Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.677
Filter
1.
Med Oncol ; 41(6): 145, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38727885

ABSTRACT

Polyelectrolytes represent a unique class of polymers abundant in ionizable functional groups. In a solution, ionized polyelectrolytes can intricately bond with oppositely charged counterparts, giving rise to a fascinating phenomenon known as a polyelectrolyte complex. These complexes arise from the interaction between oppositely charged entities, such as polymers, drugs, and combinations thereof. The polyelectrolyte complexes are highly appealing in cancer management, play an indispensable role in chemotherapy, crafting biodegradable, biocompatible 3D membranes, microcapsules, and nano-sized formulations. These versatile complexes are pivotal in designing controlled and targeted release drug delivery systems. The present review emphasizes on classification of polyelectrolyte complex along with their formation mechanisms. This review comprehensively explores the applications of polyelectrolyte complex, highlighting their efficacy in targeted drug delivery strategies for combating different forms of cancer. The innovative use of polyelectrolyte complex presents a potential breakthrough in cancer therapeutics, demonstrating their role in enhancing treatment precision and effectiveness.


Subject(s)
Antineoplastic Agents , Drug Delivery Systems , Neoplasms , Polyelectrolytes , Humans , Polyelectrolytes/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Drug Delivery Systems/methods , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/chemistry , Precision Medicine/methods
2.
Langmuir ; 40(20): 10648-10662, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38712915

ABSTRACT

This study presents new insights into the potential role of polyelectrolyte interfaces in regulating low friction and interstitial fluid pressurization of cartilage. Polymer brushes composed of hydrophilic 3-sulfopropyl methacrylate potassium salt (SPMK) tethered to a PEEK substrate (SPMK-g-PEEK) are a compelling biomimetic solution for interfacing with cartilage, inspired by the natural lubricating biopolyelectrolyte constituents of synovial fluid. These SPMK-g-PEEK surfaces exhibit a hydrated compliant layer approximately 5 µm thick, demonstrating the ability to maintain low friction coefficients (µ ∼ 0.01) across a wide speed range (0.1-200 mm/s) under physiological loads (0.75-1.2 MPa). A novel polyelectrolyte-enhanced tribological rehydration mechanism is elucidated, capable of recovering up to ∼12% cartilage strain and subsequently facilitating cartilage interstitial fluid recovery, under loads ranging from 0.25 to 2.21 MPa. This is attributed to the combined effects of fluid confinement within the contact gap and the enhanced elastohydrodynamic behavior of polymer brushes. Contrary to conventional theories that emphasize interstitial fluid pressurization in regulating cartilage lubrication, this work demonstrates that SPMK-g-PEEK's frictional behavior with cartilage is independent of these factors and provides unabating aqueous lubrication. Polyelectrolyte-enhanced tribological rehydration can occur within a static contact area and operates independently of known mechanisms of cartilage interstitial fluid recovery established for converging or migrating cartilage contacts. These findings challenge existing paradigms, proposing a novel polyelectrolyte-cartilage tribological mechanism not exclusively reliant on interstitial fluid pressurization or cartilage contact geometry. The implications of this research extend to a broader understanding of synovial joint lubrication, offering insights into the development of joint replacement materials that more accurately replicate the natural functionality of cartilage.


Subject(s)
Lubrication , Polymers , Polymers/chemistry , Animals , Polyelectrolytes/chemistry , Polyethylene Glycols/chemistry , Cartilage/chemistry , Cartilage/drug effects , Surface Properties , Benzophenones/chemistry , Cartilage, Articular/chemistry , Cartilage, Articular/physiology , Ketones/chemistry
3.
Mol Biol Rep ; 51(1): 623, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710891

ABSTRACT

BACKGROUND: An increase in cancer stem cell (CSC) populations and their resistance to common treatments could be a result of c-Myc dysregulations in certain cancer cells. In the current study, we investigated anticancer effects of c-Myc decoy ODNs loaded-poly (methacrylic acid-co-diallyl dimethyl ammonium chloride) (PMA-DDA)-coated silica nanoparticles as carriers on cancer-like stem cells (NTERA-2). METHODS AND RESULTS: The physicochemical characteristics of the synthesized nanocomposites (SiO2@PMA-DDA-DEC) were analyzed using FT-IR, DLS, and SEM techniques. UV-Vis spectrophotometer was applied to analyze the release pattern of decoy ODNs from the nanocomposite. Furthermore, uptake, cell viability, apoptosis, and cell cycle assays were used to investigate the anticancer effects of nanocomposites loaded with c-Myc decoy ODNs on NTERA-2 cancer cells. The results of physicochemical analytics demonstrated that SiO2@PMA-DDA-DEC nanocomposites were successfully synthesized. The prepared nanocomposites were taken up by NTERA-2 cells with high efficiency, and could effectively inhibit cell growth and increase apoptosis rate in the treated cells compared to the control group. Moreover, SiO2@PMA-DDA nanocomposites loaded with c-Myc decoy ODNs induced cell cycle arrest at the G0/G1 phase in the treated cells. CONCLUSIONS: The conclusion drawn from this study is that c-Myc decoy ODN-loaded SiO2@PMA-DDA nanocomposites can effectively inhibit cell growth and induce apoptosis in NTERA-2 cancer cells. Moreover, given that a metal core is incorporated into this synthetic nanocomposite, it could potentially be used in conjunction with irradiation as part of a decoy-radiotherapy combinational therapy in future investigations.


Subject(s)
Apoptosis , Cell Proliferation , Nanoparticles , Neoplastic Stem Cells , Proto-Oncogene Proteins c-myc , Humans , Apoptosis/drug effects , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Cell Proliferation/drug effects , Nanoparticles/chemistry , Cell Line, Tumor , Nanocomposites/chemistry , Polyelectrolytes/chemistry , Oligodeoxyribonucleotides/pharmacology , Oligodeoxyribonucleotides/chemistry , Cell Survival/drug effects , Silicon Dioxide/chemistry , Polyamines/chemistry , Polyamines/pharmacology , Cell Cycle/drug effects
4.
J Phys Chem B ; 128(14): 3329-3339, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38557033

ABSTRACT

In nature, DNA exists primarily in a highly compacted form. The compaction of DNA in vivo is mediated by cationic proteins: histones in somatic nuclei and protamines in sperm chromatin. The extreme, nearly crystalline packaging of DNA by protamines in spermatozoa is thought to be essential for both efficient genetic delivery as well as DNA protection against damage by mutagens and oxidative species. The protective role of protamines is required in sperm, as they are sensitive to ROS damage due to the progressive loss of DNA repair mechanisms during maturation. The degree to which DNA packaging directly relates to DNA protection in the condensed state, however, is poorly understood. Here, we utilized different polycation condensing agents to achieve varying DNA packaging densities and quantify DNA damage by free radical oxidation within the condensates. Although we see that tighter DNA packaging generally leads to better protection, the length of the polycation also plays a significant role. Molecular dynamics simulations suggest that longer polyarginine chains offer increased protection by occupying more space on the DNA surface and forming more stable interactions. Taken together, our results suggest a complex interplay among polycation properties, DNA packaging density, and DNA protection against free radical damage within condensed states.


Subject(s)
DNA , Polyelectrolytes , Semen , Male , Humans , DNA/chemistry , Chromatin , Protamines/chemistry , Spermatozoa , DNA Packaging , DNA Damage
5.
Soft Matter ; 20(16): 3499-3507, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38595066

ABSTRACT

Polymeric vesicles are perspective vehicles for fabricating enzymatic nanoreactors towards diverse biomedical and catalytic applications, yet the design of stable and permeable vesicles remains challenging. Herein, we developed polyion complex (PIC) vesicles featuring high stability and a permeable membrane for adequate enzyme loading and activation. Our design relies on co-assembly of an anionic diblock copolymer (PSS96-b-PEO113) with cationic branched poly(ethylenimine) (PEI). The polymer combination endows strong electrostatic interaction between the PSS and PEI building blocks, so their assembly can be implemented at a high salt concentration (500 mM NaCl), under which the charge interaction of the enzyme-polymer is inhibited. This control realizes the successful and safe loading of enzymes associated with the formation of stable PIC vesicles with an intrinsic permeable membrane that is favourable for enhancing enzymatic activity. The control factors for vesicle formation and enzyme loading were investigated, and the general application of loading different enzymes for cascade reaction was validated as well. Our study reveals that proper design and combination of polyelectrolytes is a facile strategy for fabricating stable and permeable polymeric PIC vesicles, which exhibit clear advantages for loading and activating enzymes, consequently boosting their diverse applications as enzymatic nanoreactors.


Subject(s)
Polyethyleneimine , Polyethyleneimine/chemistry , Permeability , Polymers/chemistry , Polyelectrolytes/chemistry
6.
Biomacromolecules ; 25(5): 3112-3121, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38651274

ABSTRACT

Responsive nanomaterials hold significant promise in the treatment of bacterial infections by recognizing internal or external stimuli to achieve stimuli-responsive behavior. In this study, we present an enzyme-responsive polyelectrolyte complex micelles (PTPMN) with α-helical cationic polypeptide as a coacervate-core for the treatment of Escherichia coli (E. coli) infection. The complex was constructed through electrostatic interaction between cationic poly(glutamic acid) derivatives and phosphorylation-modified poly(ethylene glycol)-b-poly(tyrosine) (PEG-b-PPTyr) by directly dissolving them in aqueous solution. The cationic polypeptide adopted α-helical structure and demonstrated excellent broad-spectrum antibacterial activity against both Gram-negative and Gram-positive bacteria, with a minimum inhibitory concentration (MIC) as low as 12.5 µg mL-1 against E. coli. By complexing with anionic PEG-b-PPTyr, the obtained complex formed ß-sheet structures and exhibited good biocompatibility and low hemolysis. When incubated in a bacterial environment, the complex cleaved its phosphate groups triggered by phosphatases secreted by bacteria, exposing the highly α-helical conformation and restoring its effective bactericidal ability. In vivo experiments confirmed accelerated healing in E. coli-infected wounds.


Subject(s)
Anti-Bacterial Agents , Escherichia coli , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/administration & dosage , Escherichia coli/drug effects , Animals , Microbial Sensitivity Tests , Polyelectrolytes/chemistry , Polyelectrolytes/pharmacology , Peptides/chemistry , Peptides/pharmacology , Protein Conformation, alpha-Helical , Micelles , Escherichia coli Infections/drug therapy , Hemolysis/drug effects , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Mice , Polyglutamic Acid/chemistry , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/pharmacology , Humans
7.
Int J Biol Macromol ; 268(Pt 1): 131616, 2024 May.
Article in English | MEDLINE | ID: mdl-38631592

ABSTRACT

Hydrogels have shown great potential for application in food science due to their diverse functionalities. However, most hydrogels inevitably contain toxic chemical cross-linking agent residues, posing serious food safety concerns. In this paper, a curcumin/sodium alginate/carboxymethyl chitosan hydrogels (CSCH) were prepared by self-assembly of two oppositely charged polysaccharides, carboxymethyl chitosan and sodium alginate, to form a three-dimensional network encapsulating curcumin for extending food shelf life. The network structure of the CSCH film confirmed by FTIR, XRD, and XPS was mainly formed by electrostatic interactions. The chemical stability of CSCH network encapsulated curcumin was 4.2 times greater than that of free curcumin, with excellent gas barrier, antimicrobial, antioxidant, and biosafety properties. It was found that CSCH films reduced dehydration, prevented nutrient loss, inhibited microbial growth, and lowered the respiration rate, which effectively maintained the quality of mango and prolonged its shelf-life up to 11 days. Notably, CSCH films possessed the properties of rapid recycling (10 mins) and biodegradability (53 days). This polysaccharide-based hydrogel film provides a viable strategy for the development of green and sustainable food packaging.


Subject(s)
Chitosan , Curcumin , Curcumin/chemistry , Curcumin/pharmacology , Curcumin/analogs & derivatives , Chitosan/chemistry , Chitosan/analogs & derivatives , Hydrogels/chemistry , Alginates/chemistry , Antioxidants/chemistry , Antioxidants/pharmacology , Polyelectrolytes/chemistry , Food Packaging/methods , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Methylgalactosides
8.
ACS Nano ; 18(15): 10427-10438, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38556978

ABSTRACT

Protein translocation through nanopores holds significant promise for applications in biotechnology, biomolecular analysis, and medicine. However, the interpretation of signals generated by the translocation of the protein remains challenging. In this way, it is crucial to gain a comprehensive understanding on how macromolecules translocate through a nanopore and to identify what are the critical parameters that govern the process. In this study, we investigate the interplay between protein charge regulation, orientation, and nanopore surface modifications using a theoretical framework that allows us to explicitly take into account the acid-base reactions of the titrable amino acids in the proteins and in the polyelectrolytes grafted to the nanopore surface. Our goal is to thoroughly characterize the translocation process of different proteins (GFP, ß-lactoglobulin, lysozyme, and RNase) through nanopores modified with weak polyacids. Our calculations show that the charge regulation mechanism exerts a profound effect on the translocation process. The pH-dependent interactions between proteins and charged polymers within the nanopore lead to diverse free energy landscapes with barriers, wells, and flat regions dictating translocation efficiency. Comparison of different proteins allows us to identify the significance of protein isoelectric point, size, and morphology in the translocation behavior. Taking advantage of these insights, we propose pH-responsive nanopores that can load proteins at one pH and release them at another, offering opportunities for controlled protein delivery, separation, and sensing applications.


Subject(s)
Nanopores , Polymers/chemistry , Polyelectrolytes , Proteins/chemistry , Protein Transport
9.
Nat Commun ; 15(1): 3485, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664427

ABSTRACT

Spider silk exhibits an excellent combination of high strength and toughness, which originates from the hierarchical self-assembled structure of spidroin during fiber spinning. In this work, superfine nanofibrils are established in polyelectrolyte artificial spider silk by optimizing the flexibility of polymer chains, which exhibits combination of breaking strength and toughness ranging from 1.83 GPa and 238 MJ m-3 to 0.53 GPa and 700 MJ m-3, respectively. This is achieved by introducing ions to control the dissociation of polymer chains and evaporation-induced self-assembly under external stress. In addition, the artificial spider silk possesses thermally-driven supercontraction ability. This work provides inspiration for the design of high-performance fiber materials.


Subject(s)
Nanofibers , Polyelectrolytes , Silk , Spiders , Animals , Nanofibers/chemistry , Spiders/chemistry , Silk/chemistry , Polyelectrolytes/chemistry , Tensile Strength , Muscles , Biomimetic Materials/chemistry
10.
J Chromatogr A ; 1721: 464861, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38564931

ABSTRACT

The covalent attachment of polyoxometalates (POMs) to polymers has been developed as a strategic approach for the advancement of POM-based hybrid materials with versatile applications. In this study, we utilized thiol-maleimide Michael addition to investigate the kinetics and efficacy of the "one-to-one" conjugation between Keggin type POM and polystyrene. We explored the effects of solvent polarity, catalyst, molecular weight of PS and synthetic strategies on the reaction kinetics and efficiency, by means of reverse-phase high-performance liquid chromatography (RP-HPLC). A series of comparative analysis affirmed the superior efficiency of the one-pot method, particularly when facilitated by the addition of a high-polarity solvent and an excess of maleimide. These findings offer valuable insights into the intricate interplay between reaction conditions, kinetics, and selectivity in thiol-maleimide reactions of POMs and polymers. They hold profound implications for advancing the study of POM-based multifunctional materials and the synthesis of complex hybrid molecules.


Subject(s)
Anions , Polyelectrolytes , Polymers , Sulfhydryl Compounds , Polymers/chemistry , Maleimides/chemistry , Solvents
11.
Adv Colloid Interface Sci ; 326: 103133, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38547652

ABSTRACT

DNA is a highly charged polyelectrolyte and is prone to associative phase separation driven by the presence of multivalent cations, charged surfactants, proteins, polymers and colloids. The process of DNA phase separation induced by positively charged species is often called DNA condensation. Generally, it refers to either intramolecular DNA compaction (coil-globule transition) or intermolecular DNA aggregation with macroscopic phase separation, but the formation of a DNA liquid crystalline system is also displayed. This has traditionally been described by polyelectrolyte theory and qualitative (Flory-Huggins-based) polymer theory approaches. DNA in the cell nucleus is packed into chromatin wound around the histone octamer (a protein complex comprising two copies each of the four histone proteins H2A, H2B, H3 and H4) to form nucleosomes separated by linker DNA. During the last decade, the phenomenon of the formation of biomolecular condensates (dynamic droplets) by liquid-liquid phase separation (LLPS) has emerged as a generally important mechanism for the formation of membraneless organelles from proteins, nucleic acids and their complexes. DNA and chromatin droplet formation through LLPS has recently received much attention by in vitro as well as in vivo studies that established the importance of this for compartmentalisation in the cell nucleus. Here, we review DNA and chromatin LLPS from a general colloid physical chemistry perspective. We start with a general discussion of colloidal phase separation in aqueous solutions and review the original (pre-LLPS era) work on DNA (macroscopic) phase separation for simpler systems with DNA in the presence of multivalent cations and well-defined surfactants and colloids. Following that, we discuss and illustrate the similarities of such macroscopic phase separation with the general behaviour of LLPS droplet formation by associative phase separation for DNA-protein systems, including chromatin; we also note cases of segregative association. The review ends with a discussion of chromatin LLPS in vivo and its physiological significance.


Subject(s)
Chromatin , Histones , Histones/metabolism , Polyelectrolytes , Phase Separation , DNA , Polymers/metabolism , Chemistry, Physical , Colloids , Cations/metabolism , Surface-Active Agents
12.
ACS Biomater Sci Eng ; 10(5): 3029-3040, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38551901

ABSTRACT

Polycationic polymers are widely studied antiseptics, and their efficacy is usually quantified by the solution concentration required to kill a fraction of a population of cells (e.g., by Minimum Bactericidal Concentration (MBC)). Here we describe how the response to a polycationic antimicrobial varies greatly among members of even a monoclonal population of bacteria bathed in a single common antimicrobial concentration. We use fluorescence microscopy to measure the adsorption of a labeled cationic polymer, polydiallyldimethylammmonium chloride (PDADMAC, Mw ≈ 4 × 105 g mol-1) and the time course of cell response via a cell permeability indicator for each member of an ensemble of either Escherichia coli, Staphylococcus aureus, or Pseudomonas aeruginosa cells. This is a departure from traditional methods of evaluating synthetic antimicrobials, which typically measure the overall response of a collection of cells at a particular time and therefore do not assess the diversity within a population. Cells typically die after they reach a threshold adsorption of PDADMAC, but not always. There is a substantial time lag of about 5-10 min between adsorption and death, and the time to die of an individual cell is well correlated with the rate of adsorption. The amount adsorbed and the time-to-die differ among species but follow a trend of more adsorption on more negatively charged species, as expected for a cationic polymer. The study of individual cells via time-lapse microscopy reveals additional details that are lost when measuring ensemble properties at a particular time.


Subject(s)
Escherichia coli , Pseudomonas aeruginosa , Staphylococcus aureus , Escherichia coli/drug effects , Staphylococcus aureus/drug effects , Pseudomonas aeruginosa/drug effects , Polyethylenes/chemistry , Polyethylenes/pharmacology , Quaternary Ammonium Compounds/pharmacology , Quaternary Ammonium Compounds/chemistry , Polyelectrolytes/chemistry , Polyelectrolytes/pharmacology , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Polymers/pharmacology , Polymers/chemistry , Microscopy, Fluorescence , Adsorption
13.
J Colloid Interface Sci ; 665: 801-813, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38555748

ABSTRACT

The co-assembly of polyelectrolytes (PE) with proteins offers a promising approach for designing complex structures with customizable morphologies, charge distribution, and stability for targeted cargo delivery. However, the complexity of protein structure limits our ability to predict the properties of the formed nanoparticles, and our goal is to identify the key triggers of the morphological transition in protein/PE complexes and evaluate their ability to encapsulate multivalent ionic drugs. A positively charged PE can assemble with a protein at pH above isoelectric point due to the electrostatic attraction and disassemble at pH below isoelectric point due to the repulsion. The additional hydrophilic block of the polymer should stabilize the particles in solution and enable them to encapsulate a negatively charged drug in the presence of PE excess. We demonstrated that diblock copolymers, poly(ethylene oxide)-block-poly(N,N-dimethylaminoethyl methacrylate) and poly(ethylene oxide)-block-poly(N,N,N-trimethylammonioethyl methacrylate), consisting of a polycation block and a neutral hydrophilic block, reversibly co-assemble with insulin in pH range between 5 and 8. Using small-angle neutron and X-ray scattering (SANS, SAXS), we showed that insulin arrangement within formed particles is controlled by intermolecular electrostatic forces between protein molecules, and can be tuned by varying ionic strength. For the first time, we observed by fluorescence that formed protein/PE complexes with excess of positive charges exhibited potential for encapsulating and controlled release of negatively charged bivalent drugs, protoporphyrin-IX and zinc(II) protoporphyrin-IX, enabling the development of nanocarriers for combination therapies with adjustable charge, stability, internal structure, and size.


Subject(s)
Insulin , Protoporphyrins , Polyelectrolytes , Ethylene Oxide , Scattering, Small Angle , X-Ray Diffraction , Polymers/chemistry , Proteins , Isoelectric Point
14.
J Chromatogr A ; 1720: 464802, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38507871

ABSTRACT

Successive multiple ionic-polymer layers (SMILs) have long since proved their worth in capillary electrophoresis as they ensure stable electroosmotic flow (EOF) and relatively high separation efficiency. Recently, we demonstrated that plotting the plate height (H) against the solute migration velocity (u) enabled a reliable quantitative evaluation of the coating performances in terms of separation efficiency. In this work, various physicochemical and chemical parameters of the SMIL coating were studied and optimized in order to decrease the slope of the ascending part of the H vs u curve, which is known to be controlled by the homogeneity in charge of the coating surface and by the possible residual solute adsorption onto the coating surface. SMILs based on poly(diallyldimethylammonium chloride) (PDADMAC) and poly(sodium styrene sulfonate) (PSS) were formed and the effect of each polyelectrolyte molar mass and of the number of polyelectrolyte layers (up to 21 layers) was studied. The use of polyethylene imine as an anchoring first layer was considered. More polyelectrolyte couples based on PDADMAC, polybrene, PSS, poly(vinyl sulfate), and poly(acrylic acid) were tested. Finally, zwitterionic polymers based on the poly(α-l-lysine) scaffold were synthesized and used as the last layer of SMILs, illustrating their ability to finetune the EOF, while maintaining good separation efficiency.


Subject(s)
Electrophoresis, Capillary , Polyethylenes , Polymers , Quaternary Ammonium Compounds , Polyelectrolytes , Cations , Electrophoresis, Capillary/methods , Proteins/analysis , Polyethyleneimine
15.
Langmuir ; 40(14): 7300-7309, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38536237

ABSTRACT

DNA sequencing and sensing using nanopore technology delves critically into the alterations in the measurable electrical signal as single-stranded DNA is drawn through a tiny passage. To make such precise measurements, however, slowing down the DNA in the tightly confined passage is a key requirement, which may be achieved by grafting the nanopore walls with a polyelectrolyte layer (PEL). This soft functional layer at the wall, under an off-design condition, however, may block the DNA passage completely, leading to the complete loss of output signal from the nanobio sensor. Whereas theoretical postulates have previously been put forward to explain the essential physics of DNA translocation in nanopores, these have turned out to be somewhat inadequate when confronted with the experimental findings on functionalized nanopores, including the prediction of the events of complete signal losses. Circumventing these constraints, herein we bring out a possible decisive role of the interplay between the inevitable variabilities in the ionic distribution along the nanopore axis due to its finite length as opposed to its idealized "infinite" limit as well as the differential permittivity of PEL and bulk solution that cannot be captured by the commonly used one-dimensional variant of the electrical double layer theory. Our analysis, for the first time, captures variations in the ionic concentration distribution across multidimensional physical space and delineates its impact on the DNA translocation characteristics that have hitherto remained unaddressed. Our results reveal possible complete blockages of DNA translocation as influenced by less-than-threshold permittivity values or greater-than-threshold grafting densities of the PEL. In addition, electrohydrodynamic blocking is witnessed due to the ion-selective nature of the nanopore at low ionic concentrations. Hence, our study establishes a functionally active regime over which the PEL layer in a finite-length nanopore facilitates controllable DNA translocation, enabling successful sequencing and sensing through the explicit modulation of translocation speed.


Subject(s)
Nanopores , Polyelectrolytes , DNA , DNA, Single-Stranded , Ions
16.
Int J Biol Macromol ; 264(Pt 2): 130787, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38548499

ABSTRACT

In present study, polysaccharide polyelectrolyte nanoparticles (CMAAP-CS NPs) were constructed by mixing carboxymethylated Anemarrhena asphodeloides polysaccharide (CMAAP) and chitosan (CS). CMAAP-CS NPs were applied as carrier to improve the bioavailability and stability of curcumin (Cur). The average particle size of CMAAP-CS NPs was 216.60 ± 4.21 nm and the entrapment efficiency of Cur reached 82.50 ± 2.09 %. The simulated digestion experiments in vitro confirmed that the bioavailability of Cur loaded in CMAAP-CS NPs was 59.84 ± 0.03 % after saliva, gastric and intestinal digestion, which was obvious higher than 21.57 ± 0.07 % of free Cur under the same conditions. The results of stability testing revealed that CMAAP-CS NPs could markedly reduce the degradation of Cur against storage, heating, UV light treatment, and neutral pH. This study provided promising polyelectrolyte complex loaded hydrophobic nutrients in medicine industry.


Subject(s)
Anemarrhena , Chitosan , Curcumin , Nanoparticles , Curcumin/chemistry , Drug Carriers/chemistry , Chitosan/chemistry , Polyelectrolytes , Polysaccharides/pharmacology , Nanoparticles/chemistry , Anti-Bacterial Agents/pharmacology , Particle Size
17.
Int J Biol Macromol ; 264(Pt 1): 130457, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38432265

ABSTRACT

A mucoadhesive polyelectrolyte complex (PEC) nanoparticles were developed for ocular moxifloxacin (Mox) delivery in Bacterial Keratitis (BK). Moxifloxacin-loaded G/CG-Alg NPs were prepared by an amalgamation of cationic polymers (gelatin (G)/cationized gelatin (CG)), and anionic polymer (sodium alginate (Alg)) along with Mox respectively. Mox@CG-Alg NPs were characterized for physicochemical parameters such as particle size (DLS technique), morphology (SEM analysis), DSC, XRD, encapsulation efficiency, drug loading, mucoadhesive study (by texture analyzer), mucin turbidity, and viscosity assessment. The NPs uptake and toxicity of the formulation were analyzed in the Human Corneal Epithelial (HCE) cell line and an ocular irritation study was performed on the HET-CAM. The results indicated that the CG-Alg NPs, with optimal size (217.2 ± 4 nm) and polydispersity (0.22 ± 0.05), have shown high cellular uptake in monolayer and spheroids of HCE. The drug-loaded formulation displayed mucoadhesiveness, trans-corneal permeation, and sustained the release of the Mox. The anti-bacterial efficacy studied on planktonic bacteria/biofilms of P. aeruginosa and S. aureus (in vitro) indicated that the Mox@CG-Alg NPs displayed low MIC, higher zone of bacterial growth inhibition, and cell death compared to free Mox. A significant reduction of bacterial load was observed in the BK-induced mouse model.


Subject(s)
Dieldrin/analogs & derivatives , Eye Infections, Bacterial , Keratitis , Nanoparticles , Mice , Animals , Humans , Moxifloxacin/pharmacology , Gelatin/chemistry , Polyelectrolytes , Alginates/chemistry , Staphylococcus aureus , Ophthalmic Solutions , Nanoparticles/chemistry , Keratitis/drug therapy
18.
Int J Biol Macromol ; 266(Pt 1): 131123, 2024 May.
Article in English | MEDLINE | ID: mdl-38537853

ABSTRACT

Although several bioinks have been developed for 3D bioprinting applications, the lack of optimal printability, mechanical properties, and adequate cell response has limited their practical applicability. Therefore, this work reports the development of a composite bioink consisting of bovine serum albumin (BSA), alginate, and self-assembled nanofibrous polyelectrolyte complex aggregates of gelatin and chitosan (PEC-GC). The nanofibrous PEC-GC aggregates were prepared and incorporated into the bioink in varying concentrations (0 % to 3 %). The bioink samples were bioprinted and crosslinked post-printing by calcium chloride. The average nanofiber diameter of PEC-GC was 62 ± 15 nm. It was demonstrated that PEC-GC improves the printability and cellular adhesion of the developed bioink and modulates the swelling ratio, degradation rate, and mechanical properties of the fabricated scaffold. The in vitro results revealed that the bioink with 2 % PEC-GC had the best post-printing cell viability of the encapsulated MG63 osteosarcoma cells and well oragnized stress fibers, indicating enhanced cell adhesion. The cell viability was >90 %, as observed from the MTT assay. The composite bioink also showed osteogenic potential, as confirmed by the estimation of alkaline phosphatase activity and collagen synthesis assay. This study successfully fabricated a high-shape fidelity bioink with potential in bone tissue engineering.


Subject(s)
Alginates , Bioprinting , Nanofibers , Polyelectrolytes , Printing, Three-Dimensional , Serum Albumin, Bovine , Tissue Scaffolds , Alginates/chemistry , Serum Albumin, Bovine/chemistry , Bioprinting/methods , Nanofibers/chemistry , Tissue Scaffolds/chemistry , Humans , Polyelectrolytes/chemistry , Tissue Engineering/methods , Animals , Cell Survival/drug effects , Bone and Bones/drug effects , Cattle , Cell Line, Tumor , Gelatin/chemistry , Chitosan/chemistry , Osteogenesis/drug effects , Ink , Cell Adhesion/drug effects
19.
ACS Appl Bio Mater ; 7(4): 2544-2553, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38507285

ABSTRACT

The poor accumulation of antibiotics in the cytoplasm leads to the poor eradication of intracellular bacteria. Herein, a polyelectrolyte complex (PECs@Rif) allowing direct cytosolic delivery of rifampicin (Rif) was developed for the treatment of intracellular infections by complexation of poly(α-lipoic acid) (pLA) and oligosaccharide (COS) in water and loading Rif. Due to the thiol-mediated cellular uptake, PECs@Rif delivered 3.9 times higher Rif into the cytoplasm than that of the free Rif during 8 h of incubation. After entering cells, PECs@Rif released Rif by dissociating pLA into dihydrolipoic acid (DHLA) in the presence of intracellular thioredoxin reductase (TrxR). Notably, DHLA could reduce endogenous Fe(III) to Fe(II) and provide a catalyst for the Fenton reaction to produce a large amount of reactive oxygen species (ROS), which would assist Rif in eradicating intracellular bacteria. In vitro assay showed that PECs@Rif reduced almost 2.8 orders of magnitude of intracellular bacteria, much higher than 0.7 orders of magnitude of free Rif. The bacteremia-bearing mouse models showed that PECs@Rif reduced bacterial levels in the liver, spleen, and kidney by 2.2, 3.7, and 2.3 orders of magnitude, respectively, much higher than free Rif in corresponding tissues. The direct cytosolic delivery in a thiol-mediated manner and enhanced oxidative stress proposed a feasible strategy for treating intracellular bacteria infection.


Subject(s)
Ferric Compounds , Rifampin , Mice , Animals , Rifampin/pharmacology , Polyelectrolytes , Oxidative Stress , Bacteria
20.
Proc Natl Acad Sci U S A ; 121(11): e2307809121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38437543

ABSTRACT

Rapid advances in nucleic acid therapies highlight the immense therapeutic potential of genetic therapeutics. Lipid nanoparticles (LNPs) are highly potent nonviral transfection agents that can encapsulate and deliver various nucleic acid therapeutics, including but not limited to messenger RNA (mRNA), silencing RNA (siRNA), and plasmid DNA (pDNA). However, a major challenge of targeted LNP-mediated systemic delivery is the nanoparticles' nonspecific uptake by the liver and the mononuclear phagocytic system, due partly to the adsorption of endogenous serum proteins onto LNP surfaces. Tunable LNP surface chemistries may enable efficacious delivery across a range of organs and cell types. Here, we describe a method to electrostatically adsorb bioactive polyelectrolytes onto LNPs to create layered LNPs (LLNPs). LNP cores varying in nucleic acid cargo and component lipids were stably layered with four biologically relevant polyanions: hyaluronate (HA), poly-L-aspartate (PLD), poly-L-glutamate (PLE), and polyacrylate (PAA). We further investigated the impact of the four surface polyanions on the transfection and uptake of mRNA- and pDNA-loaded LNPs in cell cultures. PLD- and PLE-LLNPs increased mRNA transfection twofold over unlayered LNPs in immune cells. HA-LLNPs increased pDNA transfection rates by more than twofold in epithelial and immune cells. In a healthy C57BL/6 murine model, PLE- and HA-LLNPs increased transfection by 1.8-fold to 2.5-fold over unlayered LNPs in the liver and spleen. These results suggest that LbL assembly is a generalizable, highly tunable platform to modify the targeting specificity, stability, and transfection efficacy of LNPs, as well as incorporate other charged targeting and therapeutic molecules into these systems.


Subject(s)
Liposomes , Nanoparticles , Animals , Mice , Polyelectrolytes , Adsorption , Static Electricity , Transfection , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Glutamic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...