Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Vaccines (Basel) ; 10(2)2022 Feb 17.
Article En | MEDLINE | ID: mdl-35214770

BACKGROUND: The ChAdOx1 nCoV-19 vaccine has been widely administered against SARS-CoV-2 infection; however, data regarding its immunogenicity, reactogenicity, and potential differences in responses among Asian populations remain scarce. METHODS: 270 participants without prior COVID-19 were enrolled to receive ChAdOx1 nCoV-19 vaccination with a prime-boost interval of 8-9 weeks. Their specific SARS-CoV-2 antibodies, neutralizing antibody titers (NT50), platelet counts, and D-dimer levels were analyzed before and after vaccination. RESULTS: The seroconversion rates of anti-RBD and anti-spike IgG at day 28 after a boost vaccination (BD28) were 100% and 95.19%, respectively. Anti-RBD and anti-spike IgG levels were highly correlated (r = 0.7891), which were 172.9 ± 170.4 and 179.3 ± 76.88 BAU/mL at BD28, respectively. The geometric mean concentrations (GMCs) of NT50 for all participants increased to 132.9 IU/mL (95% CI 120.0-147.1) at BD28 and were highly correlated with anti-RBD and anti-spike IgG levels (r = 0.8248 and 0.7474, respectively). Body weight index was statistically significantly associated with anti-RBD IgG levels (p = 0.035), while female recipients had higher anti-spike IgG levels (p = 0.038). The GMCs of NT50 declined with age (p = 0.0163) and were significantly different across age groups (159.7 IU/mL for 20-29 years, 99.4 IU/mL for ≥50 years, p = 0.0026). Injection-site pain, fever, and fatigue were the major reactogenicity, which were more pronounced after prime vaccination and in younger participants (<50 years). Platelet counts decreased and D-dimer levels increased after vaccination but were not clinically relevant. No serious adverse events or deaths were observed. CONCLUSION: The vaccine is well-tolerated and elicited robust humoral immunity against SARS-CoV-2 after standard prime-boost vaccination in Taiwanese recipients.

2.
Sci Rep ; 10(1): 12526, 2020 Jul 22.
Article En | MEDLINE | ID: mdl-32694763

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

3.
Sci Rep ; 9(1): 4606, 2019 03 14.
Article En | MEDLINE | ID: mdl-30872677

The therapeutic effects of simvastatin for renal cell carcinoma (RCC) are controversial. In this study, the effects of simvastatin on the carcinogenic properties of 3-methylcholanthrene (3MC; an aryl-hydrocarbon receptor [AhR] agonist) in human renal epithelial cells (hRECs) were investigated. We exposed in vitro and in vivo models to 3MC to induce RCC onset. 3MC upregulated the epithelial-mesenchymal transition (EMT) and tumor biomarkers; the models exhibited the reciprocal expression of histone deacetylase 1 (HDAC1) and RhoA, namely increased HDAC1 and decreased RhoA expression, through hypoxia-inducible-factor (HIF)- and AhR-dependent mechanisms. In addition to inducing EMT biomarkers, 3MC decreased von Hippel-Lindau protein levels (a risk factor for RCC) and increased CD44 expression in hRECs, which were reversed by digoxin (a HIF inhibitor) and HDAC inhibitors (suberoylanilide hydroxamic acid and trichostatin A [TSA]). Simvastatin abolished the detrimental effects of 3MC by reducing HDAC1 expression, with resulting RhoA upregulation, and reactivating RhoA in vitro and in vivo. Notably, the protective effects of simvastatin were negated by an HDAC activator (ITSA) through TSA suppression. The crucial role of RhoA in RCC carcinogenesis was verified by the overexpression of constitutively active RhoA. Collectively, these results demonstrate that simvastatin restores RhoA function through HDAC1 inhibition; therefore, simvastatin might serve as adjunct therapy for RCC induced by 3MC.


Epithelial Cells/drug effects , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Kidney/drug effects , Methylcholanthrene/adverse effects , Simvastatin/pharmacology , rhoA GTP-Binding Protein/metabolism , Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Cells, Cultured , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition/drug effects , Humans , Hyaluronan Receptors/metabolism , Hydroxamic Acids/pharmacology , Kidney/metabolism , Receptors, Aryl Hydrocarbon/metabolism
4.
Phytomedicine ; 55: 255-263, 2019 Mar 01.
Article En | MEDLINE | ID: mdl-30668437

BACKGROUND: We previously showed that 3-O-ß-D-glucopyranosyl-(3R)-hydroxybutanolide (kinsenoside), a major compound of Anoectochilus formosanus, increased lipolysis through an AMP-activated protein kinase (AMPK)-dependent pathway. PURPOSE: To extend our previous finding, we investigated the in vivo and in vitro effects of kinsenoside on lipolysis and the involvement of cyclic AMP (cAMP)-dependent protein kinase A (PKA) and AMPK in kinsenoside-mediated lipolysis. STUDY DESIGN/METHODS: Mice were fed a high-fat diet for six weeks to induce lipid deposition and then treated with 50 and 100  mg/kg kinsenoside for two weeks. The coordination of PKA and AMPK activation in lipolysis in C3H10T1/2 adipocytes was evaluated in vitro by using PKA and AMPK's corresponding inhibitors, oil-red O staining, a glycerol production assay, and Western blot analysis. RESULTS: Kinsenoside reduced body weight, fat pad mass, and hepatic lipid accumulation in obese mice, and concurrently increased the induction and activation of hormone-sensitive lipase (HSL), perilipin, adipose triglyceride lipase (ATGL), and carnitine palmitoyltransferase I (CPT1). Kinsenoside concentration-dependently increased PKA activation by increasing the phosphorylation of Ser/Thr-PKA substrates in vitro. These increases were accompanied by a reduction in fat accumulation. Using H89 and Rp-8-Br-cAMPs to inhibit PKA reduced the release of glycerol but did not alter the activation of peroxisome proliferator-activated receptor alpha or the expression of CPT1 or ATGL. By contrast, compound C, an AMPK inhibitor, inhibited CPT1 and ATGL expression in kinsenoside-treated C3H10T1/2 adipocytes. In addition, H89 caused the reactivation of AMPK downstream targets by increasing the levels of the active form of pAMPK-Thr172, suggesting that PKA negatively modulates AMPK activity. CONCLUSION: Kinsenoside increased HSL activation through PKA-mediated phosphorylation at Ser660/563 and concomitantly increased perilipin activation in lipolysis. These lipolytic effects of kinsenoside were validated using 6-Bnz-cAMPs, a PKA agonist. In this study, we demonstrated that in addition to AMPK, PKA also plays a crucial role in kinsenoside-mediated lipolysis.


4-Butyrolactone/analogs & derivatives , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Lipolysis/drug effects , Monosaccharides/metabolism , Plant Extracts/metabolism , Sterol Esterase/metabolism , 4-Butyrolactone/metabolism , Animals , Male , Mice , Orchidaceae/chemistry , Plant Extracts/chemistry
5.
J Cell Physiol ; 232(5): 1020-1029, 2017 05.
Article En | MEDLINE | ID: mdl-27442426

Endothelial nitric oxide synthase (eNOS) modulates vascular blood pressure and is predominantly expressed in endothelial cells and activated through the protein kinase B (Akt/PKB)-dependent pathway. We previously reported that 3-methylcholanthrene (3MC) activates the aryl hydrocarbon receptor (AhR) and reduces PI3K/Akt phosphorylation. This study investigated the mechanism underlying the downregulatory effects of 3-MC on nitric oxide (NO) production occurring through the AhR/RhoA/Akt-mediated mechanism. The mechanism underlying the effects of 3-MC on eNOS activity and blood pressure was examined in vitro and in vivo through genetic and pharmacological approaches. Results indicated that 3-MC modified heat shock protein 90 (HSP90), caveolin-1, dynein, and eNOS mRNA and protein expression through the AhR/RhoA-dependent mechanism in mouse cerebral vascular endothelial cells (MCVECs) and that 3-MC reduced eNOS phosphorylation through the AhR/RhoA-mediated inactivation of Akt1. The upregulation of dynein expression was associated with decreased eNOS dimer formation (eNOS dimer; an activated form of the enzyme). Coimmunoprecipitation assay results indicated that 3-MC significantly reduced the interaction between eNOS and its regulatory proteins, including Akt1 and HSP90, but increased the interaction between eNOS and caveolin-1. Immunofluorescence and Western blot analysis revealed that 3-MC reduced the amount of membrane-bound activated eNOS, and a modified Griess assay revealed that 3-MC concomitantly reduced NO production. However, simvastatin reduced 3-MC-mediated murine hypertension. Our study results indicate that AhR, RhoA, and eNOS have major roles in blood pressure regulation. Statin intervention might provide a potential therapeutic approach for reducing hypertension caused by 3-MC. J. Cell. Physiol. 232: 1020-1029, 2017. © 2016 Wiley Periodicals, Inc.


Hypertension/enzymology , Methylcholanthrene/pharmacology , Nitric Oxide Synthase Type III/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Cerebrum/blood supply , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Hypertension/pathology , Mice , Models, Biological , Nitric Oxide/metabolism , Nitroprusside/pharmacology , Nitroprusside/therapeutic use , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Simvastatin/pharmacology , Simvastatin/therapeutic use , Time Factors , rhoA GTP-Binding Protein/metabolism
6.
Arch Dermatol Res ; 306(9): 803-8, 2014 Nov.
Article En | MEDLINE | ID: mdl-25081927

Keloids is a fibroproliferative disease. The incidence of keloids among Asians has not been thoroughly studied. The objective of this study is to determine the incidence of keloids in Taiwan, which mainly consists of ethnic Chinese. Furthermore, we want to determine the comorbidity rate of other fibrosis-related diseases among keloid patients. This study was based on the National Health Insurance Research Database, which contains the data of 1 million randomly selected patients. Multivariate logistic regression analyses were employed to estimate the relative odds of keloids as a function of fibrosis-related diseases. The annual keloid incidence rate in Taiwan was 0.15 % for the general population. With a 1.33 ratio, women outnumbered men. Women with uterine leiomyoma have a 2.25-fold greater risk of keloids, compared with women without leiomyoma. We concluded that keloid incidence in Taiwan is approximately 0.15 %. Women with leiomyoma have a greater risk of keloids, this implicates that both diseases share a common etiopathological pathway.


Asian People , Keloid/ethnology , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , China/ethnology , Comorbidity , Databases, Factual , Female , Humans , Incidence , Infant , Infant, Newborn , Keloid/diagnosis , Leiomyoma/ethnology , Logistic Models , Male , Middle Aged , Multivariate Analysis , Odds Ratio , Risk Factors , Sex Factors , Taiwan/epidemiology , Time Factors , Uterine Neoplasms/ethnology , Young Adult
7.
Clin Biochem ; 46(12): 1058-1064, 2013 Aug.
Article En | MEDLINE | ID: mdl-23726813

OBJECTIVE: Toll-like receptors (TLRs) are important molecules for detecting both pathogen invasion and tissue damage. The expression of TLR4 is upregulated in ischemic stroke, at least in the subacute stage. However, the TLR downstream pathways in the context of stroke have not been well studied in previous research. The purpose of this study is to elucidate the TLR4 downstream pathways following ischemic stroke. DESIGN AND METHODS: In this study, 12 ischemic stroke patients and 12 controls were selected from among 89 ischemic stroke patients and 166 controls. The chosen subjects had the highest levels of TLR4 mRNA in the peripheral blood. The differences in the TLR downstream signaling pathways, which were studied by using an RT2 Profiler TM PCR array system (Qiagen), were analyzed. The differentially expressed genes were analyzed by using GeneSpring GX and visualized based on the TLR pathways in the Kyoto Encyclopedia of Genes and Genomes (KEGG). RESULTS: The genes upregulated in stroke patients were found to be involved in the MyD88-independent pathway and in UBE2V1-TRAF6 ubiquitin-mediated proteolysis. The genes were more expressed in extracellular space, receptor binding, and cytokine receptor binding by use of gene ontology (GO) terms than in control patients. CONCLUSIONS: We found that the MyD88-independent pathway and the ubiquitin-mediated proteolysis pathway, especially TRAF6, may be the most vital molecules among TLR downstream pathways in incidences of ischemic stroke.


Signal Transduction/genetics , Stroke/genetics , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Brain Ischemia/genetics , Brain Ischemia/pathology , Case-Control Studies , Gene Expression Profiling , Gene Expression Regulation , Humans , Proteolysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Stroke/pathology , Ubiquitin/metabolism
8.
Cancer Sci ; 103(9): 1640-50, 2012 Sep.
Article En | MEDLINE | ID: mdl-22726390

Increasing evidence suggests that PRMT5, a protein arginine methyltransferase, is involved in tumorigenesis. However, no systematic research has demonstrated the cell-transforming activity of PRMT5. We investigated the involvement of PRMT5 in tumor formation. First, we showed that PRMT5 was associated with many human cancers, through statistical analysis of microarray data in the NCBI GEO database. Overexpression of ectopic PRMT5 per se or its specific shRNA enhanced or reduced cell growth under conditions of normal or low concentrations of serum, low cell density, and poor cell attachment. A stable clone that expressed exogenous PRMT5 formed tumors in nude mice, which demonstrated that PRMT5 is a potential oncoprotein. PRMT5 accelerated cell cycle progression through G1 phase and modulated regulators of G1; for example, it upregulated cyclin-dependent kinase (CDK) 4, CDK6, and cyclins D1, D2 and E1, and inactivated retinoblastoma protein (Rb). Moreover, PRMT5 activated phosphoinositide 3-kinase (PI3K)/AKT and suppressed c-Jun N-terminal kinase (JNK)/c-Jun signaling cascades. However, only inhibition of PI3K activity, and not overexpression of JNK, blocked PRMT5-induced cell proliferation. Further analysis of PRMT5 expression in 64 samples of human lung cancer tissues by microarray and western blot analysis revealed a tight association of PRMT5 with lung cancer. Knockdown of PRMT5 retarded cell growth of lung cancer cell lines A549 and H1299. In conclusion, to the best of our knowledge, we have characterized the cell-transforming activity of PRMT5 and delineated its underlying mechanisms for the first time.


Cyclin-Dependent Kinases/metabolism , G1 Phase , Oncogene Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Nude , Oncogene Proteins/genetics , Protein-Arginine N-Methyltransferases/genetics
9.
BMC Cancer ; 11: 139, 2011 Apr 16.
Article En | MEDLINE | ID: mdl-21496277

BACKGROUND: A cross-talk between different receptor tyrosine kinases (RTKs) plays an important role in the pathogenesis of human cancers. METHODS: Both NIH-Met5 and T24-Met3 cell lines harboring an inducible human c-Met gene were established. C-Met-related RTKs were screened by RTK microarray analysis. The cross-talk of RTKs was demonstrated by Western blotting and confirmed by small interfering RNA (siRNA) silencing, followed by elucidation of the underlying mechanism. The impact of this cross-talk on biological function was demonstrated by Trans-well migration assay. Finally, the potential clinical importance was examined in a cohort of 65 cases of locally advanced and metastatic bladder cancer patients. RESULTS: A positive association of Axl or platelet-derived growth factor receptor-alpha (PDGFR-α) with c-Met expression was demonstrated at translational level, and confirmed by specific siRNA knock-down. The transactivation of c-Met on Axl or PDGFR-α in vitro was through a ras- and Src-independent activation of mitogen-activated protein kinase/extracellular signal-regulated kinase (MEK/ERK) pathway. In human bladder cancer, co-expression of these RTKs was associated with poor patient survival (p < 0.05), and overexpression of c-Met/Axl/PDGFR-α or c-Met alone showed the most significant correlation with poor survival (p < 0.01). CONCLUSIONS: In addition to c-Met, the cross-talk with Axl and/or PDGFR-α also contributes to the progression of human bladder cancer. Evaluation of Axl and PDGFR-α expression status may identify a subset of c-Met-positive bladder cancer patients who may require co-targeting therapy.


Oncogene Protein p21(ras)/metabolism , Oncogene Protein pp60(v-src)/metabolism , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Transcriptional Activation , Urinary Bladder Neoplasms/physiopathology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , NIH 3T3 Cells , Prognosis , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-met/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Signal Transduction/drug effects , Survival Analysis , Tetracycline/pharmacology , Transcriptional Activation/drug effects , Urinary Bladder Neoplasms/mortality , Axl Receptor Tyrosine Kinase
10.
Toxicol Appl Pharmacol ; 214(2): 126-35, 2006 Jul 15.
Article En | MEDLINE | ID: mdl-16545412

Benzo[a]pyrene (BaP) is one of many polycyclic aromatic hydrocarbons that have been identified as major risk factors for developing various cancers. We previously demonstrated that the liver cancer susceptibility gene glycine N-methyltransferase (GNMT) is capable of binding with BaP and protecting cells from BaP-7,8-diol 9,10-epoxide-DNA adduct formation. In this study, we used a cytotoxicity assay to demonstrate that the higher expression level of GNMT, the lower cytotoxicity occurred in the cells treated with BaP. In addition, a cDNA microarray containing 7,597 human genes was used to examine gene expression patterns in BaP-treated HepG2 (a liver cancer cell line that expresses very low levels of GNMT) and SCG2-1-1 (a stable HepG2 clone that expresses high levels of GNMT) cells. The results showed that among 6,018 readable HepG2 genes, 359 (6.0%) were up-regulated more than 1.5-fold and 768 (12.8%) were down-regulated. Overexpression of GNMT in SCG2-1-1 cells resulted in the down-regulation of genes related to the detoxification, kinase/phosphatase pathways, and oncogenes. Furthermore, real-time PCR was used to validate microarray data from 21 genes belonging to the detoxification pathway. Combining both microarray and real-time PCR data, the results showed that among 89 detoxification pathway genes analyzed, 22 (24.7%) were up-regulated and 6 (6.7%) were down-regulated in BaP-treated HepG2 cells, while in the BaP-treated SCG2-1-1 cells, 12 (13.5%) were up-regulated and 26 (29.2%) were down-regulated (P < 0.001). Therefore, GNMT sequesters BaP, diminishes BaP's effects to the liver detoxification pathway and prevents subsequent cytotoxicity.


Benzo(a)pyrene/pharmacology , Gene Expression Profiling , Glycine N-Methyltransferase/genetics , Inactivation, Metabolic/genetics , Liver/drug effects , Benzo(a)pyrene/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cluster Analysis , Down-Regulation/genetics , Glycine N-Methyltransferase/metabolism , Humans , Liver/metabolism , Liver/pathology , Liver X Receptors , Oligonucleotide Array Sequence Analysis/methods , Orphan Nuclear Receptors , Polymerase Chain Reaction/methods , Protein Binding , Receptors, Cytoplasmic and Nuclear/genetics , Up-Regulation/genetics
11.
Bioinformatics ; 20(17): 3273-6, 2004 Nov 22.
Article En | MEDLINE | ID: mdl-15217821

One possible path towards understanding the biological function of a target protein is through the discovery of how it interfaces within protein-protein interaction networks. The goal of this study was to create a virtual protein-protein interaction model using the concepts of orthologous conservation (or interologs) to elucidate the interacting networks of a particular target protein. POINT (the prediction of interactome database) is a functional database for the prediction of the human protein-protein interactome based on available orthologous interactome datasets. POINT integrates several publicly accessible databases, with emphasis placed on the extraction of a large quantity of mouse, fruit fly, worm and yeast protein-protein interactions datasets from the Database of Interacting Proteins (DIP), followed by conversion of them into a predicted human interactome. In addition, protein-protein interactions require both temporal synchronicity and precise spatial proximity. POINT therefore also incorporates correlated mRNA expression clusters obtained from cell cycle microarray databases and subcellular localization from Gene Ontology to further pinpoint the likelihood of biological relevance of each predicted interacting sets of protein partners.


Databases, Protein , Information Storage and Retrieval/methods , Protein Interaction Mapping/methods , Proteome/metabolism , Sequence Alignment/methods , Sequence Analysis, Protein/methods , User-Computer Interface , Animals , Database Management Systems , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Evolution, Molecular , Humans , Internet , Mice , Proteome/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Signal Transduction/physiology
12.
J Biol Chem ; 279(31): 32592-602, 2004 Jul 30.
Article En | MEDLINE | ID: mdl-15145941

F-box proteins, components of SCF ubiquitin-ligase complexes, are believed to be responsible for substrate recognition and recruitment in SCF-mediated proteolysis. F-box proteins that have been identified to function in the SCF complexes to date mostly have substrate-binding motifs, such as WD repeats or leucine-rich repeats in their C termini. However, many F-box proteins lack recognizable substrate-binding modules; whether they can function in the SCF complexes remains unclear. We show here that Fbx7, an F-box protein without WD repeats and leucine-rich repeats, is required for the proteasome-mediated proteolysis of the hepatoma up-regulated protein (HURP). Depletion of Fbx7 by small interfering RNA leads to depression of HURP ubiquitination and accumulation of HURP abundance. In the SCF(Fbx7) complex, Fbx7 recruits HURP through its C-terminal proline-rich region in a Cdk1-cyclin B-phosphorylation dependent manner. Mutation of the multiple Cdk1-cyclin B phosphorylation sites on HURP or the proline-rich region of Fbx7 abolishes the association between Fbx7 and HURP. Thus, Fbx7 is a functional adaptor of the SCF complex with a proline-rich region as the substrate-binding module. In addition to Fbx7, data base analyses reveal two putative mammalian proline-rich region-containing F-box proteins, KIAA1783 and RIKEN cDNA 2410015K21. Taken together, these findings further expound the diverse substrate-recognition abilities of the SCF complexes.


CDC2 Protein Kinase/metabolism , Carcinoma, Hepatocellular/metabolism , Cyclin B/metabolism , F-Box Proteins/physiology , Neoplasm Proteins/metabolism , Stem Cell Factor/metabolism , Amino Acid Motifs , Amino Acid Sequence , Binding Sites , Blotting, Western , Cell Cycle , Cell Line , Cycloheximide/pharmacology , DNA, Complementary/metabolism , Databases as Topic , Electrophoresis, Polyacrylamide Gel , Enzyme Inhibitors/pharmacology , F-Box Proteins/metabolism , Genetic Vectors , Humans , Microscopy, Fluorescence , Mitosis , Models, Biological , Models, Genetic , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Nocodazole/pharmacology , Phosphoproteins/chemistry , Phosphorylation , Precipitin Tests , Proline/chemistry , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , Protein Synthesis Inhibitors/pharmacology , RNA, Small Interfering/metabolism , Recombinant Proteins/chemistry , Recombination, Genetic , Sequence Homology, Amino Acid , Substrate Specificity , Temperature , Time Factors , Transfection , Ubiquitin/metabolism
13.
Mol Cell Proteomics ; 3(1): 93-104, 2004 Jan.
Article En | MEDLINE | ID: mdl-14602875

The increasing use of high-throughput and large-scale bioinformatics-based studies has generated a massive amount of data stored in a number of different databases. The major need now is to explore this disparate data to find biologically relevant interactions and pathways. Thus, in the post-genomic era, there is clearly a need for the development of algorithms that can accurately predict novel protein-protein interaction networks in silico. The evolutionarily conserved Aurora family kinases have been chosen as a model for the development of a method to identify novel biological networks by a comparison of human and various model organisms. Our search methodology was designed to predict and prioritize molecular targets for Aurora family kinases, so that only the most promising are subjected to empirical testing. Four potential Aurora substrates and/or interacting proteins, TACC3, survivin, Hec1, and hsNuf2, were identified and empirically validated. Together, these results justify the timely implementation of in silico biology in routine wet-lab studies and have also allowed the application of a new approach to the elucidation of protein function in the post-genomic era.


Algorithms , Computational Biology/methods , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Aurora Kinases , Cell Cycle Proteins , Cell Line , Cytoskeletal Proteins , Evolution, Molecular , Fungal Proteins/metabolism , Humans , Inhibitor of Apoptosis Proteins , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Neoplasm Proteins , Nuclear Proteins/metabolism , Sequence Alignment , Survivin , Two-Hybrid System Techniques
14.
Clin Exp Metastasis ; 20(1): 69-76, 2003.
Article En | MEDLINE | ID: mdl-12650609

Numerous genetic changes are associated with metastasis of cancer cells. Previously, we used microarray to identify that collapsin response mediator protein-1 (CRMP-1) was involved in cancer invasion and metastasis. We further characterized that CRMP-1 was a novel invasion-suppression gene. Members of the CRMP gene family are intracellular phosphoproteins involved in the mediation of semaphorin induced F-actin depolymerization and growth cone collapse. The precise mechanism by which CRMP-I inhibits invasion is not yet clear. However, CRMP-1 transfected cells had fewer filopodia and less Matrigel-invasion abilities. A low expression of CRMP-I mRNA in lung cancer tissue was significantly associated with advanced disease, lymph node metastasis, early post-operative relapse, and shorter survival. In this article, we reviewed the functions of CRMPs and semaphorins and analyzed the structure and motifs of CRMP-1 by bioinformatics. As such, we hoped to shed further light on the mechanism by which CRMP-1 suppresses the invasion of cancer cells.


Genes, Tumor Suppressor , Neoplasm Metastasis/genetics , Nerve Tissue Proteins/genetics , Phosphoproteins/genetics , Endothelial Growth Factors/metabolism , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lymphokines/metabolism , Mitosis , Nerve Tissue Proteins/chemistry , Nuclear Localization Signals , Phosphoproteins/chemistry , Protein Biosynthesis , Protein Processing, Post-Translational , Semaphorin-3A/metabolism , Semaphorins/physiology , Signal Transduction , T-Box Domain Proteins/metabolism , Transcription, Genetic , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
15.
Oncogene ; 22(2): 298-307, 2003 Jan 16.
Article En | MEDLINE | ID: mdl-12527899

An analytic strategy was followed to identify putative regulatory genes during the development of human hepatocellular carcinoma (HCC). This strategy employed a bioinformatics analysis that used a database search to identify genes, which are differentially expressed in human HCC and are also under cell cycle regulation. A novel cell cycle regulated gene (HURP) that is overexpressed in HCC was identified. Full-length cDNAs encoding the human and mouse HURP genes were isolated. They share 72 and 61% identity at the nucleotide level and amino-acid level, respectively. Endogenous levels of HURP mRNA were found to be tightly regulated during cell cycle progression as illustrated by its elevated expression in the G(2)/M phase of synchronized HeLa cells and in regenerating mouse liver after partial hepatectomy. Immunofluorescence studies revealed that hepatoma up-regulated protein (HURP) localizes to the spindle poles during mitosis. Overexpression of HURP in 293T cells resulted in an enhanced cell growth at low serum levels and at polyhema-based, anchorage-independent growth assay. Taken together, these results strongly suggest that HURP is a potential novel cell cycle regulator that may play a role in the carcinogenesis of human cancer cells.


Carcinoma, Hepatocellular/genetics , Cell Cycle/genetics , Liver Neoplasms/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Databases, Nucleic Acid , Expressed Sequence Tags , Liver/embryology , Liver/physiology , Liver Regeneration/genetics , Mice , Mice, Inbred BALB C , Mitosis , Molecular Sequence Data , Organ Specificity , Sequence Homology, Amino Acid , Spindle Apparatus/genetics
...