Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
J Enzyme Inhib Med Chem ; 38(1): 2236802, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37470394

ABSTRACT

Natural α-glucosidase inhibitors from plant-based foods such as catechins offer an attractive strategy for their potential anti-diabetic effects. In this study, infusions of three different tea types (green, white, and oolong) were investigated for their total phenolic (TPC) and catechins (EGCG, ECG, EGC, and EC) content, and for their α-glucosidase inhibitory activities. We observed that the level of TPC in white tea was significantly higher compared to oolong and green tea, which suggests higher content of EGCG and ECG catechins in fresh young leaves. Our findings showed that the higher content of such catechins in the infusion of white tea well correlated with a strong inhibition of α-glucosidase, and such inhibition was demonstrated to be more effective than the FDA-approved drug acarbose. Then, we computationally explored the molecular requirements for enzyme inhibition, especially for the most active catechins EGCG and ECG, as well as their disposition/stability within the active site.


Subject(s)
Catechin , Glycoside Hydrolase Inhibitors , alpha-Glucosidases , Catechin/chemistry , Catechin/pharmacology , Phenols , Plant Extracts/chemistry , Tea/chemistry , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/pharmacology
2.
Int J Mol Sci ; 17(8)2016 Aug 20.
Article in English | MEDLINE | ID: mdl-27556447

ABSTRACT

The HIV-1 ribonuclease H (RNase H) function of the reverse transcriptase (RT) enzyme catalyzes the selective hydrolysis of the RNA strand of the RNA:DNA heteroduplex replication intermediate, and represents a suitable target for drug development. A particularly attractive approach is constituted by the interference with the RNase H metal-dependent catalytic activity, which resides in the active site located at the C-terminus p66 subunit of RT. Herein, we report results of an in-house screening campaign that allowed us to identify 4-[4-(aryl)-1H-1,2,3-triazol-1-yl]benzenesulfonamides, prepared by the "click chemistry" approach, as novel potential HIV-1 RNase H inhibitors. Three compounds (9d, 10c, and 10d) demonstrated a selective inhibitory activity against the HIV-1 RNase H enzyme at micromolar concentrations. Drug-likeness, predicted by the calculation of a panel of physicochemical and ADME properties, putative binding modes for the active compounds, assessed by computational molecular docking, as well as a mechanistic hypothesis for this novel chemotype are reported.


Subject(s)
HIV Reverse Transcriptase/antagonists & inhibitors , Reverse Transcriptase Inhibitors/chemistry , Sulfonamides/chemistry , Sulfonamides/pharmacology , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Molecular Docking Simulation , Reverse Transcriptase Inhibitors/pharmacology , Benzenesulfonamides
3.
Mol Pharmacol ; 87(2): 323-37, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25477342

ABSTRACT

The influenza virus PA endonuclease, which cleaves capped cellular pre-mRNAs to prime viral mRNA synthesis, is a promising target for novel anti-influenza virus therapeutics. The catalytic center of this enzyme resides in the N-terminal part of PA (PA-Nter) and contains two (or possibly one or three) Mg(2+) or Mn(2+) ions, which are critical for its catalytic function. There is great interest in PA inhibitors that are optimally designed to occupy the active site and chelate the metal ions. We focused here on a series of ß-diketo acid (DKA) and DKA-bioisosteric compounds containing different scaffolds, and determined their structure-activity relationship in an enzymatic assay with PA-Nter, in order to build a three-dimensional pharmacophore model. In addition, we developed a molecular beacon (MB)-based PA-Nter assay that enabled us to compare the inhibition of Mn(2+) versus Mg(2+), the latter probably being the biologically relevant cofactor. This real-time MB assay allowed us to measure the enzyme kinetics of PA-Nter or perform high-throughput screening. Several DKA derivatives were found to cause strong inhibition of PA-Nter, with IC50 values comparable to that of the prototype L-742,001 (i.e., below 2 µM). Among the different compounds tested, L-742,001 appeared unique in having equal activity against either Mg(2+) or Mn(2+). Three compounds ( 10: , with a pyrrole scaffold, and 40: and 41: , with an indole scaffold) exhibited moderate antiviral activity in cell culture (EC99 values 64-95 µM) and were proven to affect viral RNA synthesis. Our approach of integrating complementary enzymatic, cellular, and mechanistic assays should guide ongoing development of improved influenza virus PA inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Chelating Agents/pharmacology , Drug Discovery/methods , Endonucleases/antagonists & inhibitors , Orthomyxoviridae/drug effects , Orthomyxoviridae/enzymology , Animals , Antiviral Agents/chemistry , Chelating Agents/chemistry , Dogs , Endonucleases/metabolism , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Molecular Conformation
4.
J Enzyme Inhib Med Chem ; 30(3): 466-71, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25198885

ABSTRACT

The protein arginine deiminase 4 (PAD4) is a calcium-dependent enzyme, which catalyses the irreversible conversion of peptidyl-arginines into peptidyl-citrullines and plays an important role in several diseases such as in the rheumatoid arthritis, multiple sclerosis, Alzheimer's disease, Creutzfeldt-Jacob's disease and cancer. In this study, we report the inhibition profiles and computational docking toward the PAD4 enzyme of a series of 1,2,3-triazole peptidomimetic-based derivatives incorporating the ß-phenylalanine and guanidine scaffolds. Several effective, low micromolar PAD4 inhibitors are reported in this study.


Subject(s)
Enzyme Inhibitors/pharmacology , Hydrolases/antagonists & inhibitors , Peptidomimetics/pharmacology , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Hydrolases/metabolism , Molecular Structure , Peptidomimetics/chemical synthesis , Peptidomimetics/chemistry , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases , Structure-Activity Relationship
5.
J Virol ; 87(19): 10524-38, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23824822

ABSTRACT

The influenza virus PA endonuclease, which cleaves capped host pre-mRNAs to initiate synthesis of viral mRNA, is a prime target for antiviral therapy. The diketo acid compound L-742,001 was previously identified as a potent inhibitor of the influenza virus endonuclease reaction, but information on its precise binding mode to PA or potential resistance profile is limited. Computer-assisted docking of L-742,001 into the crystal structure of inhibitor-free N-terminal PA (PA-Nter) indicated a binding orientation distinct from that seen in a recent crystallographic study with L-742,001-bound PA-Nter (R. M. DuBois et al., PLoS Pathog. 8:e1002830, 2012). A comprehensive mutational analysis was performed to determine which amino acid changes within the catalytic center of PA or its surrounding hydrophobic pockets alter the antiviral sensitivity to L-742,001 in cell culture. Marked (up to 20-fold) resistance to L-742,001 was observed for the H41A, I120T, and G81F/V/T mutant forms of PA. Two- to 3-fold resistance was seen for the T20A, L42T, and V122T mutants, and the R124Q and Y130A mutants were 3-fold more sensitive to L-742,001. Several mutations situated at noncatalytic sites in PA had no or only marginal impact on the enzymatic functionality of viral ribonucleoprotein complexes reconstituted in cell culture, consistent with the less conserved nature of these PA residues. Our data provide relevant insights into the binding mode of L-742,001 in the PA endonuclease active site. In addition, we predict some potential resistance sites that should be taken into account during optimization of PA endonuclease inhibitors toward tight binding in any of the hydrophobic pockets surrounding the catalytic center of the enzyme.


Subject(s)
Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hydroxybutyrates/pharmacology , Mutation/genetics , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae/drug effects , Piperidines/pharmacology , RNA-Dependent RNA Polymerase/genetics , Viral Proteins/genetics , Animals , Catalytic Domain , Cells, Cultured , Computational Biology , Dogs , Drug Resistance, Viral , Humans , Molecular Structure , Orthomyxoviridae/enzymology , Orthomyxoviridae/genetics , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/virology , Protein Conformation , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Virus Internalization/drug effects , Virus Replication
6.
Bioorg Med Chem Lett ; 22(18): 5801-6, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22901388

ABSTRACT

The members of a focused series of carboxylic acids and of their derivatives (esters, amides and metal complexes) have been investigated as inhibitors of the main cytosolic/transmembrane carbonic anhydrase isoforms, CA I, II, IX and XII, belonging to the mammalian α-class of CAs. These enzymes are present in red blood cells in submillimolar concentration, and typical sulfonamide CA inhibitors do not selectively inhibit any of them. Among such isozymes, the isoform-I is an 'orphan' target that mediates hemorrhagic retinal and cerebral vascular permeability, involved in retinal and cerebral disease. In the present study, we identified the first selective CA I nanomolar inhibitors, that displayed activity against other isozymes in micromolar/millimolar concentration range. Selective CA II over CA I inhibition has also been observed with some diketo acids/metal complexes. Few diketo acid derivatives showed inhibition activities against the fungal ß-class enzymes from Candida albicans and Cryptococcus neoformans in low micromolar concentration range. Prediction of drug-like properties for the most interesting compounds suggests a favorable bioavailability.


Subject(s)
Candida albicans/enzymology , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Carboxylic Acids/pharmacology , Cryptococcus neoformans/enzymology , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Carboxylic Acids/chemical synthesis , Carboxylic Acids/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship
7.
Mol Pharm ; 8(2): 507-19, 2011 Apr 04.
Article in English | MEDLINE | ID: mdl-21323359

ABSTRACT

Most active and selective strand transfer HIV-1 integrase (IN) inhibitors contain chelating functional groups that are crucial feature for the inhibition of the catalytic activities of the enzyme. In particular, diketo acids and their derivatives can coordinate one or two metal ions within the catalytic core of the enzyme. The present work is intended as a contribution to elucidate the mechanism of action of the HIV-IN inhibitors by studying the coordinative features of H2L¹ (L-708,906), an important member of the diketo acids family of inhibitors, and H2L2, a model for S-1360, another potent IN inhibitor. Magnesium(II) and manganese(II) complexes of H2L¹ and H2L² were isolated and fully characterized in solution and in the solid state. The crystal structures of the manganese complex [Mn(HL2)2(CH3OH)2]·2CH3OH were solved by X-ray diffraction analysis. Moreover, the speciation models for H2L2 with magnesium(II) and manganese(II) ions were performed and the formation constants of the complexes were measured. M(HL2)2 (M = Mg²+, Mn²+) was the most abundant species in solution at physiological pH. All the synthesized compounds were tested for their anti-IN activity, showing good results both for the ligand and the corresponding complexes. From analysis of the speciation models and of the biological data we can conclude that coordination of both metal cofactors could not be strictly necessary and that inhibitors can act as complexes and not only as free ligands.


Subject(s)
Chelating Agents/metabolism , HIV Integrase Inhibitors/metabolism , HIV Integrase/chemistry , Magnesium/metabolism , Manganese/metabolism , Organometallic Compounds/metabolism , Chelating Agents/chemistry , Crystallography, X-Ray , HIV Integrase Inhibitors/chemistry , Humans , Magnesium/chemistry , Manganese/chemistry , Models, Molecular , Molecular Structure , Organometallic Compounds/chemistry , Stereoisomerism
8.
Bioorg Med Chem Lett ; 21(8): 2515-20, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21420862

ABSTRACT

Combinated ligand- and pharmacophore-based virtual screening approaches were used to discover novel potential pharmacophores acting as carbonic anhydrase (CA, EC 4.2.1.1) inhibitors (CAIs). A free database of commercially available compounds was screened through drug-like filters using a four-point pharmacophore, and followed by docking calculation within the active site of an X-ray structure of isoform CA II. One compound, bearing a trifluoro-dihydroxy-propanone moiety, showed an interesting, selective inhibitory activity in low micromolar range against this isoform versus CA I. The chemical originality of this new pharmacophore can represent an important bioisosteric alternative to the sulfonamido-based functionalities, thus leading to the development of a new class of CAIs.


Subject(s)
Carbonic Anhydrase II/antagonists & inhibitors , Carbonic Anhydrase Inhibitors/chemistry , Carbonic Anhydrase I/antagonists & inhibitors , Carbonic Anhydrase I/metabolism , Carbonic Anhydrase II/metabolism , Carbonic Anhydrase Inhibitors/pharmacology , Catalytic Domain , Computer Simulation , Databases, Factual , Humans , Structure-Activity Relationship , Sulfonamides/chemistry , Sulfonamides/pharmacology
9.
ACS Med Chem Lett ; 11(10): 1986-1992, 2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33062183

ABSTRACT

Photoaffinity labeling (PAL) is one of the upcoming and powerful tools in the field of molecular recognition. It includes the determination of dynamic parameters, such as the identification and localization of the target protein and the site of drug binding. In this study, a photoaffinity-labeled probe for full-length human immunodeficiency virus-1 integrase (HIV-1 IN) capture was designed and synthesized, following the structure of the FDA-approved drug Raltegravir. This photoprobe was found to retain the HIV IN inhibitory potential in comparison with its parent molecule and demonstrates the ability to label the HIV-1 IN protein. Putative photoprobe/inhibitor binding sites near the catalytic site were then identified after protein digestion coupled to mass and molecular modeling analyses.

10.
ACS Med Chem Lett ; 11(5): 857-861, 2020 May 14.
Article in English | MEDLINE | ID: mdl-32435396

ABSTRACT

Gold nanoparticles (GNPs) have been proposed as carriers for drugs to improve their intrinsic therapeutic activities and to overcome pharmacokinetic problems. In this study, novel nanosystems constituted by a model ß-diketo acid (DKA) grafted to the surface of GNPs were designed and synthesized following the "multivalent high-affinity" binding strategy. These first nanoscale DKA prototypes showed improved inhibition of HIV-1 integrase (HIV-1 IN) catalytic activities as compared with free DKA ligands.

11.
Antiviral Res ; 174: 104671, 2020 02.
Article in English | MEDLINE | ID: mdl-31812637

ABSTRACT

The management of Human Immunodeficiency Virus type 1 (HIV-1) infection requires life-long treatment that is associated with chronic toxicity and possible selection of drug-resistant strains. A new opportunity for drug intervention is offered by antivirals that act as allosteric inhibitors targeting two viral functions (dual inhibitors). In this work, we investigated the effects of 5,6-dihydroxyindole-2-carboxylic acid (DHICA) derivatives on both HIV-1 Integrase (IN) and Reverse Transcriptase associated Ribonuclease H (RNase H) activities. Among the tested compounds, the dihydroxyindole-carboxamide 5 was able to inhibit in the low micromolar range (1-18 µM) multiple functions of IN, including functional IN-IN interactions, IN-LEDGF/p75 binding and IN catalytic activity. Docking and site-directed mutagenesis studies have suggested that compound 5 binds to a previously described HIV-1 IN allosteric pocket. These observations indicate that 5 is structurally and mechanistically distinct from the published allosteric HIV-1 IN inhibitors. Moreover, compound 5 also inhibited HIV-1 RNase H function, classifying this molecule as a dual HIV-1 IN and RNase H inhibitor able to impair the HIV-1 virus replication in cell culture. Overall, we identified a new scaffold as a suitable platform for the development of novel dual HIV-1 inhibitors.


Subject(s)
Carboxylic Acids/pharmacology , HIV Integrase Inhibitors/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Ribonuclease H, Human Immunodeficiency Virus/antagonists & inhibitors , Carboxylic Acids/chemistry , Cell Line , Drug Discovery , HIV Infections/virology , HIV Integrase/metabolism , HIV Integrase Inhibitors/chemistry , Humans , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship
12.
Bioorg Med Chem ; 17(7): 2925-35, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19026554

ABSTRACT

Previously, we discovered linomide analogues as novel HIV-1 integrase (IN) inhibitors. Here, to make possible structure-activity relationships, we report on the design and synthesis of a series of substituted dihydroquinoline-3-carboxylic acids. The crystal structure of the representative compound 2c has also been solved. Among the eight new analogues, 2e showed a potency in inhibiting IN strand transfer catalytic activity similar to the reference diketo acid inhibitor L-731,988 (IC(50)=0.9 microM vs. 0.54 microM, for 2e and L-731,988, respectively). Furthermore, none of the compounds showed significant cytotoxicity in two tested cancer cell lines. These compounds represent an interesting prototype of IN inhibitors, potentially involved in a metal chelating mechanism, and further optimization is warranted.


Subject(s)
Carboxylic Acids/chemical synthesis , HIV Integrase Inhibitors/chemical synthesis , HIV Integrase/chemistry , Quinolines/chemical synthesis , Carboxylic Acids/chemistry , Carboxylic Acids/toxicity , Cell Line, Tumor , Crystallography, X-Ray , Drug Design , HIV Integrase/metabolism , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/toxicity , Humans , Inhibitory Concentration 50 , Molecular Conformation , Quinolines/chemistry , Quinolines/toxicity , Structure-Activity Relationship
13.
Molecules ; 13(10): 2442-61, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18830166

ABSTRACT

HIV-1 integrase (IN) is an attractive and validated target for the development of novel therapeutics against AIDS. In the search for new IN inhibitors, we designed and synthesized three series of bis-amide and hydrazide-containing derivatives of malonic acid. We performed a docking study to investigate the potential interactions of the title compounds with essential amino acids on the IN active site.


Subject(s)
HIV Integrase Inhibitors/chemistry , Malonates/chemical synthesis , Malonates/pharmacology , Amides , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Azides , Catalytic Domain , Computer Simulation , Drug Design , HIV Integrase Inhibitors/pharmacology , Humans , Malonates/chemistry , Protein Binding , Structure-Activity Relationship
14.
Front Microbiol ; 8: 440, 2017.
Article in English | MEDLINE | ID: mdl-28373864

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1) infection, still represent a serious global health emergency. The chronic toxicity derived from the current anti-retroviral therapy limits the prolonged use of several antiretroviral agents, continuously requiring the discovery of new antiviral agents with innovative strategies of action. In particular, the development of single molecules targeting two proteins (dual inhibitors) is one of the current main goals in drug discovery. In this contest, metal-chelating molecules have been extensively explored as potential inhibitors of viral metal-dependent enzymes, resulting in some important classes of antiviral agents. Inhibition of HIV Integrase (IN) is, in this sense, paradigmatic. HIV-1 IN and Reverse Transcriptase-associated Ribonuclease H (RNase H) active sites show structural homologies, with the presence of two Mg(II) cofactors, hence it seems possible to inhibit both enzymes by means of chelating ligands with analogous structural features. Here we present a series of N'-acylhydrazone ligands with groups able to chelate the Mg(II) hard Lewis acid ions in the active sites of both the enzymes, resulting in dual inhibitors with micromolar and even nanomolar activities. The most interesting identified N'-acylhydrazone analog, compound 18, shows dual RNase H-IN inhibition and it is also able to inhibit viral replication in cell-based antiviral assays in the low micromolar range. Computational modeling studies were also conducted to explore the binding attitudes of some model ligands within the active site of both the enzymes.

15.
ACS Med Chem Lett ; 8(9): 941-946, 2017 Sep 14.
Article in English | MEDLINE | ID: mdl-28947941

ABSTRACT

We report the synthesis, biological evaluation, and structural study of a series of substituted heteroaryl-pyrazole carboxylic acid derivatives. These compounds have been developed as inhibitors of specific isoforms of carbonic anhydrase (CA), with potential as prototypes of a new class of chemotherapeutics. Both X-ray crystallography and computational modeling provide insights into the CA inhibition mechanism. Results indicate that this chemotype produces an indirect interference with the zinc ion, thus behaving differently from other related nonclassical inhibitors. Among the tested compounds, 2c with Ki = 0.21 µM toward hCA XII demonstrated significant antiproliferative activity against hypoxic tumor cell lines. Taken together, the results thus provide the basis of structural determinants for the development of novel anticancer agents.

16.
Mater Sci Eng C Mater Biol Appl ; 68: 594-602, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27524059

ABSTRACT

The bioactive flavonoid fisetin (FS) is a diet-derived antioxidant that is being increasingly investigated for its health-promoting effects. Unfortunately, the poor physicochemical and pharmacokinetic properties affect and limit the clinical application. In this study, novel polymeric nanoparticles (NPs), based on Poly-(ε-caprolactone) (PCL) and PLGA-PEG-COOH, encapsulating FS were formulated as suitable oral controlled release systems. Results showed NPs having a mean diameter of 140-200nm, and a percent loading of FS ranging from 70 to 82%. In vitro release studies revealed that NPs are able to protect and preserve the release of FS in gastric simulated conditions, also controlling the release in the intestinal medium. Moreover, the DPPH and ABTS scavenging capacity of FS, as well as α-glucosidase inhibition activity, that resulted about 20-fold higher than commercial Acarbose, were retained during nanoencapsulation process. In summary, our developed NPs can be proposed as an attractive delivery system to control the release of antioxidant and anti-hyperglycemic FS for nutraceutical and/or therapeutic application.


Subject(s)
Antioxidants , Dietary Supplements , Flavonoids , Glycoside Hydrolase Inhibitors , Nanocapsules/chemistry , Antioxidants/chemistry , Antioxidants/pharmacokinetics , Flavonoids/chemistry , Flavonoids/pharmacokinetics , Flavonols , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/pharmacokinetics , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , alpha-Glucosidases/chemistry , alpha-Glucosidases/metabolism
17.
J Med Chem ; 59(11): 5209-20, 2016 06 09.
Article in English | MEDLINE | ID: mdl-27139920

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with poor prognosis and limited therapeutic options. Therefore, there is an urgent need to identify new, safe, and targeted therapeutics for effective treatment of late as well as early stage disease. Plectin-1 (Plec-1) was recently identified as specific biomarker for detecting PDAC at an early stage. We envisioned that multivalent attachment of nanocarriers incorporating certain drugs to Plec-1-derived peptide would increase specific binding affinity and impart high specificity for PDAC cells. Previously, we discovered a novel class of compounds (e.g., quinazolinediones, QDs) that exert their cytotoxic effects by modulating ROS-mediated cell signaling. Herein, we prepared novel QD242-encapsulated polymeric nanoparticles (NPs) functionalized with a peptide to selectively bind to Plec-1. Similarly, we prepared QD-based NPs densely decorated with an isatoic anhydride derivative. Furthermore, we evaluated their impact on ligand binding and antiproliferative activity against PDAC cells. The targeted NPs were more potent than the nontargeted constructs in PDAC cells warranting further development.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Drug Delivery Systems , Nanoparticles/chemistry , Pancreatic Neoplasms/drug therapy , Quinazolinones/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Carriers/chemistry , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Pancreatic Neoplasms/pathology , Quinazolinones/chemical synthesis , Quinazolinones/chemistry , Structure-Activity Relationship
18.
Int J Nanomedicine ; 10: 6835-46, 2015.
Article in English | MEDLINE | ID: mdl-26586945

ABSTRACT

Celastrol (CL), a triterpenoid extracted from the Chinese herb Tripterygium wilfordii, has recently attracted interest for its potential antitumor effects. However, unfavorable physicochemical and pharmacokinetics properties such as low solubility, poor bioavailability, and systemic toxicity, are limiting its therapeutic application. In this context, the development of innovative nanocarriers can be useful to overcome these issues, and nanoencapsulation would represent a powerful strategy. In this study, we developed novel CL-loaded poly(ε-caprolactone) nanoparticles (NPs), and investigated their antiproliferative efficacy on prostate cancer cells. CL-NPs were prepared using a nanoprecipitation method and fully characterized by physicochemical techniques. The antiproliferative effects on LNCaP, DU-145, and PC3 cell lines of CL-NPs, compared to those of free CL at different concentrations (0.5, 1.0, and 2.0 µM), were investigated. Moreover, fluorescence microscopy was utilized to examine the cellular uptake of the nanosystems. Furthermore, to elucidate impact of nanoencapsulation on the mechanism of action, Western analyses were conducted to explore apoptosis, migration, proliferation, and angiogenesis alteration of prostate cancer cells. The results confirmed that CL-NPs inhibit proliferation dose dependently in all prostate cancer cells, with inhibitory concentration50 less than 2 µM. In particular, the NPs significantly increased cytotoxicity at lower/medium dose (0.5 and 1.0 µM) on DU145 and PC3 cell lines with respect to free CL, with modulation of apoptotic and cell cycle machinery proteins. To date, this represents the first report on the development of biocompatible polymeric NPs encapsulating CL. Our findings offer new perspectives for the exploitation of developed CL-NPs as suitable prototypes for prostate cancer treatment.


Subject(s)
Nanoparticles/chemistry , Prostatic Neoplasms/drug therapy , Triterpenes/therapeutic use , Apoptosis/drug effects , Calorimetry, Differential Scanning , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Endocytosis/drug effects , Humans , Male , Microscopy, Confocal , Nanoparticles/ultrastructure , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Particle Size , Pentacyclic Triterpenes , Prostatic Neoplasms/blood supply , Spectroscopy, Fourier Transform Infrared , Spectrum Analysis, Raman , Static Electricity , Triterpenes/chemistry , Triterpenes/pharmacology
19.
Int J Biol Macromol ; 72: 531-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25220789

ABSTRACT

The polyphenols as nutraceutical and therapeutic agents are gaining growing interest for their beneficial effects and potential in human health. In order to protect their scaffolds and functionality, and to improve the bioavailability, the microencapsulation can represent a promising strategy. This study reports on the formulation of the natural resveratrol (RSV) into microcapsules (MCs) prepared by using different concentrations of chitosan (CS) and poly(D,L-lactic-co-glycolic acid) (PLGA) as polymeric matrix. MCs were prepared by W/O/W double emulsion method and characterized in terms of morphology, size, encapsulation efficiency, physicochemical and thermal properties. RSV release behavior from MCs was evaluated under simulated gastrointestinal fluids, and the long term stability was monitored at different storage conditions. MCs resulted to have spherical shape and different morphology, with size ranging from 11 to 20 µm, and encapsulation efficiencies of 40-52%, depending on the CS concentration. Moreover, MCs containing CS exhibited a significant lower release of RSV than those containing only PLGA. Furthermore, all tested formulations were able to ensure a good retention and stability of encapsulated RSV until 6 months. In summary, CS/PLGA MCs can be proposed as an attractive delivery system to control the release and long term protection of RSV.


Subject(s)
Chitosan/pharmacology , Drug Delivery Systems , Lactic Acid/pharmacology , Stilbenes/chemistry , Capsules/chemistry , Chitosan/chemistry , Delayed-Action Preparations , Humans , Lactic Acid/chemistry , Particle Size , Polyglycolic Acid/chemistry , Polyglycolic Acid/pharmacology , Polylactic Acid-Polyglycolic Acid Copolymer , Resveratrol , Stilbenes/pharmacology
20.
J Agric Food Chem ; 63(7): 2026-32, 2015 Feb 25.
Article in English | MEDLINE | ID: mdl-25599125

ABSTRACT

With the aim to obtain controlled release and to preserve the antioxidant activity of the polyphenols, nanoencapsulation of white tea extract into polymeric nanoparticles (NPs) based on poly(ε-caprolactone) (PCL) and alginate was successfully performed. NPs were prepared by nanoprecipitation method and were characterized in terms of morphology and chemical properties. Total polyphenols and catechins contents before and after encapsulation were determined. Moreover, in vitro release profiles of encapsulated polyphenols from NPs were investigated in simulated gastrointestinal fluids. The antioxidant activity and stability of encapsulated extract were further evaluated. Interestingly, NPs released 20% of the polyphenols in simulated gastric medium, and 80% after 5 h at pH 7.4, showing a good capacity to control the polyphenols delivery. Furthermore, DPPH(•) assay confirmed that white tea extract retained its antioxidant activity and NPs protected tea polyphenols from degradation, thus opening new perspectives for the exploitation of white tea extract-loaded NPs for nutraceutical applications.


Subject(s)
Camellia sinensis/chemistry , Dietary Supplements/analysis , Drug Carriers/chemistry , Nanoparticles/chemistry , Plant Extracts/chemistry , Polyesters/chemistry , Polyphenols/chemistry , Alginates/chemistry , Drug Delivery Systems , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Particle Size
SELECTION OF CITATIONS
SEARCH DETAIL