Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 216
Filter
Add more filters

Publication year range
1.
Nano Lett ; 24(28): 8650-8657, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38949785

ABSTRACT

The ion permeability and selectivity of membranes are crucial in nanofluidic behavior, impacting industries ranging from traditional to advanced manufacturing. Herein, we demonstrate the engineering of ion-conductive membranes featuring angstrom-scale ion-transport channels by introducing ionic polyamidoamine (PAMAM) dendrimers for ion separation. The exterior quaternary ammonium-rich structure contributes to significant electrostatic charge exclusion due to enhanced local charge density; the interior protoplasmic channels of PAMAM dendrimer are assembled to provide additional degrees of free volume. This facilitates the monovalent ion transfer while maintaining continuity and efficient ion screening. The dendrimer-assembled hybrid membrane achieves high monovalent ion permeance of 2.81 mol m-2 h-1 (K+), reaching excellent mono/multivalent selectivity up to 20.1 (K+/Mg2+) and surpassing the permselectivities of state-of-the-art membranes. Both experimental results and simulating calculations suggest that the impressive ion selectivity arises from the significant disparity in transport energy barrier between mono/multivalent ions, induced by the "exterior-interior" synergistic effects of bifunctional membrane channels.

2.
Biochem Biophys Res Commun ; 705: 149729, 2024 04 23.
Article in English | MEDLINE | ID: mdl-38452515

ABSTRACT

Breast cancer resistance to chemotherapy necessitates novel combination therapeutic approaches. Linc-RoR is a long intergenic noncoding RNA that regulates stem cell differentiation and promotes metastasis and invasion in breast cancer. Herein, we report a dual delivery system employing polyamidoamine dendrimers to co-administer the natural compound curcumin and linc-RoR siRNA for breast cancer treatment. Polyamidoamine dendrimers efficiently encapsulated curcumin and formed complexes with linc-RoR siRNA at an optimal N/P ratio. In MCF-7 breast cancer cells, the dendriplexes were effectively internalized and the combination treatment synergistically enhanced cytotoxicity, arresting the cell cycle at the G1 phase and inducing apoptosis. Linc-RoR gene expression was also significantly downregulated. Individual treatments showed lower efficacy, indicating synergism between components. Mechanistic studies are warranted to define the molecular underpinnings of this synergistic interaction. Our findings suggest dual delivery of linc-RoR siRNA and curcumin via dendrimers merits further exploration as a personalized therapeutic approach for overcoming breast cancer resistance.


Subject(s)
Breast Neoplasms , Curcumin , Dendrimers , Polyamines , RNA, Long Noncoding , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , RNA, Small Interfering/genetics , Curcumin/pharmacology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Line, Tumor
3.
Int J Mol Sci ; 24(1)2023 Jan 03.
Article in English | MEDLINE | ID: mdl-36614260

ABSTRACT

Due to their unique structure, poly(amidoamine) (PAMAM) dendrimers can bind active ingredients in two ways: inside the structure or on their surface. The location of drug molecules significantly impacts the kinetics of active substance release and the mechanism of internalization into the cell. This study focuses on the effect of the protonation degree of the G4PAMAM dendrimer and the anticancer drug 5-fluorouracil (5FU) on the efficiency of complex formation. The most favorable conditions for constructing the G4PAMAM-5FU complex are a low degree of protonation of the dendrimer molecule with the drug simultaneously present in a deprotonated form. The fluorine components in the XPS spectra confirm the formation of the stable complex. Through SAXS and DLS methods, a decrease in the dendrimer's molecular size resulting from protonation changes at alkaline conditions was demonstrated. The gradual closure of the dendrimer structure observed at high pH values makes it difficult for the 5FU molecules to migrate to the interior of the support structure, thereby promoting drug immobilization on the surface. The 1H NMR and DOSY spectra indicate that electrostatic interactions determine the complex formation process. Through MD simulations, the localization profile and the number of 5FU molecules forming the complex were visualized on an atomic scale.


Subject(s)
Dendrimers , Fluorouracil , Dendrimers/chemistry , Scattering, Small Angle , X-Ray Diffraction
4.
Molecules ; 28(6)2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36985668

ABSTRACT

Camptothecin (CPT), an alkaloid with potent anticancer activity, is still not used in clinical practice due to its high hydrophobicity, toxicity, and poor active-form stability. To address these shortcomings, our research focuses on the encapsulation of this drug in the poly(amidoamine) (PAMAM) dendrimer macromolecule. The PAMAM dendrimer/CPT complex was synthesized and thoroughly characterized. The in vitro drug release study revealed that the drug was released in a slow and controlled manner in acidic and physiological conditions and that more than 80% of the drug was released after 168 h of incubation. Furthermore, it was demonstrated that CPT was released with first-order kinetics and non-Fickian transport. The studies on the hemolytic activity of the synthesized complex indicated that it is hemocompatible for potential intravenous administration at a concentration ≤ 5 µg/mL. Additionally, the developed product was shown to reduce the viability of non-small-cell lung cancer cells (A549) in a concentration- and time-dependent manner, and cancer cells were more susceptible to the complex than normal fibroblasts. Lastly, molecular modeling studies revealed that the lactone or carboxylic forms of CPT had a significant impact on the shape and stability of the complex and that its formation with the lactone form of CPT was more energetically favorable for each subsequent molecule than the carboxylic form. The report represents a systematic and structured approach to develop a PAMAM dendrimer/CPT complex that can be used as an effective drug delivery system (DDS) for the potential treatment of non-small-cell lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Dendrimers , Lung Neoplasms , Humans , Dendrimers/pharmacology , Cell Line , Camptothecin/pharmacology
5.
BMC Oral Health ; 23(1): 810, 2023 10 28.
Article in English | MEDLINE | ID: mdl-37898802

ABSTRACT

BACKGROUND: The purpose of this study was to evaluate remineralisation and its effect on microtensile bond-strength of artificially induced caries affected dentin (CAD) when treated with a commercial universal adhesive modified with poly(amidoamine) dendrimer (PAMAM) loaded mesoporous bioactive glass nanoparticles (A-PMBG). MATERIAL AND METHODS: Mesoporous bioactive glass nanoparticles (MBG) were synthesised using sol-gel process, where PAMAM was loaded (P-MBG) and added to commercial adhesive at different weight percentages (0.2, 0.5, 1 and 2 wt%). First, rheological properties of commercial and modified adhesives were evaluated. The effect of remineralization/hardness and microtensile bond-strength (MTBs) of those samples that mimicked the rheological properties of commercial adhesives were evaluated using Vickers hardness tester and universal testing machine respectively. Scanning-Electron microscope was used to visualize failed samples of MTBs and remineralization samples. Both evaluations were carried out at 1-,3 and 6-month intervals, samples being stored in stimulated salivary fluid during each time interval. RESULTS: Addition of nanoparticles altered the rheological properties. With increase in the weight percentage of nanoparticles in commercial adhesive, there was significant increase in degree of conversion, viscosity and sedimentation rate (p < 0.05). The 0.2 and 0.5 wgt% groups closely mimicked the properties of commercial adhesive and were evaluated for remineralization and MTBs. After 6 months, 0.2wgt% group showed increased MTBs (p < 0.05) and 0.5wgt% group increased remineralization/hardness (p < 0.05). CONCLUSION: The complex of PAMAM-MBG-Universal adhesive can remineralize the demineralised CAD thereby improving its bond-strength when evaluated for up to 6-months.


Subject(s)
Dental Bonding , Dental Caries , Nanoparticles , Humans , Dental Cements/therapeutic use , Dental Caries Susceptibility , Dentin , Nanoparticles/therapeutic use , Dental Caries/therapy , Tensile Strength , Materials Testing , Resin Cements/therapeutic use
6.
Mol Pharm ; 19(12): 4685-4695, 2022 12 05.
Article in English | MEDLINE | ID: mdl-36278815

ABSTRACT

Surface modification manipulates the application performance of materials, and thrombosis caused by material contact is a key risk factor of biomaterials failure in blood-contacting/implanting devices. Therefore, building a safe and effective hemocompatibility platform is still urgent. Owing to the unique properties of polyamidoamine (PAMAM) dendrimers, in this study, modified surfaces with varying dendrimer densities were interacted with elements maintaining blood homeostasis. These included the plasma proteins bovine serum albumin and fibrinogen, cells in blood (platelets and erythrocyte), as well as endothelial cells (ECs), and the objective was to evaluate the blood compatibility of the chosen materials. Whole blood test and dynamic blood circulation experiment by the arteriovenous shunt mode of rabbit were also conducted, based on the complexity and fluidity of blood. The PAMAM-modified substrates, particularly that with a high density of PAMAM (N1.0), adsorbed proteins with lessened fibrinogen adsorption, reduced platelet activation and aggregation, and suppressed clotting in whole blood and dynamic blood testing. Furthermore, the designed PAMAM dendrimer densities were safe and showed negligible erythrocyte lysis. Concurrently, PAMAM modification could maintain EC growth and did not trigger the release of procoagulant factors. These results suggest that the PAMAM-modified materials are compatible for maintaining blood homeostasis. Thus, PAMAM dendrimers can work as excellent surface modifiers for constructing a hemocompatibility platform and even a primer layer for desired functional design, promoting the service performance of blood-contacting devices.


Subject(s)
Dendrimers , Animals , Rabbits , Dendrimers/chemistry , Endothelial Cells/metabolism , Renal Dialysis , Fibrinogen
7.
Mol Pharm ; 19(7): 2573-2582, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35666687

ABSTRACT

The aim of this study was to develop a polyethylene glycol (PEG)-conjugated third-generation polyamidoamine dendrimer (PAMAM) with phosphorylated serine as an osteoid surface-targeting drug carrier for the treatment of bone diseases. We conjugated PAMAM backbones to l-serine and obtained Ser-PAMAM. Then, phosphoric acid and PEG were covalently bound to the Ser-PAMAM to generate PEGylated phosphorylated Ser-PAMAM (PEG-phosSer-PAMAM). Using osteoblast-like cells (MC3T3-E1 cells) cultured in 3D collagen gels, we showed that phosSer-PAMAM adsorbed both the hydroxyapatite and type I collagen components of the bone matrix. Fourier transform infrared spectroscopy analysis indicated that the phosphoryl side chains of phosSer-PAMAM formed electrostatic interactions and hydrogen bonds with the anionic amino acid residues of type I collagen. Mice were intravenously injected with the foregoing molecules, and a tissue distribution study disclosed that the lower limb bone took up about twice as much 111In-labeled PEG-phosSer-PAMAM as 111In-labeled nonphosphorylated PEG-Ser-PAMAM or unmodified PAMAM. An intrabone distribution experiment showed that fluorescein isothiocyanate (FITC)-labeled PEG-phosSer-PAMAM accumulated on the osteoid surfaces, which is associated with bone pathogenesis such as skeletal dysplasias and osteoporosis to a far greater extent than nonphosphorylated PEG-Ser-PAMAM. Our findings indicated that PEG-phosSer-PAMAM is a promising carrier for efficient drug targeting to osteoid surfaces.


Subject(s)
Dendrimers , Drug Carriers , Animals , Bone Matrix , Collagen Type I , Dendrimers/chemistry , Drug Carriers/chemistry , Mice , Polyamines , Polyethylene Glycols/chemistry , Serine
8.
J Nanobiotechnology ; 20(1): 43, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35062953

ABSTRACT

BACKGROUND: Chemodynamic therapy is a promising cancer treatment with specific therapeutic effect at tumor sites, as toxic hydroxyl radical (·OH) could only be generated by Fenton or Fenton-like reaction in the tumor microenvironment (TME) with low pH and high level of endogenous hydrogen peroxide. However, the low concentration of catalytic metal ions, excessive glutathione (GSH) and aggressive hypoxia at tumor site seriously restrict the curative outcomes of conventional chemodynamic therapy. RESULTS: In this study, polyethylene glycol-phenylboronic acid (PEG-PBA)-modified generation 5 (G5) poly(amidoamine) (PAMAM) dendrimers were synthesized as a targeted nanocarrier to chelate Cu(II) and then encapsulate hypoxia-sensitive drug tirapazamine (TPZ) by the formation of hydrophobic Cu(II)/TPZ complex for hypoxia-enhanced chemo/chemodynamic therapy. The formed G5.NHAc-PEG-PBA@Cu(II)/TPZ (GPPCT) nanoplatform has good stability and hemocompatibility, and could release Cu(II) ions and TPZ quickly in weakly acidic tumor sites via pH-sensitive dissociation of Cu(II)/TPZ. In vitro experiments showed that the GPPCT nanoplatforms can efficiently target murine breast cancer cells (4T1) cells overexpressing sialic acid residues, and show a significantly enhanced inhibitory effect on hypoxic cells by the activation of TPZ. The excessive GSH in tumors could be depleted by the reduction of Cu(II) to Cu(I), and abundant of toxic ·OH would be generated in tumor cells by Fenton reaction for chemodynamic therapy. In vivo experiments demonstrated that the GPPCT nanoplatform could specifically accumulate at tumors, effectively inhibit the growth and metastasis of tumors by the combination of CDT and chemotherapy, and be metabolized with no systemic toxicity. CONCLUSIONS: The targeted GPPCT nanoplatform may represent an effective model for the synergistic inhibition of different tumor types by hypoxia-enhanced chemo/chemodynamic therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Hypoxia/drug effects , Dendrimers , Nanostructures/chemistry , Tumor Microenvironment/drug effects , Animals , Dendrimers/chemistry , Dendrimers/pharmacology , Mice , Tirapazamine/pharmacology
9.
Int J Mol Sci ; 23(24)2022 Dec 17.
Article in English | MEDLINE | ID: mdl-36555742

ABSTRACT

Enzyme immobilization is a powerful strategy for enzyme stabilization and recyclability. Materials covered with multipoint molecules are very attractive for this goal, since the number of active moieties to attach the enzyme increases with respect to monofunctional linkers. This work evaluates different dendrimers supported on silica to immobilize a protease enzyme, Alcalase. Five different dendrimers were employed: two carbosilane (CBS) dendrimers of different generations (SiO2-G0Si-NH2 and SiO2-G1Si-NH2), a CBS dendrimer with a polyphenoxo core (SiO2-G1O3-NH2), and two commercial polyamidoamine (PAMAM) dendrimers of different generations (SiO2-G0PAMAM-NH2 and SiO2-G1PAMAM-NH2). The results were compared with a silica support modified with a monofunctional molecule (2-aminoethanethiol). The effect of the dendrimer generation, the immobilization conditions (immobilization time, Alcalase/SiO2 ratio, and presence of Ca2+ ions), and the digestion conditions (temperature, time, amount of support, and stirring speed) on Alcalase activity has been evaluated. Enzyme immobilization and its activity were highly affected by the kind of dendrimer and its generation, observing the most favorable behavior with SiO2-G0PAMAM-NH2. The enzyme immobilized on this support was used in two consecutive digestions and, unlike CBS supports, it did not retain peptides released in the digestion.


Subject(s)
Dendrimers , Dendrimers/chemistry , Silicon Dioxide/chemistry , Enzymes, Immobilized/chemistry
10.
AAPS PharmSciTech ; 23(7): 236, 2022 Aug 24.
Article in English | MEDLINE | ID: mdl-36002713

ABSTRACT

Piperine (PIP) is a neuroprotective phytomedicine that has profound acetylcholine esterase and reactive oxygen species inhibition effect in Alzheimer's disease (AD) model. However, the oral delivery of PIP is limited by poor aqueous solubility and low bioavailability in systemic circulation. To improve the PIP bioavailability, the polyamidoamine (PAMAM) G4 dendrimer is grafted with tocopheryl polyethylene glycol succinate-1000 (TPGS) through carbodiimide chemistry to form TPGS-PAMAM conjugate. The TPGS-PAMAM coupling was confirmed through proton NMR and FTIR techniques. PIP was encapsulated in the TPGS-PAMAM through solvent diffusion method to form PIP-TPGS-PAMAM. The particle size for PIP-TPGS-PAMAM found the less than 50 nm, whereas entrapment efficiency found to 87 ± 3.5% and 10.6 ± 2.9% drug loading. The powder differential scanning calorimetry and powder X-ray diffraction characterization were employed to evaluate the amorphous encapsulation of the PIP in TPGS-PAMAM. The PIP-TPGS-PAMAM stability was studied in the gastric fluids which showed no drastic difference in particle size and encapsulation efficiency compared to PIP-PAMAM. The in vitro release analysis revealed 37 ± 4.1% PIP release from the PIP-TPGS-PAMAM matrix, and 71 ± 4.9% PIP release from the PIP-PAMAM dendrimer was observed in 48 h. The single-dose oral gavage to Wistar rats of PIP-TPGS-PAMAM showed the AUC0-∞ 14.38 µg/mL.h, Cmax 7.77 ± 1.65 µg/mL, Tmax, 1.6 ± 0.18 h, and half-life 3.47 ± 0.64 h for PIP in systemic circulation. PIP-PAMAM and free PIP showed significantly poor AUC0-∞ compared to PIP-TPGS-PAMAM. The brain uptake studies revealed PIP-TPGS-PAMAM treated group showed 2.2 ± 0.37 µg/g PIP content compared to free PIP administered group which was 0.4 ± 0.10 µg/g. Therefore, PIP-TPGS-PAMAM can offer excellent prospect for the delivery hydrophobic drugs to brain in AD.


Subject(s)
Dendrimers , Alkaloids , Animals , Benzodioxoles , Brain , Dendrimers/chemistry , Drug Carriers/chemistry , Particle Size , Piperidines , Polyamines , Polyethylene Glycols/chemistry , Polyunsaturated Alkamides , Powders , Rats , Rats, Wistar , Succinates , Succinic Acid , Vitamin E/chemistry
11.
J Labelled Comp Radiopharm ; 64(11): 428-439, 2021 09.
Article in English | MEDLINE | ID: mdl-34330148

ABSTRACT

Amplification pretargeting has the potential to increase the tracer's accumulation in the tumor. This study aimed to develop a three-step amplification pretargeting strategy in nuclear medicine with a polymer conjugated with multiple copies of peptide nuclear acid (PNA). In this study, the tracer 18 F-labeled complementary PNA (18 F-cPNA) was prepared by click-chemistry with high radiochemical purity (>99%) and great stability in vitro. The PAMMA dendrimer generation 4 (G4) was conjugated with multiple copies of PNAs. The average number of PNA groups in the G4-PNA conjugate was determined by matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF MS) and the accessibility to the 18 F-cPNA was identified by size-exclusion high-performance liquid chromatography (SE-HPLC). There were approximately 11.7 of 64 carboxyl groups modified with PNAs, of which more than 99% were accessible to 18 F-cPNA. 18 F-cPNA was added to a mixture of CC49-cPNA and G4-PNA, and the complex exhibited a single peak on high-performance liquid chromatography (HPLC) as evidence of complete hybridization between 18 F-cPNA and CC49-cPNA/G4-PNA. The LS174T tumor cells were incubated with CC49-cPNA followed by G4-PNA as an amplification platform before 18 F-cPNA was added to hybridize with CC49-cPNA/G4-PNA. Compared with conventional pretargeting without G4-PNA, the radioactivity signal was amplified about four times, which demonstrated that the dendrimer-PNA conjugate plays a crucial role in signal amplification.


Subject(s)
Dendrimers
12.
Sensors (Basel) ; 21(19)2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34640711

ABSTRACT

Penicillins and cephalosporins belong to the ß-lactam antibiotic family, which accounts for more than half of the world market for antibiotics. Misuse of antibiotics harms human health and the environment. Here, we describe an easy, fast, and sensitive optical method for the sensing and discrimination of two penicillin and five cephalosporin antibiotics in buffered water at pH 7.4, using fifth-generation poly (amidoamine) (PAMAM) dendrimers and calcein, a commercially available macromolecular polyelectrolyte and a fluorescent dye, respectively. In aqueous solution at pH 7.4, the dendrimer and dye self-assemble to form a sensor that interacts with carboxylate-containing antibiotics through electrostatic interaction, monitored through changes in the dye's spectroscopic properties. This response was captured through absorbance, fluorescence emission, and fluorescence anisotropy. The resulting data set was processed through linear discriminant analysis (LDA), a common pattern-base recognition method, for the differentiation of cephalosporins and penicillins. By pre-hydrolysis of the ß-lactam rings under basic conditions, we were able to increase the charge density of the analytes, allowing us to discriminate the seven analytes at a concentration of 5 mM, with a limit of discrimination of 1 mM.


Subject(s)
Dendrimers , Water , Anti-Bacterial Agents/analysis , Humans , Hydrogen-Ion Concentration , beta-Lactams
13.
Int J Mol Sci ; 22(4)2021 Feb 17.
Article in English | MEDLINE | ID: mdl-33671436

ABSTRACT

Polyamidoamine PAMAM dendrimer generation 3 (G3) was modified by attachment of biotin via amide bond and glucoheptoamidated by addition of α-D-glucoheptono-1,4-lacton to obtain a series of conjugates with a variable number of biotin residues. The composition of conjugates was determined by detailed 1-D and 2-D NMR spectroscopy to reveal the number of biotin residues, which were 1, 2, 4, 6, or 8, while the number of glucoheptoamide residues substituted most of the remaining primary amine groups of PAMAM G3. The conjugates were then used as host molecules to encapsulate the 5-aminolevulinic acid. The solubility of 5-aminolevulinic acid increased twice in the presence of the 5-mM guest in water. The interaction between host and guest was accompanied by deprotonation of the carboxylic group of 5-aminolevulinic acid and proton transfer into internal ternary nitrogen atoms of the guest as evidenced by a characteristic chemical shift of resonances in the 1H NMR spectrum of associates. The guest molecules were most likely encapsulated inside inner shell voids of the host. The number of guest molecules depended on the number of biotin residues of the host, which was 15 for non-biotin-containing glucoheptoamidated G3 down to 6 for glucoheptoamidated G3 with 8 biotin residues on the host surface. The encapsulates were not cytotoxic against Caco-2 cells up to 200-µM concentration in the dark. All encapsulates were able to deliver 5-aminolevulinic acid to cells but aqueous encapsulates were more active in this regard. Simultaneously, the reactive oxygen species were detected by staining with H2DCFDA in Caco-2 cells incubated with encapsulates. The amount of PpIX was sufficient for induction of reactive oxygen species upon 30-s illumination with a 655-nm laser beam.


Subject(s)
Amides/chemistry , Aminolevulinic Acid/pharmacology , Biotin/chemistry , Dendrimers/chemistry , Drug Delivery Systems , Polyamines/chemistry , Aminolevulinic Acid/chemistry , Caco-2 Cells , Cell Death/drug effects , Cell Survival/drug effects , Dendrimers/chemical synthesis , Fluorescence , Humans , Intracellular Space/metabolism , Polyamines/chemical synthesis , Proton Magnetic Resonance Spectroscopy , Protoporphyrins/pharmacology , Reactive Oxygen Species/metabolism
14.
Int J Mol Sci ; 22(6)2021 Mar 13.
Article in English | MEDLINE | ID: mdl-33805602

ABSTRACT

Carriers of genetic material are divided into vectors of viral and non-viral origin. Viral carriers are already successfully used in experimental gene therapies, but despite advantages such as their high transfection efficiency and the wide knowledge of their practical potential, the remaining disadvantages, namely, their low capacity and complex manufacturing process, based on biological systems, are major limitations prior to their broad implementation in the clinical setting. The application of non-viral carriers in gene therapy is one of the available approaches. Poly(amidoamine) (PAMAM) dendrimers are repetitively branched, three-dimensional molecules, made of amide and amine subunits, possessing unique physiochemical properties. Surface and internal modifications improve their physicochemical properties, enabling the increase in cellular specificity and transfection efficiency and a reduction in cytotoxicity toward healthy cells. During the last 10 years of research on PAMAM dendrimers, three modification strategies have commonly been used: (1) surface modification with functional groups; (2) hybrid vector formation; (3) creation of supramolecular self-assemblies. This review describes and summarizes recent studies exploring the development of PAMAM dendrimers in anticancer gene therapies, evaluating the advantages and disadvantages of the modification approaches and the nanomedicine regulatory issues preventing their translation into the clinical setting, and highlighting important areas for further development and possible steps that seem promising in terms of development of PAMAM as a carrier of genetic material.


Subject(s)
Dendrimers/chemical synthesis , Gene Expression Regulation, Neoplastic , Gene Transfer Techniques , Genetic Therapy/methods , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/therapy , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemical synthesis , Dendrimers/administration & dosage , Government Regulation , Humans , MicroRNAs/administration & dosage , MicroRNAs/genetics , MicroRNAs/metabolism , Nanomedicine/legislation & jurisprudence , Nanomedicine/methods , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Plasmids/administration & dosage , Plasmids/chemistry , Plasmids/metabolism , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Surface Properties
15.
Drug Dev Ind Pharm ; 46(3): 412-426, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32011185

ABSTRACT

Despite poor bioavailability of the drug and in vivo stability, curcumin has been reported for many pharmacological activities. Considering the potential of dendrimers as a drug delivery system, current research work is focused on the formulation and characterization of G4 PAMAM dendrimer-Palmitic acid core-shell nanoparticle-containing curcumin as antistress therapeutics to maximize the bioavailability of curcumin. Various formulations were prepared using different concentrations of palmitic acid and an optimized ratio of dendrimer and curcumin. All formulations were investigated for evaluation of physicochemical parameters, encapsulation efficiency, and in vitro release. Particle size, PDI, zeta-potential, and encapsulation efficiency of final formulation was found to be 257.9 ± 0.365 nm, 0.10 ± 0.004, 3.59 ± 0.167 mV, and 80.87%, respectively. In vitro release studies have shown that 53.62 ± 2.431% of the drug was released after 24 h. In vivo studies pharmacokinetic parameters, drug distribution, pharmacological, and toxicological were also estimated using swiss albino mice. The findings have shown the selected formulation is better than plain curcumin formulation.


Subject(s)
Curcumin/administration & dosage , Drug Carriers/chemistry , Drug Delivery Systems , Nanoparticles , Animals , Biological Availability , Curcumin/chemistry , Curcumin/pharmacokinetics , Dendrimers/chemistry , Drug Liberation , Female , Male , Mice , Nylons/chemistry , Palmitic Acid/chemistry , Particle Size , Tissue Distribution
16.
Pharm Dev Technol ; 25(1): 9-19, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30633621

ABSTRACT

Delivery of negatively charged, high molecular weight and unstable siRNA is difficult. The present study describes the development and comparison of cationic liposomes (CLs) and polyamidoamine (PAMAM) dendrimer generation 4 (PG4) nanocarriers of gene for cancer therapy. CLs and PG4 were complexed with anticancer siRNA (siPlk1) to form siPlk1-CLs lipoplex and siPlk1-PG4 dendriplex. siPlk1-CLs/PG4 complexes were characterized for average particle size, zeta potential, fluorescence and integrity of siPlk1 by agarose gel electrophoresis, ethidium bromide intercalation assay, circular dichroism, protection against RNase and stability in serum. The complexation of CLs/siPlk1 and PG4/siPlk1 were at a 100/1 and 2/1 charge ratio respectively. The CLs and PG4 were effective in protecting siPlk1 from RNase activity, also they enhanced the siPlk1 serum stability. Additionally, siPlk1-CLs and siPlk1-PG4 were evaluated by cell culture studies. In vitro anticancer activity study using MCF-7 cells showed that siPlk1-CLs and siPlk1-PG4 causes nearly similar cell death. Both siPlk1-CLs and siPlk1-PG4 resulted in enhanced cellular uptake of siPlk1 in MDA-MB-231 cells compared to naked siPlk1 solution. Cell cycle analysis suggested that increased cell population arrest in subG1 phase by siPlk1-CLs and siPlk1-PG4 compared to naked siPlk1 solution. These observations suggested that CLs and PG4 can be a potential carrier for siPlk1 delivery in breast cancer treatment.


Subject(s)
Breast Neoplasms/drug therapy , Cations/chemistry , Cell Cycle Proteins/genetics , Dendrimers/chemistry , Liposomes/chemistry , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Breast Neoplasms/genetics , Cell Cycle/genetics , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Particle Size , Transfection/methods , Polo-Like Kinase 1
17.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 51(4): 499-504, 2020 Jul.
Article in Zh | MEDLINE | ID: mdl-32691557

ABSTRACT

OBJECTIVE: The purpose of this study was to investigate the effect of G4.5 carboxyl-terminated poly dendrimer (PAMAM-COOH) on the dentin remineralization and the matrix metalloproteinases (MMPs) activity. METHODS: The dentine samples were averagely divided into four groups: 100 mg/mL PAMAM-COOH group (A group), 10 mg/mL PAMAM-COOH group (B group), 2% (wt) chlorhexidine (CHX) group (C group) and deionized water group (Control group). MMP Activity Assay Kit was used to detect the activity of dentin endogenous MMPs in the four groups. The loss of dry mass of dentin after 30 d were measured. In situ zymography analysis was performed to detect the effects of PAMAM dendrimer in each group (except A group) on gelatinase activity in dentin. After incubation in artificial saliva for 7 and 14 d incubated, the remineralization of each group (except A group) in dentin surfaces were examined using a field emission-scanning electron microscope (FESEM). RESULTS: Compared with the control group, the dentin endogenous MMPs activity in A, B and C groups were all decreased ( P<0.05). The activity of endogenous MMPs in C group was lower than that of A and B groups ( P<0.001), but the difference between A and B groups was not statistically significant. The loss of dry mass in A, B and C groups were lower than that in control group ( P<0.05), but there were no significant difference in A, B and C groups. The in situzymography analysis showed that 48 h later, the dentin gelatinase activity in B group was inhibited compared with the control group, but the inhibitory effect was weaker than that of CHX. After 7 d and 14 d, there were no obvious mineralization in the control group, while distinct mineralization were observed in B group. The mineralization effect in group B was better than group C. CONCLUSION: G4.5 PAMAM-COOH could introduce remineralizationin and demineralizeddentin by effectively inhibiting endogenous MMPs and gelatinase, thus contributes as novel material to enhancing durability of adhesion.


Subject(s)
Dendrimers , Dentin , Matrix Metalloproteinases , Tooth Remineralization , Dendrimers/pharmacology , Dentin/enzymology , Dentin/metabolism , Enzyme Activation/drug effects , Humans , Matrix Metalloproteinases/metabolism , Saliva, Artificial
18.
Mol Pharm ; 16(4): 1541-1554, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30817166

ABSTRACT

This study involves development of a dendrimer-based nanoconstruct by conjugating α-tocopheryl succinate (α-TOS) and polyethylene glycol (PEG) on a poly(amidoamine) dendrimer (G4 PAMAM) to improve intracellular delivery of a poorly water-soluble chemotherapeutic drug, paclitaxel (PTX). The conjugates were characterized by NMR, and PTX-loaded nanocarriers (G4-TOS-PEG-PTX) were evaluated for hydrodynamic diameter, polydispersity index (PDI), zeta potential, percentage encapsulation efficiency (%EE), and percentage drug loading (%DL). A hemolysis study was performed, which indicated that the synthesized dendrimer conjugates were not toxic to red blood cells; hence, they were biocompatible. A cellular uptake study in (B16F10 and MDA MB231) monolayer cells and 3D spheroids showed that α-TOS conjugation improved the time dependent uptake of nanosized dendrimer conjugates. The cell viability assay revealed that G4-TOS-PEG-PTX enhanced the cytotoxicity of PTX as compared to free PTX and PTX-loaded G4-PEG (G4-PEG-PTX) at tested concentrations. Correspondingly, the α-TOS-anchored dendrimer induced more apoptosis as compared to free PTX and G4-PEG-PTX. Moreover, the fluorescently labeled G4-TOS-PEG penetrated deeper into MDA MB231 3D spheroids as visualized by confocal microscopy. G4-TOS-PEG-PTX showed significant growth inhibition in 3D spheroids as compared to free PTX and G4-PEG-PTX. Further, the in vivo efficacy study using B16F10 xenografted C57Bl6/J mice indicated that the G4-TOS-PEG localized in tumor sections. G4-TOS-PEG-PTX reduced the tumor growth significantly compared to free PTX and G4-PEG-PTX. G4-TOS-PEG-PTX had more apoptotic potential in tumor sections as analyzed by TUNEL assay. Hence, the newly developed dendrimer conjugate, G4-TOS-PEG, has the potential to target loaded drug to the tumor, and G4-TOS-PEG-PTX has the potential to be utilized successfully in cancer treatment.


Subject(s)
Breast Neoplasms/drug therapy , Dendrimers/chemistry , Drug Delivery Systems , Melanoma, Experimental/drug therapy , Nylons/chemistry , Paclitaxel/pharmacology , Polyamines/chemistry , alpha-Tocopherol/chemistry , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/chemistry , Apoptosis , Breast Neoplasms/pathology , Cell Proliferation , Drug Carriers/chemistry , Female , Humans , In Vitro Techniques , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Paclitaxel/chemistry , Polyethylene Glycols/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Mol Pharm ; 16(11): 4594-4609, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31593478

ABSTRACT

pH-responsive drug delivery systems are yielding opportunities to directly deliver antibiotics to the site of infection. Therefore, this study aimed to develop and evaluate novel pH-responsive lipid-dendrimer hybrid nanoparticles (LDH-NPs) for the delivery of vancomycin (VCM) to the site of infection, by intracellular bacterial pathogens. The LDH-NPs were formulated using the emulsification solvent evaporation method and were characterized by various in vitro and molecular dynamic (MD) simulation techniques. LDH-NPs were 124.4 ± 2.01 nm in size, with a zeta-potential of -7.15 ± 2.98 mV and drug entrapment efficiency of 82.70 ± 4.09%, which exhibited pH-responsive behavior by shifting the surface charge from negative at physiological pH to positive in acidic pHs, with a size increase from 124.4 ± 2.01 to 173.9 ± 13.38 nm, and 252.7 ± 3.98 nm at pHs of 7.4, 6.0, and 4.5, respectively. Results indicated that the in vitro drug release of VCM from LDH-NPs occurred faster at pH 6.0 than at pH 7.4. The antibacterial activity of LDH-NPs against methicillin-resistance Staphylococcus aureus (MRSA) showed 8-fold lower MICs at pH 6.0 and 7.4, compared to treatment with VCM only. A bacterial cell viability study showed LDH-NPs had an 84.19% killing of MRSA, compared to VCM (49.26%) at the same MIC, further confirming its efficacy. Cell uptake studies showed that LDH-NPs intracellularly accumulated in HEK 293 cells, confirming significant clearance (p < 0.0001) of intracellular bacteria. MD simulation showed that interaction between the dendrimer and oleylamine was predominantly governed by van der Waals (VdW) interactions; whereas the interaction between the dendrimer and VCM was governed by both VdW and electrostatic interactions. Therefore, this study concludes that the pH-responsive release of VCM enhanced antibacterial efficacy against MRSA and intracellular delivery of an antibiotic. Thus, LDH-NPs is a promising nanocarrier system for antibiotics with the potential to improve the treatment outcomes of bacterial infections in patients with antibiotic resistant strains.


Subject(s)
Dendrimers/chemistry , Lipids/chemistry , Nanoparticles/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Delivery Systems/methods , Drug Liberation/drug effects , HEK293 Cells , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests/methods , Particle Size , Static Electricity , Vancomycin/chemistry , Vancomycin/pharmacology
20.
Mol Pharm ; 16(6): 2376-2384, 2019 06 03.
Article in English | MEDLINE | ID: mdl-30951315

ABSTRACT

Skeletal muscle is ideally suited and highly desirable as a target for therapeutic gene delivery because of its abundance, high vascularization, and high levels of protein expression. However, efficient gene delivery to skeletal muscle remains a current challenge. Besides the major obstacle of cell-specific targeting, efficient intracellular trafficking, or the cytosolic transport of DNA to the nucleus, must be demonstrated. To overcome the challenge of cell-specific targeting, herein we develop a generation 5-polyamidoamine dendrimer (G5-PAMAM) functionalized with a skeletal muscle-targeted peptide, ASSLNIA (G5-SMTP). Specifically, to demonstrate the feasibility of our approach, we prepared a complex of our G5-SMTP dendrimer with a plasmid encoding firefly luciferase and investigated its delivery to skeletal muscle cells. Luciferase assays indicated a threefold increase in transfection efficiency of C2C12 murine skeletal muscle cells using G5-SMTP when compared with nontargeting nanocarriers using unmodified G5. To further improve the transfection yield, we employed a cationic dynein light chain 8 protein (DLC8)-binding peptide (DBP) containing an internal sequence known to bind to the DLC8 of the dynein motor protein complex. Complexation of DBP with our targeting nanocarrier, that is, G5-SMTP, and our luciferase plasmid cargo resulted in a functional nanocarrier that showed an additional sixfold increase in transfection efficiency compared with G5-SMTP transfection alone. To our knowledge, this is the first successful use of two different functional nanocarrier components that enable targeted skeletal muscle cell recognition and increased efficiency of intracellular trafficking to synergistically enhance gene delivery to skeletal muscle cells. This strategy of targeting and trafficking can also be universally applied to any cell/tissue type for which a recognition domain exists.


Subject(s)
Dendrimers/chemistry , Dyneins/chemistry , Muscle, Skeletal/metabolism , Plasmids/administration & dosage , Animals , Cell Line , Cytoplasm/metabolism , Cytoplasmic Dyneins/metabolism , Mice , Plasmids/genetics
SELECTION OF CITATIONS
SEARCH DETAIL