Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
Med Oncol ; 39(12): 243, 2022 Sep 30.
Article in English | MEDLINE | ID: mdl-36180656

ABSTRACT

Esophageal cancer is one of the most common malignancies which induces cancer-related death. Cancer metastasis and recurrence are the main obstacle faced in esophageal cancer treatment. ß-Asarone has been shown to act as an anti-cancer reagent in various cancer types. However, the anti-cancer activities of ß-Asarone in esophageal cancer have not been shown. In the current study, we show that ß-Asarone suppressed the proliferation of esophageal squamous cancer cells (ESCC) in both dose- and time-dependent manners. Moreover, ß-Asarone treatment increases activated caspase 3, caspase 9, and cleaved poly ADP-ribose polymerase, and induces apoptosis in ESCC. Additionally, ß-Asarone also suppresses epithelial-mesenchymal transition (EMT) and the invasive and migratory abilities in ESCC. Interestingly, ß-Asarone suppresses TGF-ß/Smad signaling by inhibition of TGF-ß-induced phosphorylation of Smad2 and Smad3. Importantly, we show that inhibition of TGF-ß/Smad signaling activation is critical for ß-Asarone-suppressed EMT. Our data revealed a novel role of ß-Asarone which targets invasive properties by inhibiting TGF-ß/Smad signaling activation in ESCC. Our study suggests the potential application of ß-Asarone to reduce cancer metastasis and recurrence in esophageal cancer treatment.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Adenosine Diphosphate Ribose , Allylbenzene Derivatives , Anisoles , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Caspase 3 , Caspase 9 , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Humans , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology
3.
Angew Chem Int Ed Engl ; 60(51): 26663-26670, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34614283

ABSTRACT

Targeting cereblon (CRBN) is currently one of the most frequently reported proteolysis-targeting chimera (PROTAC) approaches, owing to favorable drug-like properties of CRBN ligands, immunomodulatory imide drugs (IMiDs). However, IMiDs are known to be inherently unstable, readily undergoing hydrolysis in body fluids. Here we show that IMiDs and IMiD-based PROTACs rapidly hydrolyze in commonly utilized cell media, which significantly affects their cell efficacy. We designed novel CRBN binders, phenyl glutarimide (PG) analogues, and showed that they retained affinity for CRBN with high ligand efficiency (LE >0.48) and displayed improved chemical stability. Our efforts led to the discovery of PG PROTAC 4 c (SJ995973), a uniquely potent degrader of bromodomain and extra-terminal (BET) proteins that inhibited the viability of human acute myeloid leukemia MV4-11 cells at low picomolar concentrations (IC50 =3 pM; BRD4 DC50 =0.87 nM). These findings strongly support the utility of PG derivatives in the design of CRBN-directed PROTACs.


Subject(s)
Adaptor Proteins, Signal Transducing/chemistry , Piperidones/chemistry , Ubiquitin-Protein Ligases/chemistry , Humans , Hydrolysis , Proteolysis
4.
STAR Protoc ; 2(3): 100702, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34485934

ABSTRACT

Identification of diverse chemotypes of selective KDM4 inhibitors is important for exploring and validating the roles of KDM4s in the pathogenesis of human disease and for developing therapies. Here, we report a protocol for high-throughput screening of KDM4 inhibitors using TR-FRET demethylation functional assay. We describe this protocol for screen of KDM4B inhibitors, which can be modified to screen inhibitors of other JmjC-domain-containing KDMs. For complete details on the use and execution of this protocol, please refer to Singh et al. (2021).


Subject(s)
Drug Evaluation, Preclinical/methods , Fluorescence Resonance Energy Transfer/methods , High-Throughput Screening Assays/methods , Histone Demethylases/antagonists & inhibitors , Histones/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine , Substrate Specificity
5.
Microb Pathog ; 158: 105053, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34147587

ABSTRACT

Human parainfluenza virus type 3 (hPIV-3) entry and intrahost spread through membrane fusion are initiated by two envelope glycoproteins, hemagglutinin-neuraminidase (HN) and fusion (F) protein. Binding of HN protein to the cellular receptor via its receptor-binding sites triggers conformational changes in the F protein leading to virus-cell fusion. However, little is known about the roles of individual amino acids that comprise the receptor-binding sites in the fusion process. Here, residues R192, D216, E409, R424, R502, Y530 and E549 located within the receptor-binding site Ⅰ, and residues N551 and H552 at the putative site Ⅱ were replaced by alanine with site-directed mutagenesis. All mutants except N551A displayed statistically lower hemadsorption activities ranging from 16.4% to 80.2% of the wild-type (wt) level. With standardization of the number of bound erythrocytes, similarly, other than N551A, all mutants showed reduced fusogenic activity at three successive stages: lipid mixing (hemifusion), content mixing (full fusion) and syncytium development. Kinetic measurements of the hemifusion process showed that the initial hemifusion extent for R192A, D216A, E409A, R424A, R502A, Y530A, E549A and H552A was decreased to 69.9%, 80.6%, 71.3%, 67.3%, 50.6%, 87.4%, 84.9% and 25.1%, respectively, relative to the wt, while the initial rate of hemifusion for the E409A, R424A, R502A and H552A mutants was reduced to 69.0%, 35.4%, 62.3%, 37.0%, respectively. In addition, four mutants with reduced initial hemifusion rates also showed decreased percentages of F protein cleavage from 43.4% to 56.3% of the wt. Taken together, Mutants R192A, D216A, E409A, R424A, R502A, Y530A, E549A and H552A may lead to damage on the fusion activity at initial stage of hemifusion, of which decreased extent and rate may be associated with impaired receptor binding activity resulting in the increased activation barrier of F protein and the cleavage of it, respectively.


Subject(s)
HN Protein , Parainfluenza Virus 3, Human , Binding Sites , HN Protein/genetics , HN Protein/metabolism , Humans , Mutagenesis, Site-Directed , Parainfluenza Virus 3, Human/genetics , Protein Binding , Viral Fusion Proteins/genetics , Virus Internalization
6.
iScience ; 24(1): 101996, 2021 Jan 22.
Article in English | MEDLINE | ID: mdl-33490904

ABSTRACT

Histone lysine demethylases (KDMs) play critical roles in oncogenesis and therefore may be effective targets for anticancer therapy. Using a time-resolved fluorescence resonance energy transfer demethylation screen assay, in combination with multiple orthogonal validation approaches, we identified geldanamycin and its analog 17-DMAG as KDM inhibitors. In addition, we found that these Hsp90 inhibitors increase degradation of the alveolar rhabdomyosarcoma (aRMS) driver oncoprotein PAX3-FOXO1 and induce the repressive epigenetic mark H3K9me3 and H3K36me3 at genomic loci of PAX3-FOXO1 targets. We found that as monotherapy 17-DMAG significantly inhibits expression of PAX3-FOXO1 target genes and multiple oncogenic pathways, induces a muscle differentiation signature, delays tumor growth and extends survival in aRMS xenograft mouse models. The combination of 17-DMAG with conventional chemotherapy significantly enhances therapeutic efficacy, indicating that targeting KDM in combination with chemotherapy may serve as a therapeutic approach to PAX3-FOXO1-positive aRMS.

7.
Cancer Res ; 80(17): 3507-3518, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32651255

ABSTRACT

Inhibition of members of the bromodomain and extraterminal (BET) family of proteins has proven a valid strategy for cancer chemotherapy. All BET identified to date contain two bromodomains (BD; BD1 and BD2) that are necessary for recognition of acetylated lysine residues in the N-terminal regions of histones. Chemical matter that targets BET (BETi) also interact via these domains. Molecular and cellular data indicate that BD1 and BD2 have different biological roles depending upon their cellular context, with BD2 particularly associated with cancer. We have therefore pursued the development of BD2-selective molecules both as chemical probes and as potential leads for drug development. Here we report the structure-based generation of a novel series of tetrahydroquinoline analogs that exhibit >50-fold selectivity for BD2 versus BD1. This selective targeting resulted in engagement with BD-containing proteins in cells, resulting in modulation of MYC proteins and downstream targets. These compounds were potent cytotoxins toward numerous pediatric cancer cell lines and were minimally toxic to nontumorigenic cells. In addition, unlike the pan BETi (+)-JQ1, these BD2-selective inhibitors demonstrated no rebound expression effects. Finally, we report a pharmacokinetic-optimized, metabolically stable derivative that induced growth delay in a neuroblastoma xenograft model with minimal toxicity. We conclude that BD2-selective agents are valid candidates for antitumor drug design for pediatric malignancies driven by the MYC oncogene. SIGNIFICANCE: This study presents bromodomain-selective BET inhibitors that act as antitumor agents and demonstrates that these molecules have in vivo activity towards neuroblastoma, with essentially no toxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Design , Neoplasms , Transcription Factors/antagonists & inhibitors , Animals , Cell Line, Tumor , Child , Female , Humans , Mice , Mice, SCID , Neoplasms/genetics , Neoplasms/metabolism , Protein Domains , Proto-Oncogene Proteins c-myc/genetics , Structure-Activity Relationship , Xenograft Model Antitumor Assays
8.
Cancer Res ; 77(17): 4626-4638, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28684529

ABSTRACT

Histone lysine demethylases facilitate the activity of oncogenic transcription factors, including possibly MYC. Here we show that multiple histone demethylases influence the viability and poor prognosis of neuroblastoma cells, where MYC is often overexpressed. We also identified the approved small-molecule antifungal agent ciclopirox as a novel pan-histone demethylase inhibitor. Ciclopirox targeted several histone demethylases, including KDM4B implicated in MYC function. Accordingly, ciclopirox inhibited Myc signaling in parallel with mitochondrial oxidative phosphorylation, resulting in suppression of neuroblastoma cell viability and inhibition of tumor growth associated with an induction of differentiation. Our findings provide new insights into epigenetic regulation of MYC function and suggest a novel pharmacologic basis to target histone demethylases as an indirect MYC-targeting approach for cancer therapy. Cancer Res; 77(17); 4626-38. ©2017 AACR.


Subject(s)
Antifungal Agents/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Histone Demethylases/antagonists & inhibitors , Neuroblastoma/drug therapy , Proto-Oncogene Proteins c-myc/metabolism , Pyridones/pharmacology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Ciclopirox , Epigenesis, Genetic , Histones/metabolism , Humans , Mice , Mice, SCID , Neuroblastoma/enzymology , Neuroblastoma/pathology , Oxidative Phosphorylation/drug effects , Proto-Oncogene Proteins c-myc/genetics , RNA, Small Interfering/genetics , Transcription, Genetic/drug effects , Tumor Cells, Cultured
9.
Proc Natl Acad Sci U S A ; 113(13): 3669-74, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26976575

ABSTRACT

The influenza endonuclease is an essential subdomain of the viral RNA polymerase. It processes host pre-mRNAs to serve as primers for viral mRNA and is an attractive target for antiinfluenza drug discovery. Compound L-742,001 is a prototypical endonuclease inhibitor, and we found that repeated passaging of influenza virus in the presence of this drug did not lead to the development of resistant mutant strains. Reduced sensitivity to L-742,001 could only be induced by creating point mutations via a random mutagenesis strategy. These mutations mapped to the endonuclease active site where they can directly impact inhibitor binding. Engineered viruses containing the mutations showed resistance to L-742,001 both in vitro and in vivo, with only a modest reduction in fitness. Introduction of the mutations into a second virus also increased its resistance to the inhibitor. Using the isolated wild-type and mutant endonuclease domains, we used kinetics, inhibitor binding and crystallography to characterize how the two most significant mutations elicit resistance to L-742,001. These studies lay the foundation for the development of a new class of influenza therapeutics with reduced potential for the development of clinical endonuclease inhibitor-resistant influenza strains.


Subject(s)
Endonucleases/antagonists & inhibitors , Endonucleases/genetics , Enzyme Inhibitors/pharmacology , Hydroxybutyrates/pharmacology , Influenza A virus/drug effects , Piperidines/pharmacology , Animals , Catalytic Domain/genetics , Crystallography, X-Ray , Dogs , Drug Resistance, Viral/genetics , Endonucleases/metabolism , Genetic Variation , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H1N1 Subtype/genetics , Influenza A virus/enzymology , Influenza A virus/genetics , Kinetics , Madin Darby Canine Kidney Cells , Models, Molecular , Mutagenesis , RNA-Dependent RNA Polymerase/antagonists & inhibitors , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism
10.
Proc Natl Acad Sci U S A ; 113(12): 3275-80, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26951671

ABSTRACT

The UvsY recombination mediator protein is critical for efficient homologous recombination in bacteriophage T4 and is the functional analog of the eukaryotic Rad52 protein. During T4 homologous recombination, the UvsX recombinase has to compete with the prebound gp32 single-stranded binding protein for DNA-binding sites and UvsY stimulates this filament nucleation event. We report here the crystal structure of UvsY in four similar open-barrel heptameric assemblies and provide structural and biophysical insights into its function. The UvsY heptamer was confirmed in solution by centrifugation and light scattering, and thermodynamic analyses revealed that the UvsY-ssDNA interaction occurs within the assembly via two distinct binding modes. Using surface plasmon resonance, we also examined the binding of UvsY to both ssDNA and the ssDNA-gp32 complex. These analyses confirmed that ssDNA can bind UvsY and gp32 independently and also as a ternary complex. They also showed that residues located on the rim of the heptamer are required for optimal binding to ssDNA, thus identifying the putative ssDNA-binding surface. We propose a model in which UvsY promotes a helical ssDNA conformation that disfavors the binding of gp32 and initiates the assembly of the ssDNA-UvsX filament.


Subject(s)
Membrane Proteins/chemistry , Membrane Proteins/physiology , Viral Proteins/chemistry , Viral Proteins/physiology , Amino Acid Sequence , Models, Molecular , Molecular Sequence Data , Protein Conformation , Structure-Activity Relationship
11.
Intervirology ; 58(5): 297-309, 2015.
Article in English | MEDLINE | ID: mdl-26694747

ABSTRACT

OBJECTIVE: To investigate the effect of the leucine zipper-like motif between HRA and HRB of the human parainfluenza virus 3 fusion protein on fusion activity. METHODS: Site-directed mutagenesis was utilized to substitute the heptadic residues at 257, 264, 271, 278, 285, 292, and 299 in this motif with alanine. Additionally, 3 middle heptadic leucine residues at 271, 278, and 285 were replaced with alanine singly or in combination. A vaccinia virus-T7 RNA polymerase transient expression system was employed to express the wild-type or mutated fusion (F) proteins. Three different types of membrane fusion assays were performed to analyze the fusogenic activity, fluorescence-activated cell sorting (FACS) analysis was executed to examine the cell surface expression level, and a coimmunoprecipitation assay was conducted to probe the hemagglutinin-neuraminidase (HN)-F interaction at the cell surface. RESULTS: All of the substitutions in this motif exhibited diminished or even lost fusion activity in all stages of fusion, although they all had no effect on cell surface expression. In the coimmunoprecipitation assay, all mutants resulted in decreased detection of the HN-F complexes compared with that of the wild-type F protein. CONCLUSIONS: This motif has an important influence on fusion activity, and its integrality is indispensable for membrane fusion.


Subject(s)
Leucine Zippers , Mutation , Parainfluenza Virus 3, Human/physiology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/metabolism , Virus Internalization , Animals , Cell Line , Cricetinae , DNA Mutational Analysis , Erythrocytes , Humans , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism
12.
PLoS One ; 10(6): e0129807, 2015.
Article in English | MEDLINE | ID: mdl-26070194

ABSTRACT

The anti-apoptotic protein survivin is highly expressed in most human cancer cells, but has very low expression in normal differentiated cells. Thus survivin is considered as an attractive cancer drug target. Herein we report the design and synthesis of a series of novel survivin inhibitors based on the oxyquinoline scaffold from our recently identified hit compound UC-112. These new analogs were tested against a panel of cancer cell lines including one with multidrug-resistant phenotype. Eight of these new UC-112 analogs showed IC50 values in the nanomole range in anti-proliferative assays. The best three compounds among them along with UC-112 were submitted for NCI-60 cancer cell line screening. The results indicated that structural modification from UC-112 to our best compound 4g has improved activity by four folds (2.2 µM for UC-112 vs. 0.5 µM for 4g, average GI50 values over all cancer cell lines in the NCI-60 panel).Western blot analyses demonstrated the new compounds maintained high selectivity for survivin inhibition over other members in the inhibition of apoptosis protein family. When tested in an A375 human melanoma xenograft model, the most active compound 4g effectively suppressed tumor growth and strongly induced cancer cell apoptosis in tumor tissues. This novel scaffold is promising for the development of selective survivin inhibitors as potential anticancer agents.


Subject(s)
Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Quantitative Structure-Activity Relationship , Small Molecule Libraries/chemistry , Amino Acid Sequence , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Inhibitor of Apoptosis Proteins/chemistry , Mice , Molecular Docking Simulation , Molecular Sequence Data , Protein Binding , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/pharmacology , Survivin
13.
Viral Immunol ; 27(9): 422-9, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25226223

ABSTRACT

Residues 221-239 of rubella virus E1 glycoprotein contain antibody neutralization domains, and the solvent-exposed charged amino acids at the binding interface may be crucial for binding ability. However, the role of charged amino acid residues on the E1 epitope in peptide-antibody binding is unknown. To investigate the role of single amino acid substitutions on the important neutralizing epitope, biolayer interferometry and serological tests were performed. There are three charged residues in the neutralizing epitope: D229, R237, and H238. Substitution of D229 for amino acid A had no influence on the binding activity of the antibody to the peptide. However, substitutions of R237 or H238 for charged amino acid H or R were found to abolish the binding activity. Furthermore, substitution of an uncharged amino acid Q236 for a charged amino acid D was found to reduce the binding activity significantly. Thus, R237 and H238 are key amino acids in the rubella virus E1 neutralization epitope.


Subject(s)
Amino Acids , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Epitopes/immunology , Rubella virus/immunology , Viral Envelope Proteins/immunology , Amino Acid Substitution , Animals , DNA Mutational Analysis , Female , Interferometry , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Neutralization Tests , Protein Binding
14.
Virus Res ; 174(1-2): 137-47, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23562646

ABSTRACT

Human parainfluenza virus type 3 (hPIV-3) is a major respiratory tract pathogen that affects infants and young children. The hPIV-3 hemagglutinin-neuraminidase (HN) protein is a multifunctional protein mediating hemadsorption (HAD), neuraminidase (NA), and fusion promotion activities, each of which affects the ability of HN to promote viral fusion and entry. The hPIV-3 HN protein contains four potential sites (N308, N351, N485 and N523) for N-linked glycosylation. Electrophoretic mobility analysis of mutated HN proteins indicated that N308, N351 and N523 sites, but not the N485 site in HN protein, were targeted for the addition of glycans in BHK-21 cells. These functional glycosylation sites were systematically eliminated in various combinations from HN to form a panel of mutants in which the roles of individual carbohydrate chains and groups of carbohydrate chains could be analyzed. Removal of individual or multiple N-glycans on the hPIV-3 HN protein had no effects on transport to the cell surface, expression and NA activity. Single glycosylation site mutants (G1, G2 and G4) not only impaired fusion promotion activity but also reduced HAD activity of HN protein, which was even more obvious for all three double mutants (G12, G14 and G24) and the triple mutant (G124). In addition, every mutant protein retained F-interactive capability that was equal to the wild-type protein capability. Interestingly, the F protein that could be co-immunoprecipitated with the G12 mutated protein or immunoprecipitated with anti-F antibody was not efficiently cleaved. For G14, G24 and G124, little cleaved F protein was detected in co-immuoprecipitation F protein assay and its total amounts where in the cell lysates. The mechanism underlying hPIV-3 HN and F protein remained associated before and after receptor engagement and the strength of the HN-receptor interaction modulated the activation of F the protein which could determine the extent of fusion. Finally, we demonstrated that single or multiple N-glycosylation site mutations inhibited fusion at the earliest stages. Taken together, these results indicated that N-glycosylation of hPIV-3 HN is critical to its receptor recognition activity, cleavage of the F protein, and fusion promotion activity, but had no influence on its interaction with the homologous F protein and NA activity.


Subject(s)
HN Protein/metabolism , Parainfluenza Virus 3, Human/physiology , Protein Processing, Post-Translational , Virus Internalization , DNA Mutational Analysis , Glycosylation , HN Protein/genetics , Humans , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Parainfluenza Virus 3, Human/genetics
15.
Bing Du Xue Bao ; 29(5): 500-8, 2013 Sep.
Article in Chinese | MEDLINE | ID: mdl-24386838

ABSTRACT

To determine the functions of N-carbohydrate chains in human parainfluenza virus type 3 hemagglutinin-neuraminidase(HN) protein, a PCR-based site-directed mutagenesis method was used to obtain N-glycan mutants. Protein electrophoresis rate, cell surface expression,receptor binding activity, neuraminidase activity and cell fusion promotion activity were determined. The HN proteins of single mutants (G1, G2, and G4) and multiple mutants (G12, G14, G24 and G124) migrated faster than the wild-type (wt) HN protein on polyacrylamide gels, while G3-mutated protein and wt HN protein migrated at the same position. There was no statistic difference in cell surface expression and neuraminidase activity between wt and each mutant HN protein (P>0.05), but receptor binding activity and cell fusion promotion activity of each mutant protein was reduced to significant extent (P<0.05). G1, G2 and G4 mutants exhibited re duced receptor binding activity, which was 83.94%, 76.45% and 55.32% of the wt level, respectively. G1, G2 and G4-mutated proteins also showed reductions in fusion promotion activity, which was 80.84%, 77.83% and 64.16%, respectively. Multiple mutants with G12-, G14-, G24- and G124- substitutions could further reduce receptor binding activities, 33.07%, 20.67%, 19.96% and 15.11% of the wt HN level, respectively. G12, G14, G24 and G124 mutants exhibited levels of fusion promotion activity that were only 46.360, 12.04%, 13.43% and 4.05% of the wt amount, respectively. As N-glycans of hPIV3 HN protein play an important role in receptor binding activity and cell fusion promotion activity of HN protein. We propose that the loss of N-glycans change the conformation or orientation of globular domain that is responsible for receptor binding and lower receptor binding activity and cell fusion promotion activi ty.


Subject(s)
HN Protein/chemistry , HN Protein/metabolism , Parainfluenza Virus 3, Human/enzymology , Respirovirus Infections/virology , Glycosylation , HN Protein/genetics , Humans , Mutation , Parainfluenza Virus 3, Human/chemistry , Parainfluenza Virus 3, Human/genetics , Parainfluenza Virus 3, Human/physiology , Protein Binding , Receptors, Virus/metabolism , Respirovirus Infections/metabolism , Virus Internalization
16.
Bing Du Xue Bao ; 29(5): 578-82, 2013 Sep.
Article in Chinese | MEDLINE | ID: mdl-24386850

ABSTRACT

Rubella virus (RV), a member of the family Togaviridae, can induce apoptosis of host cells in vitro. Protein kinases of the Ras-Raf-MEK-ERK pathway and PI3K-Akt pathway play essential roles in virus multiplication, cell survival and apoptosis. Proteins p53 and TAp63 that bind to specific DNA sequences stimulate Bax in a manner to produce functional pores that facilitate release of mitochondrial cytochrome c and downstream caspase activation. In this review, the molecular mechanisms of RV-induced cell apoptosis, including RV-infected cell lines, pathological changes in cell components and apoptosis signaling pathways are summarized.


Subject(s)
Apoptosis , Rubella virus/physiology , Rubella/physiopathology , Humans , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Rubella/genetics , Rubella/metabolism , Rubella/virology , Rubella virus/genetics
17.
Article in Chinese | MEDLINE | ID: mdl-24579480

ABSTRACT

OBJECTIVE: To express the rubella virus E1-374 glycoprotein in Pichia pastoris and study the immunogenecity of the recombinant protein. METHODS: The cDNA of protein E1-374 was cloned into the expression vector pGAPZalphaA and transformed into Pichia pastoris GS115 cells by electrotransfection. The expressed protein was confirmed by indirect immunofluorescence and demonstrated immunoreactivity by Western Blot. Rubella virus IgG antibody was assayed with ELISA after mice were inmmunized by E1-374 glycoprotein. RESULTS: SDS-PAGE analysis and Western Blot analysis of E1-374 protein revealed this protein to be 46.89 x 10(3). Antiserum (1:100) and E1-374 (5.5 microg/ml) was chosen for ELISA optimization. The intra-assay coefficient of variation for the ELISA was 0.36%-12.45%. CONCLUSION: Protein E1-374 was highly expressed in Pichia pastoris cells, and it was a good choice to prepare rubella virus recombinant protein vaccines.


Subject(s)
Gene Expression , Pichia/genetics , Rubella virus/genetics , Viral Envelope Proteins/genetics , Animals , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, Inbred BALB C , Pichia/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Rubella virus/immunology , Viral Envelope Proteins/immunology
18.
Science ; 335(6072): 1110-4, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22383850

ABSTRACT

The sulfonamide antibiotics inhibit dihydropteroate synthase (DHPS), a key enzyme in the folate pathway of bacteria and primitive eukaryotes. However, resistance mutations have severely compromised the usefulness of these drugs. We report structural, computational, and mutagenesis studies on the catalytic and resistance mechanisms of DHPS. By performing the enzyme-catalyzed reaction in crystalline DHPS, we have structurally characterized key intermediates along the reaction pathway. Results support an S(N)1 reaction mechanism via formation of a novel cationic pterin intermediate. We also show that two conserved loops generate a substructure during catalysis that creates a specific binding pocket for p-aminobenzoic acid, one of the two DHPS substrates. This substructure, together with the pterin-binding pocket, explains the roles of the conserved active-site residues and reveals how sulfonamide resistance arises.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dihydropteroate Synthase/chemistry , Dihydropteroate Synthase/metabolism , Drug Resistance, Bacterial , Sulfamethoxazole/pharmacology , Sulfathiazoles/pharmacology , 4-Aminobenzoic Acid/chemistry , 4-Aminobenzoic Acid/metabolism , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Bacillus anthracis/drug effects , Bacillus anthracis/enzymology , Biocatalysis , Catalytic Domain , Crystallization , Crystallography, X-Ray , Dihydropteroate Synthase/genetics , Diphosphates/chemistry , Diphosphates/metabolism , Magnesium/chemistry , Models, Chemical , Models, Molecular , Molecular Sequence Data , Mutagenesis , Parabens/chemistry , Parabens/metabolism , Protein Conformation , Sulfamethoxazole/chemistry , Sulfamethoxazole/metabolism , Sulfathiazole , Sulfathiazoles/chemistry , Sulfathiazoles/metabolism , Yersinia pestis/drug effects , Yersinia pestis/enzymology
19.
Bioconjug Chem ; 22(10): 2110-7, 2011 Oct 19.
Article in English | MEDLINE | ID: mdl-21916405

ABSTRACT

Dihydropteroate synthase (DHPS) is the classical target of the sulfonamide class of antimicrobial agents, whose use has been limited by widespread resistance and pharmacological side effects. We have initiated a structure-based drug design approach for the development of novel DHPS inhibitors that bind to the highly conserved and structured pterin subsite rather than to the adjacent p-aminobenzoic acid binding pocket that is targeted by the sulfonamide class of antibiotics. To facilitate these studies, a robust pterin site-specific fluorescence polarization (FP) assay has been developed and is discussed herein. These studies include the design, synthesis, and characterization of two fluorescent probes, and the development and validation of a rapid DHPS FP assay. This assay has excellent DMSO tolerance and is highly reproducible as evidenced by a high Z' factor. This assay offers significant advantages over traditional radiometric or phosphate release assays against this target, and is suitable for site-specific high-throughput and fragment-based screening studies.


Subject(s)
Bacillus anthracis/enzymology , Dihydropteroate Synthase/metabolism , Fluorescence Polarization/methods , Fluorescent Dyes/chemistry , Pterins/chemistry , Binding Sites , Binding, Competitive , Dihydropteroate Synthase/chemistry , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/metabolism , Pterins/chemical synthesis , Pterins/metabolism , Sensitivity and Specificity
20.
Appl Opt ; 50(24): 4855-9, 2011 Aug 20.
Article in English | MEDLINE | ID: mdl-21857710

ABSTRACT

The paper puts forward a new method of ultrafine particle size measurement using small amounts of data of a dynamic light-scattering signal, and establishes an arithmetic model of the measurement by wavelet package transform. First, through the wavelet package transform, the ultrafine particle dynamic light-scattering signals were decomposed into multifrequency bands. Then, the noise of signals of different frequency bands were removed and the power spectrum of the wavelet packet coefficients of each frequency band was calculated. Finally, the ultrafine particle size distribution information could be deduced from inversing the power spectrum. The standard polystyrene particles of 100, 300, and 400 nm were measured using this method, and the inversion results indicated that this method can effectively remove noise and improve the accuracy of particle size measurement using small amounts of data.

SELECTION OF CITATIONS
SEARCH DETAIL
...