Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 233
Filter
1.
Toxicol Appl Pharmacol ; 410: 115338, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33217376

ABSTRACT

Modern High-Throughput Screening (HTS) techniques allow to determine in vitro bioactivity of tens of thousands of chemicals within a relatively short period of time and tested compounds are usually interpreted as either active or inactive. The interpretation is mostly based on the assumption of monotonic dose-response. This approach ignores potential abnormal dose-response relationships, such as non-monotonic dose-response (NMDR). NMDR presents a serious challenge to toxicologists and pharmacologists, since they undermine the usefulness of such concepts as lowest-observed-adverse-effect level (LOAEL) and no-observed-adverse-effect level (NOAEL). The possible presence of the NMDR in Androgen receptor (AR) agonism was examined for a structurally diverse set of chemicals (~8 300 unique compounds) from Tox21 project library. The source of activity data is Tox21 AR agonism luciferase-based HTS on the MDA-MB-453 cell line. The examination of curve fitting for 35,328 dose-response data entries was based on modified version of existing criteria for determination of NMDR. The bias that arises from compounds' cytotoxicity and interference with firefly luciferase protein was also studied. The examination has shown evidence of NMDR for several compounds, including known AR antagonists (e. g. Cyproterone acetate) and other known endocrine disruptors (e. g. Tranilast). Compounds were divided into 3 groups based on chemical class, known biological activity profile and the shape of dose-response curve. The challenges of using HTS data to determine NMDR and benefits of this analysis are discussed.


Subject(s)
Algorithms , Androgens/administration & dosage , Androgens/analysis , High-Throughput Screening Assays/methods , Dose-Response Relationship, Drug , Luciferases/antagonists & inhibitors , Metribolone/administration & dosage , Metribolone/analysis
2.
Anal Chem ; 92(11): 7393-7398, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32410446

ABSTRACT

Although bioluminescent molecular beacons designed around resonance quenchers have shown higher signal-to-noise ratios and increased sensitivity compared with fluorescent beacon systems, bioluminescence quenching is still comparatively inefficient. A more elegant solution to inefficient quenching can be realized by designing a competitive inhibitor that is structurally very similar to the native substrate, resulting in essentially complete substrate exclusion. In this work, we designed a conjugated anti-interferon-γ (IFN-γ) molecular aptamer beacon (MAB) attached to a bioluminescent protein, Gaussia luciferase (GLuc), and an inhibitor molecule with a similar structure to the native substrate coelenterazine. To prove that a MAB can be more sensitive and have a better signal-to-noise ratio, a bioluminescence-based assay was developed against IFN-γ and provided an optimized, physiologically relevant detection limit of 1.0 nM. We believe that this inhibitor approach may provide a simple alternative strategy to standard resonance quenching in the development of high-performance molecular beacon-based biosensing systems.


Subject(s)
Aptamers, Nucleotide/chemistry , Biosensing Techniques , Enzyme Inhibitors/chemistry , Imidazoles/chemistry , Luciferases/chemistry , Luminescent Proteins/chemistry , Pyrazines/chemistry , Animals , Aptamers, Nucleotide/chemical synthesis , Copepoda/enzymology , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Luciferases/antagonists & inhibitors , Luciferases/metabolism , Luminescent Measurements , Luminescent Proteins/antagonists & inhibitors , Luminescent Proteins/metabolism , Models, Molecular , Molecular Structure , Pyrazines/pharmacology , Signal-To-Noise Ratio
3.
Nanoscale ; 12(18): 10347-10360, 2020 May 14.
Article in English | MEDLINE | ID: mdl-32369076

ABSTRACT

One of the challenges for the clinical translation of RNA interference (RNAi)-based therapies concerns the deposition of therapeutically effective doses of the nucleic acids, like siRNA, at a local tissue level without severe off-target effects. To address this issue, hydrogels can be used as matrices for the local and sustained release of the siRNA cargo. In this study, the formation of polyplexes based on siRNA and poly(2-dimethylaminoethyl methacrylate) (PDMAEMA)-based polymers was investigated, followed by their loading in a thermosensitive hydrogel to promote local siRNA release. A multifunctional NPD triblock copolymer consisting of a thermosensitive poly(N-isopropylacrylamide) (PNIPAM, N), a hydrophilic poly(ethylene glycol) (PEG, P), and a cationic PDMAEMA (D) block was used to study the binding properties with siRNA taking the non-thermosensitive PD polymer as control. For both polymers, small polyplexes with sizes ranging from 10-20 nm were formed in aqueous solution (HBS buffer, 20 mM HEPES, 150 mM NaCl, pH 7.4) when prepared at a N/P charge ratio of 5 or higher. Formulating the siRNA into NPD or PD polyplexes before loading into the thermosensitive PNIPAM-PEG-PNIPAM hydrogel resulted in a more controlled and sustained release compared to free siRNA release from the hydrogel. The polyplexes were released for 128 hours in HBS, when changing the release medium twice a day, while free siRNA was completely released within 50 hours with already 40% being released after changing the release medium just once. The release of the polyplexes was dependent on the dissolution rate of the hydrogel matrix. Moreover, intact polyplexes were released from the hydrogels with a similar size as before loading, suggesting that the hydrogel material did not compromise the polyplex stability. Finally, it was shown that the released polyplexes were still biologically active and transfected FaDu cells, which was observed by siRNA-induced luciferase silencing in vitro. This study shows the development of an injectable thermosensitive hydrogel to promote local and sustained release of siRNA, which can potentially be used to deliver siRNA for various applications, such as the treatment of tumors.


Subject(s)
Hydrogels/chemistry , RNA, Small Interfering/metabolism , Cell Line, Tumor , Humans , Luciferases/antagonists & inhibitors , Luciferases/genetics , Luciferases/metabolism , Methacrylates/chemistry , Nylons/chemistry , Polyethylene Glycols/chemistry , Polymers/chemical synthesis , Polymers/chemistry , RNA Interference , RNA, Small Interfering/chemistry , Transfection
4.
Carbohydr Polym ; 229: 115451, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31826509

ABSTRACT

Oil-in-water nano-emulsions have been obtained in the HEPES 20 mM buffer solution / [Alkylamidoammonium:Kolliphor EL = 1:1] / [6 wt% ethylcellulose in ethyl acetate] system over a wide oil-to-surfactant range and above 35 wt% aqueous component at 25 °C. The nano-emulsion with an oil-to-surfactant ratio of 70/30 and 95 wt% aqueous component was used for nanoparticles preparation. These nanoparticles (mean diameter around 90 nm and zeta potential of +22 mV) were non-toxic to HeLa cells up to a concentration of 3 mM of cationic species. Successful complexation with an antisense phosphorothioate oligonucleotide targeting Renilla luciferase mRNA was achieved at cationic/anionic charge ratios above 16, as confirmed by zeta potential measurements and an electrophoretic mobility shift assay, provided that no Fetal Bovine Serum is present in the cell culture medium. Importantly, Renilla luciferase gene inhibition shows an optimum efficiency (40%) for the cationic/anionic ratio 28, which makes these complexes promising for "in vitro" cell transfection.


Subject(s)
Cellulose/analogs & derivatives , Nanoparticles/chemistry , Oligonucleotides, Antisense/genetics , Animals , Cattle , Cellulose/chemistry , Cellulose/toxicity , Gene Silencing , Gene Transfer Techniques , HeLa Cells , Humans , Luciferases/antagonists & inhibitors , Luciferases/genetics , Nanoparticles/toxicity , RNA, Messenger/genetics , Renilla/enzymology , Serum Albumin, Bovine/chemistry , Static Electricity
5.
Nucleic Acids Res ; 48(1): 75-85, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31777918

ABSTRACT

One of the major hurdles in RNAi research has been the development of safe and effective delivery systems for siRNAs. Although various chemical modifications have been proposed to improve their pharmacokinetic behaviour, their delivery to target cells and tissues presents many challenges. In this work, we implemented a receptor-targeting strategy to selectively deliver siRNAs to cancer cells using folic acid as a ligand. Folic acid is capable of binding to cell-surface folate receptors with high affinity. These receptors have become important molecular targets for cancer research as they are overexpressed in numerous cancers despite being expressed at low levels in normal tissues. Employing a post-column copper-catalyzed alkyne-azide cycloaddition (CuAAC), we report the synthesis of siRNAs bearing folic acid modifications at different positions within the sense strand. In the absence of a transfection carrier, these siRNAs were selectively taken up by cancer cells expressing folate receptors. We show that centrally modified folic acid-siRNAs display enhanced gene-silencing activity against an exogenous gene target (∼80% knockdown after 0.75 µM treatment) and low cytotoxicity. In addition, these siRNAs achieved potent dose-dependent knockdown of endogenous Bcl-2, an important anti-apoptotic gene.


Subject(s)
Folic Acid/chemistry , Gene Silencing , Gene Targeting/methods , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/genetics , Carbonates/chemistry , Cell Survival , Folate Receptors, GPI-Anchored/genetics , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/metabolism , Genes, Reporter , HT29 Cells , HeLa Cells , Humans , Luciferases/antagonists & inhibitors , Luciferases/genetics , Luciferases/metabolism , Pargyline/analogs & derivatives , Pargyline/chemistry , Potassium/chemistry , Protein Binding , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/chemical synthesis , Transfection
6.
Phytochemistry ; 155: 182-190, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30145456

ABSTRACT

Nine previously undescribed sesquiterpenoids, named magnograndins A-I, as well as fourteen known ones, were obtained from the 70% acetone extract of the leaves of Magnolia grandiflora. Their structures were ascertained based on the spectroscopic evidences. The assignment of the relative configuration of magnograndin A was further confirmed by single-crystal X-ray diffraction analysis. 1ß,10α-Epoxyparthenolide, parthenolide, and micheliolide exhibited potent cytotoxic activity against MDA-MB-468, AGS, HCT116, Hela, and MDA-MB-231 human cancer cell lines with IC50 values ranging from 1.76 to 16.11 µM. 1ß,10α-Epoxyparthenolide and micheliolide potently inhibited NF-κB transcriptional activity with IC50 of 13.92 and 8.95 µM, respectively. The expression levels of NF-κB downstream protein p65 and XIAP were clearly down-regulated in 1ß,10α-epoxyparthenolide and micheliolide treated cells, which demonstrated the inhibition of NF-κB signaling pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Enzyme Inhibitors/pharmacology , Magnolia/chemistry , Plant Leaves/chemistry , Sesquiterpenes/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Humans , Luciferases/antagonists & inhibitors , Luciferases/genetics , Luciferases/metabolism , Molecular Structure , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Sesquiterpenes/chemistry , Sesquiterpenes/isolation & purification , Structure-Activity Relationship
7.
Molecules ; 23(6)2018 Jun 07.
Article in English | MEDLINE | ID: mdl-29880739

ABSTRACT

Fargesin is a bioactive lignan from Flos Magnoliae, an herb widely used in the treatment of allergic rhinitis, sinusitis, and headache in Asia. We sought to investigate whether fargesin ameliorates experimental inflammatory bowel disease (IBD) in mice. Oral administration of fargesin significantly attenuated the symptoms of dextran sulfate sodium (DSS)-induced colitis in mice by decreasing the inflammatory infiltration and myeloperoxidase (MPO) activity, reducing tumor necrosis factor (TNF)-α secretion, and inhibiting nitric oxide (NO) production in colitis mice. The degradation of inhibitory κBα (IκBα), phosphorylation of p65, and mRNA expression of nuclear factor κB (NF-κB) target genes were inhibited by fargesin treatment in the colon of the colitis mice. In vitro, fargesin blocked the nuclear translocation of p-p65, downregulated the protein levels of inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2), and dose-dependently inhibited the activity of NF-κB-luciferase in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Taken together, for the first time, the current study demonstrated the anti-inflammatory effects of fargesin on chemically induced IBD might be associated with NF-κB signaling suppression. The findings may contribute to the development of therapies for human IBD by using fargesin or its derivatives.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Benzodioxoles/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Lignans/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Benzodioxoles/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Dextran Sulfate/administration & dosage , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/chemically induced , Lignans/pharmacology , Luciferases/antagonists & inhibitors , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/antagonists & inhibitors , Peroxidase/antagonists & inhibitors , Proteolysis , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/antagonists & inhibitors
8.
J Chem Inf Model ; 58(5): 933-942, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29667823

ABSTRACT

Firefly luciferase is an enzyme that has found ubiquitous use in biological assays in high-throughput screening (HTS) campaigns. The inhibition of luciferase in such assays could lead to a false positive result. This issue has been known for a long time, and there have been significant efforts to identify luciferase inhibitors in order to enhance recognition of false positives in screening assays. However, although a large amount of publicly accessible luciferase counterscreen data is available, to date little effort has been devoted to building a chemoinformatic model that can identify such molecules in a given data set. In this study we developed models to identify these molecules using various methods, such as molecular docking, SMARTS screening, pharmacophores, and machine learning methods. Among the structure-based methods, the pharmacophore-based method showed promising results, with a balanced accuracy of 74.2%. However, machine-learning approaches using associative neural networks outperformed all of the other methods explored, producing a final model with a balanced accuracy of 89.7%. The high predictive accuracy of this model is expected to be useful for advising which compounds are potential luciferase inhibitors present in luciferase HTS assays. The models developed in this work are freely available at the OCHEM platform at http://ochem.eu .


Subject(s)
Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Luciferases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , False Positive Reactions , Luciferases/chemistry , Luciferases/metabolism , Molecular Docking Simulation , Protein Conformation
9.
Nucleic Acid Ther ; 28(1): 44-49, 2018 02.
Article in English | MEDLINE | ID: mdl-29195056

ABSTRACT

Specific gene silencing through RNA interference (RNAi) holds great promise as the next-generation therapeutic development platform. Previously, we have shown that branched, tripodal interfering RNA (tiRNA) structures could simultaneously trigger RNAi-mediated gene silencing of three target genes with 38 nt-long guide strands associated with Argonaute 2. Herein, we show that the branched RNA structure can trigger effective gene silencing in Dicer knockout cell line, demonstrating that the Dicer-mediated processing is not required for tiRNA activity. The finding of this study confirms the flexibility of the structure of RNAi triggers as well as the length of the guide strand in RNAi-mediated gene silencing.


Subject(s)
Argonaute Proteins/genetics , DEAD-box RNA Helicases/genetics , RNA Interference , RNA, Guide, Kinetoplastida/genetics , RNA, Small Interfering/genetics , Ribonuclease III/genetics , Argonaute Proteins/metabolism , Base Sequence , Cell Line , DEAD-box RNA Helicases/deficiency , Genes, Reporter , HeLa Cells , Humans , Luciferases/antagonists & inhibitors , Luciferases/genetics , Luciferases/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Nucleic Acid Conformation , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA, Guide, Kinetoplastida/metabolism , RNA, Small Interfering/metabolism , Ribonuclease III/deficiency , Survivin/antagonists & inhibitors , Survivin/genetics , Survivin/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/genetics , beta Catenin/metabolism
10.
Phys Chem Chem Phys ; 19(15): 10163-10176, 2017 Apr 12.
Article in English | MEDLINE | ID: mdl-28374029

ABSTRACT

High-throughput screening (HTS) is widely applied in many fields ranging from drug discovery to clinical diagnostics and toxicity assessment. Firefly luciferase is commonly used as a reporter to monitor the effect of chemical compounds on the activity of a specific target or pathway in HTS. However, the false positive rate of luciferase-based HTS is relatively high because many artifacts or promiscuous compounds that have direct interaction with the luciferase reporter enzyme are usually identified as active compounds (hits). Therefore, it is necessary to develop a rapid screening method to identify these compounds that can inhibit the luciferase activity directly. In this study, a virtual screening (VS) classification model called MIEC-GBDT (MIEC: Molecular Interaction Energy Components; GBDT: Gradient Boosting Decision Tree) was developed to distinguish luciferase inhibitors from non-inhibitors. The MIECs calculated by Molecular Mechanics/Generalized Born Surface Area (MM/GBSA) free energy decomposition were used to energetically characterize the binding pattern of each small molecule at the active site of luciferase, and then the GBDT algorithm was employed to construct the classifiers based on MIECs. The predictions to the test set show that the optimized MIEC-GBDT model outperformed molecular docking and MM/GBSA rescoring. The best MIEC-GBDT model based on the MIECs with the energy terms of ΔGele, ΔGvdW, ΔGGB, and ΔGSA achieves the prediction accuracies of 87.2% and 90.3% for the inhibitors and non-inhibitors in the test sets, respectively. Moreover, the energetic analysis of the vital residues suggests that the energetic contributions of the vital residues to the binding of inhibitors are quite different from those to the binding of non-inhibitors. These results suggest that the MIEC-GBDT model is reliable and can be used as a powerful tool to identify potential interference compounds in luciferase-based HTS experiments.


Subject(s)
Luciferases/antagonists & inhibitors , Algorithms , Area Under Curve , Catalytic Domain , High-Throughput Screening Assays , Ligands , Luciferases/metabolism , Molecular Docking Simulation , Protein Structure, Tertiary , ROC Curve , Thermodynamics
11.
Nanomedicine ; 13(6): 2091-2100, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28435135

ABSTRACT

Our present study aimed to develop an antisense oligonucleotide (ASO) delivery system to achieve gene silencing in intraocular tumor via topical instillation. ASO specific for luciferase was chosen as model drug, polyamidoamine (PG5) was employed to condense ASO, and penetratin (Pene) was used to enhance cellular uptake. Nanoscale PG5/ASO/Pene polyplex was stabilized via noncovalent bonding. In vitro evaluations indicated that PG5/ASO/Pene exhibited improved cell-penetrating and gene silencing ability compared with naked ASO and PG5/ASO. Subcutaneous and orthotopic tumor models expressing luciferase were established in nude mice. After treated by PG5/ASO/Pene, immunohistochemical results of subcutaneous tumors showed significant inhibition of luciferase expression via peritumoral injection, and bioluminescence from orthotopic tumor was obviously weakened via topical instillation. To date, few works were successful in noninvasive treatment of intraocular diseases using antisense strategy, this penetratin-modified polyplex could be a promising vector to inhibit protein expression by effectively delivering ASOs into the eye.


Subject(s)
Carrier Proteins/chemistry , Cornea/metabolism , Drug Delivery Systems , Gene Silencing , Glioblastoma/metabolism , Luciferases/antagonists & inhibitors , Oligonucleotides, Antisense/pharmacology , Animals , Cell Survival/drug effects , Cell-Penetrating Peptides , Cells, Cultured , Cornea/cytology , Glioblastoma/genetics , Glioblastoma/therapy , Luciferases/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Ocular Absorption , Oligonucleotides, Antisense/administration & dosage , Polymers/chemistry
12.
ACS Chem Biol ; 12(4): 1028-1037, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28195704

ABSTRACT

Novel engineered NanoLuc (Nluc) luciferase being smaller, brighter, and superior to traditional firefly (Fluc) or Renilla (Rluc) provides a great opportunity for the development of numerous biological, biomedical, clinical, and food and environmental safety applications. This new platform created an urgent need for Nluc inhibitors that could allow selective bioluminescent suppression and multiplexing compatibility with existing luminescence or fluorescence assays. Starting from thienopyrrole carboxylate 1, a hit from a 42 000 PubChem compound library with a low micromolar IC50 against Nluc, we derivatized four different structural fragments to discover a family of potent, single digit nanomolar, cell permeable inhibitors. Further elaboration revealed a channel that allowed access to the external Nluc surface, resulting in a series of highly potent cell impermeable Nluc inhibitors with negatively charged groups likely extending to the protein surface. The permeability was evaluated by comparing EC50 shifts calculated from both live and lysed cells expressing Nluc cytosolically. Luminescence imaging further confirmed that cell permeable compounds inhibit both intracellular and extracellular Nluc, whereas less permeable compounds differentially inhibit extracellular Nluc and Nluc on the cell surface. The compounds displayed little to no toxicity to cells and high luciferase specificity, showing no activity against firefly luciferase or even the closely related NanoBit system. Looking forward, the structural motifs used to gain access to the Nluc surface can also be appended with other functional groups, and therefore interesting opportunities for developing assays based on relief-of-inhibition can be envisioned.


Subject(s)
Cell Membrane Permeability , Enzyme Inhibitors/pharmacology , Luciferases/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Inhibitory Concentration 50 , Structure-Activity Relationship
13.
Angew Chem Int Ed Engl ; 55(31): 8899-903, 2016 07 25.
Article in English | MEDLINE | ID: mdl-27356091

ABSTRACT

Supramolecular split-enzyme complementation restores enzymatic activity and allows for on-off switching. Split-luciferase fragment pairs were provided with an N-terminal FGG sequence and screened for complementation through host-guest binding to cucurbit[8]uril (Q8). Split-luciferase heterocomplex formation was induced in a Q8 concentration dependent manner, resulting in a 20-fold upregulation of luciferase activity. Supramolecular split-luciferase complementation was fully reversible, as revealed by using two types of Q8 inhibitors. Competition studies with the weak-binding FGG peptide revealed a 300-fold enhanced stability for the formation of the ternary heterocomplex compared to binding of two of the same fragments to Q8. Stochiometric binding by the potent inhibitor memantine could be used for repeated cycling of luciferase activation and deactivation in conjunction with Q8, providing a versatile module for in vitro supramolecular signaling networks.


Subject(s)
Bridged-Ring Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Luciferases/antagonists & inhibitors , Bridged-Ring Compounds/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Imidazoles/chemistry , Luciferases/metabolism , Macromolecular Substances/chemistry , Macromolecular Substances/pharmacology , Models, Molecular , Structure-Activity Relationship
14.
PLoS One ; 11(3): e0150595, 2016.
Article in English | MEDLINE | ID: mdl-26937954

ABSTRACT

Eleven new furostanol saponins, typaspidosides B-L (1-11), one new spirostanol saponin, typaspidoside M (12), and five known spirostanol saponins, 25S-atropuroside (13), neoaspidistrin (14), (25S)-pratioside D1 (15), 25S-aspidistrin (16) and 25S-neosibiricoside (17) were isolated from the rhizomes of Aspidistra typica Baill. The structures of the new compounds were established using 1D and 2D NMR (1H-1H COSY, HMQC, HMBC and ROESY) spectroscopy, high resolution mass spectrometry, and chemical methods. The aglycones of 1-3 (unusual furostanol saponins with opened E ring type), 9 and 10 (the methoxyl substituent at C-23 position) were found, identified from natural products for the first time. Moreover, the anti-HIV activities of the isolated steroidal glycosides were assessed, and compounds 13, 14, 16 and 17 exhibited high active against HIV-1.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Liliaceae/chemistry , Reassortant Viruses/drug effects , Rhizome/chemistry , Saponins/pharmacology , Sterols/pharmacology , Anti-HIV Agents/isolation & purification , Capsid Proteins/antagonists & inhibitors , Capsid Proteins/genetics , Capsid Proteins/metabolism , Gene Expression , Genes, Reporter , HEK293 Cells , HIV-1/genetics , HIV-1/growth & development , Humans , Luciferases/antagonists & inhibitors , Luciferases/genetics , Luciferases/metabolism , Magnetic Resonance Spectroscopy , Plant Extracts/chemistry , Reassortant Viruses/genetics , Reassortant Viruses/growth & development , Saponins/isolation & purification , Spirostans/isolation & purification , Spirostans/pharmacology , Steroids/isolation & purification , Steroids/pharmacology , Sterols/isolation & purification , Structure-Activity Relationship , Vesiculovirus/genetics , Virus Replication/drug effects
15.
Nanoscale ; 8(7): 4007-19, 2016 Feb 21.
Article in English | MEDLINE | ID: mdl-26819069

ABSTRACT

A new general route for siRNA delivery is presented combining porous core-shell silica nanocarriers with a modularly designed multifunctional block copolymer. Specifically, the internal storage and release of siRNA from mesoporous silica nanoparticles (MSN) with orthogonal core-shell surface chemistry was investigated as a function of pore-size, pore morphology, surface properties and pH. Very high siRNA loading capacities of up to 380 µg per mg MSN were obtained with charge-matched amino-functionalized mesoporous cores, and release profiles show up to 80% siRNA elution after 24 h. We demonstrate that adsorption and desorption of siRNA is mainly driven by electrostatic interactions, which allow for high loading capacities even in medium-sized mesopores with pore diameters down to 4 nm in a stellate pore morphology. The negatively charged MSN shell enabled the association with a block copolymer containing positively charged artificial amino acids and oleic acid blocks, which acts simultaneously as capping and endosomal release agent. The potential of this multifunctional delivery platform is demonstrated by highly effective cell transfection and siRNA delivery into KB-cells. A luciferase reporter gene knock-down of up to 80-90% was possible using extremely low cell exposures with only 2.5 µg MSN containing 0.5 µg siRNA per 100 µL well.


Subject(s)
Nanoparticles/chemistry , Polymers/chemistry , RNA, Small Interfering/metabolism , Silicon Dioxide/chemistry , Adsorption , Carbocyanines/chemistry , Cell Line, Tumor , Electrophoresis, Gel, Pulsed-Field , Fluorescent Dyes/chemistry , Genes, Reporter , Humans , Luciferases/antagonists & inhibitors , Luciferases/genetics , Luciferases/metabolism , MCF-7 Cells , Microscopy, Confocal , Microscopy, Electron, Transmission , Porosity , RNA, Small Interfering/chemistry , Transfection
16.
J Biomol Screen ; 21(4): 325-32, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26676097

ABSTRACT

Glucose homeostasis is primarily controlled by two opposing hormones, insulin and glucagon, and diabetes results when insulin fails to inhibit glucagon action. Recent efforts to control glucagon in diabetes have focused on antagonizing the glucagon receptor, which is effective in lowering blood glucose levels but leads to hyperglucogonemia in rodents. An alternative strategy would be to control glucagon production with small molecules. In pursuit of this goal, we developed a homogeneous AlphaScreen assay for measuring glucagon in cell culture media and used this in a high-throughput screen to discover synthetic compounds that inhibited glucagon secretion from an alpha cell-like cell line. Some of these compounds inhibited transcription of the glucagon gene.


Subject(s)
Glucagon-Secreting Cells/drug effects , Glucagon/antagonists & inhibitors , Hypoglycemic Agents/pharmacology , Small Molecule Libraries/pharmacology , Animals , Biotin/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Cricetinae , Gene Expression , Genes, Reporter , Glucagon/biosynthesis , Glucagon/genetics , Glucagon-Secreting Cells/cytology , Glucagon-Secreting Cells/metabolism , High-Throughput Screening Assays , Humans , Hypoglycemic Agents/chemistry , Kinetics , Luciferases/antagonists & inhibitors , Luciferases/genetics , Luciferases/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Small Molecule Libraries/chemistry , Streptavidin/chemistry
17.
Nucleic Acids Res ; 44(2): 545-57, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26704983

ABSTRACT

Large dsRNA molecules can cause potent cytotoxic and immunostimulatory effects through the activation of pattern recognition receptors; however, synthetic versions of these molecules are mostly limited to simple sequences like poly-I:C and poly-A:U. Here we show that large RNA molecules generated by rolling circle transcription fold into periodic-shRNA (p-shRNA) structures and cause potent cytotoxicity and gene silencing when delivered to cancer cells. We determined structural requirements for the dumbbell templates used to synthesize p-shRNA, and showed that these molecules likely adopt a co-transcriptionally folded structure. The cytotoxicity of p-shRNA was robustly observed across four different cancer cell lines using two different delivery systems. Despite having a considerably different folded structure than conventional dsRNA, the cytotoxicity of p-shRNA was either equal to or substantially greater than that of poly-I:C depending on the delivery vehicle. Furthermore, p-shRNA caused greater NF-κB activation in SKOV3 cells compared to poly-I:C, indicating that it is a powerful activator of innate immunity. The tuneable sequence and combined gene silencing, immunostimulatory and cytotoxic capacity of p-shRNA make it an attractive platform for cancer immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , RNA Interference , RNA, Small Interfering/pharmacology , Antineoplastic Agents/immunology , Antineoplastic Agents/metabolism , Base Sequence , Caspase 3/genetics , Caspase 3/immunology , Caspase 7/genetics , Caspase 7/immunology , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Humans , Immunity, Innate , Luciferases/antagonists & inhibitors , Luciferases/genetics , Luciferases/immunology , Molecular Sequence Data , NF-kappa B/biosynthesis , NF-kappa B/metabolism , Nucleic Acid Conformation , Poly I-C/genetics , Poly I-C/immunology , Poly I-C/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Transcription, Genetic
18.
J Ovarian Res ; 8: 80, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26626440

ABSTRACT

BACKGROUND: Recent studies have shown that microRNAs may regulate the ABCB1 gene (ATP-binding cassette, sub-family B [MDR/TAP], member 1). Computational programs have predicted that the 3'-untranslated region (3'-UTR) of ABCB1 contains a potential miRNA-binding site for miR-186. Here, we investigated the role of miR-186 in sensitizing ovarian cancer cells to paclitaxel and cisplatin. RESULTS: Human ovarian carcinoma cell lines OVCAR3, A2780, A2780/DDP, and A2780/Taxol were exposed to paclitaxel or cisplatin with or without miR-186 transfection, and cell viability was determined by MTT assay. Reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot analysis were used to assess the MDR1, GST-π, and MRP1 expression levels. Dual-luciferase reporter assay was used to reveal the correlation between miR-186 and ABCB1. Lower miR-186 while higher MDR1 and GST-π mRNA expression levels were found in the A2780/Taxol and A2780/DDP cells than in the A2780 cells. After miR-186 transfection, all the cell lines showed increased sensitivity to paclitaxel and cisplatin. MiR-186 transfection induced apoptosis while anti-miR-186 transfection reduced apoptosis. The dual-luciferase reporter assay verified that that miR-186 combined with the 3'-untranslated region (UTR) of ABCB1. MDR1 and GST-π mRNA and protein expression levels were downregulated after transfection with miR-186 but upregulated following anti-miR-186 transfection compared to the mock and negative control cancer cells; however, the MRP1 expression levels did not significantly differ among the groups. CONCLUSION: Our results are the first to demonstrate that miR-186 may sensitize ovarian cancer cell to paclitaxel and cisplatin by targeting ABCB1 and modulating the expression of GST-π.


Subject(s)
Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , MicroRNAs/physiology , Ovarian Neoplasms/drug therapy , Paclitaxel/therapeutic use , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Down-Regulation/physiology , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Female , Glutathione Transferase/metabolism , Humans , Luciferases/antagonists & inhibitors , MicroRNAs/metabolism , Transfection
19.
J Med Chem ; 58(4): 1717-35, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25680029

ABSTRACT

WNT signaling is frequently deregulated in malignancy, particularly in colon cancer, and plays a key role in the generation and maintenance of cancer stem cells. We report the discovery and optimization of a 3,4,5-trisubstituted pyridine 9 using a high-throughput cell-based reporter assay of WNT pathway activity. We demonstrate a twisted conformation about the pyridine-piperidine bond of 9 by small-molecule X-ray crystallography. Medicinal chemistry optimization to maintain this twisted conformation, cognisant of physicochemical properties likely to maintain good cell permeability, led to 74 (CCT251545), a potent small-molecule inhibitor of WNT signaling with good oral pharmacokinetics. We demonstrate inhibition of WNT pathway activity in a solid human tumor xenograft model with evidence for tumor growth inhibition following oral dosing. This work provides a successful example of hypothesis-driven medicinal chemistry optimization from a singleton hit against a cell-based pathway assay without knowledge of the biochemical target.


Subject(s)
Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Drug Evaluation, Preclinical/methods , Luciferases/antagonists & inhibitors , Pyridines/pharmacology , Small Molecule Libraries/pharmacology , Spiro Compounds/pharmacology , Wnt Signaling Pathway/drug effects , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Biological Assay/methods , Biological Availability , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Crystallography, X-Ray , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Luciferases/metabolism , Mice , Models, Molecular , Molecular Structure , Pyridines/administration & dosage , Pyridines/chemistry , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , Spiro Compounds/administration & dosage , Spiro Compounds/chemistry , Structure-Activity Relationship , Xenograft Model Antitumor Assays
20.
Bioorg Med Chem ; 22(24): 6776-80, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25468041

ABSTRACT

α-Aminoisobutyric acid (Aib)-containing peptide analogs derived from TV-XIIa, a cell-penetrating peptide (CPP), were synthesized to explore structure-activity relationships. The replacement of Aib at position 1, 5, or 9 in the TV-XIIa amino acid sequence with alanine (Ala) suppressed the cellular uptake,whereas the simultaneous substitution of the two proline (Pro) residues at positions 6 and 10 with Aib(P-IV) considerably increased the cellular uptake. In order to explore the potential use of the Aib-containing peptide analogs for the cellular delivery of oligonucleotides (ODNs), we synthesized a covalent conjugate (P-IV-AON) of a 15-mer antisense ODN, which is complementary to luciferase gene, with P-IV, and the antisense effect of the P-IV-AON conjugate on luciferase expression in A549 cells was examined. Luciferase expression was decreased in the presence of the conjugate upon treatment with the reaction buffer at the concentrations of 5 and 10 µM.


Subject(s)
Aminoisobutyric Acids/chemistry , Cell-Penetrating Peptides/metabolism , Oligonucleotides/metabolism , Peptides/metabolism , Amino Acid Sequence , Cell Line, Tumor , Cell-Penetrating Peptides/chemical synthesis , Cell-Penetrating Peptides/chemistry , Genes, Reporter , Humans , Luciferases/antagonists & inhibitors , Luciferases/genetics , Luciferases/metabolism , Oligonucleotides/chemistry , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/metabolism , Peptides/chemical synthesis , Peptides/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...