Network


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

Hotspot


Dive into the research topics where Yu-g Lin is active.

Publication


Featured researches published by Yu-g Lin.


Nucleic Acids Research | 2014

miRTarBase update 2014: an information resource for experimentally validated miRNA-target interactions

Sheng-Da Hsu; Yu-Ting Tseng; Sirjana Shrestha; Yu-Ling Lin; Anas Khaleel; Chih-Hung Chou; Chao-Fang Chu; Hsi-Yuan Huang; Ching-Min Lin; Shu-Yi Ho; Ting-Yan Jian; Feng-Mao Lin; Tzu-Hao Chang; Shun-Long Weng; Kuang-Wen Liao; I-En Liao; Chun-Chi Liu; Hsien-Da Huang

MicroRNAs (miRNAs) are small non-coding RNA molecules capable of negatively regulating gene expression to control many cellular mechanisms. The miRTarBase database (http://mirtarbase.mbc.nctu.edu.tw/) provides the most current and comprehensive information of experimentally validated miRNA-target interactions. The database was launched in 2010 with data sources for >100 published studies in the identification of miRNA targets, molecular networks of miRNA targets and systems biology, and the current release (2013, version 4) includes significant expansions and enhancements over the initial release (2010, version 1). This article reports the current status of and recent improvements to the database, including (i) a 14-fold increase to miRNA-target interaction entries, (ii) a miRNA-target network, (iii) expression profile of miRNA and its target gene, (iv) miRNA target-associated diseases and (v) additional utilities including an upgrade reminder and an error reporting/user feedback system.


Nucleic Acids Research | 2016

miRTarBase 2016: updates to the experimentally validated miRNA-target interactions database

Chih-Hung Chou; Nai-Wen Chang; Sirjana Shrestha; Sheng-Da Hsu; Yu-Ling Lin; Wei-Hsiang Lee; Chi-Dung Yang; Hsiao-Chin Hong; Ting-Yen Wei; Siang-Jyun Tu; Tzi-Ren Tsai; Shu-Yi Ho; Ting-Yan Jian; Hsin-Yi Wu; Pin-Rong Chen; Nai-Chieh Lin; Hsin-Tzu Huang; Tzu-Ling Yang; Chung-Yuan Pai; Chun-San Tai; Wen-Liang Chen; Chia-Yen Huang; Chun-Chi Liu; Shun-Long Weng; Kuang-Wen Liao; Wen-Lian Hsu; Hsien-Da Huang

MicroRNAs (miRNAs) are small non-coding RNAs of approximately 22 nucleotides, which negatively regulate the gene expression at the post-transcriptional level. This study describes an update of the miRTarBase (http://miRTarBase.mbc.nctu.edu.tw/) that provides information about experimentally validated miRNA-target interactions (MTIs). The latest update of the miRTarBase expanded it to identify systematically Argonaute-miRNA-RNA interactions from 138 crosslinking and immunoprecipitation sequencing (CLIP-seq) data sets that were generated by 21 independent studies. The database contains 4966 articles, 7439 strongly validated MTIs (using reporter assays or western blots) and 348 007 MTIs from CLIP-seq. The number of MTIs in the miRTarBase has increased around 7-fold since the 2014 miRTarBase update. The miRNA and gene expression profiles from The Cancer Genome Atlas (TCGA) are integrated to provide an effective overview of this exponential growth in the miRNA experimental data. These improvements make the miRTarBase one of the more comprehensively annotated, experimentally validated miRNA-target interactions databases and motivate additional miRNA research efforts.


Nanomedicine: Nanotechnology, Biology and Medicine | 2012

A Lipo-PEG-PEI complex for encapsulating curcumin that enhances its antitumor effects on curcumin-sensitive and curcumin-resistance cells

Yu-Ling Lin; Yen-Ku Liu; Nu-Man Tsai; Jui-Hung Hsieh; Chia-Hung Chen; Ching-Min Lin; Kuang-Wen Liao

A cationic liposome-PEG-PEI complex (LPPC) was used as a carrier for the encapsulation of hydrophobic curcumin to give curcumin/LPPC. Curcumin/LPPC had an average size less than 270 nm and a zeta potential of approximately 40 mV. The LPPC encapsulation efficiency for curcumin was about 45%. The authors found it surprising that the cytotoxic activity of the curcumin/LPPC was fivefold higher than curcumin when tested on curcumin-sensitive cells and 20-fold more active against curcumin-resistant cells. Curcumin/LPPC treatment caused a cell cycle arrest at G2/M phase, which rapidly resulted in apoptosis. The increased cytotoxic activity of curcumin/LPPC is likely attributable to its rapid accumulation in the cell. In vivo, administration of curcumin/LPPC inhibited about 60 - 90% of tumor growth in mice bearing CT-26 or B16F10 cells. These results demonstrate LPPC encapsulation technology is able to enhance the effects of antitumor drugs. Use of this technology may provide a new tool for cancer therapy, especially for drug-resistant cancer. From the Clinical Editor: This team of investigators used a cationic liposome-PEG-PEI complex (LPPC) to encapsulate curcumin. The different delivery method resulted in the five-fold increase of cytotoxic activity against curcumin-sensitive cells and twenty-fold against curcumin-resistant cells.


Nucleic Acids Research | 2018

miRTarBase update 2018: a resource for experimentally validated microRNA-target interactions

Chih-Hung Chou; Sirjana Shrestha; Chi-Dung Yang; Nai-Wen Chang; Yu-Ling Lin; Kuang-Wen Liao; Wei-Chih Huang; Ting-Hsuan Sun; Siang-Jyun Tu; Wei-Hsiang Lee; Men-Yee Chiew; Chun-San Tai; Ting-Yen Wei; Tzi-Ren Tsai; Hsin-Tzu Huang; Chung-Yu Wang; Hsin-Yi Wu; Shu-Yi Ho; Pin-Rong Chen; Cheng-Hsun Chuang; Pei-Jung Hsieh; Yi-Shin Wu; Wen-Liang Chen; Meng-Ju Li; Yu-chun Wu; Xin-Yi Huang; Fung Ling Ng; Waradee Buddhakosai; Pei-Chun Huang; Kuan-Chun Lan

Abstract MicroRNAs (miRNAs) are small non-coding RNAs of ∼ 22 nucleotides that are involved in negative regulation of mRNA at the post-transcriptional level. Previously, we developed miRTarBase which provides information about experimentally validated miRNA-target interactions (MTIs). Here, we describe an updated database containing 422 517 curated MTIs from 4076 miRNAs and 23 054 target genes collected from over 8500 articles. The number of MTIs curated by strong evidence has increased ∼1.4-fold since the last update in 2016. In this updated version, target sites validated by reporter assay that are available in the literature can be downloaded. The target site sequence can extract new features for analysis via a machine learning approach which can help to evaluate the performance of miRNA-target prediction tools. Furthermore, different ways of browsing enhance user browsing specific MTIs. With these improvements, miRTarBase serves as more comprehensively annotated, experimentally validated miRNA-target interactions databases in the field of miRNA related research. miRTarBase is available at http://miRTarBase.mbc.nctu.edu.tw/.


International Journal of Nanomedicine | 2012

Liposome-based polymer complex as a novel adjuvant: enhancement of specific antibody production and isotype switch.

Chia-Hung Chen; Yu-Ling Lin; Yen-Ku Liu; Pei-Juin He; Ching-Min Lin; Chang-Jer Wu; Tian-Lu Cheng; Shih-Jen Liu; Kuang-Wen Liao

The aim of vaccination is to induce appropriate immunity against pathogens. Antibody-mediated immunity is critical for protection against many virus diseases, although it is becoming more evident that coordinated, multifunctional immune responses lead to the most effective defense. Specific antibody (Ab) isotypes are more efficient at protecting against pathogen invasion in different locations in the body. For example, compared to other Ab isotypes, immunoglobulin (Ig) A provides more protection at mucosal areas. In this study, we developed a cationic lipopolymer (liposome-polyethylene glycol-polyethyleneimine complex [LPPC]) adjuvant that strongly adsorbs antigens or immunomodulators onto its surface to enhance or switch immune responses. The results demonstrate that LPPC enhances uptake ability, surface marker expression, proinflammatory cytokine release, and antigen presentation in mouse phagocytes. In contrast to Freund’s adjuvant, LPPC preferentially activates Th1- immunity against antigens in vivo. With lipopolysaccharides or CpG oligodeoxynucleotides, LPPC dramatically enhances the IgA or IgG2A proportion of total Ig, even in hosts that have developed Th2 immunities and high IgG1 serum titers. Taken together, the results demonstrate that the LPPC adjuvant not only increases the immunogenicity of antigens but also modulates host immunity to produce an appropriate Ab isotype by combining with immunomodulators.


Biotechnology and Bioengineering | 2011

A unique and potent protein binding nature of liposome containing polyethylenimine and polyethylene Glycol: A nondisplaceable property

Yen-Ku Liu; Yu-Ling Lin; Chia-Hung Chen; Ching-Min Lin; Kuan-Lun Ma; Fu‐Hsuan Chou; Jin-Sheng Tsai; Hsin-Yu Lin; Fu-Rong Chen; Tian-Lu Cheng; Chia-Ching Chang; Kuang-Wen Liao

Most of the currently available targeting vectors are produced via the linkage of targeting molecules. However, the coupling process is complicated, and the covalent linkage may attenuate the activity of certain targeting molecules. In this study, we have developed a cationic liposome complexed with polyethylenimine and polyethylene glycol polymers (LPPC) that can capture various proteins without covalent conjugation. Characterizations of prepared LPPC revealed that the maximal‐binding capacity was about 170 µg of bovine serum albumin to 40 µg of sphere‐shaped LPPC (180 nm). The proteins were essentially located at or near the surface when analyzed by atomic force or transmission electron microscopy. We demonstrate that polyethylenimine was an essential component to bind the proteins. Upon the saturation of captured proteins, a given protein could not be displaced by other additional proteins and still retained its biological activity. Using a variety of functional proteins, we show some typical examples of the utility of incorporated beta‐glucuronidase and antibodies onto the LPPC. The beta‐glucuronidase can be used for the study of antigen–antibody interactions, whereas in studies with the antibody complex, we used anti‐CD3 as an agonist to stimulate the proliferation of peripheral blood mononuclear cells via a receptor‐mediated mechanism and anti‐VEGFR for cell staining. In conclusion, the prepared LPPC can provide a platform to capture biologically and biochemically functional proteins on its surface for various applications, such as cell signaling, cell profiling, noncovalent enzyme‐linked immunoassays, and others not mentioned. Biotechnol. Bioeng. 2011; 108:1318–1327.


Journal of Agricultural and Food Chemistry | 2012

Inhibitory Effects of Chloroform Extracts Derived from Corbicula fluminea on the Release of Pro-inflammatory Cytokines

Ching-Min Lin; Yu-Ling Lin; Nu-Man Tsai; Hsin-Yi Wu; Shu-Yi Ho; Chia-Hung Chen; Yen-Ku Liu; Li-Ping Ho; Ru-Ping Lee; Kuang-Wen Liao

Corbicula fluminea, the primary freshwater bivalve cultivated in Taiwan, was formerly used as a remedy for hepatitis. Recent reports indicate that C. fluminea has many bioactivities, but it remains unknown whether C. fluminea affects inflammation. This study explored the anti-inflammatory activity of C. fluminea. C. fluminea was first treated with chloroform to obtain clam chloroform extracts (CCEs). On the basis of the assay for the release of pro-inflammatory cytokines in vitro and in vivo, the results show that the CCEs significantly lowered the release of lipopolysaccharide (LPS)-induced pro-inflammatory cytokines. Additionally, the CCEs reduced LPS-induced organ damage. Real-time polymerase chain reaction analysis suggested that CCEs inhibit the LPS-induced mRNA expression of interleukin-1β and tumor necrosis factor-α. Western blot analysis indicated that the CCEs increased expression of IκB and attenuated the phosphorylation of IκB. Gas chromatography-mass spectrometry suggests that phytosterols and fatty acids are responsible for the anti-inflammatory properties of CCEs. Taken together, CCEs have the potential to be developed as an anti-inflammatory functional food.


Phytotherapy Research | 2012

7,7′′-Dimethoxyagastisflavone-induced Apoptotic or Autophagic Cell Death in Different Cancer Cells

Chia-Hsiang Hwang; Yu-Ling Lin; Yen-Ku Liu; Chia-Hung Chen; Hsin-Yi Wu; Cheng-Chang Chang; Chao-Yuan Chang; Yu-Kuo Chang; Kuang-Wen Liao; Yiu-Kay Lai

7,7′′‐Dimethoxyagastisflavone (DMGF), a biflavonoid isolated from the needles of Taxus × media cv. Hicksii, was evaluated for its antiproliferative and antineoplastic effects in three human cancer cell lines. Interestingly, DMGF caused cell death via different pathways in different cancer cells. DMGF induced apoptosis, activated caspase‐3 activity and changed the mitochondrial membrane potential in HT‐29 human colon cancer cells. However, the apoptotic pathway is not the major pathway involved in DMGF‐induced cell death in A549 human lung cancer cells and HepG2 human hepatoma cells. Treatment with 3‐MA, an inhibitor of autophagy, significantly decreased DMGF‐induced cell death in HepG2 and A549 cells, but did not affect DMGF‐induced cell death in HT‐29 cells. Following DMGF treatment, the HepG2 cells increased expression of LC3B‐II, a marker used to monitor autophagy in cells. Thus, DMGF induced apoptotic cell death in HT‐29 cells, triggered both apoptotic and autophagic death in A549 cells and induced autophagic cell death in HepG2 cells. Copyright


Cytokine | 2010

The treatment of propofol induced the TGF-β1 expression in human endothelial cells to suppress endocytosis activities of monocytes.

Chi-Han Li; Ru-Ping Lee; Yu-Ling Lin; Chen-Si Lin; Bang-Gee Hsu; Feng-Jen Tseng; Yu-Cheng Chen; Kuang-Wen Liao; Fwu-Lin Yang

Propofol anesthesia and sedation are known to downregulate the functions of many hematopoietic cells, such as macrophages and neutrophils, in vivo. However, the effects of propofol on secretion of the regulatory cytokine transforming growth factor β1 (TGF-β1) in vivo are unknown. In this study, the effects of propofol on TGF-β1 expression in human peripheral blood mononuclear cells, umbilical vein endothelial cells (HUVECs), lymphocytes (Jurkat) and monocytes (THP-1) were tested. Moreover, these sera were also tested for regulatory activity on monocyte endocytosis with or without treatment with the TGF-β1 pathway inhibitor SB431542. Propofol raised levels of both total and activated TGF-β1 in propofol-treated patient sera after surgical operations. Furthermore, propofol induced secretion of latent TGF-β1 in HUVEC cells and enhanced TGF-β1 activation in THP-1 and Jurkat cells in vitro. Additionally, sera from propofol-treated patients suppressed monocyte endocytosis ex vivo, an effect that was abrogated by the TGF-β1 pathway inhibitor SB431542.


International Journal of Nanomedicine | 2015

Liposomal n -butylidenephthalide protects the drug from oxidation and enhances its antitumor effects in glioblastoma multiforme

Yu-Ling Lin; Kai-Fu Chang; Xiao-Fan Huang; Che-Lun Hung; Shyh-Chang Chen; Wan-Ru Chao; Kuang-Wen Liao; Nu-Man Tsai

Background The natural compound n-butylidenephthalide (BP) can pass through the blood–brain barrier to inhibit the growth of glioblastoma multiforme tumors. However, BP has an unstable structure that reduces its antitumor activity and half-life in vivo. Objective The aim of this study is to design a drug delivery system to encapsulate BP to enhance its efficacy by improving its protection and delivery. Methods To protect its structural stability against protein-rich and peroxide solutions, BP was encapsulated into a lipo-PEG-PEI complex (LPPC). Then, the cytotoxicity of BP/LPPC following preincubation in protein-rich, acid/alkaline, and peroxide solutions was analyzed by MTT. Cell uptake of BP/LPPC was also measured by confocal microscopy. The therapeutic effects of BP/LPPC were analyzed in xenograft mice following intratumoral and intravenous injections. Results When BP was encapsulated in LPPC, its cytotoxicity was maintained following preincubation in protein-rich, acid/alkaline, and peroxide solutions. The cytotoxic activity of encapsulated BP was higher than that of free BP (~4.5- to 8.5-fold). This increased cytotoxic activity of BP/LPPC is attributable to its rapid transport across the cell membrane. In an animal study, a subcutaneously xenografted glioblastoma multiforme mouse that was treated with BP by intratumoral and intravenous administration showed inhibited tumor growth. The same dose of BP/LPPC was significantly more effective in terms of tumor inhibition. Conclusion LPPC encapsulation technology is able to protect BP’s structural stability and enhance its antitumor effects, thus providing a better tool for use in cancer therapy.

Collaboration


Dive into the Yu-g Lin's collaboration.

Top Co-Authors

Avatar

Kuang-Wen Liao

National Chiao Tung University

View shared research outputs
Top Co-Authors

Avatar

Yen-Ku Liu

National Chiao Tung University

View shared research outputs
Top Co-Authors

Avatar

Chia-Hung Chen

National Chiao Tung University

View shared research outputs
Top Co-Authors

Avatar

Nu-Man Tsai

Chung Shan Medical University

View shared research outputs
Top Co-Authors

Avatar

Shu-Yi Ho

National Chiao Tung University

View shared research outputs
Top Co-Authors

Avatar

Ching-Min Lin

National Chiao Tung University

View shared research outputs
Top Co-Authors

Avatar

Hsin-Yi Wu

National Chiao Tung University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kuang Wen Liao

National Chiao Tung University

View shared research outputs
Top Co-Authors

Avatar

Pin-Rong Chen

National Chiao Tung University

View shared research outputs
Researchain Logo
Decentralizing Knowledge