uptakes. Protein corona analysis revealed three transporter proteins, such as afamin, apolipoprotein E, and basigin, could contribute to BBB penetration, validating the possible mechanism of N4-RMSN50@PEG/THPMP crossing the BBB. With this simple approach, MSNs with critical negative charge and size could overcome the BBB-limiting characteristics of therapeutic drug molecules; furthermore, their use may also cause drug sustained-release in the brain, decreasing peripheral toxicity.
- Published
- 2022
9. Anticancer polypyrrole-polyethylenimine drug-free nanozyme for precise B-cell lymphoma therapy
- Author
-
Thi Thuy Nguyen, Er-Yuan Chuang, Ya-Ping Chen, Po-Chun Tseng, Ming-Kai Jhan, Chun-Yi Lai, Yung-Ting Wang, Yu-Ping Hung, Chun Austin Changou, Chi-Ming Lee, Chia-Ling Chen, and Chiou-Feng Lin
- Subjects
Pharmacology ,General Medicine - Published
- 2023
10. Type-3 Hyaluronan Synthase Attenuates Tumor Cells Invasion in Human Mammary Parenchymal Tissues
- Author
-
Tzu Chun Cheng, Li Ching Chen, Ching Chuan Kuo, Hang Lung Chang, Ming Thau Sheu, Juo Han Lin, Chih Hsiung Wu, Shih Hsin Tu, Hui Wen Chang, Yuan Soon Ho, Chun A. Changou, and Wen Jui Lee
- Subjects
autophagy ,Xenotransplantation ,medicine.medical_treatment ,Pharmaceutical Science ,Organic chemistry ,Breast Neoplasms ,Article ,Analytical Chemistry ,Metastasis ,chemistry.chemical_compound ,Mice ,Breast cancer ,breast cancer ,QD241-441 ,hyaluronan synthase 3 ,Drug Discovery ,Hyaluronic acid ,medicine ,Tumor Cells, Cultured ,Animals ,Humans ,tumor microenvironment ,RNA, Messenger ,Physical and Theoretical Chemistry ,Parenchymal Tissue ,Mice, Knockout ,Tumor microenvironment ,biology ,CD44 ,Mammary Neoplasms, Experimental ,medicine.disease ,Mice, Inbred C57BL ,Hyaluronan synthase ,chemistry ,Chemistry (miscellaneous) ,Cancer cell ,biology.protein ,Cancer research ,Molecular Medicine ,tubulin acetylation ,Female ,Hyaluronan Synthases - Abstract
The microenvironment for tumor growth and developing metastasis should be essential. This study demonstrated that the hyaluronic acid synthase 3 (HAS3) protein and its enzymatic product hyaluronic acid (HA) encompassed in the subcutaneous extracellular matrix can attenuate the invasion of human breast tumor cells. Decreased HA levels in subcutaneous Has3-KO mouse tissues promoted orthotopic breast cancer (E0771) cell-derived allograft tumor growth. MDA-MB-231 cells premixed with higher concentration HA attenuate tumor growth in xenografted nude mice. Human patient-derived xenotransplantation (PDX) experiments found that HA selected the highly migratory breast cancer cells with CD44 expression accumulated in the tumor/stroma junction. In conclusion, HAS3 and HA were detected in the stroma breast tissues at a high level attenuates effects for induced breast cancer cell death, and inhibit the cancer cells invasion at the initial stage. However, the highly migratory cancer cells were resistant to the HA-mediated effects with unknown mechanisms.
- Published
- 2021
11. Alteration of mesenchymal stem cells polarity by laminar shear stimulation promoting β-catenin nuclear localization
- Author
-
Wei Tse Hsu, Oscar K. Lee, Yu-Ju Chen, Chun A. Changou, Tzu Hao Chang, Chia Li Han, Meng Hua Yen, Wei Ta Chen, Jennifer Hui Chun Ho, and Ji-Yen Cheng
- Subjects
Angiogenesis ,Biophysics ,Bioengineering ,02 engineering and technology ,Cell junction ,Biomaterials ,03 medical and health sciences ,Paracrine signalling ,Human Umbilical Vein Endothelial Cells ,Humans ,Protein Interaction Maps ,Wnt Signaling Pathway ,Cells, Cultured ,beta Catenin ,030304 developmental biology ,Cell Nucleus ,0303 health sciences ,Chemistry ,Mesenchymal stem cell ,Wnt signaling pathway ,Cell Polarity ,Mesenchymal Stem Cells ,021001 nanoscience & nanotechnology ,Actins ,Cell biology ,Endothelial stem cell ,Mechanics of Materials ,Catenin ,Ceramics and Composites ,Mechanosensitive channels ,Stress, Mechanical ,0210 nano-technology - Abstract
Mesenchymal stem cell (MSC) is mechanosensitive and the respond to mechanical force is pattern specific. We previously reported that oscillatory shear stress at 0.5 ± 4 dyne/cm 2 guided MSCs polarity vertical to net flow direction before apolaric stage at 30 min resulting in phosphorylation of β-catenin and inhibition of Wnt signaling . This time, we explored laminar shear stress (LS) at 0.5 dyne/cm 2 polarized MSCs by guiding F-actin orientation parallel to the flow direction before apolarity at 30 min accompanied with activation of Wnt signaling. Time-dependent microarray analysis supported cell-cell junctional complex of MSCs was the major mechanosensor on MSCs to respond 0.5 dyne/cm 2 LS. Three-dimensional immunofluorescence image confirmed LS promoting β-catenin nuclear localization during 15 min to 1 h with a peak at 30 min. Functional analysis of proteomic study on MSC with 30 min LS stimulation indicated that upregulation of β-catenin downstream proteins related to cardiovascular development, endothelial cell protection and angiogenesis . Conditioned medium from MSCs with 30 min LS stimulation improved the viability of human endothelial cells from oxidative damage . In conclusion, 0.5 dyne/cm 2 LS on MSCs for 30 min guides MSCs lack of polarity and promotes β-catenin nuclear translocation favoring Wnt activation and paracrine cardiovascular support.
- Published
- 2019
12. Platelet autophagic machinery involved in thrombosis through a novel linkage of AMPK-MTOR to sphingolipid metabolism
- Author
-
Cheng Yang Lee, Kuan Hung Lin, Tzu Yin Lee, Thanasekaran Jayakumar, Wan-Jung Lu, Tzu Hao Chang, Joen Rong Sheu, Chao Chien Chang, Ray Jade Chen, Nguyen Thi Thu Trang, Chun A. Changou, Cheng Ying Hsieh, Yuan Chin Hsiung, and Chih Hao Yang
- Subjects
0301 basic medicine ,Blood Platelets ,Sphingomyelin phosphodiesterase ,Biology ,AMP-Activated Protein Kinases ,03 medical and health sciences ,Mice ,Phosphatidylinositol 3-Kinases ,Sequestosome 1 ,Tandem Mass Spectrometry ,Autophagy ,Animals ,Platelet activation ,Mean platelet volume ,education ,Molecular Biology ,Mechanistic target of rapamycin ,education.field_of_study ,Sphingolipids ,030102 biochemistry & molecular biology ,TOR Serine-Threonine Kinases ,AMPK ,Thrombosis ,Cell Biology ,BECN1 ,Hydrogen Peroxide ,Molecular biology ,030104 developmental biology ,biology.protein ,Chromatography, Liquid ,Research Paper - Abstract
Basal macroautophagy/autophagy has recently been found in anucleate platelets. Platelet autophagy is involved in platelet activation and thrombus formation. However, the mechanism underlying autophagy in anucleate platelets require further clarification. Our data revealed that LC3-II formation and SQSTM1/p62 degradation were noted in H(2)O(2)-activated human platelets, which could be blocked by 3-methyladenine and bafilomycin A(1), indicating that platelet activation may cause platelet autophagy. AMPK phosphorylation and MTOR dephosphorylation were also detected, and block of AMPK activity by the AMPK inhibitor dorsomorphin reversed SQSTM1 degradation and LC3-II formation. Moreover, autophagosome formation was observed through transmission electron microscopy and deconvolution microscopy. These findings suggest that platelet autophagy was induced partly through the AMPK-MTOR pathway. In addition, increased LC3-II expression occurred only in H(2)O(2)-treated Atg5(f/f) platelets, but not in H(2)O(2)-treated atg5(−/−) platelets, suggesting that platelet autophagy occurs during platelet activation. atg5(−/−) platelets also exhibited a lower aggregation in response to agonists, and platelet-specific atg5(−/−) mice exhibited delayed thrombus formation in mesenteric microvessles and decreased mortality rate due to pulmonary thrombosis. Notably, metabolic analysis revealed that sphingolipid metabolism is involved in platelet activation, as evidenced by observed several altered metabolites, which could be reversed by dorsomorphin. Therefore, platelet autophagy and platelet activation are positively correlated, partly through the interconnected network of sphingolipid metabolism. In conclusion, this study for the first time demonstrated that AMPK-MTOR signaling could regulate platelet autophagy. A novel linkage between AMPK-MTOR and sphingolipid metabolism in anucleate platelet autophagy was also identified: platelet autophagy and platelet activation are positively correlated. Abbreviations: 3-MA: 3-methyladenine; A.C.D.: citric acid/sod. citrate/glucose; ADP: adenosine diphosphate; AKT: AKT serine/threonine kinase; AMPK: AMP-activated protein kinase; ANOVA: analysis of variance; ATG: autophagy-related; B4GALT/LacCS: beta-1,4-galactosyltransferase; Baf-A1: bafilomycin A(1); BECN1: beclin 1; BHT: butylate hydrooxytoluene; BSA: bovine serum albumin; DAG: diacylglycerol; ECL: enhanced chemiluminescence; EDTA: ethylenediamine tetraacetic acid; ELISA: enzyme-linked immunosorbent assay; GALC/GCDase: galactosylceramidase; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GBA/GluSDase: glucosylceramidase beta; GPI: glycosylphosphatidylinositol; H(2)O(2): hydrogen peroxide; HMDB: human metabolome database; HRP: horseradish peroxidase; IF: immunofluorescence; IgG: immunoglobulin G; KEGG: Kyoto Encyclopedia of Genes and Genomes; LAMP1: lysosomal associated membrane protein 1; LC-MS/MS: liquid chromatography-tandem mass spectrometry; mAb: monoclonal antibody; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MPV: mean platelet volume; MTOR: mechanistic target of rapamycin kinase; ox-LDL: oxidized low-density lipoprotein; pAb: polyclonal antibody; PC: phosphatidylcholine; PCR: polymerase chain reaction; PI3K: phosphoinositide 3-kinase; PLS-DA: partial least-squares discriminant analysis; PRP: platelet-rich plasma; Q-TOF: quadrupole-time of flight; RBC: red blood cell; ROS: reactive oxygen species; RPS6KB/p70S6K: ribosomal protein S6 kinase B; SDS: sodium dodecyl sulfate; S.E.M.: standard error of the mean; SEM: scanning electron microscopy; SGMS: sphingomyelin synthase; SM: sphingomyelin; SMPD/SMase: sphingomyelin phosphodiesterase; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; UGT8/CGT: UDP glycosyltransferase 8; UGCG/GCS: UDP-glucose ceramide glucosyltransferase; ULK1: unc-51 like autophagy activating kinase 1; UPLC: ultra-performance liquid chromatography; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PtdIns3P: phosphatidylinositol-3-phosphate; WBC: white blood cell; WT: wild type
- Published
- 2021
13. Arginine starvation elicits chromatin leakage and cGAS-STING activation via epigenetic silencing of metabolic and DNA-repair genes
- Author
-
Chia Lin Chen, Shauh Der Yeh, Chih Pin Chuu, Hung Jung Wang, Yen Ling Yu, Mei Ling Cheng, Tse Chun Kuo, Chun A. Changou, Hongwu Chen, Cheng Ying Chu, Cheng Chin Kuo, Hsing Jien Kung, Sheng Chieh Hsu, Lu Hai Wang, David K. Ann, Yun Yen, and Chien-Feng Li
- Subjects
0301 basic medicine ,Male ,Aging ,Arginine ,DNA Repair ,Arginine starvation ,Medicine (miscellaneous) ,Castration-Resistant ,0302 clinical medicine ,2.1 Biological and endogenous factors ,Aetiology ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Cancer ,Prostate Cancer ,Nucleotidyltransferases ,Chromatin ,Cell biology ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,Prostatic Neoplasms, Castration-Resistant ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,PC-3 Cells ,HIV/AIDS ,Histone Demethylases ,Development of treatments and therapeutic interventions ,Research Paper ,Epigenetic gene silencing ,Urologic Diseases ,Programmed cell death ,DNA damage ,DNA repair ,1.1 Normal biological development and functioning ,Oncology and Carcinogenesis ,Biology ,03 medical and health sciences ,Downregulation and upregulation ,Underpinning research ,Genetics ,Humans ,Epigenetics ,Gene Silencing ,Nutrition ,Neoplastic ,Membrane Proteins ,Prostatic Neoplasms ,030104 developmental biology ,Gene Expression Regulation ,DNA leakage ,cGAS-STING activation - Abstract
Rationale: One of the most common metabolic defects in cancers is the deficiency in arginine synthesis, which has been exploited therapeutically. Yet, challenges remain, and the mechanisms of arginine-starvation induced killing are largely unclear. Here, we sought to demonstrate the underlying mechanisms by which arginine starvation-induced cell death and to develop a dietary arginine-restriction xenograft model to study the in vivo effects. Methods: Multiple castration-resistant prostate cancer cell lines were treated with arginine starvation followed by comprehensive analysis of microarray, RNA-seq and ChIP-seq were to identify the molecular and epigenetic pathways affected by arginine starvation. Metabolomics and Seahorse Flux analyses were used to determine the metabolic profiles. A dietary arginine-restriction xenograft mouse model was developed to assess the effects of arginine starvation on tumor growth and inflammatory responses. Results: We showed that arginine starvation coordinately and epigenetically suppressed gene expressions, including those involved in oxidative phosphorylation and DNA repair, resulting in DNA damage, chromatin-leakage and cGAS-STING activation, accompanied by the upregulation of type I interferon response. We further demonstrated that arginine starvation-caused depletion of α-ketoglutarate and inactivation of histone demethylases are the underlying causes of epigenetic silencing. Significantly, our dietary arginine-restriction model showed that arginine starvation suppressed prostate cancer growth in vivo, with evidence of enhanced interferon responses and recruitment of immune cells. Conclusions: Arginine-starvation induces tumor cell killing by metabolite depletion and epigenetic silencing of metabolic genes, leading to DNA damage and chromatin leakage. The resulting cGAS-STING activation may further enhance these killing effects.
- Published
- 2021
14. Heteronemin and tetrac derivatives suppress non-small cell lung cancer growth via ERK1/2 inhibition
- Author
-
Cheng-Chin Chung, Tung-Yung Huang, Hung-Ru Chu, Roberto De Luca, Elena Candelotti, Chi-Hung Huang, Yu-Chen S.H. Yang, Sandra Incerpi, Jens Z. Pedersen, Chi-Yu Lin, Haw-Ming Huang, Sheng-Yang Lee, Zi-Lin Li, Chun A. ChangOu, Wen-Shan Li, Paul J. Davis, Hung-Yun Lin, Jacqueline Whang-Peng, and Kuan Wang
- Subjects
Gene Expression Regulation, Neoplastic ,Thyroxine ,Lung Neoplasms ,Terpenes ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Humans ,Antineoplastic Agents ,Drug Therapy, Combination ,General Medicine ,Extracellular Signal-Regulated MAP Kinases ,Toxicology ,Food Science - Abstract
The most common cancer, lung cancer, causes deaths worldwide. Most lung cancer patients have non-small cell lung carcinomas (NSCLCs) with a poor prognosis. The chemotherapies frequently cause resistance therefore search for new effective drugs for NSCLC patients is an urgent and essential issue. Deaminated thyroxine, tetraiodothyroacetic acid (tetrac), and its nano-analogue (NDAT) exhibit antiproliferative properties in several types of cancers. On the other hand, the most abundant secondary metabolite in the sponge Hippospongia sp., heteronemin, shows effective cytotoxic activity against different types of cancer cells. In the current study, we investigated the anticancer effects of heteronemin against two NSCLC cell lines, A549 and H1299 cells in vitro. Combined treatment with heteronemin and tetrac derivatives synergistically inhibited cancer cell growth and significantly modulated the ERK1/2 and STAT3 pathways in A549 cells but only ERK1/2 in H1299 cells. The combination treatments induce apoptosis via the caspases pathway in A549 cells but promote cell cycle arrest via CCND1 and PCNA inhibition in H1299 cells. In summary, these results suggest that combined treatment with heteronemin and tetrac derivatives could suppress signal transduction pathways essential for NSCLC cell growth. The synergetic effects can be used potentially as a therapeutic procedure for NSCLC patients.
- Published
- 2022
15. NDAT Targets PI3K-Mediated PD-L1 Upregulation to Reduce Proliferation in Gefitinib-Resistant Colorectal Cancer
- Author
-
Hung Ru Chu, Chun A. Changou, Shih Jiuan Chiu, Hung Yun Lin, Shwu Huey Wang, Ya Jung Shih, Ema Dwi Hastuti, Tung Cheng Chang, R. Holland Cheng, Zi Lin Li, Yi Shin Pan, Yi Ru Chen, Feng Cheng Liu, Wong Jin Chang, Kuan Wang, Jacqueline Whang-Peng, Shaker A. Mousa, Yu Tang Chin, Alexander T.H. Wu, Tung Yung Huang, and Paul J. Davis
- Subjects
0301 basic medicine ,Colorectal cancer ,Drug Resistance ,gefitinib ,PI3K ,B7-H1 Antigen ,Metastasis ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Tumor Cells, Cultured ,lcsh:QH301-705.5 ,Polyglactin 910 ,Phosphoinositide-3 Kinase Inhibitors ,Cancer ,Cultured ,Chemistry ,General Medicine ,Tumor Cells ,Colo-Rectal Cancer ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,Colorectal Neoplasms ,HT29 Cells ,medicine.drug ,PD-L1 ,NDAT ,Antineoplastic Agents ,colorectal cancer ,colorectal cancer (CRC) ,Keywords: colorectal cancer (CRC), gefitinib ,Article ,03 medical and health sciences ,Gefitinib ,medicine ,Animals ,Humans ,MTT assay ,neoplasms ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Neoplastic ,Cell growth ,medicine.disease ,HCT116 Cells ,Xenograft Model Antitumor Assays ,Thyroxine ,030104 developmental biology ,lcsh:Biology (General) ,Gene Expression Regulation ,Drug Resistance, Neoplasm ,Cancer cell ,Cancer research ,Neoplasm ,Digestive Diseases - Abstract
The property of drug-resistance may attenuate clinical therapy in cancer cells, such as chemoresistance to gefitinib in colon cancer cells. In previous studies, overexpression of PD-L1 causes proliferation and metastasis in cancer cells, therefore, the PD-L1 pathway allows tumor cells to exert an adaptive resistance mechanism in vivo. Nano-diamino-tetrac (NDAT) has been shown to enhance the anti-proliferative effect induced by first-line chemotherapy in various types of cancer, including colorectal cancer (CRC). In this work, we attempted to explore whether NDAT could enhance the anti-proliferative effect of gefitinib in CRC and clarified the mechanism of their interaction. The MTT assay was utilized to detect a reduction in cell proliferation in four primary culture tumor cells treated with gefitinib or NDAT. The gene expression of PD-L1 and other tumor growth-related molecules were quantified by quantitative polymerase chain reaction (qPCR). Furthermore, the identification of PI3K and PD-L1 in treated CRC cells were detected by western blotting analysis. PD-L1 presentation in HCT116 xenograft tumors was characterized by specialized immunohistochemistry (IHC) and the hematoxylin and eosin stain (H&, E stain). The correlations between the change in PD-L1 expression and tumorigenic characteristics were also analyzed. (3) The PD-L1 was highly expressed in Colo_160224 rather than in the other three primary CRC cells and HCT-116 cells. Moreover, the PD-L1 expression was decreased by gefitinib (1 µ, M and 10 µ, M) in two cells (Colo_150624 and 160426), but 10 µ, M gefitinib stimulated PD-L1 expression in gefitinib-resistant primary CRC Colo_160224 cells. Inactivated PI3K reduced PD-L1 expression and proliferation in CRC Colo_160224 cells. Gefitinib didn&rsquo, t inhibit PD-L1 expression and PI3K activation in gefitinib-resistant Colo_160224 cells. However, NDAT inhibited PI3K activation as well as PD-L1 accumulation in gefitinib-resistant Colo_160224 cells. The combined treatment of NDAT and gefitinib inhibited pPI3K and PD-L1 expression and cell proliferation. Additionally, NDAT reduced PD-L1 accumulation and tumor growth in the HCT116 (K-RAS mutant) xenograft experiment. (4) Gefitinib might suppress PD-L1 expression but did not inhibit proliferation through PI3K in gefitinib-resistant primary CRC cells. However, NDAT not only down-regulated PD-L1 expression via blocking PI3K activation but also inhibited cell proliferation in gefitinib-resistant CRCs.
- Published
- 2020
16. CCL5 promotion of bioenergy metabolism is crucial for hippocampal synapse complex and memory formation
- Author
-
Hui Min Chen, Yuan Chin Hsiung, Yuan Hao Chen, Cheng Yang Lee, Wen Chang Chang, Man Hau Ho, Chiu Jing-Yuan, Yung Hsiao Chiang, Szu Yi Chou, Tzu Hao Chang, Yun Wang, Chun A. Changou, You Yin Chen, Barry J. Hoffer, Reni Ajoy, and Yu Chun Lo
- Subjects
0301 basic medicine ,Physiology ,Long-Term Potentiation ,Hippocampus ,Carbohydrate metabolism ,Hippocampal formation ,Article ,Synapse ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,stomatognathic system ,Memory ,Premovement neuronal activity ,Animals ,Glycolysis ,Molecular Biology ,Mice, Knockout ,Neuronal Plasticity ,Chemistry ,Glutamate receptor ,Long-term potentiation ,Cell biology ,Psychiatry and Mental health ,stomatognathic diseases ,030104 developmental biology ,Synapses ,030217 neurology & neurosurgery ,Neuroscience - Abstract
Glucoregulatory efficiency and ATP production are key regulators for neuronal plasticity and memory formation. Besides its chemotactic and neuroinflammatory functions, the CC chemokine––CCL5 displays neurotrophic activity. We found impaired learning-memory and cognition in CCL5-knockout mice at 4 months of age correlated with reduced hippocampal long-term potentiation and impaired synapse structure. Re-expressing CCL5 in knockout mouse hippocampus restored synaptic protein expression, neuronal connectivity and cognitive function. Using metabolomics coupled with FDG-PET imaging and seahorse analysis, we found that CCL5 participates in hippocampal fructose and mannose degradation, glycolysis, gluconeogenesis as well as glutamate and purine metabolism. CCL5 additionally supports mitochondrial structural integrity, purine synthesis, ATP generation, and subsequent aerobic glucose metabolism. Overexpressing CCL5 in WT mice also enhanced memory-cognition performance as well as hippocampal neuronal activity and connectivity through promotion of de novo purine and glutamate metabolism. Thus, CCL5 actions on glucose aerobic metabolism are critical for mitochondrial function which contribute to hippocampal spine and synapse formation, improving learning and memory., In addition to neuroinflammatory functions, chemokine CCL5 shows strong neurotrophic properties. In this study, we found that CCL5 facilitates hippocampal glucoregulatory efficiency. CCL5 promotes aerobic glucose metabolism, mitochondrial ATP generation and purine synthesis, improving synaptic plasticity and enhancing memory and cognition functions.
- Published
- 2020
17. A Phase II Clinical Trial on the Combination Therapy of PHY906 Plus Capecitabine in Hepatocellular Carcinoma
- Author
-
Servina Liu, Shwu-Huey Liu, Yung-Chi Cheng, Li-Tzong Chen, Yun Yen, Frank Luh, Her Shyong Shiah, and Chun A. Changou
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Carcinoma, Hepatocellular ,Combination therapy ,Hepatocellular carcinoma ,Salvage therapy ,Capecitabine ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,Adverse effect ,Survival rate ,business.industry ,Clinical Trial Results ,Liver Neoplasms ,medicine.disease ,Clinical trial ,Treatment Outcome ,030220 oncology & carcinogenesis ,030211 gastroenterology & hepatology ,Fluorouracil ,Chinese herbal medicine ,business ,PHY906 ,medicine.drug ,Drugs, Chinese Herbal - Abstract
Trial Information ClinicalTrials.gov Identifier: NCT00076609 Sponsor: Yiviva Inc. Principal Investigator: Yun Yen IRB Approved: Yes Lessons Learned A PHY906 and capecitabine combination could be effective as a salvage therapy for patients with hepatocellular carcinoma (HCC) previously treated with multiple systemic therapies. This traditional Chinese medicine formulation can work with Western cancer chemotherapeutic agents to improve clinical outcomes or alleviate side effects for patients with advanced HCC. Background This study aimed to evaluate efficacy and safety of capecitabine combined with a PHY906 (a pharmaceutical-grade formulation of four traditional Chinese herbs) in the treatment of advanced hepatocellular carcinoma (HCC) in Asian patients who were positive for hepatitis B virus (HBV). Methods This study was an open-label, phase II safety and efficacy clinical trial of PHY906 and capecitabine in patients with advanced HCC. Patients received 750 mg/m2 capecitabine b.i.d. 14 days plus 800 mg of PHY906 b.i.d. on days 1–4 and days 8–11 every 21-day cycle. The primary endpoint was 6-month survival rate, and secondary endpoints were progression-free survival, overall survival, disease control rate, and safety. Results Thirty-nine subjects completed the study with a 46.2% stable disease rate. The median progression-free survival was 1.5 months, and median overall survival (mOS) was 6 months with a 51.3% 6-month survival rate. The most common adverse events included lower hemoglobin, diarrhea, pain, abdomen (not otherwise specified), fatigue, increased aspartate aminotransferase, and bilirubin. Patients who (a) had not received previous chemotherapies or targeted therapy or (b) had lower starting alpha-fetoprotein (AFP) levels or (c) had HBV infection showed better clinical outcome. Conclusion Our data showed that PHY906 increases the therapeutic index of capecitabine by enhancing its antitumor activity and reduces its toxicity profile in advanced HCC.
- Published
- 2020
18. Clinical-grade cryopreserved doxorubicin-loaded platelets: role of cancer cells and platelet extracellular vesicles activation loop
- Author
-
Hadi Goubran, Long Sheng Lu, Thierry Burnouf, Yu Wen Wu, Chun A. Changou, and Cheng Chain Huang
- Subjects
Blood Platelets ,0301 basic medicine ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,lcsh:Medicine ,Extracellular Vesicles ,03 medical and health sciences ,Tissue factor ,0302 clinical medicine ,Thrombin ,polycyclic compounds ,medicine ,Humans ,Pharmacology (medical) ,Platelet ,Doxorubicin ,Molecular Biology ,Cancer ,Cryopreservation ,Liposome ,Chemistry ,Research ,lcsh:R ,Biochemistry (medical) ,Cell Biology ,General Medicine ,Cryopreserved platelet ,Neoplastic Cells, Circulating ,Molecular biology ,carbohydrates (lipids) ,030104 developmental biology ,Platelet extracellular vesicles ,Targeted drug delivery ,030220 oncology & carcinogenesis ,Drug delivery ,Cancer cell ,medicine.drug - Abstract
Background Human platelets (PLT) and PLT-extracellular vesicles (PEV) released upon thrombin activation express receptors that interact with tumour cells and, thus, can serve as a delivery platform of anti-cancer agents. Drug-loaded nanoparticles coated with PLT membranes were demonstrated to have improved targeting efficiency to tumours, but remain impractical for clinical translation. PLT and PEV targeted drug delivery vehicles should facilitate clinical developments if clinical-grade procedures can be developed. Methods PLT from therapeutic-grade PLT concentrate (PC; N > 50) were loaded with doxorubicin (DOX) and stored at − 80 °C (DOX-loaded PLT) with 6% dimethyl sulfoxide (cryopreserved DOX-loaded PLT). Surface markers and function of cryopreserved DOX-loaded PLT was confirmed by Western blot and thromboelastography, respectively. The morphology of fresh and cryopreserved naïve and DOX-loaded PLT was observed by scanning electron microscopy. The content of tissue factor-expressing cancer-derived extracellular vesicles (TF-EV) present in conditioned medium (CM) of breast cancer cells cultures was measured. The drug release by fresh and cryopreserved DOX-loaded PLT triggered by various pH and CM was determined by high performance liquid chromatography. The thrombin activated PEV was analyzed by nanoparticle tracking analysis. The cellular uptake of DOX from PLT was observed by deconvolution microscopy. The cytotoxicities of DOX-loaded PLT, cryopreserved DOX-loaded PLT, DOX and liposomal DOX on breast, lung and colon cancer cells were analyzed by CCK-8 assay. Results 15~36 × 106 molecules of DOX could be loaded in each PLT within 3 to 9 days after collection. The characterization and bioreactivity of cryopreserved DOX-loaded PLT were preserved, as evidenced by (a) microscopic observations, (b) preservation of important PLT membrane markers CD41, CD61, protease activated receptor-1, (c) functional activity, (d) reactivity to TF-EV, and (e) efficient generation of PEV upon thrombin activation. The transfer of DOX from cryopreserved PLT to cancer cells was achieved within 90 min, and stimulated by TF-EV and low pH. The cryopreserved DOX-loaded PLT formulation was 7~23-times more toxic to three cancer cells than liposomal DOX. Conclusions Cryopreserved DOX-loaded PLT can be prepared under clinically compliant conditions preserving the membrane functionality for anti-cancer therapy. These findings open perspectives for translational applications of PLT-based drug delivery systems.
- Published
- 2020
19. Upregulation of Protein Synthesis and Proteasome Degradation Confers Sensitivity to Proteasome Inhibitor Bortezomib in Myc-Atypical Teratoid/Rhabdoid Tumors
- Author
-
Yun Ru Liu, Feng Chi Chang, Muh Lii Liang, Shih-Chieh Lin, Wen Chang Huang, Tsung Han Hsieh, Donald Ming-Tak Ho, Kevin Li Chun Hsieh, Hsin Hung Chen, Meng En Chao, Huy Minh Tran, Yi Yen Lee, Yen Lin Liu, Wan Chen, Chun A. Changou, Che Chang Chang, Min Lan Tsai, Shian Ying Sung, Tai-Tong Wong, Kuo Sheng Wu, Hsin Lun Lee, Alice L. Yu, Yun Yen, and Shing Shung Chu
- Subjects
0301 basic medicine ,p53 ,Cancer Research ,protein synthesis ,Oncology and Carcinogenesis ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,Rare Diseases ,0302 clinical medicine ,Downregulation and upregulation ,Gene expression ,Genetics ,medicine ,2.1 Biological and endogenous factors ,Aetiology ,Multiple myeloma ,Cancer ,Pediatric ,Oncogene ,Chemistry ,Bortezomib ,bortezomib ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,proteasome degradation ,Orphan Drug ,030104 developmental biology ,Oncology ,5.1 Pharmaceuticals ,Apoptosis ,Cell culture ,030220 oncology & carcinogenesis ,Proteasome inhibitor ,Cancer research ,Myc-ATRTs ,Development of treatments and therapeutic interventions ,Biotechnology ,medicine.drug - Abstract
Atypical teratoid rhabdoid tumors (ATRTs) are among the most malignant brain tumors in early childhood and remain incurable. Myc-ATRT is driven by the Myc oncogene, which directly controls the intracellular protein synthesis rate. Proteasome inhibitor bortezomib (BTZ) was approved by the Food and Drug Administration as a primary treatment for multiple myeloma. This study aimed to determine whether the upregulation of protein synthesis and proteasome degradation in Myc-ATRTs increases tumor cell sensitivity to BTZ. We performed differential gene expression and gene set enrichment analysis on matched primary and recurrent patient-derived xenograft (PDX) samples from an infant with ATRT. Concomitant upregulation of the Myc pathway, protein synthesis and proteasome degradation were identified in recurrent ATRTs. Additionally, we found the proteasome-encoding genes were highly expressed in ATRTs compared with in normal brain tissues, correlated with the malignancy of tumor cells and were essential for tumor cell survival. BTZ inhibited proliferation and induced apoptosis through the accumulation of p53 in three human Myc-ATRT cell lines (PDX-derived tumor cell line Re1-P6, BT-12 and CHLA-266). Furthermore, BTZ inhibited tumor growth and prolonged survival in Myc-ATRT orthotopic xenograft mice. Our findings suggest that BTZ may be a promising targeted therapy for Myc-ATRTs.
- Published
- 2020
- Full Text
- View/download PDF
20. Synthesis and characterization of Gd-DTPA/fucoidan/peptide complex nanoparticle and in vitro magnetic resonance imaging of inflamed endothelial cells
- Author
-
Hsueh Liang Chu, Min Lang Tsai, Rou Li, Chun A. Changou, Kun Ying Lu, Che Chang Chang, Fwu Long Mi, Chun-Hua Hsu, Lee Hsin Chang, Tsai Mu Cheng, and Yu Chieh Jill Kao
- Subjects
Gadolinium DTPA ,Materials science ,Endothelium ,MRI contrast agent ,Contrast Media ,Bioengineering ,02 engineering and technology ,010402 general chemistry ,01 natural sciences ,Biomaterials ,chemistry.chemical_compound ,Polysaccharides ,medicine ,Cytotoxicity ,biology ,Fucoidan ,Endothelial Cells ,021001 nanoscience & nanotechnology ,Protamine ,Magnetic Resonance Imaging ,In vitro ,0104 chemical sciences ,medicine.anatomical_structure ,chemistry ,Mechanics of Materials ,Drug delivery ,biology.protein ,Cell-penetrating peptide ,Biophysics ,Nanoparticles ,0210 nano-technology ,Peptides - Abstract
P-selectin overexpressed on activated endothelial cells and platelets is a new target for treatment of cancers and cardiovascular diseases such as atherosclerosis and thrombosis. In this study, depolymerized low molecular weight fucoidan (LMWF8775) and a thermolysin-hydrolyzed protamine peptide (TPP1880) were prepared. TPP1880 and LMWF8775 were able to form self-assembled complex nanoparticles (CNPs). The formation of TPP1880/LMWF8775 CNPs was characterized by Fourier-transform infrared spectra, circular dichroism spectra and isothermal titration calorimetry. The CNPs selectively targeted PMA-stimulated, inflamed endothelial cells (HUVECs) with high expression of P-selectin. Gd-DTPA MRI contrast agent was successfully loaded in the CNPs with better T1 relaxivity and selectively accumulated in the activated HUVECs with increased MRI intensity and reduced cytotoxicity as compared to free Gd-DTPA. Our results suggest that the TPP1880/LMWF8775 CNPs may have potential in future for early diagnosis of cardiovascular diseases and cancers in which the endothelium is inflamed or activated.
- Published
- 2020
21. Resveratrol inhibits human leiomyoma cell proliferation via crosstalk between integrin αvβ3 and IGF-1R
- Author
-
Ya Jung Shih, Yu Tang Chin, Shwu Jiuan Lin, Heng Yuan Tang, Hung Yun Lin, Yi Ru Chen, Hsuan Liang Liu, Paul J. Davis, Szu Yi Chou, Jacqueline Whang-Peng, Yih Ho, Yu Chen Sh Yang, and Chun A. Changou
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Integrin ,Resveratrol ,Toxicology ,Receptor, IGF Type 1 ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cyclin D1 ,Stilbenes ,In Situ Nick-End Labeling ,medicine ,Humans ,Phosphorylation ,Receptor ,Cell Proliferation ,Leiomyoma ,biology ,Cell growth ,Growth factor ,food and beverages ,Receptor Cross-Talk ,General Medicine ,Flow Cytometry ,Integrin alphaVbeta3 ,Proliferating cell nuclear antigen ,030104 developmental biology ,chemistry ,Cell culture ,030220 oncology & carcinogenesis ,Uterine Neoplasms ,biology.protein ,Cancer research ,Female ,Food Science - Abstract
Leiomyomas (myomas) are the most common benign smooth muscle cell tumor of the myometrium. Resveratrol, a stilbene, has been used as an anti-inflammatory and antitumor agent. In the current study, we investigated the inhibitory effect of resveratrol on the proliferation of primary human myoma cell cultures. Resveratrol arrested cell proliferation via integrin αvβ3. It also inhibited integrin αvβ3 expression and protein accumulation. Concurrently, constitutive AKT phosphorylation in myoma cells was inhibited by resveratrol. Expressions of proapoptotic genes, such as cyclooxygenase (COX)-2, p21 and CDKN2, were induced by resveratrol in myoma cells. On the other hand, expressions of proliferative (anti-apoptotic) genes were either inhibited, as in BCL2, or unchanged, as in cyclin D1 and proliferating cell nuclear antigen (PCNA). The accumulation of insulin-like growth factor (IGF)-1 receptor (IGF-1R) was inhibited by resveratrol in primary myoma cells. IGF-1-induced cell proliferation was inhibited by co-incubation with resveratrol. Therefore, growth modulation of myoma cells occurs via mechanisms dependent on cross-talk between integrin αvβ3 and IGF-1R. Our findings suggest that resveratrol can be considered an alternative therapeutic agent for myomas.
- Published
- 2018
22. Enhancement by Nano-Diamino-Tetrac of Antiproliferative Action of Gefitinib on Colorectal Cancer Cells: Mediation by EGFR Sialylation and PI3K Activation
- Author
-
Hung Yun Lin, Shwu Huey Wang, Yi Ru Chen, Chih Hsiung Wu, Liang Shun Wang, André Wendindondé Nana, Yu Chen Sh Yang, Ya Jung Shih, Tung Cheng Chang, Jacqueline Whang-Peng, Ai Shih, Yu Tang Chin, Kuan Wang, Chun A. Changou, Yu Min Liao, Steven C. Stain, and Paul J. Davis
- Subjects
0301 basic medicine ,Cancer Research ,Colorectal cancer ,Endocrinology, Diabetes and Metabolism ,Mutant ,Mice, Nude ,Apoptosis ,Drug resistance ,03 medical and health sciences ,HT29 Cells ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Endocrinology ,Gefitinib ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,heterocyclic compounds ,skin and connective tissue diseases ,neoplasms ,Polyglactin 910 ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Original Paper ,Endocrine and Autonomic Systems ,Chemistry ,Cell growth ,medicine.disease ,HCT116 Cells ,Xenograft Model Antitumor Assays ,In vitro ,respiratory tract diseases ,Enzyme Activation ,ErbB Receptors ,Thyroxine ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Colorectal Neoplasms ,medicine.drug - Abstract
Drug resistance complicates the clinical use of gefitinib. Tetraiodothyroacetic acid (tetrac) and nano-diamino-tetrac (NDAT) have been shown in vitro and in xenografts to have antiproliferative/angiogenic properties and to potentiate antiproliferative activity of other anticancer agents. In the current study, we investigated the effects of NDAT on the anticancer activities of gefitinib in human colorectal cancer cells. β-Galactoside α-2,6-sialyltransferase 1 (ST6Gal1) catalyzes EGFR sialylation that is associated with gefitinib resistance in colorectal cancers, and this was also investigated. Gefitinib inhibited cell proliferation of HT-29 cells (K-ras wild-type), and NDAT significantly enhanced the antiproliferative action of gefitinib. Gefitinib inhibited cell proliferation of HCT116 cells (K-ras mutant) only in high concentration, and this was further enhanced by NDAT. NDAT enhancedd gefitinib-induced antiproliferation in gefitinib-resistant colorectal cancer cells by inhibiting ST6Gal1 activity and PI3K activation. Furthermore, NDAT enhanced gefitinib-induced anticancer activity additively in colorectal cancer HCT116 cell xenograft-bearing nude mice. Results suggest that NDAT may have an application with gefitinib as combination colorectal cancer therapy.
- Published
- 2018
23. The role of sentrin-specific protease 2 substrate recognition in TGF-β-induced tumorigenesis
- Author
-
Chia Ju Lin, Tsai Ling Ho, Yen Sung Huang, Ruei Ting Chang, Hsiu-Ming Shih, Jen Chong Jeng, Ying Mei Lin, Shin Mei Liu, Chun Chen Ho, Chun A. Changou, and Che Chang Chang
- Subjects
0301 basic medicine ,Cellular pathology ,Carcinogenesis ,SUMO protein ,lcsh:Medicine ,Matrix metalloproteinase ,medicine.disease_cause ,Article ,Substrate Specificity ,03 medical and health sciences ,Cell Movement ,Transforming Growth Factor beta ,Spheroids, Cellular ,medicine ,Humans ,Author Correction ,lcsh:Science ,Psychological repression ,Smad4 Protein ,Multidisciplinary ,biology ,Chemistry ,lcsh:R ,Sumoylation ,Cell migration ,Transforming growth factor beta ,Cell biology ,Cysteine Endopeptidases ,030104 developmental biology ,biology.protein ,lcsh:Q ,Transforming growth factor ,Protein Binding ,Signal Transduction - Abstract
Smad4, a common-mediator of Smads, plays a central role in forming complexes with receptor-phosphorylated Smads, and then transduces transforming growth factor (TGF)-β signals into the nuclei. Although many cellular factors are involved in TGF-β induced epithelial-to-mesenchymal transition (EMT) and cell migration, very little is known with the mechanism of Smad4 regulation on pro-oncogenes response by TGF-β. Herein, we demonstrate the interaction of Sentrin-specific protease 2 (SENP2) with Smad4 through SENP2 residue 363~400. The same segment is also important for desumoylation of Smad4, and able to relieve sumoylation-mediated TGF-β repression. The SENP2363~400 segment is critical for TGF-β-induced cell migration, which is correlated with SENP2363~400 deletion mutant failed to increase matrix metalloproteinase (MMP)-9 and EMT marker gene expression. Moreover, our results suggest that the interaction and desumoylation between SENP2 and Smad4 promote cell migration in triple-negative breast cancer cells. Altogether, our data show how SENP2 regulates its substrate for desumoylation, and also the role of SENP2 in TGF-β induced cancer cell migration.
- Published
- 2018
24. Thyroxine inhibits resveratrol-caused apoptosis by PD-L1 in ovarian cancer cells
- Author
-
Chun A. Changou, Yu Tang Chin, Yih Ho, Yu Chen Sh Yang, Meng Ti Hsieh, André Wendindondé Nana, Ya Jung Shih, Po Li Wei, Paul J. Davis, Aleck Hercbergs, Hung Yun Lin, and Yi Ru Chen
- Subjects
0301 basic medicine ,Cancer Research ,Endocrinology, Diabetes and Metabolism ,Apoptosis ,Resveratrol ,Transfection ,B7-H1 Antigen ,Small hairpin RNA ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Endocrinology ,Gene expression ,medicine ,Humans ,Ovarian Neoplasms ,Gene knockdown ,Chemistry ,Cell growth ,medicine.disease ,Thyroxine ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Female ,Ovarian cancer - Abstract
Thyroid hormone,l-thyroxine (T4), has been shown to promote ovarian cancer cell proliferation via a receptor on plasma membrane integrin αvβ3 and to induce the activation of ERK1/2 and expression of programmed death-ligand 1 (PD-L1) in cancer cells. In contrast, resveratrol binds to integrin αvβ3 at a discrete site and induces p53-dependent antiproliferation in malignant neoplastic cells. The mechanism of resveratrol action requires nuclear accumulation of inducible cyclooxygenase (COX)-2 and its complexation with phosphorylated ERK1/2. In this study, we examined the mechanism by which T4impairs resveratrol-induced antiproliferation in human ovarian cancer cells and found that T4inhibited resveratrol-induced nuclear accumulation of COX-2. Furthermore, T4increased expression and cytoplasmic accumulation of PD-L1, which in turn acted to retain inducible COX-2 in the cytoplasm. Knockdown ofPD-L1by small hairpin RNA (shRNA) relieved the inhibitory effect of T4on resveratrol-induced nuclear accumulation of COX-2- and COX-2/p53-dependent gene expression. Thus, T4inhibits COX-2-dependent apoptosis in ovarian cancer cells by retaining inducible COX-2 with PD-L1 in the cytoplasm. These findings provide new insights into the antagonizing effect of T4on resveratrol’s anticancer properties.
- Published
- 2018
25. Dual Inhibition of PIK3C3 and FGFR as a New Therapeutic Approach to Treat Bladder Cancer
- Author
-
Kai Cheng Hsu, Yu Chen Lin, Hao Ching Wang, Yan Ni Lo, Yun Yen, Chun A. Changou, Cheng Ying Chu, Yu Ching Lee, Chun Han Chen, Jing Ping Liou, Tsung Han Hsieh, and Yun Ru Liu
- Subjects
0301 basic medicine ,Cancer Research ,Programmed cell death ,DNA damage ,Drug Evaluation, Preclinical ,Vacuole ,Autophagy-Related Protein 5 ,Small hairpin RNA ,Gene Knockout Techniques ,03 medical and health sciences ,Cell Line, Tumor ,Autophagy ,medicine ,Humans ,Triazines ,Chemistry ,Phenylurea Compounds ,Cancer ,medicine.disease ,Class III Phosphatidylinositol 3-Kinases ,Receptors, Fibroblast Growth Factor ,Cell biology ,Molecular Docking Simulation ,Pyrimidines ,030104 developmental biology ,Urinary Bladder Neoplasms ,Oncology ,Drug Resistance, Neoplasm ,Cell culture ,Ectopic expression ,Cisplatin ,Protein Binding - Abstract
Purpose: MPT0L145 has been developed as a FGFR inhibitor exhibiting significant anti-bladder cancer activity in vitro and in vivo via promoting autophagy-dependent cell death. Here, we aim to elucidate the underlying mechanisms. Experimental Design: Autophagy flux, morphology, and intracellular organelles were evaluated by Western blotting, transmission electron microscope, and fluorescence microscope. Molecular docking and surface plasmon resonance assay were performed to identify drug–protein interaction. Lentiviral delivery of cDNA or shRNA and CRISPR/Cas9-mediated genome editing was used to modulate gene expression. Mitochondrial oxygen consumption rate was measured by a Seahorse XFe24 extracellular flux analyzer, and ROS level was measured by flow cytometry. Results: MPT0L145 persistently increased incomplete autophagy and phase-lucent vacuoles at the perinuclear region, which were identified as enlarged and alkalinized late-endosomes. Screening of a panel of lipid kinases revealed that MPT0L145 strongly inhibits PIK3C3 with a Kd value of 0.53 nmol/L. Ectopic expression of PIK3C3 reversed MPT0L145-increased cell death and incomplete autophagy. Four residues (Y670, F684, I760, D761) at the ATP-binding site of PIK3C3 are important for the binding of MPT0L145. In addition, MPT0L145 promotes mitochondrial dysfunction, ROS production, and DNA damage, which may in part, contribute to cell death. ATG5-knockout rescued MPT0L145-induced cell death, suggesting simultaneous induction of autophagy is crucial to its anticancer activity. Finally, our data demonstrated that MPT0L145 is able to overcome cisplatin resistance in bladder cancer cells. Conclusions: MPT0L145 is a first-in-class PIK3C3/FGFR inhibitor, providing an innovative strategy to design new compounds that increase autophagy, but simultaneously perturb its process to promote bladder cancer cell death. Clin Cancer Res; 24(5); 1176–89. ©2017 AACR.
- Published
- 2018
26. Leptin OB3 peptide suppresses leptin-induced signaling and progression in ovarian cancer cells
- Author
-
Yu Tang Chin, Earl Fu, Le Ming Wang, André Wendindondé Nana, Meng Ti Hsieh, Hung Yun Lin, Ya Jung Shih, Chun A. Changou, Yu Chen S.H. Yang, Heng Yuan Tang, Paul J. Davis, and Hsien Chung Chiu
- Subjects
0301 basic medicine ,Male ,Leptin ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Estrogen receptor ,Mice, Nude ,lcsh:Medicine ,03 medical and health sciences ,0302 clinical medicine ,Ovarian cancer ,Internal medicine ,medicine ,Animals ,Humans ,Pharmacology (medical) ,Obesity ,Molecular Biology ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Ovarian Neoplasms ,Mice, Inbred BALB C ,OB3-leptin peptide ,Leptin receptor ,Chemistry ,Kinase ,Research ,Biochemistry (medical) ,digestive, oral, and skin physiology ,lcsh:R ,Cell Biology ,General Medicine ,medicine.disease ,Peptide Fragments ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Endocrinology ,030220 oncology & carcinogenesis ,Cancer cell ,Female ,Signal transduction ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
Background Obesity and its comorbidities constitute a serious health burden worldwide. Leptin plays an important role in diet control; however, it has a stimulatory potential on cancer cell proliferation. The OB3 peptide, a synthetic peptide, was shown to be more active than leptin in regulating metabolism but with no mitogenic effects in cancer cells. Methods In this study, we investigated the proliferative effects, gene expressions and signaling pathways modulated by leptin and OB3 in human ovarian cancer cells. In addition, an animal study was performed. Results Leptin, but not OB3, induced the proliferation of ovarian cancer cells. Interestingly, OB3 blocked the leptin-induced proliferative effect when it was co-applied with leptin. Both leptin and OB3 activated the phosphatidylinositol-3-kinase (PI3K) signal transduction pathway. In addition, leptin stimulated the phosphorylation of signal transducer and activator of transcription-3 (STAT3) Tyr-705 as well as estrogen receptor (ER)α, and the expression of ERα-responsive genes. Interestingly, all leptin-induced signal activation and gene expressions were blocked by the co-incubation with OB3 and the inhibition of extracellular signal-regulated kinase (ERK)1/2. Coincidently, leptin, but not OB3, increased circulating levels of follicle-stimulating hormone (FSH) which is known to play important roles in the initiation and proliferation of ovarian cancer cells. Conclusions In summary, our findings suggest that the OB3 peptide may prevent leptin-induced ovarian cancer initiation and progression by disrupting leptin-induced proliferative signals via STAT3 phosphorylation and ERα activation. Therefore, the OB3 peptide is a potential anticancer agent that might be employed to prevent leptin-induced cancers in obese people.
- Published
- 2017
27. Long noncoding RNA LncHIFCAR/MIR31HG is a HIF-1α co-activator driving oral cancer progression
- Author
-
Ming Heng Wu, Hsing Jien Kung, Yu Wen Hung, Wen Chang Wang, Chun A. Changou, Ling Yu Wang, Cheng Ying Chu, Wei Fan Chiang, Yun Yen, Alexander T.H. Wu, Jing Wen Shih, Chiu-Lien Hung, and Yen Ling Yu
- Subjects
0301 basic medicine ,Science ,Mice, Nude ,General Physics and Astronomy ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Transactivation ,Downregulation and upregulation ,Biomarkers, Tumor ,Animals ,Humans ,Proportional Hazards Models ,Regulation of gene expression ,Genetics ,Gene knockdown ,Multidisciplinary ,RNA ,Promoter ,General Chemistry ,Middle Aged ,Gene signature ,Hypoxia-Inducible Factor 1, alpha Subunit ,Prognosis ,Survival Analysis ,Xenograft Model Antitumor Assays ,Long non-coding RNA ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Cancer research ,Tumor Hypoxia ,Female ,Mouth Neoplasms ,RNA, Long Noncoding - Abstract
Long noncoding RNAs (lncRNAs) have been implicated in hypoxia/HIF-1-associated cancer progression through largely unknown mechanisms. Here we identify MIR31HG as a hypoxia-inducible lncRNA and therefore we name it LncHIFCAR (long noncoding HIF-1α co-activating RNA); we describe its oncogenic role as a HIF-1α co-activator that regulates the HIF-1 transcriptional network, crucial for cancer development. Extensive analyses of clinical data indicate LncHIFCAR level is substantially upregulated in oral carcinoma, significantly associated with poor clinical outcomes and representing an independent prognostic predictor. Overexpression of LncHIFCAR induces pseudo-hypoxic gene signature, whereas knockdown of LncHIFCAR impairs the hypoxia-induced HIF-1α transactivation, sphere-forming ability, metabolic shift and metastatic potential in vitro and in vivo. Mechanistically, LncHIFCAR forms a complex with HIF-1α via direct binding and facilitates the recruitment of HIF-1α and p300 cofactor to the target promoters. Our results uncover an lncRNA-mediated mechanism for HIF-1 activation and establish the clinical values of LncHIFCAR in prognosis and potential therapeutic strategy for oral carcinoma., Cancer cells adapt to the changing microenvironment by activating different pathways through multiple mechanisms. Here the authors identify long noncoding RNA MIR31HG as a HIF-1α co-activator required for the induction of the hypoxic response and show its oncogenic role in oral carcinogenesis.
- Published
- 2017
28. The CD9, CD81, and CD151 EC2 domains bind to the classical RGD-binding site of integrin αvβ3
- Author
-
Chun A. Changou, Yoko K. Takada, Jessica Yu, Yoshikazu Takada, Chia Ying Lee, and Dora M. Cedano-Prieto
- Subjects
Protein Conformation, alpha-Helical ,0301 basic medicine ,Integrin ,Gene Expression ,CHO Cells ,Tetraspanin 24 ,Biochemistry ,Tetraspanin 29 ,Tetraspanin 28 ,03 medical and health sciences ,Cricetulus ,0302 clinical medicine ,Tetraspanin ,Escherichia coli ,Animals ,Protein Interaction Domains and Motifs ,Amino Acid Sequence ,Cloning, Molecular ,Binding site ,Receptor ,Cell adhesion ,Molecular Biology ,CD151 ,Binding Sites ,Sequence Homology, Amino Acid ,biology ,Antibodies, Monoclonal ,Cell Biology ,Integrin alphaVbeta3 ,Recombinant Proteins ,Cell biology ,Molecular Docking Simulation ,Kinetics ,030104 developmental biology ,Docking (molecular) ,030220 oncology & carcinogenesis ,embryonic structures ,biology.protein ,Protein Conformation, beta-Strand ,Sequence Alignment ,Protein Binding ,CD81 - Abstract
Tetraspanins play important roles in normal (e.g. cell adhesion, motility, activation, and proliferation) and pathological conditions (e.g. metastasis and viral infection). Tetraspanins interact with integrins and regulate integrin functions, but the specifics of tetraspanin–integrin interactions are unclear. Using co-immunoprecipitation with integrins as a sole method to detect interaction between integrins and full-length tetraspanins, it has been proposed that the variable region (helices D and E) of the extracellular-2 (EC2) domain of tetraspanins laterally associates with a non-ligand-binding site of integrins. We describe that, using adhesion assays, the EC2 domain of CD81, CD9, and CD151 bound to integrin αvβ3, and this binding was suppressed by cRGDfV, a specific inhibitor of αvβ3, and antibody 7E3, which is mapped to the ligand-binding site of β3. We also present evidence that the specificity loop of β3 directly bound to the EC2 domains. This suggests that the EC2 domains specifically bind to the classical ligand-binding site of αvβ3. αvβ3 was a more effective receptor for the EC2 domains than the previously known tetraspanin receptors α3β1, α4β1, and α6β1. Docking simulation predicted that the helices A and B of CD81 EC2 bind to the RGD-binding site of αvβ3. Substituting Lys residues at positions 116 and 144/148 of CD81 EC2 in the predicted integrin-binding interface reduced the binding of CD81 EC2 to αvβ3, consistent with the docking model. These findings suggest that, in contrast with previous models, the ligand-binding site of integrin αvβ3, a new tetraspanin receptor, binds to the constant region (helices A and B) of the EC2 domain.
- Published
- 2017
29. Author Correction: The role of sentrin-specific protease 2 substrate recognition in TGF-β-induced tumorigenesis
- Author
-
Jen Chong Jeng, Tsai Ling Ho, Yen Sung Huang, Ruei Ting Chang, Ying Mei Lin, Chia Ju Lin, Hsiu-Ming Shih, Che Chang Chang, Shin Mei Liu, Chun A. Changou, and Chun Chen Ho
- Subjects
Multidisciplinary ,Protease ,Chemistry ,medicine.medical_treatment ,lcsh:R ,lcsh:Medicine ,Substrate recognition ,medicine.disease_cause ,Cell biology ,medicine ,lcsh:Q ,Carcinogenesis ,lcsh:Science ,Transforming growth factor - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2020
30. Host immune response to anti-cancer camptothecin conjugated cyclodextrin-based polymers
- Author
-
Mark E. Davis, Chun A. Changou, Yi Fan Chen, Yen Hsin Wang, Cing Syuan Lei, and Yun Yen
- Subjects
0301 basic medicine ,Endocrinology, Diabetes and Metabolism ,T cell ,Clinical Biochemistry ,Central nervous system ,lcsh:Medicine ,Antineoplastic Agents ,Review ,Immune responses ,Adaptive Immunity ,Pharmacology ,Efficacy ,Mice ,03 medical and health sciences ,Nanoparticle ,0302 clinical medicine ,Immune system ,Animals ,Medicine ,Pharmacology (medical) ,Cellulose ,Adverse effect ,Molecular Biology ,Cyclodextrins ,business.industry ,lcsh:R ,Biochemistry (medical) ,Cancer ,Cell Biology ,General Medicine ,medicine.disease ,Immunity, Innate ,Specific Pathogen-Free Organisms ,Brain tumor ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Cancer cell ,Nanoparticles ,Camptothecin ,business ,medicine.drug - Abstract
Introduction Efficacy and safety are critical concerns when designing drug carriers. Nanoparticles are a particular type of carrier that has gained recent attention in cancer therapeutics. Methods In this study, we assess the safety profile of IT-101, a nanoparticle formed by self-assembly of camptothecin (CPT) conjugated cyclodextrin-based polymers. IT-101 delivers CPT to target cancer cells in animal models of numerous human cancers and in humans. Previous data from preclinical and clinical trials indicate that IT-101 has no notable immunological side effects. However, there have been no published studies focused on evaluating the effects of IT-101 on host immune systems. Results In this work, we demonstrate that IT-101 diminished initial host immune response following first injection of the nanopharmaceutical and induced NK cell activation and T cell proliferation upon further IT-101 exposure. Additionally, IT-101 could attenuate tumor growth more efficiently than CPT treatment only. Conclusions Drugs administration in whole-body circulation may lead to poorly bioavailable in central nervous system and often has toxic effects on peripheral tissues. Conjugated with cyclodextrin-based polymers not only reduce adverse effects but also modulate the immune responses to elevate drug efficacy. These immune responses may potentially facilitate actions of immune blockage, such as PD1/PDL1 in cancer treatment.
- Published
- 2019
31. NDAT suppresses pro-inflammatory gene expression to enhance resveratrol-induced anti-proliferation in oral cancer cells
- Author
-
Yih Ho, Shaker A. Mousa, Hung Yun Lin, Yu Tang Chin, Chien Yi Wu, Tung Yung Huang, Kuan Wang, Sheng Yang Lee, Paul J. Davis, Ya Jung Shih, Chun A. Changou, Yi Shin Pan, Zi Lin Li, Jaqulene Whang-Peng, Po Yu Su, Dana R. Crawford, Kuan Wei Su, Yu Chen S.H. Yang, Yi Ru Chen, and Hsien Chung Chiu
- Subjects
STAT3 Transcription Factor ,Gene Expression ,Resveratrol ,Toxicology ,B7-H1 Antigen ,03 medical and health sciences ,chemistry.chemical_compound ,0404 agricultural biotechnology ,Cyclin D1 ,Cell Line, Tumor ,Gene expression ,Humans ,STAT3 ,Gene ,Polyglactin 910 ,Inflammatory genes ,030304 developmental biology ,Cell Proliferation ,0303 health sciences ,biology ,Drug Synergism ,04 agricultural and veterinary sciences ,General Medicine ,Anti proliferative ,040401 food science ,Thyroxine ,chemistry ,Cyclooxygenase 2 ,Cancer cell ,Cancer research ,biology.protein ,Mouth Neoplasms ,Food Science - Abstract
Nano-diamino-tetrac (NDAT), a tetraiodothyroxine deaminated nano-particulated analog, has shown to inhibit expression of pro-inflammatory genes. NDAT inhibits expression of programmed death-ligand 1 (PD-L1). On the other hand, in addition to inhibiting inflammatory effect, the stilbene, resveratrol induces expression of cyclooxygenase-2 (COX-2) and its accumulation. Sequentially, inducible COX-2 complexes with p53 and induces p53-dependent anti-proliferation. In current study, we investigated mechanisms involved in combined treatment of NDAT and resveratrol on anti-proliferation in human oral cancer cells. Both resveratrol and NDAT inhibited expression of pro-inflammatory IL-1β and TNF-α. They also inhibited expression of CCND1 and PD-L1. Both resveratrol and NDAT induced BAD expression but only resveratrol induced COX-2 expression in both OEC-M1 and SCC-25 cells. Combined treatment attenuated gene expression significantly compared with resveratrol treatment in both cancer cell lines. Resveratrol reduced nuclear PD-L1 accumulation which was enhanced by a STAT3 inhibitor, S31-201 or NDAT suggesting that NDAT may inactivate STAT3 to inhibit PD-L1 accumulation. In the presence of T4, NDAT further enhanced resveratrol-induced anti-proliferation in both cancer cell lines. These findings provide a novel understanding of the inhibition of NDAT in thyroxine-induced pro-inflammatory effect on resveratrol-induced anticancer properties.
- Published
- 2019
32. Characterization of FN1–FGFR1 and novel FN1–FGF1 fusion genes in a large series of phosphaturic mesenchymal tumors
- Author
-
Sheng Yao Su, Andrew L. Folpe, Michael T. Collins, Eiichi Konishi, Yoshinao Oda, Thomas O. Carpenter, Steven D. Billings, Eric S. Orwoll, Shyang-Rong Shih, Kesavan Sittampalam, Rong-Sen Yang, Shu Hwa Chen, Fredrik Petersson, Jen-Chieh Lee, Cheng-Han Lee, G. Petur Nielsen, Hsuan-Ying Huang, Chung Yen Lin, Chun A. Changou, Keh-Sung Tsai, and Chien-Feng Li
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Oncogene Proteins, Fusion ,Angiogenesis ,Bone Neoplasms ,Soft Tissue Neoplasms ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,Fusion gene ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Receptor, Fibroblast Growth Factor, Type 1 ,Gene ,Fibroblast growth factor receptor 1 ,Mesenchymal stem cell ,Phosphaturic mesenchymal tumor ,Fibronectins ,Fibroblast Growth Factors ,Fibroblast Growth Factor-23 ,stomatognathic diseases ,030104 developmental biology ,030220 oncology & carcinogenesis ,Fibroblast Growth Factor 1 ,Immunohistochemistry ,Carcinogenesis - Abstract
Phosphaturic mesenchymal tumors typically cause paraneoplastic osteomalacia, chiefly as a result of FGF23 secretion. In a prior study, we identified FN1-FGFR1 fusion in 9 of 15 phosphaturic mesenchymal tumors. In this study, a total of 66 phosphaturic mesenchymal tumors and 7 tumors resembling phosphaturic mesenchymal tumor but without known phosphaturia were studied. A novel FN1-FGF1 fusion gene was identified in two cases without FN1-FGFR1 fusion by RNA sequencing and cross-validated with direct sequencing and western blot. Fluorescence in situ hybridization analyses revealed FN1-FGFR1 fusion in 16 of 39 (41%) phosphaturic mesenchymal tumors and identified an additional case with FN1-FGF1 fusion. The two fusion genes were mutually exclusive. Combined with previous data, the overall prevalence of FN1-FGFR1 and FN1-FGF1 fusions was 42% (21/50) and 6% (3/50), respectively. FGFR1 immunohistochemistry was positive in 82% (45/55) of phosphaturic mesenchymal tumors regardless of fusion status. By contrast, 121 cases of potential morphologic mimics (belonging to 13 tumor types) rarely expressed FGFR1, the main exceptions being solitary fibrous tumors (positive in 40%), chondroblastomas (40%), and giant cell tumors of bone (38%), suggesting a possible role for FGFR1 immunohistochemistry in the diagnosis of phosphaturic mesenchymal tumor. With the exception of one case reported in our prior study, none of the remaining tumors resembling phosphaturic mesenchymal tumor had either fusion type or expressed significant FGFR1. Our findings provide insight into possible mechanisms underlying the pathogenesis of phosphaturic mesenchymal tumor and imply a central role of the FGF1-FGFR1 signaling pathway. The novel FN1-FGF1 protein is expected to be secreted and serves as a ligand that binds and activates FGFR1 to achieve an autocrine loop. Further study is required to determine the functions of these fusion proteins.
- Published
- 2016
33. Genomewide copy number analysis of Müllerian adenosarcoma identified chromosomal instability in the aggressive subgroup
- Author
-
Alexandra Lauria, Tzu-Pin Lu, Ming-Chieh Lin, Hsuan-Ying Huang, Chin-Yao Lin, Jen-Chieh Lee, Ying-Cheng Chiang, Chun A. Changou, Hsien-Neng Huang, Cher-Wei Liang, Kuan-Ting Kuo, and Ben Davidson
- Subjects
Adult ,0301 basic medicine ,medicine.medical_specialty ,Pathology ,DNA Copy Number Variations ,Gene Dosage ,Copy number analysis ,Biology ,Molecular Inversion Probe ,Chromosome Painting ,Pathology and Forensic Medicine ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,CDKN2A ,Chromosome instability ,Biomarkers, Tumor ,medicine ,Humans ,Genetic Predisposition to Disease ,Copy-number variation ,Mullerian Ducts ,Chromosome 12 ,Aged ,Chromothripsis ,Paraffin Embedding ,Adenosarcoma ,Cytogenetics ,Middle Aged ,Immunohistochemistry ,Phenotype ,030104 developmental biology ,030220 oncology & carcinogenesis ,Uterine Neoplasms ,Female ,Genome-Wide Association Study - Abstract
Müllerian adenosarcomas are malignant gynecologic neoplasms. Advanced staging and sarcomatous overgrowth predict poor prognosis. Because the genomic landscape remains poorly understood, we conducted this study to characterize the genomewide copy number variations in adenosarcomas. Sixteen tumors, including eight with and eight without sarcomatous overgrowth, were subjected to a molecular inversion probe array analysis. Copy number variations, particularly losses, were significantly higher in cases with sarcomatous overgrowth. Frequent gains of chromosomal 12q were noted, often involving cancer-associated genes CDK4 (six cases), MDM2, CPM, YEATS4, DDIT3, GLI1 (five each), HMGA2 and STAT6 (four), without association with sarcomatous overgrowth status. The most frequent losses involved chromosomes 13q (five cases), 9p, 16q and 17q (four cases each) and were almost limited to cases with sarcomatous overgrowth. MDM2 and CDK4 amplification, as well as losses of RB1 (observed in two cases) and CDKN2A/B (one case), was verified by FISH. By immunohistochemistry, all MDM2/CDK4-coamplified cases were confirmed to overexpress both encoded proteins, whereas all four cases with (plus an additional four without) gain of HMGA2 overexpressed the HMGA2 protein. Both cases with RB1 loss were negative for the immunostaining of the encoded protein. Chromothripsis-like copy number profiles involving chromosome 12 or 14 were observed in three fatal cases, all of which harbored sarcomatous overgrowth. With whole chromosome painting and deconvolution fluorescent microscopy, dividing tumor cells in all three cases were shown to have scattered extrachromosomal materials derived from chromosomes involved by chromothripsis, suggesting that this phenomenon may serve as visual evidence for chromothripsis in paraffin tissue. In conclusion, we identified frequent chromosome 12q amplifications, including loci containing potential pharmacological targets. Global chromosomal instability and chromothripsis were more frequent in cases with sarcomatous overgrowth. To our knowledge, this is the first time that evidence of chromothripsis has been demonstrated in paraffin-embedded clinical tissues and in adenosarcomas.
- Published
- 2016
34. Molecular-Clinical Correlation in Pediatric Medulloblastoma: A Cohort Series Study of 52 Cases in Taiwan
- Author
-
Shing Shung Chu, Muh Lii Liang, Yi Yen Lee, Feng Chi Chang, Wen Chang Huang, Che Chang Chang, Huy Minh Tran, Tai-Tong Wong, Yun Ru Liu, Er Chieh Cho, Yen Lin Liu, Wan Chen, Hsin Hung Chen, Shih-Chieh Lin, Kuo Sheng Wu, Meng En Chao, Donald Ming-Tak Ho, Alice L. Yu, Tsung Han Hsieh, Chun A. Changou, Min Lan Tsai, Shian Ying Sung, Kevin Li Chun Hsieh, Hsin Lun Lee, Shiann-Tarng Jou, Kung Hao Liang, and Yi Wei Chen
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,risk stratification ,DNA damage response ,Metastasis ,0302 clinical medicine ,2.1 Biological and endogenous factors ,RNA-Seq ,Aetiology ,Exome sequencing ,Cancer ,Pediatric ,screening and diagnosis ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Detection ,030220 oncology & carcinogenesis ,Cohort ,DNA methylation ,Pediatric Research Initiative ,medicine.medical_specialty ,Pediatric Cancer ,Oncology and Carcinogenesis ,medulloblastoma ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,Rare Diseases ,Germline mutation ,Clinical Research ,Internal medicine ,Genetics ,molecular-clinical correlation ,medicine ,Genetic predisposition ,Genetic Testing ,Gene ,Medulloblastoma ,business.industry ,Human Genome ,medicine.disease ,Brain Disorders ,4.1 Discovery and preclinical testing of markers and technologies ,Brain Cancer ,Good Health and Well Being ,molecular–clinical correlation ,030104 developmental biology ,somatic mutations ,business ,genetic predisposition - Abstract
In 2016, a project was initiated in Taiwan to adopt molecular diagnosis of childhood medulloblastoma (MB). In this study, we aimed to identify a molecular-clinical correlation and somatic mutation for exploring risk-adapted treatment, drug targets, and potential genetic predisposition. In total, 52 frozen tumor tissues of childhood MBs were collected. RNA sequencing (RNA-Seq) and DNA methylation array data were generated. Molecular subgrouping and clinical correlation analysis were performed. An adjusted Heidelberg risk stratification scheme was defined for updated clinical risk stratification. We selected 51 genes for somatic variant calling using RNA-Seq data. Relevant clinical findings were defined. Potential drug targets and genetic predispositions were explored. Four core molecular subgroups (WNT, SHH, Group 3, and Group 4) were identified. Genetic backgrounds of metastasis at diagnosis and extent of tumor resection were observed. The adjusted Heidelberg scheme showed its applicability. Potential drug targets were detected in the pathways of DNA damage response. Among the 10 patients with SHH MBs analyzed using whole exome sequencing studies, five patients exhibited potential genetic predispositions and four patients had relevant germline mutations. The findings of this study provide valuable information for updated risk adapted treatment and personalized care of childhood MBs in our cohort series and in Taiwan.
- Published
- 2020
35. Optical imaging of ovarian cancer using a matrix metalloproteinase-3-sensitive near-infrared fluorescent probe
- Author
-
Kuo Hwa Wang, Kuan Chou Chen, Yu Hui Tsai, Tze Chien Chen, Yung Ming Wang, Chun S. Zuo, Li Hsuan Chiu, Chun A. Changou, and Wen Fu T. Lai
- Subjects
0301 basic medicine ,Fluorescence-lifetime imaging microscopy ,endocrine system diseases ,lcsh:Medicine ,Biochemistry ,Diagnostic Radiology ,0302 clinical medicine ,Animal Cells ,Medicine and Health Sciences ,lcsh:Science ,Connective Tissue Cells ,Liquid Chromatography ,Metalloproteinase ,Multidisciplinary ,Chemistry ,Radiology and Imaging ,Chromatographic Techniques ,Proteases ,Magnetic Resonance Imaging ,female genital diseases and pregnancy complications ,Ovarian Cancer ,Enzymes ,In Vivo Imaging ,Oncology ,Connective Tissue ,030220 oncology & carcinogenesis ,Cellular Types ,Anatomy ,Preclinical imaging ,Research Article ,Stromal cell ,Imaging Techniques ,Research and Analysis Methods ,03 medical and health sciences ,In vivo ,Diagnostic Medicine ,Fluorescence Imaging ,medicine ,Cancer Detection and Diagnosis ,lcsh:R ,Cancer ,Cancers and Neoplasms ,Biology and Life Sciences ,Proteins ,Cell Biology ,medicine.disease ,High Performance Liquid Chromatography ,030104 developmental biology ,Biological Tissue ,Cancer research ,Enzymology ,Metalloproteases ,lcsh:Q ,Molecular imaging ,Stromal Cells ,Ovarian cancer ,Gynecological Tumors - Abstract
Epithelial ovarian cancer (EOC) is the seventh most common cancer among women worldwide. The 5-year survival rate for women with EOC is only 30%-50%, which is largely due to the typically late diagnosis of this condition. EOC is difficult to detect in its early stage because of its asymptomatic nature. Recently, near-infrared fluorescent (NIRF) imaging has been developed as a potential tool for detecting EOC at the molecular level. In this study, a NIRF-sensitive probe was designed to detect matrix metalloproteinase (MMP) activity in ovarian cancer cells. A cyanine fluorochrome was conjugated to the amino terminus of a peptide substrate with enzymatic specificity for MMP-3. To analyze the novel MMP-3 probe, an in vivo EOC model was established by subcutaneously implanting SKOV3 cells, a serous-type EOC cell line, in mice. This novel MMP-3-sensitive probe specifically reacted with only the active MMP-3 enzyme, resulting in a significantly enhanced NIRF emission intensity. Histological analysis demonstrated that MMP-3 expression and activity were enhanced in the stromal cells surrounding the ovarian cancer cells. These studies establish a molecular imaging reporter for diagnosing early-stage EOC. Additional studies are required to confirm the early-stage activity of MMP-3 in EOC and its diagnostic and prognostic significance.
- Published
- 2018
36. Tetrac downregulates β-catenin and HMGA2 to promote the effect of resveratrol in colon cancer
- Author
-
Ya Jung Shih, Yu Tang Chin, Wen Shan Li, Hung Yun Lin, James A. Bennett, Earl Fu, Leroy F. Liu, Jens Z. Pedersen, Chi-Yu Lin, Sandra Incerpi, Chun A. Changou, Jacqueline Whang-Peng, Shaker A. Mousa, Paul J. Davis, André Wendindondé Nana, Yih Ho, Yi Ru Chen, Nana, André Wendindondé, Chin, Yu-Tang, Lin, Chi-Yu, Ho, Yih, Bennett, James A, Shih, Ya-Jung, Chen, Yi-Ru, Changou, Chun A, Pedersen, Jens Z, Incerpi, Sandra, Liu, Leroy F, Whang-Peng, Jacqueline, Fu, Earl, Li, Wen-Shan, Mousa, Shaker A, Lin, Hung-Yun, and Davis, Paul J
- Subjects
0301 basic medicine ,Cancer Research ,Beta-catenin ,HMGA2 ,Endocrinology, Diabetes and Metabolism ,Down-Regulation ,Mice, Nude ,colorectal cancer ,Antineoplastic Agents ,Resveratrol ,resveratrol ,Settore BIO/09 ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Endocrinology ,Cell Line, Tumor ,Animals ,Humans ,tetraiodothyroacetic acid ,beta Catenin ,Cell Proliferation ,biology ,Cell growth ,HMGA2 Protein ,Wnt signaling pathway ,Drug Synergism ,β-catenin ,colorectal cancers ,Gene Expression Regulation, Neoplastic ,Thyroxine ,030104 developmental biology ,Oncology ,chemistry ,Apoptosis ,030220 oncology & carcinogenesis ,Catenin ,Cancer cell ,Colonic Neoplasms ,biology.protein ,Cancer research ,Female - Abstract
The molecular pathogenesis of colorectal cancer encompasses the activation of several oncogenic signaling pathways that include the Wnt/β-catenin pathway and the overexpression of high mobility group protein A2 (HMGA2). Resveratrol – the polyphenolic phytoalexin – binds to integrin αvβ3 to induce apoptosis in cancer cellsviacyclooxygenase 2 (COX-2) nuclear accumulation and p53-dependent apoptosis. Tetraiodothyroacetic acid (tetrac) is a de-aminated derivative ofl-thyroxine (T4), which – in contrast to the parental hormone – impairs cancer cell proliferation. In the current study, we found that tetrac promoted resveratrol-induced anti-proliferation in colon cancer cell lines, in primary cultures of colon cancer cells, andin vivo. The mechanisms implicated in this action involved the downregulation of nuclear β-catenin and HMGA2, which are capable of compromising resveratrol-induced COX-2 nuclear translocation. Silencing of either β-catenin or HMGA2 promoted resveratrol-induced anti-proliferation and COX-2 nuclear accumulation which is essential for integrin αvβ3-mediated-resveratrol-induced apoptosis in cancer cells. Concurrently, tetrac enhanced nuclear abundance of chibby family member 1, the nuclear β-catenin antagonist, which may further compromise the nuclear β-catenin-dependent gene expression and proliferation. Taken together, these results suggest that tetrac targets β-catenin and HMGA2 to promote resveratrol-induced-anti-proliferation in colon cancers, highlighting its potential in anti-cancer combination therapy.
- Published
- 2017
37. Live Images of GLUT4 Protein Trafficking in Mouse Primary Hypothalamic Neurons Using Deconvolution Microscopy
- Author
-
Szu-Yi Chou, Reni Ajoy, and Chun A. Changou
- Subjects
0301 basic medicine ,Male ,General Chemical Engineering ,medicine.medical_treatment ,Hypothalamus ,030209 endocrinology & metabolism ,Inflammation ,Stimulation ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Neurobiology ,liposome based transfection ,medicine ,deconvolution microscopy ,Animals ,Neurons ,Microscopy ,Glucose Transporter Type 4 ,General Immunology and Microbiology ,biology ,General Neuroscience ,Insulin ,Issue 130 ,Cell biology ,Insulin receptor ,live image recording ,Protein Transport ,030104 developmental biology ,medicine.anatomical_structure ,Knockout mouse ,biology.protein ,Primary mouse hypothalamic neuron culture ,Tumor necrosis factor alpha ,Neuron ,medicine.symptom ,GLUT4 - Abstract
Type 2 diabetes mellitus (T2DM) is a global health crisis which is characterized by insulin signaling impairment and chronic inflammation in peripheral tissues. The hypothalamus in the central nervous system (CNS) is the control center for energy and insulin signal response regulation. Chronic inflammation in peripheral tissues and imbalances of certain chemokines (such as CCL5, TNFα, and IL-6) contribute to diabetes and obesity. However, the functional mechanism(s) connecting chemokines and hypothalamic insulin signal regulation still remain unclear. In vitro primary neuron culture models are convenient and simple models which can be used to investigate insulin signal regulation in hypothalamic neurons. In this study, we introduced exogeneous GLUT4 protein conjugated with GFP (GFP-GLUT4) into primary hypothalamic neurons to track GLUT4 membrane translocation upon insulin stimulation. Time-lapse images of GFP-GLUT4 protein trafficking were recorded by deconvolution microscopy, which allowed users to generate high-speed, high-resolution images without damaging the neurons significantly while conducting the experiment. The contribution of CCR5 in insulin regulated GLUT4 translocation was observed in CCR5 deficient hypothalamic neurons, which were isolated and cultured from CCR5 knockout mice. Our results demonstrated that the GLUT4 membrane translocation efficiency was reduced in CCR5 deficient hypothalamic neurons after insulin stimulation.
- Published
- 2017
38. Integrin β3 and LKB1 are independently involved in the inhibition of proliferation by lovastatin in human intrahepatic cholangiocarcinoma
- Author
-
Chun A. Changou, Jinghan Wang, Yun Ru Liu, Chun Han Chen, Hung Yun Lin, Xiaoqing Jiang, Frank Luh, Yun Yen, and Sheng Huei Yang
- Subjects
0301 basic medicine ,Cell ,Cholangiocarcinoma ,0302 clinical medicine ,AMP-Activated Protein Kinase Kinases ,Cell Movement ,TGF-β1 ,polycyclic compounds ,beta Catenin ,Microscopy, Confocal ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Integrin beta3 ,Cell migration ,Intercellular adhesion molecule ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,RNA Interference ,lipids (amino acids, peptides, and proteins) ,Lovastatin ,Signal transduction ,Signal Transduction ,Research Paper ,medicine.drug ,LKB1 ,integrin ,Immunoblotting ,Integrin ,Protein Serine-Threonine Kinases ,HMG-CoA reductase inhibitor ,03 medical and health sciences ,Cell Line, Tumor ,Cell Adhesion ,medicine ,Humans ,Cell adhesion ,Cell Proliferation ,Cell growth ,business.industry ,nutritional and metabolic diseases ,Bile duct cancer ,030104 developmental biology ,Bile Duct Neoplasms ,Microscopy, Fluorescence ,Immunology ,Cancer research ,biology.protein ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,business - Abstract
Human intrahepatic cholangiocarcinomas are one of the most difficult cancers to treat. In our study, Lovastatin, a 3-hydroxy-3-methylglutaryl-coenzyme-CoA (HMG-CoA) reductase inhibitor, demonstrated anticancer properties by inhibiting cancer cell proliferation, cell migration and cell adhesion. Lovastatin inhibited the expressions of transforming growth factor (TGF)-β1, cyclooxygenase (COX)-2, and intercellular adhesion molecule (ICAM)-1. Furthermore, lovastatin inhibited the expressions of integrin β1 and integrin β3 but not integrin αv or integrin β5. While Lovastatin's inhibitory effects on TGFβ1, COX2, and ICAM-1 expression were independently controlled by the tumor suppressor LKB1, integrin β3 expression was not affected. Lovastatin's inhibitory effect on cell adhesion was associated with the decreased expression of integrin β3 and cell surface heterodimer integrin αvβ3. Quantitative real time PCR, fluorescent microscopy, and cell migration assays all confirmed that Lovastatin inhibits integrin αvβ3 downstream signaling including FAK activation, and β-catenin, vimentin, ZO-1, and β-actin. Overall, Lovastatin reduced tumor cell proliferation and migration by modifying the expression of genes involved in cell adhesion and other critical cellular processes. Our study highlights novel anti-cancer properties of Lovastatin and supports further exploration of statins in the context of cholangiocarcinoma therapy.
- Published
- 2015
39. Mechanisms of dihydrotestosterone action on resveratrol-induced anti-proliferation in breast cancer cells with different ERα status
- Author
-
Tung Cheng Chang, Paul J. Davis, Leroy F. Liu, Sheng Huei Yang, Chun A. Changou, Earl Fu, Yu Tang Chin, Hung Yun Lin, Wei Chun HuangFu, and Hsuan Yu Lai
- Subjects
endocrine system ,medicine.medical_specialty ,DHT ,Apoptosis ,Breast Neoplasms ,resveratrol ,Resveratrol ,Biology ,urologic and male genital diseases ,chemistry.chemical_compound ,breast cancer ,estrogen receptor-α ,Cell Line, Tumor ,Internal medicine ,Stilbenes ,polycyclic compounds ,medicine ,Humans ,Drug Interactions ,Phosphorylation ,skin and connective tissue diseases ,Receptor ,Cell Proliferation ,Cell growth ,integrin αvβ3 ,Estrogen Receptor alpha ,Dihydrotestosterone ,Integrin alphaVbeta3 ,Cell biology ,Endocrinology ,Oncology ,chemistry ,Cancer cell ,MCF-7 Cells ,Female ,Signal transduction ,Estrogen receptor alpha ,hormones, hormone substitutes, and hormone antagonists ,Research Paper ,Signal Transduction ,medicine.drug - Abstract
Dihydrotestosterone (DHT) has been shown to promote breast cancer growth via different mechanisms. In addition to binding to ERα, the DHT membrane receptor exists on integrin αvβ3. Resveratrol induces p53-dependent apoptosis via plasma membrane integrin αvβ3. Resveratrol and DHT signals are both transduced by activated ERK1/2; however, DHT promotes cell proliferation in cancer cells, whereas resveratrol is pro-apoptotic. In this study, we examined the mechanism by which DHT inhibits resveratrol-induced apoptosis in human ERα positive (MCF-7) and negative (MDA-MB-231) breast cancer cells. DHT inhibited resveratrol-stimulated phosphorylation of Ser-15 of p53 in a concentration-dependent manner. These effects of DHT on resveratrol action were blocked by an ERα antagonist, ICI 182,780, in MCF-7 breast cancer cells. DHT inhibited resveratrol-induced nuclear complex of p53-COX-2 formation which is required p53-dependent apoptosis. ChIP studies of COX-2/p53 binding to DNA and expression of p53-responsive genes indicated that DHT inhibited resveratrol-induced p53-directed transcriptional activity. In addition, DHT did inhibit resveratrol-induced COX-2/p53-dependent gene expression. These results suggest that DHT inhibits p53-dependent apoptosis in breast cancer cells by interfering with nuclear COX-2 accumulation which is essential for stimulation of apoptotic pathways. Thus, the surface receptor sites for resveratrol and DHT are discrete and activate ERK1/2-dependent downstream effects on apoptosis that are distinctive. These studies provide new insights into the antagonizing effects of resveratrol versus DHT, an important step toward better understanding and eventually treating breast cancer. It also indicates the complex pathways by which apoptosis is induced by resveratrol in DHT-depleted and -repleted environments.
- Published
- 2015
40. Arginine starvation-associated atypical cellular death involves mitochondrial dysfunction, nuclear DNA leakage, and chromatin autophagy
- Author
-
Hsing Jien Kung, Chun A. Changou, Li Xing, Yun-Ru Chen, Yun Yen, Frank Y. S. Chuang, R. Holland Cheng, David K. Ann, and Richard J. Bold
- Subjects
Programmed cell death ,Multidisciplinary ,Arginine ,ATG5 ,Autophagy ,Autophagosome membrane ,Argininosuccinate synthase ,biology.protein ,Mitochondrion ,Biology ,Molecular biology ,Arginine deiminase ,Cell biology - Abstract
Autophagy is the principal catabolic prosurvival pathway during nutritional starvation. However, excessive autophagy could be cytotoxic, contributing to cell death, but its mechanism remains elusive. Arginine starvation has emerged as a potential therapy for several types of cancers, owing to their tumor-selective deficiency of the arginine metabolism. We demonstrated here that arginine depletion by arginine deiminase induces a cytotoxic autophagy in argininosuccinate synthetase (ASS1)-deficient prostate cancer cells. Advanced microscopic analyses of arginine-deprived dying cells revealed a novel phenotype with giant autophagosome formation, nucleus membrane rupture, and histone-associated DNA leakage encaptured by autophagosomes, which we shall refer to as chromatin autophagy, or chromatophagy. In addition, nuclear inner membrane (lamin A/C) underwent localized rearrangement and outer membrane (NUP98) partially fused with autophagosome membrane. Further analysis showed that prolonged arginine depletion impaired mitochondrial oxidative phosphorylation function and depolarized mitochondrial membrane potential. Thus, reactive oxygen species (ROS) production significantly increased in both cytosolic and mitochondrial fractions, presumably leading to DNA damage accumulation. Addition of ROS scavenger N-acetyl cysteine or knockdown of ATG5 or BECLIN1 attenuated the chromatophagy phenotype. Our data uncover an atypical autophagy-related death pathway and suggest that mitochondrial damage is central to linking arginine starvation and chromatophagy in two distinct cellular compartments.
- Published
- 2014
41. Thyroid hormone-induced expression of inflammatory cytokines interfere with resveratrol-induced anti-proliferation of oral cancer cells
- Author
-
Yi Ru Chen, Hsien Chung Chiu, Ya Jung Shih, Yu Chen S.H. Yang, Jacqueline Whang-Peng, Hung Yun Lin, Shaker A. Mousa, Yu Tang Chin, Po Yu Su, Shwu Huey Wang, Leroy F. Liu, Yu Shen Chen, Paul J. Davis, Sheng Yang Lee, Yun Hsuan Wu, Zi Lin Li, Kuan Wang, and Chun A. Changou
- Subjects
STAT3 Transcription Factor ,Gene Expression ,Resveratrol ,Toxicology ,Proinflammatory cytokine ,03 medical and health sciences ,chemistry.chemical_compound ,0404 agricultural biotechnology ,Cell Line, Tumor ,Gene expression ,medicine ,Humans ,STAT3 ,Cell Proliferation ,030304 developmental biology ,Cell Nucleus ,0303 health sciences ,biology ,Cancer ,04 agricultural and veterinary sciences ,General Medicine ,medicine.disease ,040401 food science ,Thyroxine ,chemistry ,Cyclooxygenase 2 ,Cancer cell ,biology.protein ,STAT protein ,Cancer research ,Cytokines ,Mouth Neoplasms ,Inflammation Mediators ,Signal transduction ,Food Science - Abstract
Thyroid hormone, L-thyroxine (T4), induces inflammatory genes expressions and promotes cancer growth. It also induces expression of the checkpoint programmed death-ligand 1 (PD-L1), which plays a vital role in cancer progression. On the other hand, resveratrol inhibits inflammatory genes expressions. Moreover, resveratrol increases nuclear inducible cyclooxygenase (COX)-2 accumulation, complexes with p53, and induces p53-dependent anti-proliferation. In this study, we investigated the effect of T4 on resveratrol-induced anti-proliferation in oral cancer. T4 increased the expression and cytoplasmic accumulation of PD-L1. Increased expressions of pro-inflammatory genes, interleukin (IL)-1β and transforming growth factor (TGF)-β1, were shown to stimulate PD-L1 expression. T4 stimulated pro-inflammatory and proliferative genes expressions, and oral cancer cells proliferation. In contrast, resveratrol inhibited those genes and activated anti-proliferative genes. T4 retained resveratrol-induced COX-2 in cytoplasm and prevented COX-2 nuclear accumulation when resveratrol treated cancer cells. A specific signal transducer and activator of transcription 3 (STAT3) inhibitor, S31-201, blocked T4-induced inhibition and restored resveratrol-induced nuclear COX-2 accumulation. By inhibiting the T4-activated STAT3 signal transduction axis with S31-201, resveratrol was able to sequentially reestablish COX-2/p53-dependent gene expressions and anti-proliferation. These findings provide a novel understanding of the inhibitory effects of T4 on resveratrol-induced anticancer properties via the sequential expression of PD-L1 and inflammatory genes.
- Published
- 2019
42. Hepatitis C Virus Non-Structural Protein 5A (NS5A) Disrupts Mitochondrial Dynamics and Induces Mitophagy
- Author
-
Christopher D. Richardson, Alagie Jassey, Hsue-Yin Hsu, Chun A. Changou, Ching-Hsuan Liu, and Liang Tzung Lin
- Subjects
0301 basic medicine ,Ubiquitin-Protein Ligases ,viruses ,Hepacivirus ,Viral Nonstructural Proteins ,Mitochondrion ,NS5A ,Article ,Parkin ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,Mitophagy ,Autophagy ,Humans ,lcsh:QH301-705.5 ,Membrane Potential, Mitochondrial ,chemistry.chemical_classification ,Reactive oxygen species ,Effector ,Chemistry ,virus diseases ,Bafilomycin ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,Lipids ,mitochondrial dynamics ,digestive system diseases ,Mitochondria ,Cell biology ,Protein Transport ,mitophagy ,030104 developmental biology ,lcsh:Biology (General) ,030220 oncology & carcinogenesis ,HCV ,Replicon ,Reactive Oxygen Species ,Microtubule-Associated Proteins - Abstract
Mitophagy is a selective form of autophagy, targeting damaged mitochondria for lysosomal degradation. Although HCV infection has been shown to induce mitophagy, the precise underlying mechanism and the effector protein responsible remain unclear. Herein, we demonstrated that the HCV non-structural protein 5A (NS5A) plays a key role in regulating cellular mitophagy. Specifically, the expression of HCV NS5A in the hepatoma cells triggered hallmarks of mitophagy including mitochondrial fragmentation, loss of mitochondrial membrane potential, and Parkin translocation to the mitochondria. Furthermore, mitophagy induction through the expression of NS5A led to an increase in autophagic flux as demonstrated by an accumulation of LC3II in the presence of bafilomycin and a time-dependent decrease in p62 protein level. Intriguingly, the expression of NS5A concomitantly enhanced reactive oxygen species (ROS) production, and treatment with an antioxidant attenuated the NS5A-induced mitophagy event. These phenomena are similarly recapitulated in the NS5A-expressing HCV subgenomic replicon cells. Finally, we demonstrated that expression of HCV core, which has been documented to inhibit mitophagy, blocked the mitophagy induction both in cells harboring HCV replicating subgenomes or expressing NS5A alone. Our results, therefore, identified a new role for NS5A as an important regulator of HCV-induced mitophagy and have implications to broadening our understanding of the HCV-mitophagy interplay.
- Published
- 2019
43. Chromatophagy: Autophagy goes nuclear and captures broken chromatin during arginine-starvation
- Author
-
Hsing Jien Kung, Chien-Feng Li, Chun A. Changou, and David K. Ann
- Subjects
Southern taiwan ,Library science ,ASS1 ,Biology ,Arginine ,Autophagic Puncta ,Cell Line, Tumor ,Arginine metabolism ,Autophagy ,Humans ,ADI-PEG20 ,China ,Molecular Biology ,arginine auxotrophy ,Cell Nucleus ,Genetics ,National health ,ROS ,DNA ,Cell Biology ,Chromatin ,humanities ,chromatophagy ,DNA metabolism ,ADI, arginine deiminase ,ADI-PEG20, pegylated PADI ,ASS1, argininosuccinate synthase 1 ,MN, micronuclei ,NAC, N-acetylcysteine ,PMN, piecemeal microautophagy of the nucleus ,ROS, reactive oxygen species ,DNA Damage - Abstract
Chromatophagy: Autophagy goes nuclear and captures broken chromatin during arginine-starvation Hsing-Jien Kung, Chun A Changou, Chien-Feng Li & David K Ann a Department of Biochemistry & Molecular Medicine; UC Davis Comprehensive Cancer Center; Sacramento, CA USA; b Integrated Laboratory; Center of Translational Medicine; Taipei Medical University; Taipei, Taiwan, Republic of China; c Graduate Institute of Translational Medicine; Taipei Medical University; Taipei, Taiwan, Republic of China; d National Health Research Institutes; Taiwan; Republic of China; e Department of Pathology; Chi-Mei Medical Center; Tainan, Taiwan, Republic of China; f National Institute of Cancer Research; National Health Research Institutes; Tainan, Taiwan, Republic of China; g Department of Biotechnology; Southern Taiwan University of Science and Technology; Tainan, Taiwan, Republic of China; h Department of Metabolic Disorders Research; Beckman Research Institute; City of Hope, Duarte, CA USA Accepted author version posted online: 04 Feb 2015.
- Published
- 2015
44. CCL5/RANTES contributes to hypothalamic insulin signaling for systemic insulin responsiveness through CCR5
- Author
-
Chun A. Changou, Yang Kao Wang, Barry J. Hoffer, Ya Ting Hsieh, Szu-Yi Chou, and Reni Ajoy
- Subjects
0301 basic medicine ,medicine.medical_specialty ,medicine.medical_treatment ,Hypothalamus ,Carbohydrate metabolism ,Article ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Diabetes mellitus ,medicine ,Animals ,Humans ,Insulin ,Phosphorylation ,Receptor ,Chemokine CCL5 ,Neurons ,Glucose Transporter Type 4 ,Multidisciplinary ,biology ,business.industry ,Arcuate Nucleus of Hypothalamus ,virus diseases ,Type 2 Diabetes Mellitus ,medicine.disease ,Receptor, Insulin ,Disease Models, Animal ,Insulin receptor ,030104 developmental biology ,Endocrinology ,Diabetes Mellitus, Type 2 ,biology.protein ,Signal transduction ,Energy Metabolism ,business ,030217 neurology & neurosurgery ,GLUT4 ,Signal Transduction - Abstract
Many neurodegenerative diseases are accompanied by metabolic disorders. CCL5/RANTES, and its receptor CCR5 are known to contribute to neuronal function as well as to metabolic disorders such as type 2 diabetes mellitus, obesity, atherosclerosis and metabolic changes after HIV infection. Herein, we found that the lack of CCR5 or CCL5 in mice impaired regulation of energy metabolism in hypothalamus. Immunostaining and co-immunoprecipitation revealed the specific expression of CCR5, associated with insulin receptors, in the hypothalamic arcuate nucleus (ARC). Both ex vivo stimulation and in vitro tissue culture studies demonstrated that the activation of insulin, and PI3K-Akt pathways were impaired in CCR5 and CCL5 deficient hypothalamus. The inhibitory phosphorylation of insulin response substrate-1 at Ser302 (IRS-1S302) but not IRS-2, by insulin was markedly increased in CCR5 and CCL5 deficient animals. Elevating CCR5/CCL5 activity induced GLUT4 membrane translocation and reduced phospho-IRS-1S302 through AMPKα-S6 Kinase. Blocking CCR5 using the antagonist, MetCCL5, abolished the de-phosphorylation of IRS-1S302 and insulin signal activation. In addition, intracerebroventricular delivery of MetCCL5 interrupted hypothalamic insulin signaling and elicited peripheral insulin responsiveness and glucose intolerance. Taken together, our data suggest that CCR5 regulates insulin signaling in hypothalamus which contributes to systemic insulin sensitivity and glucose metabolism.
- Published
- 2016
- Full Text
- View/download PDF
45. Crosstalk between integrin αvβ3 and ERα contributes to thyroid hormone-induced proliferation of ovarian cancer cells
- Author
-
Hung Yun Lin, Le Ming Wang, Sheng Yang Lee, Yu Tang Chin, Hsuan Yu Lai, Meng Ti Hsieh, Yu Chen S.H. Yang, Yung Ning Yang, Paul J. Davis, Chun A. Changou, Leroy F. Liu, Jacqueline Whang-Peng, and Shaker A. Mousa
- Subjects
0301 basic medicine ,Thyroid Hormones ,Estrogen receptor ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Medicine ,Humans ,Cell Proliferation ,Mitogen-Activated Protein Kinase 1 ,Ovarian Neoplasms ,Integrin alphaVbeta3 ,Mitogen-Activated Protein Kinase 3 ,Fulvestrant ,business.industry ,Kinase ,MEK inhibitor ,Thyroid ,Estrogen Receptor alpha ,integrin αvβ3 ,thyroid hormone ,030104 developmental biology ,medicine.anatomical_structure ,ovarian cancer ,Oncology ,ERα crosstalk ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Female ,Signal transduction ,business ,Estrogen receptor alpha ,medicine.drug ,Protein Binding ,Signal Transduction ,Research Paper - Abstract
// Meng-Ti Hsieh 1, 2, * , Le-Ming Wang 3, * , Chun A. Changou 2, 4, 5 , Yu-Tang Chin 1, 6, 7, 8 , Yu-Chen S.H. Yang 9 , Hsuan-Yu Lai 1 , Sheng-Yang Lee 6, 7, 8 , Yung-Ning Yang 10 , Jacqueline Whang-Peng 1 , Leroy F. Liu 1 , Hung-Yun Lin 1, 2 , Shaker A. Mousa 11 , Paul J. Davis 11, 12 1 Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan 2 The PhD Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan 3 Department of Obstetrics and Gynecology, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan 4 Integrated Laboratory, Center of Translational Medicine, Taipei Medical University, Taipei, Taiwan 5 Core Facility, Taipei Medical University, Taipei, Taiwan 6 Department of Dentistry, Wan-Fang Medical Center, Taipei Medical University, Taipei, Taiwan 7 School of Dentistry, Taipei Medical University, Taipei, Taiwan 8 Center for Teeth Bank and Dental Stem Cell Technology, Taipei Medical University, Taipei, Taiwan 9 Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan 10 Department of Pediatrics, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan 11 Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, New York, USA 12 Department of Medicine, Albany Medical College, Albany, New York, USA * These authors contributed equally to this work Correspondence to: Yung-Ning Yang, email: ancaly@yahoo.com.tw Hung-Yun Lin, email: linhy@tmu.edu.tw Keywords: thyroid hormone, integrin αvβ3, ERα crosstalk, ovarian cancer Received: April 23, 2016 Accepted: July 10, 2016 Published: July 21, 2016 ABSTRACT Ovarian cancer is the leading cause of death in gynecological diseases. Thyroid hormone promotes proliferation of ovarian cancer cells via cell surface receptor integrin αvβ3 that activates extracellular regulated kinase (ERK1/2). However, the mechanisms are still not fully understood. Thyroxine (T 4 ) at a physiologic total hormone concentration (10 –7 M) significantly increased proliferating cell nuclear antigen (PCNA) abundance in these cell lines, as did 3, 5, 3’-triiodo-L-thyronine (T 3 ) at a supraphysiologic concentration. Thyroid hormone (T 4 and T 3 ) treatment of human ovarian cancer cells resulted in enhanced activation of the Ras/MAPK(ERK1/2) signal transduction pathway. An MEK inhibitor (PD98059) blocked hormone-induced cell proliferation but not ER phosphorylation. Knock-down of either integrin αv or β3 by RNAi blocked thyroid hormone-induced phosphorylation of ERK1/2. We also found that thyroid hormone causes elevated phosphorylation and nuclear enrichment of estrogen receptor α (ERα). Confocal microscopy indicated that both T4 and estradiol (E 2 ) caused nuclear translocation of integrin αv and phosphorylation of ERα. The specific ERα antagonist (ICI 182,780; fulvestrant) blocked T 4 -induced ERK1/2 activation, ERα phosphorylation, PCNA expression and proliferation. The nuclear co-localization of integrin αv and phosphorylated ERα was inhibited by ICI. ICI time-course studies indicated that mechanisms involved in T 4 - and E 2 -induced nuclear co-localization of phosphorylated ERα and integrin αv are dissimilar. Chromatin immunoprecipitation results showed that T 4 -induced binding of integrin αv monomer to ERα promoter and this was reduced by ICI. In summary, thyroid hormone stimulates proliferation of ovarian cancer cells via crosstalk between integrin αv and ERα, mimicking functions of E 2 .
- Published
- 2016
46. PYCR1 and PYCR2 Interact and Collaborate with RRM2B to Protect Cells from Overt Oxidative Stress
- Author
-
Mabel Bin Er Lee, Chun A. Changou, Leila Su, Michelle Tang, Mingming Su, Yun Yen, Shuya Hu, Wei Jia, Willem den Besten, Mei-Ling Kuo, Sonja Hess, Chih Ming Chou, and Michael J. Sweredoski
- Subjects
0301 basic medicine ,Recombinant Fusion Proteins ,Cell Cycle Proteins ,Biology ,Reductase ,Mitochondrion ,medicine.disease_cause ,Article ,Antioxidants ,Mass Spectrometry ,Cell Line ,03 medical and health sciences ,Protein Interaction Mapping ,Ribonucleotide Reductases ,medicine ,Animals ,Humans ,Gene silencing ,Gene Silencing ,Fragmentation (cell biology) ,Telomerase ,Zebrafish ,Multidisciplinary ,Cell Cycle Checkpoints ,Mitochondria ,Isoenzymes ,Oxidative Stress ,Protein Transport ,030104 developmental biology ,Ribonucleotide reductase ,Biochemistry ,Cell culture ,Gene Knockdown Techniques ,Multiprotein Complexes ,Pyrroline Carboxylate Reductases ,Tumor Suppressor Protein p53 ,Signal transduction ,Oxidative stress ,Protein Binding ,Signal Transduction - Abstract
Ribonucleotide reductase small subunit B (RRM2B) is a stress response protein that protects normal human fibroblasts from oxidative stress. However, the underlying mechanism that governs this function is not entirely understood. To identify factors that interact with RRM2B and mediate anti-oxidation function, large-scale purification of human Flag-tagged RRM2B complexes was performed. Pyrroline-5-carboxylate reductase 1 and 2 (PYCR1, PYCR2) were identified by mass spectrometry analysis as components of RRM2B complexes. Silencing of both PYCR1 and PYCR2 by expressing short hairpin RNAs induced defects in cell proliferation, partial fragmentation of the mitochondrial network and hypersensitivity to oxidative stress in hTERT-immortalized human foreskin fibroblasts (HFF-hTERT). Moderate overexpression of RRM2B, comparable to stress-induced level, protected cells from oxidative stress. Silencing of both PYCR1 and PYCR2 completely abolished anti-oxidation activity of RRM2B, demonstrating a functional collaboration of these metabolic enzymes in response to oxidative stress.
- Published
- 2016
47. Optical imaging of ovarian cancer using a matrix metalloproteinase-3-sensitive near-infrared fluorescent probe
- Author
-
Tsai Yu-Hui, Chen Tze-Chien, Chiu Li-Hsuan, Chun S. Zuo, T. Lai Wen-Fu, Yun-Ming Wang, Chen Kuan-Chou, Wang Kuo-Hwa, and Chun A. Changou
- Subjects
Matrix Metalloproteinase 3 ,Materials science ,lcsh:Medicine ,Mice ,Optical imaging ,Nuclear magnetic resonance ,Cell Line, Tumor ,medicine ,Animals ,Humans ,lcsh:Science ,Fluorescent Dyes ,Ovarian Neoplasms ,Spectroscopy, Near-Infrared ,Multidisciplinary ,lcsh:R ,Optical Imaging ,Near-infrared spectroscopy ,Correction ,medicine.disease ,Fluorescence ,Coculture Techniques ,Heterografts ,lcsh:Q ,Female ,Ovarian cancer - Abstract
Epithelial ovarian cancer (EOC) is the seventh most common cancer among women worldwide. The 5-year survival rate for women with EOC is only 30%-50%, which is largely due to the typically late diagnosis of this condition. EOC is difficult to detect in its early stage because of its asymptomatic nature. Recently, near-infrared fluorescent (NIRF) imaging has been developed as a potential tool for detecting EOC at the molecular level. In this study, a NIRF-sensitive probe was designed to detect matrix metalloproteinase (MMP) activity in ovarian cancer cells. A cyanine fluorochrome was conjugated to the amino terminus of a peptide substrate with enzymatic specificity for MMP-3. To analyze the novel MMP-3 probe, an in vivo EOC model was established by subcutaneously implanting SKOV3 cells, a serous-type EOC cell line, in mice. This novel MMP-3-sensitive probe specifically reacted with only the active MMP-3 enzyme, resulting in a significantly enhanced NIRF emission intensity. Histological analysis demonstrated that MMP-3 expression and activity were enhanced in the stromal cells surrounding the ovarian cancer cells. These studies establish a molecular imaging reporter for diagnosing early-stage EOC. Additional studies are required to confirm the early-stage activity of MMP-3 in EOC and its diagnostic and prognostic significance.
- Published
- 2018
48. Abstract 2042: Nano-tetraiodothyroacetic acid (NDAT) potentiates gefitinib-induced antiproliferation in colorectal cancer cells by inhibiting EGFR sialylation and PI3K activation
- Author
-
Hung Yun Lin, Jacqueline Whang-Peng, Chi-Yu Lin, Chun A. Changou, Yu-Tang Chin, and Paul J. Davis
- Subjects
Cancer Research ,Gefitinib ,Oncology ,Biochemistry ,Colorectal cancer ,Chemistry ,Tetraiodothyroacetic acid ,medicine ,Cancer research ,medicine.disease ,PI3K/AKT/mTOR pathway ,medicine.drug - Abstract
The resistance of gefitinib has been revealed to complicate cancer therapy. Tetraiodothyroacetic acid (tetrac) and its nanoparticulate derivative (NDAT, nano-tetrac) have been proved in vitro and in vivo xenograft demonstrating anti-proliferative and anti-angiogenetic activities. It also indicates that they potentiate anti-cancer agent-induced anti-proliferation in cancer cells. In this study, we investigated the effects of NDAT on gefitinib-induced anti-cancer activities in human colorectal cancer cells. Gefitinib inhibited cell proliferation at concentration 1 μM in K-ras wild type HT29 cell and NDAT enhanced the anti-proliferation-induced gefitinib significantly. Meanwhile, both inhibited proliferative and metastatic genes expression in HT29 cells. On the other hand, 10 μM gefitinib inhibited cell proliferation in K-ras mutant HCT116 cell which was further enhanced by NDAT. Different from results in HT-29 cells, only NDAT inhibited proliferative and metastatic genes expression significantly and enhanced the effect of gefitinib in HCT116 cells. ST6Gal-1 catalyzes sialylation of EGFR and induces gefitinib-resistant in colorectal cancers. In addition, NDAT did reduced not only ST6Gal-1 gene expression, but also its protein production. However, the inhibition of ST6Gal-1 expression may not be sufficient to induced anti-proliferation in colorectal cancer cells. PI3K inhibitor, LY294002, was able to potentiate the gefitinib-induced anti-proliferation in HCT116 cells suggesting that constitutive activation of PI3K may play a key role on gefitinib-resistance in HCT116 cells. In summary, NDAT potentiated gefitinib-induced anti-proliferation via inhibiting the activity of ST6Gal-1 and PI3K activation in gefitinib-resistant colorectal cancer cells. Citation Format: Yu-Tang Chin, Chi-Yu Lin, Chun A. Changou, Jacqueline Whang-Peng, Paul J. Davis, Hung-Yun Lin. Nano-tetraiodothyroacetic acid (NDAT) potentiates gefitinib-induced antiproliferation in colorectal cancer cells by inhibiting EGFR sialylation and PI3K activation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 2042. doi:10.1158/1538-7445.AM2017-2042
- Published
- 2017
49. Quantitative analysis of autophagy using advanced 3D fluorescence microscopy
- Author
-
Deanna Wolfson, Chun A. Changou, Richard J. Bold, Hsing Jien Kung, Balpreet Singh Ahluwalia, and Frank Y. S. Chuang
- Subjects
Autophagosome ,Male ,General Chemical Engineering ,Cell ,Argininosuccinate synthase ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Prostate cancer ,Imaging, Three-Dimensional ,Live cell imaging ,Lysosome ,Cell Line, Tumor ,medicine ,Autophagy ,Humans ,Fluorescent Dyes ,General Immunology and Microbiology ,General Neuroscience ,Prostatic Neoplasms ,medicine.disease ,Cell biology ,Cellular Biology ,medicine.anatomical_structure ,Microscopy, Fluorescence ,Cancer cell ,biology.protein ,Lysosomes - Abstract
Prostate cancer is the leading form of malignancies among men in the U.S. While surgery carries a significant risk of impotence and incontinence, traditional chemotherapeutic approaches have been largely unsuccessful. Hormone therapy is effective at early stage, but often fails with the eventual development of hormone-refractory tumors. We have been interested in developing therapeutics targeting specific metabolic deficiency of tumor cells. We recently showed that prostate tumor cells specifically lack an enzyme (argininosuccinate synthase, or ASS) involved in the synthesis of the amino acid arginine(1). This condition causes the tumor cells to become dependent on exogenous arginine, and they undergo metabolic stress when free arginine is depleted by arginine deiminase (ADI)(1,10). Indeed, we have shown that human prostate cancer cells CWR22Rv1 are effectively killed by ADI with caspase-independent apoptosis and aggressive autophagy (or macroautophagy)(1,2,3). Autophagy is an evolutionarily-conserved process that allows cells to metabolize unwanted proteins by lysosomal breakdown during nutritional starvation(4,5). Although the essential components of this pathway are well-characterized(6,7,8,9), many aspects of the molecular mechanism are still unclear - in particular, what is the role of autophagy in the death-response of prostate cancer cells after ADI treatment? In order to address this question, we required an experimental method to measure the level and extent of autophagic response in cells - and since there are no known molecular markers that can accurately track this process, we chose to develop an imaging-based approach, using quantitative 3D fluorescence microscopy(11,12). Using CWR22Rv1 cells specifically-labeled with fluorescent probes for autophagosomes and lysosomes, we show that 3D image stacks acquired with either widefield deconvolution microscopy (and later, with super-resolution, structured-illumination microscopy) can clearly capture the early stages of autophagy induction. With commercially available digital image analysis applications, we can readily obtain statistical information about autophagosome and lysosome number, size, distribution, and degree of colocalization from any imaged cell. This information allows us to precisely track the progress of autophagy in living cells and enables our continued investigation into the role of autophagy in cancer chemotherapy.
- Published
- 2013
50. Microscopic analysis of cell death by metabolic stress-induced autophagy in prostate cancer
- Author
-
Frank Y. S. Chuang, R. Holland Cheng, Richard J. Bold, Hsing Jien Kung, and Chun A. Changou
- Subjects
chemistry.chemical_classification ,Programmed cell death ,Reactive oxygen species ,Autophagy ,medicine.disease ,Cell biology ,Prostate cancer ,Cell nucleus ,medicine.anatomical_structure ,chemistry ,Apoptosis ,medicine ,Arginine deiminase ,Intracellular - Abstract
Autophagy is an intracellular recycling mechanism that helps cells to survive against environmental stress and nutritional starvation. We have recently shown that prostate cancers undergo metabolic stress and caspase-independent cell death following exposure to arginine deiminase (ADI, an enzyme that degrades arginine in tissue). The aims of our current investigation into the application of ADI as a novel cancer therapy are to identify the components mediating tumor cell death, and to determine the role of autophagy (stimulated by ADI and/or rapamycin) on cell death. Using advanced fluorescence microscopy techniques including 3D deconvolution and superresolution structured-illumination microscopy (SIM), we show that prostate tumor cells that are killed after exposure to ADI for extended periods, exhibit a morphology that is distinct from caspase-dependent apoptosis; and that autophagosomes forming as a result of ADI stimulation contain DAPI-stained nuclear material. Fluorescence imaging (as well as cryo-electron microscopy) show a breakdown of both the inner and outer nuclear membranes at the interface between the cell nucleus and aggregated autophagolysosomes. Finally, the addition of N-acetyl cysteine (or NAC, a scavenger for reactive oxygen species) effectively abolishes the appearance of autophagolysosomes containing nuclear material. We hope to continue this research to understand the processes that govern the survival or death of these tumor cells, in order to develop methods to improve the efficacy of cancer pharmacotherapy.
- Published
- 2013
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.