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










Publication year range
1.
Sci Rep ; 9(1): 9398, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31253836

ABSTRACT

The bovine lactoferricin L6 (RRWQWR) has been previously identified as a novel cell-penetrating peptide (CPP) that is able to efficiently internalize into human cells. L6 interacts with quantum dots (QDs) noncovalently to generate stable L6/QD complexes that enter cells by endocytosis. In this study, we demonstrate a modified L6 (HL6; CHHHHHRRWQWRHHHHHC), in which short polyhistidine peptides are introduced into both flanks of L6, has enhanced cell-penetrating ability in human bronchoalveolar carcinoma A549 cells. The mechanism of cellular uptake of HL6/QD complexes is primarily direct membrane translocation rather than endocytosis. Dimethyl sulfoxide (DMSO), but not pyrenebutyrate (PB), ethanol, oleic acid, or 1,2-benzisothiazol-3(2 H)-one (BIT), slightly enhances HL6-mediated protein transduction efficiency. Neither HL6 nor HL6/QD complexes are cytotoxic to A549 or HeLa cells. These results indicate that HL6 could be a more efficient drug carrier than L6 for biomedical as well as biotechnological applications, and that the function of polyhistidine peptides is critical to CPP-mediated protein transduction.


Subject(s)
Cell Membrane/drug effects , Cell Membrane/metabolism , Cell-Penetrating Peptides/metabolism , Histidine , Amino Acid Sequence , Cell Line, Tumor , Cell-Penetrating Peptides/chemistry , Drug Carriers/chemistry , Drug Delivery Systems , Histidine/administration & dosage , Histidine/chemistry , Humans
2.
J Nanosci Nanotechnol ; 19(2): 613-621, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30360131

ABSTRACT

Cell-penetrating peptides (CPPs) containing a preponderance of basic amino acids are able to deliver biologically active macromolecules and nanomaterials into live cells. Quantum dots (QDs) are nanoparticles with unique fluorescence properties that have found wide application in biomedical imaging. In this study, we demonstrate transduction of an L6 CPP (RRWQWR) derived from bovine lactoferricin (LFcin) into human lung cancer cells. L6 noncovalently interacts with QDs to form stable complexes. L6/QD complexes enter cells most efficiently when prepared at a nitrogen/phosphate ratio of 60. Mechanistic studies indicate that L6/QD complexes enter cells by endocytosis. Treatment with 1,2-benzisothiazol-3(2H)-one (BIT), an industrial preservative that enhances uptake of certain CPPs, does not affect L6 CPP-mediated protein transduction efficiency. L6 and L6/QD complexes are not cytotoxic. These results indicate that L6 LFcin might be an efficient and safe nanoshuttle for nanoparticles or nanomedicines in biomedical applications.


Subject(s)
Cell-Penetrating Peptides , Nanoparticles , Quantum Dots , Animals , Cattle , Cell-Penetrating Peptides/metabolism , Cell-Penetrating Peptides/pharmacology , Endocytosis , Humans , Lactoferrin , Nanoparticles/toxicity
3.
Curr Pharm Biotechnol ; 18(7): 569-584, 2017.
Article in English | MEDLINE | ID: mdl-28828981

ABSTRACT

BACKGROUND: Development of effective drug delivery systems (DDS) is a critical issue in health care and medicine. Advances in molecular biology and nanotechnology have allowed the introduction of nanomaterial-based drug delivery systems. Cell-penetrating peptides (CPPs) can form the basis of drug delivery systems by virtue of their ability to support the transport of cargoes into the cell. Potential cargoes include proteins, DNA, RNA, liposomes, and nanomaterials. These cargoes generally retain their bioactivities upon entering cells. METHOD: In the present study, the smallest, fully-active lactoferricin-derived CPP, L5a is used to demonstrate the primary contributor of cellular internalization. RESULTS: The secondary helical structure of L5a encompasses symmetrical positive charges around the periphery. The contributions of cell-specificity, peptide length, concentration, zeta potential, particle size, and spatial structure of the peptides were examined, but only zeta potential and spatial structure affected protein transduction efficiency. FITC-labeled L5a appeared to enter cells via direct membrane translocation insofar as endocytic modulators did not block FITC-L5a entry. This is the same mechanism of protein transduction active in Cy5 labeled DNA delivery mediated by FITC-L5a. A significant reduction of transduction efficiency was observed with structurally incomplete FITC-L5a formed by tryptic destruction, in which case the mechanism of internalization switched to a classical energydependent endocytosis pathway. CONCLUSION: These results support the continued development of the non-cytotoxic L5a as an efficient tool for drug delivery.


Subject(s)
Cell-Penetrating Peptides/metabolism , Drug Delivery Systems/methods , Endocytosis/physiology , Nanoparticles/metabolism , Amino Acid Sequence , Cell Line, Tumor , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/genetics , Flow Cytometry , Fluorescein-5-isothiocyanate , Humans , Lactoferrin/chemistry , Liposomes/pharmacology , Microscopy, Confocal , Microscopy, Fluorescence , Nanoparticles/chemistry , Particle Size , Plasmids , Surface Properties
4.
PLoS One ; 11(3): e0150439, 2016.
Article in English | MEDLINE | ID: mdl-26942714

ABSTRACT

Cell-penetrating peptides (CPPs) have been shown to deliver cargos, including protein, DNA, RNA, and nanomaterials, in fully active forms into live cells. Most of the CPP sequences in use today are based on non-native proteins that may be immunogenic. Here we demonstrate that the L5a CPP (RRWQW) from bovine lactoferricin (LFcin), stably and noncovalently complexed with plasmid DNA and prepared at an optimal nitrogen/phosphate ratio of 12, is able to efficiently enter into human lung cancer A549 cells. The L5a CPP delivered a plasmid containing the enhanced green fluorescent protein (EGFP) coding sequence that was subsequently expressed in cells, as revealed by real-time PCR and fluorescent microscopy at the mRNA and protein levels, respectively. Treatment with calcium chloride increased the level of gene expression, without affecting CPP-mediated transfection efficiency. Zeta-potential analysis revealed that positively electrostatic interactions of CPP/DNA complexes correlated with CPP-mediated transport. The L5a and L5a/DNA complexes were not cytotoxic. This biomimetic LFcin L5a represents one of the shortest effective CPPs and could be a promising lead peptide with less immunogenic for DNA delivery in gene therapy.


Subject(s)
Cell-Penetrating Peptides/metabolism , DNA/metabolism , Gene Transfer Techniques , Lactoferrin/chemistry , Amino Acid Sequence , Animals , Calcium Chloride/pharmacology , Cattle , Cell Line, Tumor , Cell-Penetrating Peptides/chemistry , Endocytosis/drug effects , Fluorescein-5-isothiocyanate/metabolism , Genes, Reporter , Humans , Microscopy, Fluorescence , Molecular Sequence Data , Particle Size , Plasmids/metabolism , Real-Time Polymerase Chain Reaction , Static Electricity , Transfection
5.
J Nanosci Nanotechnol ; 15(3): 2067-78, 2015 Mar.
Article in English | MEDLINE | ID: mdl-26413622

ABSTRACT

Nanoparticles, such as semiconductor quantum dots (QDs), have been found increasing use in biomedical diagnosis and therapeutics because of their unique properties, including quantum confinement, surface plasmon resonance, and superparamagnetism. Cell-penetrating peptides (CPPs) represent an efficient mechanism to overcome plasma membrane barriers and deliver biologically active molecules into cells. In this study, we demonstrate that three arginine-rich CPPs (SR9, HR9, and PR9) can noncovalently complex with red light emitting QDs, dramatically increasing their deliv- ery into living cells. Zeta-potential and size analyses highlight the importance of electrostatic interactions between positive-charged CPP/QD complexes and negative-charged plasma membranes indicating the efficiency of transmembrane complex transport. Subcellular colocalization indicates associations of QD with early endosomes and lysosomes following PR9-mediated delivery. Our study demonstrates that nontoxic CPPs of varied composition provide an effective vehicle for the design of optimized drug delivery systems.


Subject(s)
Arginine , Cell-Penetrating Peptides/chemistry , Quantum Dots/chemistry , Quantum Dots/metabolism , Biological Transport , Cell Line, Tumor , Color , Humans , Intracellular Space/metabolism
6.
PLoS One ; 10(9): e0138463, 2015.
Article in English | MEDLINE | ID: mdl-26384023

ABSTRACT

The purpose of this study was to evaluate the extent of constitutive activity among orphan class-A G protein coupled receptors within the cAMP signaling pathway. Constitutive signaling was revealed by changes in gene expression under control of the cAMP response element. Gene expression was measured in Chinese hamster ovary cells transiently co-transfected with plasmids containing a luciferase reporter and orphan receptor. Criteria adopted for defining constitutive activation were: 1) 200% elevation over baseline reporter gene expression; 2) 40% inhibition of baseline expression; and 3) 40% inhibition of expression stimulated by 3 µM forskolin. Five patterns of activity were noted: 1) inhibition under both baseline and forskolin stimulated expression (GPR15, GPR17, GPR18, GPR20, GPR25, GPR27, GPR31, GPR32, GPR45, GPR57, GPR68, GPR83, GPR84, GPR132, GPR150, GPR176); 2) no effect on baseline expression, but inhibition of forskolin stimulated expression (GPR4, GPR26, GPR61, GPR62, GPR78, GPR101, GPR119); 3) elevation of baseline signaling coupled with inhibition of forskolin stimulated expression (GPR6, GPR12); 4) elevation of baseline signaling without inhibition of forskolin stimulated expression (GPR3, GPR21, GPR52, GPR65); and 5) no effect on expression (GPR1, GPR19, GPR22, GPR34, GPR35, GPR39, GPR63, GPR82, GPR85, GPR87). Constitutive activity was observed in 75% of the orphan class-A receptors examined (30 of 40). This constitutive signaling cannot be explained by simple overexpression of the receptor. Inhibition of cAMP mediated expression was far more common (65%) than stimulation of expression (15%). Orphan receptors that were closely related based on amino acid homology tended to have similar effects on gene expression. These results suggest that identification of inverse agonists may be a fruitful approach for categorizing these orphan receptors and targeting them for pharmacological intervention.


Subject(s)
Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Animals , CHO Cells , Cell Line , Colforsin/pharmacology , Cricetulus , Cyclic AMP/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology
7.
Biomed Res Int ; 2015: 834079, 2015.
Article in English | MEDLINE | ID: mdl-25883975

ABSTRACT

Many viral and nonviral systems have been developed to aid delivery of biologically active molecules into cells. Among these, cell-penetrating peptides (CPPs) have received increasing attention in the past two decades for biomedical applications. In this review, we focus on opportunities and challenges associated with CPP delivery of nucleic acids and nanomaterials. We first describe the nature of versatile CPPs and their interactions with various types of cargoes. We then discuss in vivo and in vitro delivery of nucleic acids and nanomaterials by CPPs. Studies on the mechanisms of cellular entry and limitations in the methods used are detailed.


Subject(s)
Cell-Penetrating Peptides/pharmacology , Drug Delivery Systems/methods , Nanostructures , Nucleic Acids/pharmacology , Animals , Humans
8.
J Membr Biol ; 248(2): 355-68, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25655108

ABSTRACT

Bacterial and archaeal cell envelopes are complex multilayered barriers that serve to protect these microorganisms from their extremely harsh and often hostile environments. Import of exogenous proteins and nanoparticles into cells is important for biotechnological applications in prokaryotes. In this report, we demonstrate that cell-penetrating peptides (CPPs), both bacteria-expressed nona-arginine peptide (R9) and synthetic R9 (SR9), are able to deliver noncovalently associated proteins or quantum dots into four representative species of prokaryotes: cyanobacteria (Synechocystis sp. PCC 6803), bacteria (Escherichia coli DH5α and Arthrobacter ilicis D-50), and archaea (Thermus aquaticus). Although energy-dependent endocytosis is generally accepted as a hallmark that distinguishes eukaryotes from prokaryotes, cellular uptake of uncomplexed green fluorescent protein (GFP) by cyanobacteria was mediated by classical endocytosis. Mechanistic studies revealed that macropinocytosis plays a critical and major role in CPP-mediated protein transduction in all four prokaryotes. Membrane damage was not observed when cyanobacterial cells were treated with R9/GFP complexes, nor was cytotoxicity detected when bacteria or archaea were treated with SR9/QD complexes in the presence of macropinocytic inhibitors. These results indicate that the uptake of protein is not due to a compromise of membrane integrity in cyanobacteria, and that CPP can be an effective and safe carrier for membrane trafficking in prokaryotic cells. Our investigation provides important new insights into the transport of exogenous proteins and nanoparticles across the complex membrane systems of prokaryotes.


Subject(s)
Cell-Penetrating Peptides/metabolism , Endocytosis , Prokaryotic Cells/physiology , Archaea/metabolism , Bacteria/metabolism , Cell Membrane/metabolism , Cell-Penetrating Peptides/genetics , Cell-Penetrating Peptides/toxicity , Microscopy, Fluorescence , Permeability , Protein Transport
9.
Curr Pharm Biotechnol ; 15(3): 267-75, 2014.
Article in English | MEDLINE | ID: mdl-24938892

ABSTRACT

Cellular and nuclear delivery of biomolecules is limited by low membrane permeability. Cell-penetrating peptides (CPPs) can be covalently linked to cargos to improve cellular internalization. Our work indicates that arginine-rich CPPs are also able to interact with a variety of cargos, including DNA, RNA, proteins and nanomaterials, in a noncovalent manner and subsequently effect their delivery into cells. The advantages of noncovalent attachment in CPP-mediated transduction are multiple: ease of use, ease of production, and versatility with respect to both cargo composition and functional delivery (i.e., the cargo is not chemically modified). We have extended this approach to achieve simultaneous transduction of covalently and noncovalently associated complexes, opening a new method for delivering multiple types of cargos, including proteins, fluorescent nanomaterials, nucleic acid and others. These novel variations of CPP-mediated transport should be of broad utility in the transport of genes, small interfering RNAs, proteins and nanoparticles in biomedical research and therapeutic intervention.


Subject(s)
Cell-Penetrating Peptides/pharmacokinetics , Drug Delivery Systems , Animals , Biological Transport , Cell-Penetrating Peptides/chemistry , DNA/administration & dosage , DNA/pharmacokinetics , Humans , Nanostructures/administration & dosage , Proteins/administration & dosage , Proteins/pharmacokinetics , RNA/administration & dosage , RNA/pharmacokinetics
10.
Chem Biol Interact ; 206(2): 319-26, 2013 Nov 25.
Article in English | MEDLINE | ID: mdl-24120544

ABSTRACT

A clear understanding of physicochemical factors governing nanoparticle toxicity is still in its infancy. We used a systematic approach to delineate physicochemical properties of nanoparticles that govern cytotoxicity. The cytotoxicity of fourth period metal oxide nanoparticles (NPs): TiO2, Cr2O3, Mn2O3, Fe2O3, NiO, CuO, and ZnO increases with the atomic number of the transition metal oxide. This trend was not cell-type specific, as observed in non-transformed human lung cells (BEAS-2B) and human bronchoalveolar carcinoma-derived cells (A549). Addition of NPs to the cell culture medium did not significantly alter pH. Physiochemical properties were assessed to discover the determinants of cytotoxicity: (1) point-of-zero charge (PZC) (i.e., isoelectric point) described the surface charge of NPs in cytosolic and lysosomal compartments; (2) relative number of available binding sites on the NP surface quantified by X-ray photoelectron spectroscopy was used to estimate the probability of biomolecular interactions on the particle surface; (3) band-gap energy measurements to predict electron abstraction from NPs which might lead to oxidative stress and subsequent cell death; and (4) ion dissolution. Our results indicate that cytotoxicity is a function of particle surface charge, the relative number of available surface binding sites, and metal ion dissolution from NPs. These findings provide a physicochemical basis for both risk assessment and the design of safer nanomaterials.


Subject(s)
Apoptosis/drug effects , Metal Nanoparticles/toxicity , Transition Elements/chemistry , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Humans , Hydrogen-Ion Concentration , Metal Nanoparticles/chemistry , Oxides/chemistry , Surface Properties
11.
J Biomed Sci ; 20: 48, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23844974

ABSTRACT

BACKGROUND: Oxidative stress increases the cytosolic content of calcium in the cytoplasm through a combination of effects on calcium pumps, exchangers, channels and binding proteins. In this study, oxidative stress was produced by exposure to tert-butyl hydroperoxide (tBHP); cell viability was assessed using a dye reduction assay; receptor binding was characterized using [3H]N-methylscopolamine ([3H]MS); and cytosolic and luminal endoplasmic reticulum (ER) calcium concentrations ([Ca2+]i and [Ca2+]L, respectively) were measured by fluorescent imaging. RESULTS: Activation of M3 muscarinic receptors induced a biphasic increase in [Ca2+]i: an initial, inositol trisphosphate (IP3)-mediated release of Ca2+ from endoplasmic reticulum (ER) stores followed by a sustained phase of Ca2+ entry (i.e., store-operated calcium entry; SOCE). Under non-cytotoxic conditions, tBHP increased resting [Ca2+]i; a 90 minute exposure to tBHP (0.5-10 mM ) increased [Ca2+]i from 26 to up to 127 nM and decreased [Ca2+]L by 55%. The initial response to 10 µM carbamylcholine was depressed by tBHP in the absence, but not the presence, of extracellular calcium. SOCE, however, was depressed in both the presence and absence of extracellular calcium. Acute exposure to tBHP did not block calcium influx through open SOCE channels. Activation of SOCE following thapsigargin-induced depletion of ER calcium was depressed by tBHP exposure. In calcium-free media, tBHP depressed both SOCE and the extent of thapsigargin-induced release of Ca2+ from the ER. M3 receptor binding parameters (ligand affinity, guanine nucleotide sensitivity, allosteric modulation) were not affected by exposure to tBHP. CONCLUSIONS: Oxidative stress induced by tBHP affected several aspects of M3 receptor signaling pathway in CHO cells, including resting [Ca2+]i, [Ca2+]L, IP3 receptor mediated release of calcium from the ER, and calcium entry through the SOCE. tBHP had little effect on M3 receptor binding or G protein coupling. Thus, oxidative stress affects multiple aspects of calcium homeostasis and calcium dependent signaling.


Subject(s)
Calcium Signaling/genetics , Endoplasmic Reticulum/metabolism , Ion Transport/genetics , Oxidative Stress/drug effects , Animals , CHO Cells , Calcium/metabolism , Carrier Proteins/metabolism , Cell Survival/genetics , Cricetinae , Cricetulus , Cytoplasm/metabolism , Endoplasmic Reticulum/drug effects , Signal Transduction/genetics , tert-Butylhydroperoxide/toxicity
12.
PLoS One ; 8(6): e67100, 2013.
Article in English | MEDLINE | ID: mdl-23840594

ABSTRACT

Cell-penetrating peptides (CPPs) can traverse cellular membranes and deliver biologically active molecules into cells. In this study, we demonstrate that CPPs comprised of nona-arginine (R9) and a penetration accelerating peptide sequence (Pas) that facilitates escape from endocytic lysosomes, denoted as PR9, greatly enhance the delivery of noncovalently associated quantum dots (QDs) into human A549 cells. Mechanistic studies, intracellular trafficking analysis and a functional gene assay reveal that endocytosis is the main route for intracellular delivery of PR9/QD complexes. Endocytic trafficking of PR9/QD complexes was monitored using both confocal and transmission electron microscopy (TEM). Zeta-potential and size analyses indicate the importance of electrostatic forces in the interaction of PR9/QD complexes with plasma membranes. Circular dichroism (CD) spectroscopy reveals that the secondary structural elements of PR9 have similar conformations in aqueous buffer at pH 7 and 5. This study of nontoxic PR9 provides a basis for the design of optimized cargo delivery that allows escape from endocytic vesicles.


Subject(s)
Arginine/chemistry , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/metabolism , Drug Carriers/chemistry , Drug Carriers/metabolism , Endocytosis , Nanoparticles , A549 Cells , Actins/metabolism , Amino Acid Sequence , Humans , Intracellular Space/metabolism , Kinetics , Lysosomes/metabolism , Nanoparticles/chemistry , Particle Size , Protein Transport , Quantum Dots/chemistry
13.
PLoS One ; 8(5): e64205, 2013.
Article in English | MEDLINE | ID: mdl-23724035

ABSTRACT

Cell-penetrating peptides (CPPs) comprised of basic amino residues are able to cross cytoplasmic membranes and are able to deliver biologically active molecules inside cells. However, CPP/cargo entrapment in endosome limits biomedical utility as cargoes are destroyed in the acidic environment. In this study, we demonstrate protein transduction of a novel CPP comprised of an INF7 fusion peptide and nona-arginine (designated IR9). IR9 noncovalently interacts with quantum dots (QDs) and DNAs to form stable IR9/QD and IR9/DNA complexes which are capable of entering human A549 cells. Zeta-potentials were a better predictor of transduction efficiency than gel shift analysis, emphasizing the importance of electrostatic interactions of CPP/cargo complexes with plasma membranes. Mechanistic studies revealed that IR9, IR9/QD and IR9/DNA complexes may enter cells by endocytosis. Further, IR9, IR9/QD and IR9/DNA complexes were not cytotoxic at concentrations below 30, 5 and 20.1 µM, respectively. Without labor intensive production of fusion proteins from prokaryotes, these results indicate that IR9 could be a safe carrier of genes and drugs in biomedical applications.


Subject(s)
Cell-Penetrating Peptides/pharmacology , DNA/metabolism , Drug Delivery Systems , Intracellular Space/metabolism , Nanoparticles/chemistry , Cell Death/drug effects , Endocytosis/drug effects , Flow Cytometry , Humans , Intracellular Space/drug effects , Microscopy, Confocal , Microscopy, Fluorescence , Plasmids/metabolism , Quantum Dots , Static Electricity
14.
Colloids Surf B Biointerfaces ; 111: 162-70, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23792556

ABSTRACT

Semiconductor nanoparticles, also known as quantum dots (QDs), are widely used in biomedical imaging studies and pharmaceutical research. Cell-penetrating peptides (CPPs) are a group of small peptides that are able to traverse cell membrane and deliver a variety of cargoes into living cells. CPPs deliver QDs into cells with minimal nonspecific absorption and toxic effect. In this study, water-soluble, monodisperse, carboxyl-functionalized indium phosphide (InP)/zinc sulfide (ZnS) QDs coated with polyethylene glycol lipids (designated QInP) were synthesized for the first time. The physicochemical properties (optical absorption, fluorescence and charging state) and cellular internalization of QInP and CPP/QInP complexes were characterized. CPPs noncovalently interact with QInP in vitro to form stable CPP/QInP complexes, which can then efficiently deliver QInP into human A549 cells. The introduction of 500nM of CPP/QInP complexes and QInP at concentrations of less than 1µM did not reduce cell viability. These results indicate that carboxylated and polyethylene-glycolylated (PEGylated) bifunctionalized QInP are biocompatible nanoparticles with potential for use in biomedical imaging studies and drug delivery applications.


Subject(s)
Cell-Penetrating Peptides/chemistry , Endocytosis , Phosphines/chemical synthesis , Polyethylene Glycols/chemical synthesis , Quantum Dots/chemistry , Sulfides/chemical synthesis , Zinc Compounds/chemical synthesis , Cell Line, Tumor , Endocytosis/drug effects , Humans , Indium/chemistry , Intracellular Space/chemistry , Intracellular Space/drug effects , Phosphines/chemistry , Polyethylene Glycols/chemistry , Quantum Dots/toxicity , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Sulfides/chemistry , Sulfides/toxicity , Zinc Compounds/chemistry , Zinc Compounds/toxicity
15.
J Biomed Sci ; 20: 11, 2013 Feb 23.
Article in English | MEDLINE | ID: mdl-23432810

ABSTRACT

BACKGROUND: Honokiol, a cell-permeable phenolic compound derived from the bark of magnolia trees and present in Asian herbal teas, has a unique array of pharmacological actions, including the inhibition of multiple autonomic responses. We determined the effects of honokiol on calcium signaling underlying transmission mediated by human M3 muscarinic receptors expressed in Chinese hamster ovary (CHO) cells. Receptor binding was determined in radiolabelled ligand binding assays; changes in intracellular calcium concentrations were determined using a fura-2 ratiometric imaging protocol; cytotoxicity was determined using a dye reduction assay. RESULTS: Honokiol had a potent (EC50 ≈ 5 µmol/l) inhibitory effect on store operated calcium entry (SOCE) that was induced by activation of the M3 receptors. This effect was specific, rapid and partially reversible, and was seen at concentrations not associated with cytotoxicity, inhibition of IP3 receptor-mediated calcium release, depletion of ER calcium stores, or disruption of M3 receptor binding. CONCLUSIONS: It is likely that an inhibition of SOCE contributes to honokiol disruption of parasympathetic motor functions, as well as many of its beneficial pharmacological properties.


Subject(s)
Biphenyl Compounds/administration & dosage , Calcium Signaling/drug effects , Calcium/metabolism , Lignans/administration & dosage , Receptor, Muscarinic M3/metabolism , Animals , CHO Cells , Cricetinae , Cytoplasm/drug effects , Cytoplasm/metabolism , Gene Expression Regulation/drug effects , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Ion Transport/drug effects
16.
J Nanosci Nanotechnol ; 10(10): 6534-43, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21137758

ABSTRACT

Protein transduction domains comprised of basic amino acid-rich peptides, can efficiently deliver covalently fused macromolecules into cells. Quantum dots (QDs) are luminescent semiconductor nanocrystals that are finding increasing application in biological imaging. Previous studies showed that protein transduction domains mediate the internalization of covalently attached QDs. In this study, we demonstrate that arginine-rich intracellular delivery peptides (cell-penetrating peptides; CPPs), analogs of naturally-occuring protein transduction domains, deliver noncovalently associated QDs into living cells; CPPs dramatically increase the rate and efficiency of cellular uptake of QD probes. The optimal molecular ratio between arginine-rich CPPs and QD cargoes for cellular internalization is approximately 60:1. Upon entry into cells, the QDs are concentrated in the perinuclear region. There is no cytotoxicity following transport of QDs present at concentrations up to 200 nM. The mechanism for arginine-rich CPP/QD complexes to traverse cell membrane appears to involve a combination of internalization pathways. These results provide insight into the mechanism of arginine-rich CPP delivery of noncovalently attached cargoes, and may provide a powerful tool for imaging in vivo.


Subject(s)
Cell-Penetrating Peptides/pharmacokinetics , Peptides/pharmacokinetics , Quantum Dots , Analysis of Variance , Cell Line, Tumor , Cell Survival/drug effects , Cell-Penetrating Peptides/chemistry , Electrophoretic Mobility Shift Assay , Endocytosis/drug effects , Humans , Particle Size , Peptides/chemistry , Protein Transport/drug effects , Spectrometry, Fluorescence
17.
Toxicol In Vitro ; 24(7): 1953-61, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20708676

ABSTRACT

The influences of ZnO nanoparticles on cellular responses to activation of muscarinic receptors were studied in Chinese hamster ovary cells expressing the human M3 muscarinic acetylcholine receptor. ZnO particles (20 nm) induced cytotoxicity in a time and concentration-dependent manner: following a 24h exposure, toxicity was minimal at concentrations below 20 µg/ml but virtually complete at concentrations above 28 µg/ml. ZnO particles did not affect antagonist binding to M3 receptors or allosteric ligand effects, but increased agonist binding affinity while eliminating guanine nucleotide sensitivity. At a noncytotoxic concentration (10 µg/ml), ZnO increased resting [Ca(2+)](i) from 40 to 130 nM without compromising calcium homeostatic mechanisms. ZnO particles had minimal effects on IP3- or thapsigargin-mediated release of intracellular calcium from the endoplasmic reticulum, but strongly inhibited store-operated calcium entry (capacitive calcium entry). The latter effect was seen as (1) a decrease in the plateau phase of the response and (2) a decrease in Ca(2+) entry upon introduction of calcium to the extracellular medium following thapsigargin-induced depletion of calcium from the endoplasmic reticulum (EC50's ≈ 2 µg/ml). Thus, ZnO nanoparticles interfere with two specific aspects of the M3 signaling pathway, agonist binding and store-operated calcium entry.


Subject(s)
Calcium/metabolism , Nanoparticles , Receptor, Muscarinic M3/metabolism , Zinc Oxide/toxicity , Animals , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Homeostasis/drug effects , Humans , Protein Binding/drug effects , Signal Transduction/drug effects , Thapsigargin/pharmacology , Time Factors , Zinc Oxide/administration & dosage
18.
Toxicol In Vitro ; 24(1): 45-55, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19755143

ABSTRACT

The influence of 20nm ZnO nanoparticles on cytotoxicity, oxidative stress, intracellular calcium homeostasis, and gene expression was studied in human bronchial epithelial cells (BEAS-2B). ZnO caused a concentration- and time-dependent cytotoxicity while elevating oxidative stress and causing membrane damage (cellular LDH release). There was a remarkably steep relationship between concentration and toxicity at concentrations from 5 to 10microg/ml. Cytotoxicity was completely abolished by the antioxidant N-acetylcysteine (NAC). Exposure to ZnO also increased intracellular calcium levels ([Ca(2+)](in)) in a concentration- and time-dependent manner that was partially attenuated by NAC. Nifedipine, a calcium channel blocker, partially attenuated the elevated [Ca(2+)](in), indicating that some of the excess [Ca(2+)](in) is a result of influx from outside the cell. The relationships between oxidative stress, [Ca(2+)](in), and cytotoxicity are discussed. Exposure to a sublethal concentration of ZnO increased the expression of four genes that are involved in apoptosis and oxidative stress responses BNIP, PRDX3, PRNP, and TXRND1, by at least 2.5-fold. Thus, ZnO alters transcriptional regulation in BEAS-2B cells.


Subject(s)
Calcium/metabolism , Epithelial Cells/metabolism , Gene Expression/drug effects , Lung/metabolism , Nanoparticles/toxicity , Oxidative Stress/drug effects , Zinc Oxide/toxicity , Acetylcysteine/pharmacology , Calcium Channel Blockers/pharmacology , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Free Radical Scavengers/pharmacology , Homeostasis/drug effects , Humans , L-Lactate Dehydrogenase/metabolism , Lung/cytology , Lung/drug effects , Nifedipine/pharmacology , Reactive Oxygen Species/metabolism
19.
Materials (Basel) ; 3(10): 4842-4859, 2010 Oct 25.
Article in English | MEDLINE | ID: mdl-28883356

ABSTRACT

Nanotechnology has evolved to play a prominent role in our economy. Increased use of nanomaterials poses potential human health risk. It is therefore critical to understand the nature and origin of the toxicity imposed by nanomaterials (nanotoxicity). In this article we review the toxicity of the transition metal oxides in the 4th period that are widely used in industry and biotechnology. Nanoparticle toxicity is compellingly related to oxidative stress and alteration of calcium homeostasis, gene expression, pro-inflammatory responses, and cellular signaling events. The precise physicochemical properties that dictate the toxicity of nanoparticles have yet to be defined, but may include element-specific surface catalytic activity (e.g., metallic, semiconducting properties), nanoparticle uptake, or nanoparticle dissolution. These in vitro studies substantially advance our understanding in mechanisms of toxicity, which may lead to safer design of nanomaterials.

20.
BMC Pharmacol ; 9: 7, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19368719

ABSTRACT

BACKGROUND: While doxorubicin (DOX) is widely used in cancer chemotherapy, long-term severe cardiotoxicity limits its use. This is the first report of the chemoprotective efficacy of a relatively new thiol antioxidant, N-acetylcysteine amide (NACA), on DOX-induced cell death in cardiomyocytes. We hypothesized that NACA would protect H9c2 cardiomyocytes from DOX-induced toxicity by reducing oxidative stress. Accordingly, we determined the ability of NACA to mitigate the cytotoxicity of DOX in H9c2 cells and correlated these effects with the production of indicators of oxidative stress. RESULTS: DOX at 5 microM induced cardiotoxicity while 1) increasing the generation of reactive oxygen species (ROS), 2) decreasing levels and activities of antioxidants and antioxidant enzymes (catalase, glutathione peroxidase, glutathione reductase) and 3) increasing lipid peroxidation. NACA at 750 microM substantially reduced the levels of ROS and lipid peroxidation, as well as increased both GSH level and GSH/GSSG ratio. However, treating H9c2 cells with NACA did little to protect H9c2 cells from DOX-induced cell death. CONCLUSION: Although NACA effectively reduced oxidative stress in DOX-treated H9c2 cells, it had minimal effects on DOX-induced cell death. NACA prevented oxidative stress by elevation of GSH and CYS, reduction of ROS and lipid peroxidation, and restoration of antioxidant enzyme activities. Further studies to identify oxidative stress-independent pathways that lead to DOX-induced cell death in H9c2 are warranted.


Subject(s)
Acetylcysteine/analogs & derivatives , Doxorubicin/toxicity , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Acetylcysteine/pharmacology , Animals , Antibiotics, Antineoplastic/toxicity , Catalase/metabolism , Cell Line , Cell Survival/drug effects , Cysteine/metabolism , Dose-Response Relationship, Drug , Glutathione/metabolism , Glutathione Disulfide/metabolism , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Lipid Peroxidation/drug effects , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...