89 results on '"Tsai-Kun Li"'
Search Results
2. Supplementary Figure 4 from Mitoxantrone Inhibits HIF-1α Expression in a Topoisomerase II–Independent Pathway
- Author
-
Tsai-Kun Li and Yng-Miin Toh
- Abstract
PDF file - 124K
- Published
- 2023
3. Supplementary Figure 5 from Mitoxantrone Inhibits HIF-1α Expression in a Topoisomerase II–Independent Pathway
- Author
-
Tsai-Kun Li and Yng-Miin Toh
- Abstract
PDF file - 270K
- Published
- 2023
4. Supplementary Figure 4 from DNA Topoisomerase III Alpha Regulates p53-Mediated Tumor Suppression
- Author
-
Tsai-Kun Li, Yen-Hsiu Yeh, Shu-Chun Teng, Tang-Long Shen, Hsiang-Chin Chen, Han-Wen Chang, Jia-Rong Fan, and Mei-Yi Hsieh
- Abstract
PDF file 2097K, Knockdown of hTOP3alpha and p53, but not BLM, reduced cellular senescence and enhanced anchorage-independent growth of AGS cells
- Published
- 2023
5. Supplementary Figure 1 from Mitoxantrone Inhibits HIF-1α Expression in a Topoisomerase II–Independent Pathway
- Author
-
Tsai-Kun Li and Yng-Miin Toh
- Abstract
PDF file - 93.5K
- Published
- 2023
6. Supplementary Figure 1 from DNA Topoisomerase III Alpha Regulates p53-Mediated Tumor Suppression
- Author
-
Tsai-Kun Li, Yen-Hsiu Yeh, Shu-Chun Teng, Tang-Long Shen, Hsiang-Chin Chen, Han-Wen Chang, Jia-Rong Fan, and Mei-Yi Hsieh
- Abstract
PDF file 2053K, DNA damage-induced expression of p53, p5315P and p21 is greatly reduced in hTOP3α-deficient cells
- Published
- 2023
7. Data from Mitoxantrone Inhibits HIF-1α Expression in a Topoisomerase II–Independent Pathway
- Author
-
Tsai-Kun Li and Yng-Miin Toh
- Abstract
Purpose: Solid tumors encounter a growth-limiting hypoxic microenvironment as they develop. Hypoxia-inducible factors (HIF) play important roles in hypoxia-associated tumor development and therapeutic resistance. Targeting the HIF pathway (especially HIF-1α) represents a promising cancer treatment strategy. Here, we report a novel class of HIF-1α inhibitors and the possible molecular basis of inhibition.Experimental Design: We analyzed the inhibitory effects of clinically used topoisomerase II (TOP2)–targeting drugs on HIF-1α expression with a primary focus on mitoxantrone. The potential role of TOP2 in mitoxantrone-inhibited HIF-1α expression was studied using pharmacologic inhibition, a knockdown approach, and TOP2 mutant cells. Moreover, involvement of mitoxantrone in proteasome-mediated degradation, transcription, and translation of HIF-1α was examined.Results: The TOP2-targeting mitoxantrone, but neither doxorubicin nor etoposide (VP-16), strongly inhibited HIF-1α expression under hypoxic conditions in a dose- and time-dependent manner. Surprisingly, the mitoxantrone-mediated inhibition of HIF-1α expression was largely independent of two TOP2 isozymes, proteasomal degradation, and transcription. Furthermore, mitoxantrone inhibited HIF-1α expression and function in a similar fashion as cycloheximide, suggesting that mitoxantrone might inhibit HIF-1α via a blockage at its translation step. In vitro translation experiments using HIF-1α mRNA further confirmed inhibition of HIF-1α translation by mitoxantrone. Interestingly, levels of the polysome-bound HIF-1α and VEGF-A mRNA were elevated and decreased after mitoxantrone treatment, respectively.Conclusions: We have identified the TOP2-targeting compound, mitoxantrone, as an HIF-1α inhibitor possibly through a translation inhibition mechanism, suggesting the possibility of an additional anticancer activity for mitoxantrone. Clin Cancer Res; 17(15); 5026–37. ©2011 AACR.
- Published
- 2023
8. Supplementary Methods, Figure Legends 1-5 from Mitoxantrone Inhibits HIF-1α Expression in a Topoisomerase II–Independent Pathway
- Author
-
Tsai-Kun Li and Yng-Miin Toh
- Abstract
PDF file - 97.5K
- Published
- 2023
9. Supplementary Figure 3 from DNA Topoisomerase III Alpha Regulates p53-Mediated Tumor Suppression
- Author
-
Tsai-Kun Li, Yen-Hsiu Yeh, Shu-Chun Teng, Tang-Long Shen, Hsiang-Chin Chen, Han-Wen Chang, Jia-Rong Fan, and Mei-Yi Hsieh
- Abstract
PDF file 1548KB, hTOP3alpha regulates TP53 mRNA and p53 protein stability
- Published
- 2023
10. Supplementary Figure 5 from DNA Topoisomerase III Alpha Regulates p53-Mediated Tumor Suppression
- Author
-
Tsai-Kun Li, Yen-Hsiu Yeh, Shu-Chun Teng, Tang-Long Shen, Hsiang-Chin Chen, Han-Wen Chang, Jia-Rong Fan, and Mei-Yi Hsieh
- Abstract
PDF file 3495K, Reduced expression of hTOP3alpha in renal and stomach tumor samples compared to the matched normal tissues
- Published
- 2023
11. Data from DNA Topoisomerase III Alpha Regulates p53-Mediated Tumor Suppression
- Author
-
Tsai-Kun Li, Yen-Hsiu Yeh, Shu-Chun Teng, Tang-Long Shen, Hsiang-Chin Chen, Han-Wen Chang, Jia-Rong Fan, and Mei-Yi Hsieh
- Abstract
Purpose: Human DNA topoisomerase III alpha (hTOP3α) is involved in DNA repair surveillance and cell-cycle checkpoints possibly through formatting complex with tumor suppressors. However, its role in cancer development remained unsolved.Experimental Design: Coimmunoprecipitation, sucrose gradient, chromatin immunoprecipitation (ChIP), real time PCR, and immunoblotting analyses were performed to determine interactions of hTOP3α with p53. Paired cell lines with different hTOP3α levels were generated via ectopic expression and short hairpin RNA (shRNA)-mediated knockdown approaches. Cellular tumorigenic properties were analyzed using cell counting, colony formation, senescence, soft agar assays, and mouse xenograft models.Results: The hTOP3α isozyme binds to p53 and cofractionizes with p53 in gradients differing from fractions containing hTOP3α and BLM. Knockdown of hTOP3α expression (sh-hTOP3α) caused a higher anchorage-independent growth of nontumorigenic RHEK-1 cells. Similarly, sh-hTOP3α and ectopic expression of hTOP3α in cancer cell lines caused increased and reduced tumorigenic abilities, respectively. Genetic and mutation experiments revealed that functional hTOP3α, p53, and p21 are required for this tumor-suppressive activity. Mechanism-wise, ChIP data revealed that hTOP3α binds to the p53 and p21 promoters and positively regulates their expression. Two proteins affect promoter recruitments of each other and collaborate in p21 expression. Moreover, sh-hTOP3α and sh-p53 in AGS cells caused a similar reduction in senescence and hTOP3α mRNA levels were lower in gastric and renal tumor samples.Conclusion: We concluded that hTOP3α interacts with p53, regulates p53 and p21 expression, and contributes to the p53-mediated tumor suppression. Clin Cancer Res; 20(6); 1489–501. ©2014 AACR.
- Published
- 2023
12. Supplementary Figure 3 from Mitoxantrone Inhibits HIF-1α Expression in a Topoisomerase II–Independent Pathway
- Author
-
Tsai-Kun Li and Yng-Miin Toh
- Abstract
PDF file - 172K
- Published
- 2023
13. Supplementary Figure 2 from Mitoxantrone Inhibits HIF-1α Expression in a Topoisomerase II–Independent Pathway
- Author
-
Tsai-Kun Li and Yng-Miin Toh
- Abstract
PDF file - 57.5K
- Published
- 2023
14. Supplementary Figure 2 from DNA Topoisomerase III Alpha Regulates p53-Mediated Tumor Suppression
- Author
-
Tsai-Kun Li, Yen-Hsiu Yeh, Shu-Chun Teng, Tang-Long Shen, Hsiang-Chin Chen, Han-Wen Chang, Jia-Rong Fan, and Mei-Yi Hsieh
- Abstract
PDF file 1796K, Forced expression of hTOP3alpha induced expression of p5315P, p21 and {gamma}H2AX, but not ATM1981P under DNA damaging-agent stimulation
- Published
- 2023
15. Activation of multiple proteolysis systems contributes to acute cadmium cytotoxicity
- Author
-
Yen-Hsiu Yeh, Chia-Chih Tsai, Tien-Wen Chen, Chieh-Hua Lee, Wei-Jer Chang, Mei-Yi Hsieh, and Tsai-Kun Li
- Subjects
A549 Cells ,Cytotoxins ,Proteolysis ,Clinical Biochemistry ,Humans ,Cell Biology ,General Medicine ,Reactive Oxygen Species ,Molecular Biology ,DNA Topoisomerases ,Cadmium ,HeLa Cells - Abstract
Cadmium exhibits both toxic and carcinogenic effects, and its cytotoxicity is linked to various cellular pathways, such as oxidative stress, ubiquitin-proteasome, and p53-mediated response pathways. The molecular mechanism(s) underlying cadmium cytotoxicity appears to be complex, but remains largely unclear. Here, we examined the effects of cadmium on the protein catabolism using two surrogate markers, DNA topoisomerases I and II alpha and its contribution to cytotoxicity. We have found that cadmium exposure induced time- and concentration-dependent decreases in the protein level of surrogate markers and therefore suggest that cadmium may be involved in proteolysis system activation. A pharmacological study further revealed the novel role(s) of these proteolytic activities and reactive oxygen species (ROS) in the cadmium-induced acute toxicity: (i) Proteasome inhibition only partially relieved the cadmium-induced proteolysis of topoisomerases; (ii) Moreover, we report for the first time that the activation of metalloproteases, serine proteases, and cysteine proteases contributes to the acute cadmium cytotoxicity; (iii) Consistent with the notion that both ROS generation and proteolysis system activation contribute to the cadmium-induced proteolysis and cytotoxicity, the scavenger N-acetylcysteine and aforementioned protease inhibition not only reduced the cadmium-induced topoisomerase degradation but also alleviated the cadmium-induced cell killing. Taken together, acute cadmium exposure may activate multiple proteolytic systems and ROS formation, subsequently leading to intracellular damage and cytotoxicity. Thus, our results provide a novel insight into potential action mechanism(s) by which cadmium exerts its cytotoxic effect and suggest potential strategies to prevent cadmium-associated acute toxicity.
- Published
- 2022
16. The role of extracellular vesicles in prostate cancer with clinical applications
- Author
-
Chun-Jung Lin, Tang-Long Shen, Jer Tsong Hsieh, Yu Ling Tai, Benjamin P C Chen, and Tsai-Kun Li
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Stromal cell ,Endosome ,Endocrinology, Diabetes and Metabolism ,Biology ,Extracellular Vesicles ,03 medical and health sciences ,Prostate cancer ,Paracrine signalling ,0302 clinical medicine ,Endocrinology ,Biomarkers, Tumor ,medicine ,Humans ,Tumor microenvironment ,Prostatic Neoplasms ,Prognosis ,medicine.disease ,Precision medicine ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,Intracellular - Abstract
In mammalian cells, extracellular vesicles (EVs) derived from the endosomal system carry many different kinds of bioactive molecule to deliver to recipient cells in a paracrine or endocrine manner. EVs can mediate local and systemic intercellular communications, including reeducating stromal cells, remodeling the architecture of the tumor microenvironment, modulating cancer metabolism and metastases, or even conferring drug resistance. Because the molecular and functional characteristics of prostate cancer (PCa) evolve over time, the bioactive molecule profiles/signatures of tumor-derived EVs (TDEs) reflect the real-time status of cancer cells. TDEs appear to be valuable diagnostic and prognostic biomarkers as well as potential therapeutic vehicles, suggesting their essential role in precision medicine of disease management. We summarized critical aspects of TDEs in PCa and discussed their potential clinical applications.
- Published
- 2020
17. Promoters of ASCL1- and NEUROD1-dependent genes are specific targets of lurbinectedin in SCLC cells
- Author
-
Federico Costanzo, Marta Martínez Diez, Gema Santamaría Nuñez, Juan Ignacio Díaz‐Hernandéz, Carlos Mario Genes Robles, Javier Díez Pérez, Emmanuel Compe, Romeo Ricci, Tsai‐Kun Li, Frédéric Coin, Juan Fernando Martínez Leal, Eva Maria Garrido‐Martin, and Jean Marc Egly
- Subjects
Repressor Proteins ,Lung Neoplasms ,Cell Line, Tumor ,Basic Helix-Loop-Helix Transcription Factors ,Molecular Medicine ,Humans ,Promoter Regions, Genetic ,Heterocyclic Compounds, 4 or More Rings ,Small Cell Lung Carcinoma ,Carbolines - Abstract
Small-Cell Lung Cancer (SCLC) is an aggressive neuroendocrine malignancy with a poor prognosis. Here, we focus on the neuroendocrine SCLC subtypes, SCLC-A and SCLC-N, whose transcription addiction was driven by ASCL1 and NEUROD1 transcription factors which target E-box motifs to activate up to 40% of total genes, the promoters of which are maintained in a steadily open chromatin environment according to ATAC and H3K27Ac signatures. This leverage is used by the marine agent lurbinectedin, which preferentially targets the CpG islands located downstream of the transcription start site, thus arresting elongating RNAPII and promoting its degradation. This abrogates the expression of ASCL1 and NEUROD1 and of their dependent genes, such as BCL2, INSM1, MYC, and AURKA, which are responsible for relevant SCLC tumorigenic properties such as inhibition of apoptosis and cell survival, as well as for a part of its neuroendocrine features. In summary, we show how the transcription addiction of these cells becomes their Achilles's heel, and how this is effectively exploited by lurbinectedin as a novel SCLC therapeutic endeavor.
- Published
- 2022
18. The paracrine induction of prostate cancer progression by caveolin-1
- Author
-
Ping Mu, Andrew Dang, Debabrata Saha, Jer Tsong Hsieh, Yu-An Chen, Chih Ho Lai, Tang-Long Shen, U-Ging Lo, Yu-Ling Tai, Ho Lin, Eun-Jin Yun, Elizabeth Hernandez, Su Deng, Tsai-Kun Li, and Chun-Jung Lin
- Subjects
Male ,Cancer Research ,Immunology ,Population ,Caveolin 1 ,Mice, SCID ,Biology ,Exosomes ,Neuroendocrine differentiation ,Article ,Metastasis ,Cellular and Molecular Neuroscience ,Prostate cancer ,Mice ,Cancer stem cell ,Cell Line, Tumor ,Paracrine Communication ,medicine ,Animals ,Humans ,lcsh:QH573-671 ,education ,education.field_of_study ,Cancer stem cells ,lcsh:Cytology ,Cancer ,Cell Biology ,medicine.disease ,Neoplasm Proteins ,Prostatic Neoplasms, Castration-Resistant ,Cancer cell ,Cancer research ,Stem cell ,Signal Transduction - Abstract
A subpopulation of cancer stem cells (CSCs) plays a critical role of cancer progression, recurrence, and therapeutic resistance. Many studies have indicated that castration-resistant prostate cancer (CRPC) is associated with stem cell phenotypes, which could further promote neuroendocrine transdifferentiation. Although only a small subset of genetically pre-programmed cells in each organ has stem cell capability, CSCs appear to be inducible among a heterogeneous cancer cell population. However, the inductive mechanism(s) leading to the emergence of these CSCs are not fully understood in CRPC. Tumor cells actively produce, release, and utilize exosomes to promote cancer development and metastasis, cancer immune evasion as well as chemotherapeutic resistance; the impact of tumor-derived exosomes (TDE) and its cargo on prostate cancer (PCa) development is still unclear. In this study, we demonstrate that the presence of Cav-1 in TDE acts as a potent driver to induce CSC phenotypes and epithelial–mesenchymal transition in PCa undergoing neuroendocrine differentiation through NFκB signaling pathway. Furthermore, Cav-1 in mCRPC-derived exosomes is capable of inducing radio- and chemo-resistance in recipient cells. Collectively, these data support Cav-1 as a critical driver for mCRPC progression.
- Published
- 2019
19. Selected ellipticine derivatives, known to target topoisomerase II, suppress the alternative lengthening of telomere (ALT) pathway in telomerase-negative cells
- Author
-
Sevil, Zencir, Meng-Hsun, Hsieh, Joel-Sean, Hsu, Yavuz, Ergun, Guan-Ling, Chou, Tsai-Kun, Li, Shu-Chun, Teng, and Zeki, Topcu
- Subjects
Fluorescent Antibody Technique ,Humans ,Telomere Homeostasis ,Topoisomerase II Inhibitors ,Antineoplastic Agents ,Ellipticines ,Telomerase ,In Situ Hybridization, Fluorescence ,Cell Line - Abstract
DNA topoisomerase and telomerase enzymes are popular targets of several anti-tumor drugs. Smooth proceeding of telomeric recombination requires Topoisomerase II (Top2), which is involved in telomere-telomere recombination through functioning in relaxation of positive supercoils among the cells adopting telomerase-independent Alternative lengthening of telomere (ALT) pathway. Most of the inhibitors reported so far have been designed to targetsolely telomerase-positive cells, which can potentially lead to therapeutic failure because tumor cells treated with telomerase inhibitors can activate the ALT pathway for telomere maintenance. Knowing that ALT cells are more sensitive against a Top2 inhibitor, ICRF-93 agent, compared to telomerase-positive cells, we analyzed two selected ellipticine derivatives that we recently reported as TopII-targeting compounds, to assess their effects on the formation of DNA breaks and suppression of ALT pathway.Cell viability, Comet, C-Circle assays, dot blot, immunofluorescence staining, and telomere fluorescence in situ hybridization (FISH) staining were used for determining the effect of the compounds on ALT status of tumor cells.Treatment of ALT cells with ellipticine derivatives resulted in the formation of DNA breaks and suppression of ALT-associated phenotypes in vitro. Our results will contribute to the development of therapeutic strategies combining telomerase and ALT pathway inhibitors.
- Published
- 2020
20. Selected Ellipticine Derivatives, Known To Target Topoisomerase Ii, Suppress The Alternative Lengthening Of Telomere (Alt) Pathway In Telomerase-Negative Cells
- Author
-
Joel-Sean Hsu, Yavuz Ergun, Sevil Zencir, Zeki Topcu, Guan-Ling Chou, Meng-Hsun Hsieh, Shu-Chun Teng, Tsai-Kun Li, and Ege Üniversitesi
- Subjects
0301 basic medicine ,Cancer Research ,Telomerase ,MTS assay ,telomerase inhibitor ,Fluorescent Antibody Technique ,0302 clinical medicine ,ellipticine derivative ,telomere length ,Topoisomerase II Inhibitors ,genetics ,ALT cell line ,Ellipticines ,telomere homeostasis ,antineoplastic agent ,In Situ Hybridization, Fluorescence ,gyrase inhibitor ,chemistry.chemical_classification ,Ellipticine derivatives ,DNA strand breakage ,SaOS-2 cell line ,biology ,medicine.diagnostic_test ,drug effect ,General Medicine ,Alternative lengthening of telomere ,unclassified drug ,female ,Oncology ,priority journal ,030220 oncology & carcinogenesis ,DNA supercoil ,cytotoxicity ,DNA topoisomerase II ,phenotype ,Antineoplastic Agents ,telomerase ,chemistry ,Article ,Cell Line ,03 medical and health sciences ,comet assay ,medicine ,Humans ,controlled study ,Viability assay ,human ,DNA topoisomerase (ATP hydrolysing) ,drug selectivity ,immunofluorescence ,fluorescence in situ hybridization ,cell viability ,Topoisomerase ,human cell ,icrf 93 ,Anti-cancer therapeutics ,In vitro ,Telomere ,030104 developmental biology ,Enzyme ,Cancer research ,biology.protein ,ellipticine ,Fluorescence in situ hybridization - Abstract
Background DNA topoisomerase and telomerase enzymes are popular targets of several anti-tumor drugs. Smooth proceeding of telomeric recombination requires Topoisomerase II (Top2), which is involved in telomere-telomere recombination through functioning in relaxation of positive supercoils among the cells adopting telomerase-independent Alternative lengthening of telomere (ALT) pathway. Most of the inhibitors reported so far have been designed to targetsolely telomerase-positive cells, which can potentially lead to therapeutic failure because tumor cells treated with telomerase inhibitors can activate the ALT pathway for telomere maintenance. Knowing that ALT cells are more sensitive against a Top2 inhibitor, ICRF-93 agent, compared to telomerase-positive cells, we analyzed two selected ellipticine derivatives that we recently reported as TopII-targeting compounds, to assess their effects on the formation of DNA breaks and suppression of ALT pathway. Methods Cell viability, Comet, C-Circle assays, dot blot, immunofluorescence staining, and telomere fluorescence in situ hybridization (FISH) staining were used for determining the effect of the compounds on ALT status of tumor cells. Results and conclusions Treatment of ALT cells with ellipticine derivatives resulted in the formation of DNA breaks and suppression of ALT-associated phenotypes in vitro. Our results will contribute to the development of therapeutic strategies combining telomerase and ALT pathway inhibitors., Turkish Scientific and Technical Research AssemblyTurkiye Bilimsel ve Teknolojik Arastirma Kurumu (TUBITAK) [115Z349], This research was supported by Grant 115Z349 (Z.T. and S.Z.) from the Turkish Scientific and Technical Research Assembly. the cell lines used in this study were provided by Drs. Shu-Chun Teng and Tsai-Kun Li (College of Medicine, National Taiwan University, Taipei, Taiwan).
- Published
- 2020
21. Design, synthesis, and biological evaluation of heterotetracyclic quinolinone derivatives as anticancer agents targeting topoisomerases
- Author
-
Tsung Chih Chen, Ting Yu Chang, Yi Song Chen, Mei Hsiang Lin, Jiann Fong Lee, Tsai-Kun Li, Yen Hsiu Yeh, Hao Syun Chou, Sung Bau Lee, Nian Zhe Lee, and Zheng Fang Liu
- Subjects
Pharmacology ,0303 health sciences ,biology ,010405 organic chemistry ,Chemistry ,DNA damage ,Topoisomerase ,Organic Chemistry ,Cell ,General Medicine ,Cleavage (embryo) ,01 natural sciences ,Molecular biology ,0104 chemical sciences ,03 medical and health sciences ,chemistry.chemical_compound ,medicine.anatomical_structure ,Apoptosis ,Drug Discovery ,Cancer cell ,medicine ,biology.protein ,Gene ,DNA ,030304 developmental biology - Abstract
A series of thiochromeno [2,3-c]quinolin-12-one derivatives with various substitutions were synthesized and evaluated as topoisomerase (Topo) inhibitors. Six (8, 10, 12, 14, 19, and 26) of 23 compounds showed strong inhibitory activities against Topo-mediated DNA relaxation and proliferation of five human cell lines including breast (MDA-MB-231, MDA-MB-468 and MCF7), colorectal (HCT116) and non-small cell lung (H1299) cancers. Among these, compounds 14 and 26 exhibited full inhibitory activities against Topo I at 3 μM and Topo IIα at 1 μM. Cancer cells treated with 26 accumulated DNA damage and were arrested at the G2/M phase. With time, cells proceeded to apoptosis, as revealed by increased amounts of cells with fragmented DNA and cleavage of caspase-8 and -9. In contrast, normal breast epithelial cells showed low sensitivity to 26. Taken together, our study identifies 26 as a potent Topo dual-inhibitor with low toxicity to normal cells, and elucidates that the terminal amino group of N-2-aminoethylamino or N-3-aminopropylamino at the 6th position and 8,10-di-halogen substituents on thiochromeno [2,3-c]quinolin-12-one are critical for the Topo-inhibiting and cancer-killing activities.
- Published
- 2019
22. Retraction notice to 'Design, synthesis, and biological evaluation of heterotetracyclic quinolinone derivatives as anticancer agents targeting topoisomerases' [Eur. J. Med. Chem. 190 (2020) 112074]
- Author
-
Ting Yu Chang, Hao Syun Chou, Sung Bau Lee, Yen Hsiu Yeh, Tsai-Kun Li, Tsung Chih Chen, Nian Zhe Lee, Yi Song Chen, Jiann Fong Lee, Zheng Fang Liu, and Mei Hsiang Lin
- Subjects
Pharmacology ,biology ,Design synthesis ,Notice ,Chemistry ,Topoisomerase ,Organic Chemistry ,Drug Discovery ,biology.protein ,General Medicine ,Combinatorial chemistry ,Biological evaluation - Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the authors. The authors regret to inform that they would like to withdraw this accepted article, due to serious errors in authorship, affiliations, material sources and supporting grant names/numbers. The authors sincerely apologize for these oversights and miscommunications the study caused.
- Published
- 2020
23. Regioselective synthesis and biological evaluation of N-substituted 2-aminoquinazolin-4-ones
- Author
-
Wen Hsiung Yeh, Pen Yuan Liao, Ming Hsuan Lu, Ming Chih Hsu, Chia Chi Lin, Tsai-Kun Li, Yi Hung Chen, Ying Cheng Chen, Yu Ting Liu, Tun Cheng Chien, Tsung Han Hsieh, Zhen Yuan Liao, and Yi Song Chen
- Subjects
Aqueous solution ,Molecular Structure ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Regioselectivity ,Antineoplastic Agents ,010402 general chemistry ,01 natural sciences ,Biochemistry ,Dimroth rearrangement ,Medicinal chemistry ,0104 chemical sciences ,chemistry.chemical_compound ,Sodium hydroxide ,Cyclization ,Cell Line, Tumor ,Nitriles ,Structural isomer ,Humans ,ortho-Aminobenzoates ,Physical and Theoretical Chemistry ,Biological evaluation ,Quinazolinones - Abstract
The reaction of methyl anthranilates with N-arylcyanamides in the presence of p-TsOH in t-BuOH under reflux afforded predominantly 3-arylquinazolin-4-ones. In contrast, the reaction of the same reactants with TMSCl in t-BuOH at 60 °C followed by the Dimroth rearrangement in aqueous ethanolic sodium hydroxide gave exclusively the regioisomers, 2-(N-arylamino)quinazolin-4-ones. The regioselective synthesis of N-aryl-substituted 2-aminoquinazolin-4-ones can be further applied to the synthesis of benzimidazo[2,1-b]quinazolin-12-ones.
- Published
- 2018
24. Inflammatory interferon activates HIF-1α-mediated epithelial-to-mesenchymal transition via PI3K/AKT/mTOR pathway
- Author
-
Tsai-Kun Li, Ho-Fu Hsiao, Yen-Cheng Yeh, Yen-Hsiu Yeh, and Tien-Wen Chen
- Subjects
0301 basic medicine ,Cancer Research ,Epithelial-Mesenchymal Transition ,MAP Kinase Signaling System ,HIF-1α ,Apoptosis ,medicine.disease_cause ,lcsh:RC254-282 ,Models, Biological ,03 medical and health sciences ,Phosphatidylinositol 3-Kinases ,Neoplasms ,medicine ,Tumor Microenvironment ,Humans ,Vasculogenic mimicry ,Epithelial–mesenchymal transition ,Hypoxia ,Protein kinase B ,Protein Kinase Inhibitors ,PI3K/AKT/mTOR pathway ,Oncogenesis ,Janus Kinases ,Tumor microenvironment ,Chemistry ,Research ,TOR Serine-Threonine Kinases ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Hypoxia-Inducible Factor 1, alpha Subunit ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,Oncology ,Epithelial-to-mesenchymal transition ,Inflammatory hypoxia ,Cancer cell ,Cancer research ,ras Proteins ,Interferon ,Interferons ,Signal transduction ,Carcinogenesis ,Proto-Oncogene Proteins c-akt ,Biomarkers ,Signal Transduction - Abstract
Background Tumor microenvironments (TMEs) activate various axes/pathways, predominantly inflammatory and hypoxic responses, impact tumorigenesis, metastasis and therapeutic resistance significantly. Although molecular pathways of individual TME are extensively studied, evidence showing interaction and crosstalk between hypoxia and inflammation remain unclear. Thus, we examined whether interferon (IFN) could modulate both inflammatory and hypoxic responses under normoxia and its relation with cancer development. Methods IFN was used to induce inflammation response and HIF-1α expression in various cancer cell lines. Corresponding signaling pathways were then analyzed by a combination of pharmacological inhibitors, immunoblotting, GST-Raf pull-down assays, dominant-negative and short-hairpin RNA-mediated knockdown approaches. Specifically, roles of functional HIF-1α in the IFN-induced epithelial-mesenchymal transition (EMT) and other tumorigenic propensities were examined by knockdown, pharmacological inhibition, luciferase reporter, clonogenic, anchorage-independent growth, wound-healing, vasculogenic mimicry, invasion and sphere-formation assays as well as cellular morphology observation. Results We showed for the first time that IFN induced functional HIF-1α expression in a time- and dose- dependent manner in various cancer cell lines under both hypoxic and normoxic conditions, and then leading to an activated HIF-1α pathway in an IFN-mediated pro-inflammatory TME. IFN regulates anti-apoptosis activity, cellular metastasis, EMT and vasculogenic mimicry by a novel mechanism through mainly the activation of PI3K/AKT/mTOR axis. Subsequently, pharmacological and genetic modulations of HIF-1α, JAK, PI3K/AKT/mTOR or p38 pathways efficiently abrogate above IFN-induced tumorigenic propensities. Moreover, HIF-1α is required for the IFN-induced invasiveness, tumorigenesis and vasculogenic mimicry. Further supports for the HIF-1α-dependent tumorigenesis were obtained from results of xenograft mouse model and sphere-formation assay. Conclusions Our mechanistic study showed an induction of HIF-1α and EMT ability in an IFN-mediated inflammatory TME and thus demonstrating a novel interaction between inflammatory and hypoxic TMEs. Moreover, targeting HIF-1α may be a potential target for inhibiting tumor tumorigenesis and EMT by decreasing cancer cells wound healing and anchorage-independent colony growth. Our results also lead to rationale guidance for developing new therapeutic strategies to prevent relapse via targeting TME-providing IFN signaling and HIF-1α programming. Electronic supplementary material The online version of this article (10.1186/s13046-018-0730-6) contains supplementary material, which is available to authorized users.
- Published
- 2018
25. Additional file 5: of Inflammatory interferon activates HIF-1α-mediated epithelial-to-mesenchymal transition via PI3K/AKT/mTOR pathway
- Author
-
Yen-Hsiu Yeh, Ho-Fu Hsiao, Yen-Cheng Yeh, Tien-Wen Chen, and Tsai-Kun Li
- Abstract
Figure S5. The IFN-α not only attenuated MX-induced apoptosis, but also promote PI3K- and MAPK-P38-dependent invasion activity.(A) IFN-α co-treatment reduced the MX-induced apoptotic cleavage of PARP1.(B) LY294002 (LY) and SB203580 (SB) could both effectively inhibit the IFN-α-induced invasion abilities. (PPT 237 kb)
- Published
- 2018
- Full Text
- View/download PDF
26. Additional file 3: of Inflammatory interferon activates HIF-1α-mediated epithelial-to-mesenchymal transition via PI3K/AKT/mTOR pathway
- Author
-
Yen-Hsiu Yeh, Ho-Fu Hsiao, Yen-Cheng Yeh, Tien-Wen Chen, and Tsai-Kun Li
- Abstract
Figure S3. The JAK/PI-3 K, AKT/GSK3β and p38/ERK/JNK axes contributed to the IFN-α-induced HIF-1α expression. (A) Knockdown of STAT1 (si-STAT1) expression has no effect on the IFN-α-induced HIF-1α expression. (B) Ectopic expression of PTEN antagonized the IFN-α-activated AKT/GSK3β pathway. (C-D) β-catenin inhibition by FH535 treatment not only decreased the IFN-α-induced expression of HIF-1α (C), but also reduced the IFN-α-induced active β-catenin (D). (PPT 221 kb)
- Published
- 2018
- Full Text
- View/download PDF
27. Additional file 1: of Inflammatory interferon activates HIF-1α-mediated epithelial-to-mesenchymal transition via PI3K/AKT/mTOR pathway
- Author
-
Yen-Hsiu Yeh, Ho-Fu Hsiao, Yen-Cheng Yeh, Tien-Wen Chen, and Tsai-Kun Li
- Abstract
Figure S1. IFN-α-induced HIF-1α expression in transcription- and translation-dependent, but 26S proteasome-independent. (A) RNA transcription is required for IFN-α mediated HIF-1α induction. 769-P cells were pre-treated 30 min. With or without 100 μM DRB and subsequently exposed for 24 h with either IFN-α (1000 units/ml) or desferoxamine (DFX, 260 μM), and protein labels determined by immunoblotting analysis with indicated antibodies. (B-C) Interferon-α treatment promotes the de novo synthesis of HIF-1α expression (B) and does not impact on the stability of HIF-1α. (C) Cells were treated with IFN-α before addition of (DRB, 100 μM), cycloheximide (CHX, 100 μM) or MG-132 (10 μM) (D) IFN-α stimulated HIF-1α expression similarly in the VHL-deficient 769-P cells with or without ectopic expression of functional VHL. (PPT 251 kb)
- Published
- 2018
- Full Text
- View/download PDF
28. Additional file 4: of Inflammatory interferon activates HIF-1α-mediated epithelial-to-mesenchymal transition via PI3K/AKT/mTOR pathway
- Author
-
Yen-Hsiu Yeh, Ho-Fu Hsiao, Yen-Cheng Yeh, Tien-Wen Chen, and Tsai-Kun Li
- Subjects
enzymes and coenzymes (carbohydrates) - Abstract
Figure S4. NF-κB is minimally involved in the IFN-α mediated HIF-1α accumulation. (A) IFN-α slightly activated IKK as suggested by a minimal increase in IkBaS32 phosphorylation. (B-D) Targeting IKK/IkBα/NF-κB pathway by Sulfasalazine (Sulfa, B), IkBα-M mutant (C) and si-p65 (D) do not alter much of IFN-α-induced HIF-1α expression. (PPT 201 kb)
- Published
- 2018
- Full Text
- View/download PDF
29. Additional file 2: of Inflammatory interferon activates HIF-1α-mediated epithelial-to-mesenchymal transition via PI3K/AKT/mTOR pathway
- Author
-
Yen-Hsiu Yeh, Ho-Fu Hsiao, Yen-Cheng Yeh, Tien-Wen Chen, and Tsai-Kun Li
- Abstract
Figure S2. IFN-α could up-regulate HIF-1α expression in the presence of 1% O2 with a different induction kinetics. Cells were treated with or without IFN-α, exposed to hypoxia (1% O2) and then harvested at indicated time points (3, 6, 9 and 12 h) for immunoblotting. (PPT 162 kb)
- Published
- 2018
- Full Text
- View/download PDF
30. Additional file 6: of Inflammatory interferon activates HIF-1α-mediated epithelial-to-mesenchymal transition via PI3K/AKT/mTOR pathway
- Author
-
Yen-Hsiu Yeh, Ho-Fu Hsiao, Yen-Cheng Yeh, Tien-Wen Chen, and Tsai-Kun Li
- Abstract
Figure S6. Direct effects of IFN-α on the expression of EMT and stemness biomarkers. (A-B) Cells were treated with 0.5, 1, 2.5 and 5 mg of anti-IFN-α antibodies and their impacts on the expression of EMT marker vimentin (A) and stemness marker Bmi1 genes (B) were determined by immunoblotting analysis. (PPT 133 kb)
- Published
- 2018
- Full Text
- View/download PDF
31. Involvement of p38 MAPK in the Anticancer Activity of Cultivated Cordyceps militaris
- Author
-
Wan-Jung Lai, Tang-Long Shen, Yen-Hsun Chen, Richard Chu, Shang-Min Chou, Tsai-Kun Li, Cheng-Hsiang Kao, Sheng-Hong Tsai, and Tzu-Wen Hong
- Subjects
Programmed cell death ,Poly ADP ribose polymerase ,Gene Expression ,HL-60 Cells ,p38 Mitogen-Activated Protein Kinases ,Histones ,Cordyceps militaris ,medicine ,Humans ,Cytotoxic T cell ,Protein kinase B ,Leukemia ,biology ,General Medicine ,biology.organism_classification ,medicine.disease ,Antineoplastic Agents, Phytogenic ,Up-Regulation ,Cell biology ,Complementary and alternative medicine ,Apoptosis ,Cordyceps ,Cancer cell ,Cancer research ,Drug Screening Assays, Antitumor ,DNA Damage ,Drugs, Chinese Herbal ,Signal Transduction - Abstract
Cordyceps militaris is a traditional Chinese medicine frequently used for tonic and therapeutic purposes. Reports from our laboratory and others have demonstrated that extracts of the cultivated fruiting bodies of C. militaris (CM) exhibit a potent cytotoxic effect against many cancer cell lines, especially human leukemia cells. Here, we further investigated the underlying mechanism through which CM is cytotoxic to cancer cells. The CM-mediated induction of PARP cleavage and its related DNA damage signal (γH2AX) was diminished by caspase inhibitor I. In contrast, a ROS scavenger failed to prevent CM-mediated leukemia cell death. Moreover, two signaling molecules, AKT and p38 MAPK, were activated during the course of apoptosis induction. Employing MTT analysis, we found that a p38 MAPK inhibitor but not an AKT inhibitor could rescue cells from CM-mediated cell death, as well as inhibit the cleavage of PARP, formation of apoptotic bodies and up-regulation of the γH2AX signal. These results suggest that CM-mediated leukemia cell death occurs through the activation of the p38 MAPK pathway, indicating its potential therapeutic effects against human leukemia.
- Published
- 2015
32. Producing irreversible topoisomerase II-mediated DNA breaks by site-specific Pt(II)-methionine coordination chemistry
- Author
-
Tsai-Kun Li, Shin Fu Chen, Ying Ren Wang, Yi Song Chen, Chyuan Chuan Wu, Yi Wen Liao, Tun Cheng Chien, Nei-Li Chan, Te-Sheng Lin, and Ko Ting Liu
- Subjects
0301 basic medicine ,Organoplatinum Compounds ,Stereochemistry ,Protein Conformation ,Antineoplastic Agents ,HL-60 Cells ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Protein structure ,Methionine ,Structural Biology ,Antigens, Neoplasm ,Cell Line, Tumor ,medicine ,Genetics ,Moiety ,Humans ,Topoisomerase II Inhibitors ,Poly-ADP-Ribose Binding Proteins ,Etoposide ,Podophyllotoxin ,chemistry.chemical_classification ,Topoisomerase ,DNA Breaks ,DNA ,DNA-Binding Proteins ,030104 developmental biology ,Enzyme ,DNA Topoisomerases, Type II ,Biochemistry ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Corrigendum ,medicine.drug - Abstract
Human type II topoisomerase (Top2) isoforms, hTop2α and hTop2β, are targeted by some of the most successful anticancer drugs. These drugs induce Top2-mediated DNA cleavage to trigger cell-death pathways. The potency of these drugs correlates positively with their efficacy in stabilizing the enzyme-mediated DNA breaks. Structural analysis of hTop2α and hTop2β revealed the presence of methionine residues in the drug-binding pocket, we therefore tested whether a tighter Top2-drug association may be accomplished by introducing a methionine-reactive Pt2+ into a drug to further stabilize the DNA break. Herein, we synthesized an organoplatinum compound, etoplatin-N2β, by replacing the methionine-juxtaposing group of the drug etoposide with a cis-dichlorodiammineplatinum(II) moiety. Compared to etoposide, etoplatin-N2β more potently inhibits both human Top2s. While the DNA breaks arrested by etoposide can be rejoined, those captured by etoplatin-N2β are practically irreversible. Crystallographic analyses of hTop2β complexed with DNA and etoplatin-N2β demonstrate coordinate bond formation between Pt2+ and a flanking methionine. Notably, this stable coordinate tether can be loosened by disrupting the structural integrity of drug-binding pocket, suggesting that Pt2+ coordination chemistry may allow for the development of potent inhibitors with protein conformation-dependent reversibility. This approach may be exploited to achieve isoform-specific targeting of human Top2s.
- Published
- 2017
33. Correction: DNA Topoisomerase II Is Involved in Regulation of Cyst Wall Protein Genes and Differentiation in Giardia lamblia
- Author
-
Bo-Chi Lin, Li-Hsin Su, Shih-Che Weng, Yu-Jiao Pan, Nei-Li Chan, Tsai-Kun Li, Hsin-Chih Wang, and Chin-Hung Sun
- Subjects
Chromatin Immunoprecipitation ,lcsh:Arctic medicine. Tropical medicine ,lcsh:RC955-962 ,lcsh:Public aspects of medicine ,Gene Expression Profiling ,DNA Mutational Analysis ,Public Health, Environmental and Occupational Health ,Oocysts ,Protozoan Proteins ,Correction ,lcsh:RA1-1270 ,Microarray Analysis ,Infectious Diseases ,DNA Topoisomerases, Type II ,Gene Expression Regulation ,Humans ,Giardia lamblia ,Promoter Regions, Genetic ,Protein Binding - Abstract
The protozoan Giardia lamblia differentiates into infectious cysts within the human intestinal tract for disease transmission. Expression of the cyst wall protein (cwp) genes increases with similar kinetics during encystation. However, little is known how their gene regulation shares common mechanisms. DNA topoisomerases maintain normal topology of genomic DNA. They are necessary for cell proliferation and tissue development as they are involved in transcription, DNA replication, and chromosome condensation. A putative topoisomerase II (topo II) gene has been identified in the G. lamblia genome. We asked whether Topo II could regulate Giardia encystation. We found that Topo II was present in cell nuclei and its gene was up-regulated during encystation. Topo II has typical ATPase and DNA cleavage activity of type II topoisomerases. Mutation analysis revealed that the catalytic important Tyr residue and cleavage domain are important for Topo II function. We used etoposide-mediated topoisomerase immunoprecipitation assays to confirm the binding of Topo II to the cwp promoters in vivo. Interestingly, Topo II overexpression increased the levels of cwp gene expression and cyst formation. Microarray analysis identified up-regulation of cwp and specific vsp genes by Topo II. We also found that the type II topoisomerase inhibitor etoposide has growth inhibition effect on Giardia. Addition of etoposide significantly decreased the levels of cwp gene expression and cyst formation. Our results suggest that Topo II has been functionally conserved during evolution and that Topo II plays important roles in induction of the cwp genes, which is key to Giardia differentiation into cysts.
- Published
- 2017
34. Synergistic property of cordycepin in cultivated Cordyceps militaris-mediated apoptosis in human leukemia cells
- Author
-
Shang-Min Chou, Tzu-Wen Hong, Wan-Jung Lai, Jui-Ya Lai, Sz Hsien Yu, Tang-Long Shen, Tsai-Kun Li, Richard Chu, Cheng-Hsiang Kao, Sheng-Hong Tsai, Shih-Torng Ding, and Yen-Hsun Chen
- Subjects
Pharmaceutical Science ,Antineoplastic Agents ,Apoptosis ,HL-60 Cells ,DNA laddering ,chemistry.chemical_compound ,Drug Discovery ,Cordyceps militaris ,Humans ,Cytotoxic T cell ,Cell Proliferation ,Pharmacology ,Cordyceps ,Deoxyadenosines ,biology ,Cordycepin ,biology.organism_classification ,Apoptotic body ,Molecular biology ,Complementary and alternative medicine ,Biochemistry ,chemistry ,Cancer cell ,Molecular Medicine - Abstract
Cordyceps militaris is a well-known Chinese traditional medicinal mushroom frequently used for tonics and recently of a potential interest for cancer intervention. Here, we explored the cancer cell killing activity of the hot water extracts of C. militaris cultured mycelia (CM(MY)) and cultivated fruiting bodies (CM(FB)). We found that CM(FB) exhibited a greater cytotoxic effect against various cancer cells over CM(MY). Apoptotic phenotypes including apoptotic body formation, DNA laddering, caspase 3 activation and cleavage of PARP proteins were induced by CM(FB) treatment but only slightly induced by same concentration of CM(MY) treatment in human HL-60 leukemia cells. Cordycepin in CM(FB) (10.47 mg/g) is significantly higher (∼ 15.2 times) than that of CM(MY) (0.69 mg/g). Using isobolographic analysis, the synergy of cytotoxicity was observed across different combined concentrations of CM(MY) and cordycepin. By complementing cordycepin into CM(MY) to the level comparable with CM(FB), we observed that CM(MY) (500 μg/ml) with cordycepin (4.8 μg/ml) induced apoptosis to a level similar to that induced by CM(FB) (500 μg/ml). Together, our results suggest that cordycepin possesses a synergistic cytotoxic effect with Cordyceps militaris-mediated apoptosis in human leukemia cells and therefore explaining a better anti-proliferating activity of CM(FB) over CM(MY).
- Published
- 2014
35. DNA Topoisomerase III Alpha Regulates p53-Mediated Tumor Suppression
- Author
-
Han-Wen Chang, Tsai-Kun Li, Jia-Rong Fan, Hsiang-Chin Chen, Tang-Long Shen, Mei Yi Hsieh, Yen-Hsiu Yeh, and Shu-Chun Teng
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Chromatin Immunoprecipitation ,Cancer Research ,DNA repair ,Immunoblotting ,Mice, SCID ,Biology ,Real-Time Polymerase Chain Reaction ,medicine.disease_cause ,Small hairpin RNA ,Mice ,Mice, Inbred NOD ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Immunoprecipitation ,Regulation of gene expression ,Gene knockdown ,Mutation ,Molecular biology ,Gene Expression Regulation, Neoplastic ,Real-time polymerase chain reaction ,DNA Topoisomerases, Type I ,Oncology ,Heterografts ,Ectopic expression ,Tumor Suppressor Protein p53 ,Chromatin immunoprecipitation - Abstract
Purpose: Human DNA topoisomerase III alpha (hTOP3α) is involved in DNA repair surveillance and cell-cycle checkpoints possibly through formatting complex with tumor suppressors. However, its role in cancer development remained unsolved. Experimental Design: Coimmunoprecipitation, sucrose gradient, chromatin immunoprecipitation (ChIP), real time PCR, and immunoblotting analyses were performed to determine interactions of hTOP3α with p53. Paired cell lines with different hTOP3α levels were generated via ectopic expression and short hairpin RNA (shRNA)-mediated knockdown approaches. Cellular tumorigenic properties were analyzed using cell counting, colony formation, senescence, soft agar assays, and mouse xenograft models. Results: The hTOP3α isozyme binds to p53 and cofractionizes with p53 in gradients differing from fractions containing hTOP3α and BLM. Knockdown of hTOP3α expression (sh-hTOP3α) caused a higher anchorage-independent growth of nontumorigenic RHEK-1 cells. Similarly, sh-hTOP3α and ectopic expression of hTOP3α in cancer cell lines caused increased and reduced tumorigenic abilities, respectively. Genetic and mutation experiments revealed that functional hTOP3α, p53, and p21 are required for this tumor-suppressive activity. Mechanism-wise, ChIP data revealed that hTOP3α binds to the p53 and p21 promoters and positively regulates their expression. Two proteins affect promoter recruitments of each other and collaborate in p21 expression. Moreover, sh-hTOP3α and sh-p53 in AGS cells caused a similar reduction in senescence and hTOP3α mRNA levels were lower in gastric and renal tumor samples. Conclusion: We concluded that hTOP3α interacts with p53, regulates p53 and p21 expression, and contributes to the p53-mediated tumor suppression. Clin Cancer Res; 20(6); 1489–501. ©2014 AACR.
- Published
- 2014
36. A Negative Feedback of the HIF-1α Pathway via Interferon-Stimulated Gene 15 and ISGylation
- Author
-
Tsai-Kun Li, Mei Yi Hsieh, Yen-Hsiu Yeh, Yu-Chen Yang, and Yen-Cheng Yeh
- Subjects
Cancer Research ,Biology ,Response Elements ,Cell Line, Tumor ,Gene expression ,Humans ,Protein Interaction Domains and Motifs ,Hypoxia ,Ubiquitins ,Transcription factor ,Cell Proliferation ,Regulation of gene expression ,Gene knockdown ,Interferon-stimulated gene ,Hypoxia-Inducible Factor 1, alpha Subunit ,ISG15 ,Molecular biology ,Cell biology ,Reverse transcription polymerase chain reaction ,Cell Transformation, Neoplastic ,Gene Expression Regulation ,Oncology ,Cytokines ,Interferons ,Signal transduction ,Protein Binding ,Signal Transduction - Abstract
Purpose: The IFN-stimulated gene 15 (ISG15)- and ubiquitin-conjugation pathways play roles in mediating hypoxic and inflammatory responses. To identify interaction(s) between these two tumor microenvironments, we investigated the effect of ISG15 on the activity of the master hypoxic transcription factor HIF-1α. Experimental Design: IFN and desferoxamine treatments were used to induce the expression of ISGs and HIF-1α, respectively. Interactions between HIF-1α and the ISG15 and ISGylation system were studied using knockdown of mRNA expression, immunoblotting, coimmunoprecipitation, and pull-down analyses. Effects of the ISG15 and ISGylation system on the HIF-1α–directed processes were examined using reporter, reverse transcription polymerase chain reaction (RT-PCR), and tumorigenic growth assays. Results: We found that the level of the free form of HIF-1α is differentially regulated by IFN treatment, and that the free ISG15 level is lower under hypoxia. Mechanism-directed studies have shown that HIF-1α not only interacts physically with ISG15, but is also ISGylated in multiple domains. ISG15 expression disrupts the functional dimerization of HIF-1α and -1β. Subsequently, expression of the ISG15 and/or ISGylation system attenuates HIF-1α–mediated gene expression and tumorigenic growth. Conclusion: In summary, our results revealed cross-talk between inflammatory and hypoxic pathways through the ISGylation of HIF-1α. On the basis of these results, we propose a novel negative feedback loop for the HIF-1α–mediated pathway involving the regulation of HIF-1α via IFN-induced ISGylation. Clin Cancer Res; 19(21); 5927–39. ©2013 AACR.
- Published
- 2013
37. On the structural basis and design guidelines for type II topoisomerase-targeting anticancer drugs
- Author
-
Nei-Li Chan, Yi-Ching Li, Ying-Ren Wang, Tsai-Kun Li, and C.-K. Wu
- Subjects
Drug ,Amsacrine ,Models, Molecular ,media_common.quotation_subject ,Antineoplastic Agents ,Guidelines as Topic ,Drug action ,Cleavage (embryo) ,chemistry.chemical_compound ,Structure-Activity Relationship ,Structural Biology ,Antigens, Neoplasm ,Genetics ,Humans ,Topoisomerase II Inhibitors ,Poly-ADP-Ribose Binding Proteins ,media_common ,biology ,Topoisomerase ,DNA-Binding Proteins ,DNA Topoisomerases, Type II ,Biochemistry ,chemistry ,Drug Design ,biology.protein ,Biophysics ,Topoisomerase-II Inhibitor ,Mitoxantrone ,Type II topoisomerase ,DNA - Abstract
Type II topoisomerases (Top2s) alter DNA topology via the formation of an enzyme–DNA adduct termed cleavage complex, which harbors a transient double-strand break in one DNA to allow the passage of another. Agents targeting human Top2s are clinically active anticancer drugs whose trapping of Top2-mediated DNA breakage effectively induces genome fragmentation and cell death. To understand the structural basis of this drug action, we previously determined the structure of human Top2 β-isoform forming a cleavage complex with the drug etoposide and DNA, and described the insertion of drug into DNA cleavage site and drug-induced decoupling of catalytic groups. By developing a post-crystallization drug replacement procedure that simplifies structural characterization of drug-stabilized cleavage complexes, we have extended the analysis toward other structurally distinct drugs, m-AMSA and mitoxantrone. Besides the expected drug intercalation, a switch in ribose puckering in the 3′-nucleotide of the cleavage site was robustly observed in the new structures, representing a new mechanism for trapping the Top2 cleavage complex. Analysis of drug-binding modes and the conformational landscapes of the drug-binding pockets provide rationalization of the drugs’ structural-activity relationships and explain why Top2 mutants exhibit differential effects toward each drug. Drug design guidelines were proposed to facilitate the development of isoform-specific Top2-targeting anticancer agents.
- Published
- 2013
38. Topoisomerase II-Mediated DNA Cleavage and Mutagenesis Activated by Nitric Oxide Underlie the Inflammation-Associated Tumorigenesis
- Author
-
Han-Yi Elizabeth Chou, Yu-Chen Yang, Tsai-Kun Li, Wei-Jer Chang, Pei-Han Tai, and Tang-Long Shen
- Subjects
Genome instability ,Pyridines ,Physiology ,DNA damage ,9,10-Dimethyl-1,2-benzanthracene ,Clinical Biochemistry ,HL-60 Cells ,Nitric Oxide ,medicine.disease_cause ,Biochemistry ,Cell Line ,Nitric oxide ,S-Nitrosoglutathione ,Mice ,chemistry.chemical_compound ,medicine ,Animals ,Humans ,Nitric Oxide Donors ,DNA Cleavage ,Molecular Biology ,Etoposide ,General Environmental Science ,Inflammation ,Mice, Knockout ,biology ,Topoisomerase ,Mutagenesis ,Cell Biology ,HCT116 Cells ,Molecular biology ,Coculture Techniques ,Cell Transformation, Neoplastic ,DNA Topoisomerases, Type II ,chemistry ,Cancer research ,biology.protein ,General Earth and Planetary Sciences ,Carcinogenesis ,medicine.drug - Abstract
Both cancer-suppressing and cancer-promoting properties of reactive nitrogen and oxygen species (RNOS) have been suggested to play a role in tumor pathology, particularly those activities associated with chronic inflammation. Here, we address the impact of nitric oxide (NO) on the induction of DNA damage and genome instability with a specific focus on the involvement of topoisomerase II (TOP2). We also investigate the contribution of NO to the formation of skin melanoma in mice.Similar to the TOP2-targeting drug, etoposide (VP-16), the NO-donor, S-nitrosoglutathione (GSNO), induces skin melanomas formation in 7,12-dimethyl- benz[a]anthracene (DMBA)-initiated mice. To explore the mechanism(s) underlying this NO-induced tumorigenesis, we use a co-culture model system to demonstrate that inflamed macrophages with inducible NO synthase (iNOS) expression cause γ-H2AX activation, p53 phosphorylation, and chromosome DNA breaks in the target cells. Inhibitor experiments revealed that NO and TOP2 isozymes are responsible for the above described cellular phenotypes. Notably, NO, unlike VP-16, preferentially induces the formation of TOP2β cleavable complexes (TOP2βcc) in cells. Moreover, GSNO induced TOP2-dependent DNA sequence rearrangements and cytotoxicity. Furthermore, the incidences of GSNO- and VP-16-induced skin melanomas were also observed to be lower in the skin-specific top2β-knockout mice. Our results suggest that TOP2 isozymes contribute to NO-induced mutagenesis and subsequent cancer development during chronic inflammation.We provide the first experimental evidence for the functional role of TOP2 in NO-caused DNA damage, mutagenesis, and carcinogenesis. Notably, these studies contribute to our molecular understanding of the cancer-promoting actions of RNOS during chronic inflammation.
- Published
- 2013
39. microRNA-183 Mediates Protective Postconditioning of the Liver by Repressing Apaf-1
- Author
-
Han-Chen Lin, Er-Yen Yen, I-Rue Lai, Tsai-Kun Li, and Shin-Yun Liu
- Subjects
0301 basic medicine ,Male ,Programmed cell death ,Physiology ,Mrna expression ,Clinical Biochemistry ,Clone (cell biology) ,Gene Expression ,Apoptosis ,Biology ,Pharmacology ,Biochemistry ,03 medical and health sciences ,Mice ,0302 clinical medicine ,microRNA ,Animals ,Ischemic Postconditioning ,Molecular Biology ,General Environmental Science ,Microarray analysis techniques ,Cell Biology ,Rats ,Oxygen ,MicroRNAs ,Oxidative Stress ,030104 developmental biology ,Apoptotic Protease-Activating Factor 1 ,Glucose ,Gene Expression Regulation ,Liver ,030220 oncology & carcinogenesis ,Reperfusion Injury ,General Earth and Planetary Sciences ,RNA Interference ,Injury model ,In vivo experiment ,Signal Transduction - Abstract
Ischemic postconditioning (iPoC) is known to mitigate ischemia-reperfusion (IR) injury of the liver, the mechanisms of which remain to be elucidated. This study explored the role of microRNA-183 (miR-183) in the protective mechanism of iPoC.Microarray analysis showed miR-183 was robustly expressed in rats' livers with iPoC. miR-183 repressed the mRNA expression of Apaf-1, which is an apoptosis promoting factor. Using an oxygen-glucose deprivation (OGD) injury model in Clone 9 cells, hypoxic postconditioning (HPoC) and an miR-183 mimetic significantly decreased cell death after OGD, but miR-183 inhibitors eliminated the protection of HPoC. The increased expression of Apaf-1 and the downstream activation of capsase-3/9 after OGD were mitigated by HPoC or the addition of miR-183 mimetics, whereas miR-183 inhibitor diminished the effect of HPoC on Apaf-1-caspase signaling. In the in vivo experiment, iPoC and agomiR-183 decreased the expression of serum ALT after liver IR in the mice, but antagomiR-183 mitigated the effect of iPoC. The results of hematoxylin and eosin and TUNEL staining were compatible with the biochemical assay. Moreover, iPoC and agomiR-183 decreased the expression of Apaf-1 and 4-HNE after IR injury in mouse livers, whereas the antagomiR-mediated prevention of miR-183 expression led to increased protein expression of Apaf-1 and 4-HNE in the postischemic livers.Our experiment showed the first time that miR-183 was induced in protective postconditioning and reduced reperfusion injury of the livers via the targeting of apoptotic signaling.miR-183 mediated the tolerance induced by iPoC in livers via Apaf-1 repressing. Antioxid. Redox Signal. 26, 583-597.
- Published
- 2016
40. Trichodermin induces c-Jun N-terminal kinase-dependent apoptosis caused by mitotic arrest and DNA damage in human p53-mutated pancreatic cancer cells and xenografts
- Author
-
Yung Wei Lin, Po Chuan Wang, Michael Hsiao, Yen Hsiu Yeh, Jih Jung Chen, Ming Hsien Chien, Tsai-Kun Li, Jyh Ming Chow, Tzong-Huei Lee, Wei Jiunn Lee, Shih-Wei Wang, and Kuo Tai Hua
- Subjects
0301 basic medicine ,Male ,Cancer Research ,DNA damage ,Trichodermin ,Mitosis ,Apoptosis ,Mice, SCID ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Pancreatic cancer ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Kinase ,c-jun ,Intrinsic apoptosis ,JNK Mitogen-Activated Protein Kinases ,Cancer ,medicine.disease ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,chemistry ,Cancer research ,Tumor Suppressor Protein p53 ,DNA Damage ,Signal Transduction - Abstract
Pancreatic cancer is an aggressive malignancy, which generally responds poorly to chemotherapy. In this study, trichodermin, an endophytic fungal metabolite from Nalanthamala psidii, was identified as a potent and selective antitumor agent in human pancreatic cancer. Trichodermin exhibited antiproliferative effects against pancreatic cancer cells, especially p53-mutated cells (MIA PaCa-2 and BxPC-3) rather than normal pancreatic epithelial cells. We found that trichodermin induced caspase-dependent and mitochondrial intrinsic apoptosis. Trichodermin also increased apoptosis through mitotic arrest by activating Cdc2/cyclin B1 complex activity. Moreover, trichodermin promoted the activation of c-Jun N-terminal kinase (JNK), and inhibition of JNK by its inhibitor, shRNA, or siRNA significantly reversed trichodermin-mediated caspase-dependent apoptosis. Trichodermin triggered DNA damage stress to activate p53 function for executing apoptosis in p53-mutated cells. Importantly, we demonstrated that trichodermin with efficacy similar to gemcitabine, profoundly suppressed tumor growth through inducing intratumoral DNA damage and JNK activation in orthotopic pancreatic cancer model. Based on these findings, trichodermin is a potential therapeutic agent worthy of further development into a clinical trial candidate for treating cancer, especially the mutant p53 pancreatic cancer.
- Published
- 2016
41. Rottlerin potentiates camptothecin-induced cytotoxicity in human hormone refractory prostate cancers through increased formation and stabilization of topoisomerase I-DNA cleavage complexes in a PKCδ-independent pathway
- Author
-
Ching-Shih Chen, Tsai-Kun Li, Yunn-Fang Ho, Lih-Ching Hsu, Jui-Ling Hsu, and Jih-Hwa Guh
- Subjects
Male ,Neoplasms, Hormone-Dependent ,Cell Survival ,DNA repair ,DNA damage ,Blotting, Western ,Cell Culture Techniques ,Apoptosis ,Topoisomerase-I Inhibitor ,Transfection ,Biochemistry ,Article ,chemistry.chemical_compound ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,In Situ Nick-End Labeling ,medicine ,Humans ,Benzopyrans ,heterocyclic compounds ,DNA Cleavage ,RNA, Small Interfering ,Pharmacology ,biology ,Topoisomerase ,Acetophenones ,Prostatic Neoplasms ,Drug Synergism ,Flow Cytometry ,Molecular biology ,Protein Kinase C-delta ,DNA Topoisomerases, Type I ,chemistry ,Cancer research ,biology.protein ,DNA fragmentation ,Camptothecin ,Topoisomerase I Inhibitors ,Rottlerin ,medicine.drug - Abstract
Combination therapy, which can optimize killing activity to cancers and minimize drug resistance, is a mainstream therapy against hormone-refractory prostate cancers (HRPCs). Rottlerin, a natural polyphenolic component, synergistically increased PC-3 (a HRPC cell line) apoptosis induced by camptothecin (a topoisomerase I inhibitor). Using siRNA technique to knockdown protein kinase C-δ (PKCδ), the data showed that rottlerin-mediated synergistic effect was PKCδ-independent, although rottlerin has been used as a PKCδ inhibitor. Rottlerin potentiated camptothecin-induced DNA fragmentation at S phase and ATM phosphorylation at Ser1981. The effect was correlated to apoptosis (r2 = 0.9). To detect upstream signals, the data showed that camptothecin acted on and stabilized topoisomerase I-DNA complex, leading to the formation of camptothecin-trapped cleavage complexes (TOP1cc). The effect was potentiated by rottlerin. To determine DNA repair capability, the time-related γH2A.X formation was examined after camptothecin removal. Consequently, rottlerin significantly inhibited camptothecin removal-mediated decline of γH2A.X formation at S phase, indicating the impairment of DNA repair activity in the presence of rottlerin. The combinatory treatment of camptothecin and rottlerin induced conformational change and activation of Bax and formation of truncated Bad, suggesting the contribution of mitochondria stress to apoptosis. In summary, the data suggest that rottlerin-mediated camptothecin sensitization is through the augmented stabilization of TOP1cc, leading to an increase of DNA damage stress and, possibly, an impairment of DNA repair capability. Subsequently, mitochondria-involved apoptosis is triggered through Bax activation and truncated Bad formation. The novel discovery may provide an anticancer approach of combinatory use between rottlerin and camptothecin for the treatment of HRPCs.
- Published
- 2012
42. Multiple Domains of the Tobacco mosaic virus p126 Protein Can Independently Suppress Local and Systemic RNA Silencing
- Author
-
Shih-Shun Lin, Ting-Hsuan Hung, Tang-Long Shen, Li-Ya Wang, Tsai-Kun Li, and Nai-Chun Lin
- Subjects
RNA-induced transcriptional silencing ,Physiology ,RNA-induced silencing complex ,DNA Mutational Analysis ,Green Fluorescent Proteins ,Trans-acting siRNA ,Viral Proteins ,Tobacco ,Tobacco mosaic virus ,Gene silencing ,Plant Diseases ,Genetics ,biology ,DNA Helicases ,Helicase ,Methyltransferases ,General Medicine ,Argonaute ,Protein Structure, Tertiary ,Plant Leaves ,Tobacco Mosaic Virus ,RNA silencing ,Gene Expression Regulation ,biology.protein ,RNA Interference ,Agronomy and Crop Science - Abstract
Small RNA-mediated RNA silencing is a widespread antiviral mechanism in plants and other organisms. Many viruses encode suppressors of RNA silencing for counter-defense. The p126 protein encoded by Tobacco mosaic virus (TMV) has been reported to be a suppressor of RNA silencing but the mechanism of its function remains unclear. This protein is unique among the known plant viral silencing suppressors because of its large size and multiple domains. Here, we report that the methyltransferase, helicase, and nonconserved region II (NONII) of p126 each has silencing-suppressor function. The silencing-suppression activities of methyltransferase and helicase can be uncoupled from their enzyme activities. Specific amino acids in NONII previously shown to be crucial for viral accumulation and symptom development are also crucial for silencing suppression. These results suggest that some viral proteins have evolved to possess modular structural domains that can independently interfere with host silencing, and that this may be an effective mechanism of increasing the robustness of a virus.
- Published
- 2012
43. Mitoxantrone Inhibits HIF-1α Expression in a Topoisomerase II–Independent Pathway
- Author
-
Yng-Miin Toh and Tsai-Kun Li
- Subjects
Proteasome Endopeptidase Complex ,Cancer Research ,Antineoplastic Agents ,Cycloheximide ,Biology ,Pharmacology ,Gene Knockout Techniques ,chemistry.chemical_compound ,Cell Line, Tumor ,medicine ,Humans ,Doxorubicin ,RNA, Messenger ,Etoposide ,Gene knockdown ,Mitoxantrone ,Topoisomerase ,Hypoxia-Inducible Factor 1, alpha Subunit ,Kidney Neoplasms ,DNA Topoisomerases, Type II ,Oncology ,chemistry ,Cell culture ,Protein Biosynthesis ,Mutation ,biology.protein ,Signal transduction ,Colorectal Neoplasms ,Signal Transduction ,medicine.drug - Abstract
Purpose: Solid tumors encounter a growth-limiting hypoxic microenvironment as they develop. Hypoxia-inducible factors (HIF) play important roles in hypoxia-associated tumor development and therapeutic resistance. Targeting the HIF pathway (especially HIF-1α) represents a promising cancer treatment strategy. Here, we report a novel class of HIF-1α inhibitors and the possible molecular basis of inhibition. Experimental Design: We analyzed the inhibitory effects of clinically used topoisomerase II (TOP2)–targeting drugs on HIF-1α expression with a primary focus on mitoxantrone. The potential role of TOP2 in mitoxantrone-inhibited HIF-1α expression was studied using pharmacologic inhibition, a knockdown approach, and TOP2 mutant cells. Moreover, involvement of mitoxantrone in proteasome-mediated degradation, transcription, and translation of HIF-1α was examined. Results: The TOP2-targeting mitoxantrone, but neither doxorubicin nor etoposide (VP-16), strongly inhibited HIF-1α expression under hypoxic conditions in a dose- and time-dependent manner. Surprisingly, the mitoxantrone-mediated inhibition of HIF-1α expression was largely independent of two TOP2 isozymes, proteasomal degradation, and transcription. Furthermore, mitoxantrone inhibited HIF-1α expression and function in a similar fashion as cycloheximide, suggesting that mitoxantrone might inhibit HIF-1α via a blockage at its translation step. In vitro translation experiments using HIF-1α mRNA further confirmed inhibition of HIF-1α translation by mitoxantrone. Interestingly, levels of the polysome-bound HIF-1α and VEGF-A mRNA were elevated and decreased after mitoxantrone treatment, respectively. Conclusions: We have identified the TOP2-targeting compound, mitoxantrone, as an HIF-1α inhibitor possibly through a translation inhibition mechanism, suggesting the possibility of an additional anticancer activity for mitoxantrone. Clin Cancer Res; 17(15); 5026–37. ©2011 AACR.
- Published
- 2011
44. Structural Basis of Type II Topoisomerase Inhibition by the Anticancer Drug Etoposide
- Author
-
Yu-Jen Yu, Tien-Jui Yen, Nei-Li Chan, Chia-Wang Chiang, Tsai-Kun Li, Li-Ying Lin, Te-Sheng Lin, Lynn Farh, and C.-K. Wu
- Subjects
Models, Molecular ,Base pair ,Population ,Crystallography, X-Ray ,Cleavage (embryo) ,Structure-Activity Relationship ,chemistry.chemical_compound ,Catalytic Domain ,medicine ,Humans ,Topoisomerase II Inhibitors ,A-DNA ,Protein Structure, Quaternary ,education ,Base Pairing ,Etoposide ,education.field_of_study ,Multidisciplinary ,biology ,Topoisomerase ,DNA ,Protein Structure, Tertiary ,DNA-Binding Proteins ,DNA Topoisomerases, Type II ,Biochemistry ,chemistry ,Drug Resistance, Neoplasm ,Mutation ,biology.protein ,Mutant Proteins ,Protein Multimerization ,Type II topoisomerase ,medicine.drug - Abstract
Type II topoisomerases (TOP2s) resolve the topological problems of DNA by transiently cleaving both strands of a DNA duplex to form a cleavage complex through which another DNA segment can be transported. Several widely prescribed anticancer drugs increase the population of TOP2 cleavage complex, which leads to TOP2-mediated chromosome DNA breakage and death of cancer cells. We present the crystal structure of a large fragment of human TOP2β complexed to DNA and to the anticancer drug etoposide to reveal structural details of drug-induced stabilization of a cleavage complex. The interplay between the protein, the DNA, and the drug explains the structure-activity relations of etoposide derivatives and the molecular basis of drug-resistant mutations. The analysis of protein-drug interactions provides information applicable for developing an isoform-specific TOP2-targeting strategy.
- Published
- 2011
45. Sodium salicylate acts through direct inhibition of phosphoinositide 3-kinase-like kinases to modulate topoisomerase-mediated DNA damage responses
- Author
-
Tsai-Kun Li, Tang-Long Shen, Ting-Hsiang Huang, Jia-Rong Fan, and Chen-Yu Wen
- Subjects
DNA damage ,Sodium Salicylate ,Cell Cycle Proteins ,Ataxia Telangiectasia Mutated Proteins ,DNA-Activated Protein Kinase ,Protein Serine-Threonine Kinases ,Pharmacology ,MAP2K7 ,chemistry.chemical_compound ,Replication Protein A ,Humans ,Drug Interactions ,ASK1 ,Phosphorylation ,Kinase activity ,Protein kinase A ,Sodium salicylate ,Cell Death ,Cyclooxygenase 2 Inhibitors ,biology ,Tumor Suppressor Proteins ,Anti-Inflammatory Agents, Non-Steroidal ,Cyclin-dependent kinase 2 ,NF-kappa B ,HCT116 Cells ,DNA-Binding Proteins ,chemistry ,Biochemistry ,biology.protein ,Camptothecin ,Cyclin-dependent kinase 9 ,Tumor Suppressor Protein p53 ,DNA Topoisomerases ,DNA Damage ,Signal Transduction - Abstract
Chemopreventive non-steroidal anti-inflammatory drugs (NSAIDs) exhibit diverse pharmacological and biological activities mainly through their inhibitory effect on cyclooxygenase (COX). However, COX-independent mechanisms involving kinase inhibition have been proposed to explain certain therapeutic effects of NSAIDs. Here, we explored the potential relationship between chemopreventive NSAIDs and DNA damage responses induced by treatment with topoisomerase-targeting drugs. (1) Sodium salicylate, a non-COX-selective NSAID, was shown to reduce DNA damage-induced RPA and p53 phosphorylation. (2) The formation of enzyme cleavable complexes by topoisomerase-targeting drugs was not affected in the presence of sodium salicylate. (3) The attenuating effect of NSAIDs on the DNA damage responses is COX-2-independent, since COX-2-selective inhibitors failed to inhibit DNA damage-induced phosphorylation of replication protein A (RPA) and p53. (4) This COX-2-independent attenuating effect was mediated through interference of neither nuclear factor kappa B nor extracellular signal-regulated kinase pathways. (5) The activation of ataxia telangiectasia mutated (ATM) kinase and DNA-dependent protein kinase (DNA-PK), two key signal transducers upstream of RPA and p53, was found to be significantly reduced with sodium salicylate treatment. (6) Most importantly, sodium salicylate and other NSAIDs directly inhibited kinase activity of ATM and DNA-PK. The extent of inhibition on the kinase activity also correlated with the degree of attenuation on the DNA damage responses. (7) Unexpectedly, sodium salicylate showed a p53-independent protection effect on topoisomerase-mediated cell killing. Together, our study provides evidence that NSAIDs exhibit a novel COX-independent modulating activity of NSAIDs on the DNA damage responses and it is through inhibition of phosphoinositide 3-kinase-like kinases.
- Published
- 2010
46. Discovery of a Novel Series of Quinolone and Naphthyridine Derivatives as Potential Topoisomerase I Inhibitors by Scaffold Modification
- Author
-
Xungui He, Ji-Wang Chern, Qidong You, Qinglong Guo, Xiaojian Wang, Chiung-Hua Huang, Zhiyu Li, Qian Yang, Tsai-Kun Li, and Xiao-Guang Chen
- Subjects
Models, Molecular ,Programmed cell death ,Transplantation, Heterologous ,Molecular Conformation ,Mice, Nude ,Antineoplastic Agents ,Quinolones ,Topoisomerase-I Inhibitor ,Mice ,In vivo ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,medicine ,Animals ,Humans ,Enzyme Inhibitors ,Naphthyridines ,Cytotoxicity ,Cell Proliferation ,biology ,Chemistry ,Topoisomerase ,DNA ,Cell biology ,Biochemistry ,Cell culture ,Apoptosis ,biology.protein ,Molecular Medicine ,Female ,Topoisomerase I Inhibitors ,Camptothecin ,medicine.drug - Abstract
A novel series of topoisomerase I (Top I) inhibitors were designed on the basis of camptothecin using scaffold modification strategy. Thirty-one new compounds were synthesized and evaluated for anticell proliferation activity. The most potent compound 26 presented a significant inhibitory effect on Top I, leading to Top I-mediated cleavage and influences on Top I expression at the cellular level. Moreover, 26 was proved to induce cell death via apoptosis and accelerated DNA strand breaks without significant alteration in cell cycle populations. All of the experimental results herein indicated that 26 could interact with DNA-Top I complex and induce cancer cell apoptosis to produce antitumor effects. The in vivo evaluation of 26 on the growth of HT-29 tumor xenografts in nude mice suggested its therapeutic potential for further development.
- Published
- 2009
47. Synthesis, DNA binding, and cytotoxicity of 1,4-bis(2-amino-ethylamino)anthraquinone–amino acid conjugates
- Author
-
Chieh-Hua Lee, On Lee, Ming-Show Yang, Pi-Hung Kao, Ling-Wei Hsin, Wan-Ru Chen, Hui-Po Wang, Tsai-Kun Li, Ya-Ling Chan, Hung-Wei Chen, Jih-Hwa Guh, and Chin-Ping His
- Subjects
Stereochemistry ,Clinical Biochemistry ,Pharmaceutical Science ,Anthraquinones ,Antineoplastic Agents ,Ametantrone ,Biochemistry ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,Combinatorial Chemistry Techniques ,Humans ,Cytotoxic T cell ,Structure–activity relationship ,Amino Acid Sequence ,Amino Acids ,Cytotoxicity ,Molecular Biology ,Peptide sequence ,chemistry.chemical_classification ,Molecular Structure ,Organic Chemistry ,Stereoisomerism ,DNA ,In vitro ,Amino acid ,chemistry ,Molecular Medicine ,Drug Screening Assays, Antitumor - Abstract
Two series of 1,4-bis(2-amino-ethylamino)anthraquinone-amino acid conjugates (BACs), ametantrone (AT)-amino acid conjugates (AACs) and mitoxantrone (MX)-amino acid conjugates (MACs), were designed and synthesized. The DNA binding of BACs was evaluated by DNA thermal denaturation experiment. In the series, the methionine-substituted BACs had the weakest DNA binding, while the lysine-substituted BACs had the highest T(m) values. The abilities of BACs to inhibit the growth of MCF-7, NCI-H460, SF-268, and PC-3 cell lines were determined. l-Met-MAC 16 and l-Lys-MAC 20 were the most potent growth inhibitors. MAC 16 was more cytotoxic than MX, whereas the T(m) of MAC 16 was much lower than that of MX. In contrast to MAC 16, l-Lys-MAC 20 demonstrated higher T(m) than MX. These data suggested that Met-BACs possessed a different pharmacological profile, in which the ability to stabilize DNA is not parallel to the ability to kill cancer cells, from that of AT and MX. The primary mechanism of cytotoxicity for MAC 16 was most likely through TOP2 poisoning. Therefore, MAC 16 may provide a lead for the development of novel generations of anthraquinone-type antitumor agents.
- Published
- 2008
48. Genistein induces apoptosis in human hepatocellular carcinomas via interaction of endoplasmic reticulum stress and mitochondrial insult
- Author
-
Tsai-Kun Li, Jui-Ling Hsu, Jih-Hwa Guh, Shih-Wei Wang, Chieh Yu Peng, Ting Chun Yeh, Po Cheng Chiang, and Chun-Jung Lin
- Subjects
medicine.medical_specialty ,Programmed cell death ,Carcinoma, Hepatocellular ,Genistein ,Antineoplastic Agents ,Apoptosis ,Mitochondrion ,Biology ,Endoplasmic Reticulum ,Biochemistry ,chemistry.chemical_compound ,Cell Line, Tumor ,Cyclosporin a ,Internal medicine ,medicine ,Humans ,Endoplasmic Reticulum Chaperone BiP ,Heat-Shock Proteins ,Pharmacology ,Endoplasmic reticulum ,Liver Neoplasms ,Mitochondria ,Apoptotic Protease-Activating Factor 1 ,Endocrinology ,Mitochondrial permeability transition pore ,chemistry ,Caspases ,Unfolded protein response ,Cancer research ,Reactive Oxygen Species ,Transcription Factor CHOP ,Molecular Chaperones - Abstract
Hepatocellular carcinoma is a very common malignancy and is chemoresistant to currently available chemotherapeutic agents. Endoplasmic reticulum (ER) stress-induced apoptotic pathway is suggested to be less affected by the resistance mechanisms, becoming a potential target of chemotherapeutic strategy. The anticancer effects and expression of GADD153, a transcription factor induced by ER stress, were examined in hepatocellular carcinoma Hep3B cells. The correlation between these two parameters was constructed under flavonoid stimulation with a correlation coefficient (r) of 0.8. The data also showed that genistein (isoflavone) was the most effective one. Genistein induced the activation of several ER stress-relevant regulators, including m-calpain, GADD153, GRP78 and caspase-12. Furthermore, genistein-induced effect was inhibited in cells transfected with antisense GADD153 cDNA, indicating a functional role of GADD153. Notably, genistein induced the activation of caspase-2, whereas did not cause the DNA damage. It also triggered the production of ROS. The antioxidant trolox significantly reduced ROS accumulation, but did not modify genistein-induced apoptotic cell death. The long-term exposure (48 h) of cells to genistein caused Mcl-1 down-regulation and Bad cleavage; furthermore, cyclosporin A (an inhibitor of mitochondrial permeability transition pore) almost completely abolished genistein-induced loss of mitochondrial membrane potential, and induced a 30% reverse of apoptosis caused by long-term treatment (48 h) of genistein, suggesting the involvement of mitochondrial stress in the late phase of genistein-induced effect. Taken together, it is suggested that genistein induces the anticancer effect through a mechanism initiated by ER stress and facilitated by mitochondrial insult in Hep3B cells.
- Published
- 2007
49. Characterization of the Uracil-DNA Glycosylase Activity of Epstein-Barr Virus BKRF3 and Its Role in Lytic Viral DNA Replication
- Author
-
Chih-Chung Lu, Geir Slupphaug, Ho-Ting Huang, Chung-Pei Lee, Meng-Chuan Wu, Yi Chun Chen, Mei-Ru Chen, Tsai-Kun Li, and Jiin-Tarng Wang
- Subjects
DNA Replication ,Herpesvirus 4, Human ,biology ,DNA polymerase ,DNA polymerase II ,Immunology ,DNA replication ,Eukaryotic DNA replication ,Virus Replication ,Microbiology ,DNA polymerase delta ,Molecular biology ,Genome Replication and Regulation of Viral Gene Expression ,Replication factor C ,Control of chromosome duplication ,Virology ,Insect Science ,DNA, Viral ,biology.protein ,Humans ,Uracil-DNA Glycosidase ,Replication protein A ,Cells, Cultured ,HeLa Cells - Abstract
Uracil-DNA glycosylases (UDGs) of the uracil- N -glycosylase (UNG) family are the primary DNA repair enzymes responsible for removal of inappropriate uracil from DNA. Recent studies further suggest that the nuclear human UNG2 and the UDGs of large DNA viruses may coordinate with their DNA polymerase accessory factors to enhance DNA replication. Based on its amino acid sequence, the putative UDG of Epstein-Barr virus (EBV), BKRF3, belongs to the UNG family of proteins, and it was demonstrated previously to enhance oriLyt-dependent DNA replication in a cotransfection replication assay. However, the expression and enzyme activity of EBV BKRF3 have not yet been characterized. In this study, His-BKRF3 was expressed in bacteria and purified for biochemical analysis. Similar to the case for the Escherichia coli and human UNG enzymes, His-BKRF3 excised uracil from single-stranded DNA more efficiently than from double-stranded DNA and was inhibited by the purified bacteriophage PBS1 inhibitor Ugi. In addition, BKRF3 was able to complement an E. coli ung mutant in rifampin and nalidixic acid resistance mutator assays. The expression kinetics and subcellular localization of BKRF3 products were detected in EBV-positive lymphoid and epithelial cells by using BKRF3-specific mouse antibodies. Expression of BKRF3 is regulated mainly by the immediate-early transcription activator Rta. The efficiency of EBV lytic DNA replication was slightly affected by BKRF3 small interfering RNA (siRNA), whereas cellular UNG2 siRNA or inhibition of cellular and viral UNG activities by expressing Ugi repressed EBV lytic DNA replication. Taking these results together, we demonstrate the UNG activity of BKRF3 in vitro and in vivo and suggest that UNGs may participate in DNA replication or repair and thereby promote efficient production of viral DNA.
- Published
- 2007
50. Rhapontigenin inhibits TGF-β-mediated epithelial‑mesenchymal transition via the PI3K/AKT/mTOR pathway and is not associated with HIF-1α degradation
- Author
-
Tsai-Kun Li, Shih-Wei Wang, Yen-Cheng Yeh, Ho-Fu Hsiao, and Yen-Hsiu Yeh
- Subjects
0301 basic medicine ,Cancer Research ,Proteasome Endopeptidase Complex ,Epithelial-Mesenchymal Transition ,Antineoplastic Agents ,Biology ,Bioinformatics ,Transforming Growth Factor beta1 ,03 medical and health sciences ,Phosphatidylinositol 3-Kinases ,Stilbenes ,Humans ,Epithelial–mesenchymal transition ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Oncogene ,TOR Serine-Threonine Kinases ,Ubiquitination ,General Medicine ,Cell cycle ,Hypoxia-Inducible Factor 1, alpha Subunit ,030104 developmental biology ,Oncology ,Cancer cell ,Proteolysis ,Cancer research ,Snail Family Transcription Factors ,Signal transduction ,Drug Screening Assays, Antitumor ,Proto-Oncogene Proteins c-akt ,Transforming growth factor ,HeLa Cells ,Signal Transduction - Abstract
The epithelial-mesenchymal transition (EMT) is a pivotal event in cancer cell invasion and metastasis. Emerging evidence suggests that rhapontigenin (Rha) may impede the progression of cancer by disrupting angiogenesis and the EMT. However, the underlying mechanism of Rha has not yet been clarified. In this study, we used transforming growth factor β (TGF-β) to trigger EMT in diverse types of cancer cells and revealed that Rha inhibited TGF-β-induced EMT and derived‑cell invasiveness. The effects of TGF-β were blocked by Rha via interference with the PI3K/AKT/mTOR/GSK3β/β‑catenin signaling pathway. Furthermore, Rha also inhibited TGF-β‑induced expression of transcription regulators Snail and hypoxia-inducible factor 1α (HIF-1α) by causing their degradation by the 26S proteasome. Surprisingly, although HIF-1α was degraded with Snail as a result of Rha exposure, HIF-1α was not a key factor involved in TGF-β-mediated EMT induced by Rha. Knocking-down Snail expression, but not HIF-1α expression, by RNA interference dramatically reversed TGF-β-mediated EMT. Moreover, Rha abolished TGF-β-triggered cell invasiveness. Our results demonstrate that Rha inhibits TGF-β-induced EMT in cancer cells by suppressing the activity of the PI3K/AKT/mTOR pathway. Therefore, Rha may represent a new route for therapeutic intervention in cancer patients and merits future studies to assess its potential.
- Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.